Administrative Appointments


  • Faculty Director, Stanford Cancer Center Proteomics, Stanford Cancer Center (2015 - Present)
  • Faculty Director, Stanford University Mass Spectrometry, Stanford University (2014 - Present)

Honors & Awards


  • Merck Fellow, Life Sciences Research Foundation (1991)
  • Baxter Award, Baxter Foundation (1997)
  • Lutje-Stubbs Scholar, Stanford University (1998)
  • Hume Faculty Scholar, Stanford University (1999)
  • William Cohen Lecturer, Dana-Farber Cancer Institute (1999)
  • Scholar, Kirsch Foundation (2003)
  • Pluto Society, AAUP (2005)
  • Staff Scientist, Genentech (2005-2013)
  • Fellow, American Association for the Advancement of Science (2008)
  • Fellow, Sigma Xi (2017)

Professional Education


  • Fellow, Harvard Medical School, Cell Biology, Cell Cycle (1994)
  • Fellow, UCSF, Biochemistry & Biophysics (1993)
  • Graduate Student, Whitehead Institute, MIT, Cancer Biiology (1989)
  • Ph.D., Harvard University, Biophysics (1989)
  • B. A., Yale College, Mathematics, Economics (1982)

Current Research and Scholarly Interests


Cell cycle control of DNA replication in embryonic and somatic cells: cyclins and the cell cycle in Xenopus embryos.

2023-24 Courses


Stanford Advisees


All Publications


  • The IFT81-IFT74 complex acts as an unconventional RabL2 GTPase-activating protein during intraflagellar transport. The EMBO journal Boegholm, N., Petriman, N. A., Loureiro-Lopez, M., Wang, J., Vela, M. I., Liu, B., Kanie, T., Ng, R., Jackson, P. K., Andersen, J. S., Lorentzen, E. 2023: e111807

    Abstract

    Cilia are important cellular organelles for signaling and motility and are constructed via intraflagellar transport (IFT). RabL2 is a small GTPase that localizes to the basal body of cilia via an interaction with the centriolar protein CEP19 before downstream association with the IFT machinery, which is followed by initiation of IFT. We reconstituted and purified RabL2 with CEP19 or IFT proteins to show that a reconstituted pentameric IFT complex containing IFT81/74 enhances the GTP hydrolysis rate of RabL2. The binding site on IFT81/74 that promotes GTP hydrolysis in RabL2 was mapped to a 70-amino-acid-long coiled-coil region of IFT81/74. We present structural models for RabL2-containing IFT complexes that we validate in vitro and in cellulo and demonstrate that Chlamydomonas IFT81/74 enhances GTP hydrolysis of human RabL2, suggesting an ancient evolutionarily conserved activity. Our results provide an architectural understanding of how RabL2 is incorporated into the IFT complex and a molecular rationale for why RabL2 dissociates from anterograde IFT trains soon after departure from the ciliary base.

    View details for DOI 10.15252/embj.2022111807

    View details for PubMedID 37606072

  • UHRF1 is a mediator of KRAS driven oncogenesis in lung adenocarcinoma. Nature communications Kostyrko, K., Román, M., Lee, A. G., Simpson, D. R., Dinh, P. T., Leung, S. G., Marini, K. D., Kelly, M. R., Broyde, J., Califano, A., Jackson, P. K., Alejandro Sweet-Cordero, E. 2023; 14 (1): 3966

    Abstract

    KRAS is a frequent driver in lung cancer. To identify KRAS-specific vulnerabilities in lung cancer, we performed RNAi screens in primary spheroids derived from a Kras mutant mouse lung cancer model and discovered an epigenetic regulator Ubiquitin-like containing PHD and RING finger domains 1 (UHRF1). In human lung cancer models UHRF1 knock-out selectively impaired growth and induced apoptosis only in KRAS mutant cells. Genome-wide methylation and gene expression analysis of UHRF1-depleted KRAS mutant cells revealed global DNA hypomethylation leading to upregulation of tumor suppressor genes (TSGs). A focused CRISPR/Cas9 screen validated several of these TSGs as mediators of UHRF1-driven tumorigenesis. In vivo, UHRF1 knock-out inhibited tumor growth of KRAS-driven mouse lung cancer models. Finally, in lung cancer patients high UHRF1 expression is anti-correlated with TSG expression and predicts worse outcomes for patients with KRAS mutant tumors. These results nominate UHRF1 as a KRAS-specific vulnerability and potential target for therapeutic intervention.

    View details for DOI 10.1038/s41467-023-39591-2

    View details for PubMedID 37407562

    View details for PubMedCentralID 7367715

  • Multiplexed screens identify RAS paralogues HRAS and NRAS as suppressors of KRAS-driven lung cancer growth. Nature cell biology Tang, R., Shuldiner, E. G., Kelly, M., Murray, C. W., Hebert, J. D., Andrejka, L., Tsai, M. K., Hughes, N. W., Parker, M. I., Cai, H., Li, Y. C., Wahl, G. M., Dunbrack, R. L., Jackson, P. K., Petrov, D. A., Winslow, M. M. 2023

    Abstract

    Oncogenic KRAS mutations occur in approximately 30% of lung adenocarcinoma. Despite several decades of effort, oncogenic KRAS-driven lung cancer remains difficult to treat, and our understanding of the regulators of RAS signalling is incomplete. Here to uncover the impact of diverse KRAS-interacting proteins on lung cancer growth, we combined multiplexed somatic CRISPR/Cas9-based genome editing in genetically engineered mouse models with tumour barcoding and high-throughput barcode sequencing. Through a series of CRISPR/Cas9 screens in autochthonous lung cancer models, we show that HRAS and NRAS are suppressors of KRASG12D-driven tumour growth in vivo and confirm these effects in oncogenic KRAS-driven human lung cancer cell lines. Mechanistically, RAS paralogues interact with oncogenic KRAS, suppress KRAS-KRAS interactions, and reduce downstream ERK signalling. Furthermore, HRAS and NRAS mutations identified in oncogenic KRAS-driven human tumours partially abolished this effect. By comparing the tumour-suppressive effects of HRAS and NRAS in oncogenic KRAS- and oncogenic BRAF-driven lung cancer models, we confirm that RAS paralogues are specific suppressors of KRAS-driven lung cancer in vivo. Our study outlines a technological avenue to uncover positive and negative regulators of oncogenic KRAS-driven cancer in a multiplexed manner in vivo and highlights the role RAS paralogue imbalance in oncogenic KRAS-driven lung cancer.

    View details for DOI 10.1038/s41556-022-01049-w

    View details for PubMedID 36635501

  • SARS-CoV-2 replication in airway epithelia requires motile cilia and microvillar reprogramming. Cell Wu, C., Lidsky, P. V., Xiao, Y., Cheng, R., Lee, I. T., Nakayama, T., Jiang, S., He, W., Demeter, J., Knight, M. G., Turn, R. E., Rojas-Hernandez, L. S., Ye, C., Chiem, K., Shon, J., Martinez-Sobrido, L., Bertozzi, C. R., Nolan, G. P., Nayak, J. V., Milla, C., Andino, R., Jackson, P. K. 2022

    Abstract

    How SARS-CoV-2 penetrates the airway barrier of mucus and periciliary mucins to infect nasal epithelium remains unclear. Using primary nasal epithelial organoid cultures, we found that the virus attaches to motile cilia via the ACE2 receptor. SARS-CoV-2 traverses the mucus layer, using motile cilia as tracks to access the cell body. Depleting cilia blocks infection for SARS-CoV-2 and other respiratory viruses. SARS-CoV-2 progeny attach to airway microvilli 24h post-infection and trigger formation of apically extended and highly branched microvilli that organize viral egress from the microvilli back into the mucus layer, supporting a model of virus dispersion throughout airway tissue via mucociliary transport. Phosphoproteomics and kinase inhibition reveal that microvillar remodeling is regulated by p21-activated kinases (PAK). Importantly, Omicron variants bind with higher affinity to motile cilia and show accelerated viral entry. Our work suggests that motile cilia, microvilli, and mucociliary-dependent mucus flow are critical for efficient virus replication in nasal epithelia.

    View details for DOI 10.1016/j.cell.2022.11.030

    View details for PubMedID 36580912

  • Oxaliplatin disrupts nucleolar function through biophysical disintegration. Cell reports Schmidt, H. B., Jaafar, Z. A., Wulff, B. E., Rodencal, J. J., Hong, K., Aziz-Zanjani, M. O., Jackson, P. K., Leonetti, M. D., Dixon, S. J., Rohatgi, R., Brandman, O. 2022; 41 (6): 111629

    Abstract

    Platinum (Pt) compounds such as oxaliplatin are among the most commonly prescribed anti-cancer drugs. Despite their considerable clinical impact, the molecular basis of platinum cytotoxicity and cancer specificity remain unclear. Here we show that oxaliplatin, a backbone for the treatment of colorectal cancer, causes liquid-liquid demixing of nucleoli at clinically relevant concentrations. Our data suggest that this biophysical defect leads to cell-cycle arrest, shutdown of Pol I-mediated transcription, and ultimately cell death. We propose that instead of targeting a single molecule, oxaliplatin preferentially partitions into nucleoli, where it modifies nucleolar RNA and proteins. This mechanism provides a general approach for drugging the increasing number of cellular processes linked to biomolecular condensates.

    View details for DOI 10.1016/j.celrep.2022.111629

    View details for PubMedID 36351392

  • The Mettl3 epitranscriptomic writer amplifies p53 stress responses. Molecular cell Raj, N., Wang, M., Seoane, J. A., Zhao, R. L., Kaiser, A. M., Moonie, N. A., Demeter, J., Boutelle, A. M., Kerr, C. H., Mulligan, A. S., Moffatt, C., Zeng, S. X., Lu, H., Barna, M., Curtis, C., Chang, H. Y., Jackson, P. K., Attardi, L. D. 2022

    Abstract

    The p53 transcription factor drives anti-proliferative gene expression programs in response to diverse stressors, including DNA damage and oncogenic signaling. Here, we seek to uncover new mechanisms through which p53 regulates gene expression using tandem affinity purification/mass spectrometry to identify p53-interacting proteins. This approach identified METTL3, an m6A RNA-methyltransferase complex (MTC) constituent, as a p53 interactor. We find that METTL3 promotes p53 protein stabilization and target gene expression in response to DNA damage and oncogenic signals, by both catalytic activity-dependent and independent mechanisms. METTL3 also enhances p53 tumor suppressor activity in invivo mouse cancer models and human cancer cells. Notably, METTL3 only promotes tumor suppression in the context of intact p53. Analysis of human cancer genome data further supports the notion that the MTC reinforces p53 function in human cancer. Together, these studies reveal a fundamental role for METTL3 in amplifying p53 signaling in response to cellular stress.

    View details for DOI 10.1016/j.molcel.2022.04.010

    View details for PubMedID 35512709

  • LKB1 drives stasis and C/EBP-mediated reprogramming to an alveolar type II fate in lung cancer. Nature communications Murray, C. W., Brady, J. J., Han, M., Cai, H., Tsai, M. K., Pierce, S. E., Cheng, R., Demeter, J., Feldser, D. M., Jackson, P. K., Shackelford, D. B., Winslow, M. M. 2022; 13 (1): 1090

    Abstract

    LKB1 is among the most frequently altered tumor suppressors in lung adenocarcinoma. Inactivation of Lkb1 accelerates the growth and progression of oncogenic KRAS-driven lung tumors in mouse models. However, the molecular mechanisms by which LKB1 constrains lung tumorigenesis and whether the cancer state that stems from Lkb1 deficiency can be reverted remains unknown. To identify the processes governed by LKB1 in vivo, we generated an allele which enables Lkb1 inactivation at tumor initiation and subsequent Lkb1 restoration in established tumors. Restoration of Lkb1 in oncogenic KRAS-driven lung tumors suppressed proliferation and led to tumor stasis. Lkb1 restoration activated targets of C/EBP transcription factors and drove neoplastic cells from a progenitor-like state to a less proliferative alveolar type II cell-like state. We show that C/EBP transcription factors govern a subset of genes that are induced by LKB1 and depend upon NKX2-1. We also demonstrate that a defining factor of the alveolar type II lineage, C/EBPalpha, constrains oncogenic KRAS-driven lung tumor growth in vivo. Thus, this key tumor suppressor regulates lineage-specific transcription factors, thereby constraining lung tumor development through enforced differentiation.

    View details for DOI 10.1038/s41467-022-28619-8

    View details for PubMedID 35228570

  • Multi-omic analysis reveals divergent molecular events in scarring and regenerative wound healing. Cell stem cell Mascharak, S., Talbott, H. E., Januszyk, M., Griffin, M., Chen, K., Davitt, M. F., Demeter, J., Henn, D., Bonham, C. A., Foster, D. S., Mooney, N., Cheng, R., Jackson, P. K., Wan, D. C., Gurtner, G. C., Longaker, M. T. 1800

    Abstract

    Regeneration is the holy grail of tissue repair, but skin injury typically yields fibrotic, non-functional scars. Developing pro-regenerative therapies requires rigorous understanding of the molecular progression from injury to fibrosis or regeneration. Here, we report the divergent molecular events driving skin wound cells toward scarring or regenerative fates. We profile scarring versus YAP-inhibition-induced wound regeneration at the transcriptional (single-cell RNA sequencing), protein (timsTOF proteomics), and tissue (extracellular matrix ultrastructural analysis) levels. Using cell-surface barcoding, we integrate these data to reveal fibrotic and regenerative "molecular trajectories" of healing. We show that disrupting YAP mechanotransduction yields regenerative repair by fibroblasts with activated Trps1 and Wnt signaling. Finally, via invivo gene knockdown and overexpression in wounds, we identify Trps1 as a key regulatory gene that is necessary and partially sufficient for wound regeneration. Our findings serve as a multi-omic map of wound regeneration and could have therapeutic implications for pathologic fibroses.

    View details for DOI 10.1016/j.stem.2021.12.011

    View details for PubMedID 35077667

  • Primary cilia on muscle stem cells are critical to maintain regenerative capacity and are lost during aging Nature Communications Palla , A. R., Hilgendorf, K. I., Yang, A. V., Kerr, J. P., Hinken, A. C., Demeter, J., Kraft, P., Mooney, N. A., Yucel, N., Burns, D. M., Wang, Y. X., Jackson, P. K., Blau, H. M. 2022; 13(1):1439
  • A defective viral genome strategy elicits broad protective immunity against respiratory viruses. Cell Xiao, Y., Lidsky, P. V., Shirogane, Y., Aviner, R., Wu, C., Li, W., Zheng, W., Talbot, D., Catching, A., Doitsh, G., Su, W., Gekko, C. E., Nayak, A., Ernst, J. D., Brodsky, L., Brodsky, E., Rousseau, E., Capponi, S., Bianco, S., Nakamura, R., Jackson, P. K., Frydman, J., Andino, R. 2021

    Abstract

    RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.

    View details for DOI 10.1016/j.cell.2021.11.023

    View details for PubMedID 34852237

  • Identifying cancer drivers SCIENCE Cheng, R., Jackson, P. K. 2021; 374 (6563): 38-39
  • Determinants of SARS-CoV-2 entry and replication in airway mucosal tissue and susceptibility in smokers. Cell reports. Medicine Nakayama, T., Lee, I. T., Jiang, S., Matter, M. S., Yan, C. H., Overdevest, J. B., Wu, C., Goltsev, Y., Shih, L., Liao, C., Zhu, B., Bai, Y., Lidsky, P., Xiao, Y., Zarabanda, D., Yang, A., Easwaran, M., Schurch, C. M., Chu, P., Chen, H., Stalder, A. K., McIlwain, D. R., Borchard, N. A., Gall, P. A., Dholakia, S. S., Le, W., Xu, L., Tai, C., Yeh, T., Erickson-Direnzo, E., Duran, J. M., Mertz, K. D., Hwang, P. H., Haslbauer, J. D., Jackson, P. K., Menter, T., Andino, R., Canoll, P. D., DeConde, A. S., Patel, Z. M., Tzankov, A., Nolan, G. P., Nayak, J. V. 2021: 100421

    Abstract

    Understanding viral tropism is an essential step towards reducing SARS-CoV-2 transmission, decreasing mortality from COVID-19, and limiting opportunities for mutant strains to arise. Currently, little is known about the extent to which distinct tissue sites in the human head & neck region and proximal respiratory tract selectively permit SARS-CoV-2 infection and replication. In this translational study, we discover key variabilities in the expression of ACE2 and TMPRSS2, essential SARS-CoV-2 entry factors, among the mucosal tissues of the human proximal airways. We show that SARS-CoV-2 infection is present in all examined head & neck tissues, with a notable tropism for the nasal cavity and tracheal mucosa. Finally, we uncover an association between smoking and higher SARS-CoV-2 viral infection in the human proximal airway, which may explain the increased susceptibility of smokers to developing severe COVID-19. This is at least partially explained by differences in IFN-beta1 levels between smokers and non-smokers.

    View details for DOI 10.1016/j.xcrm.2021.100421

    View details for PubMedID 34604819

  • Ethacridine inhibits SARS-CoV-2 by inactivating viral particles. PLoS pathogens Li, X., Lidsky, P., Xiao, Y., Wu, C., Garcia-Knight, M., Yang, J., Nakayama, T., Nayak, J. V., Jackson, P. K., Andino, R., Shu, X. 2021; 17 (9): e1009898

    Abstract

    The respiratory disease COVID-19 is caused by the coronavirus SARS-CoV-2. Here we report the discovery of ethacridine as a potent drug against SARS-CoV-2 (EC50 ~ 0.08 muM). Ethacridine was identified via high-throughput screening of an FDA-approved drug library in living cells using a fluorescence assay. Plaque assays, RT-PCR and immunofluorescence imaging at various stages of viral infection demonstrate that the main mode of action of ethacridine is through inactivation of viral particles, preventing their binding to the host cells. Consistently, ethacridine is effective in various cell types, including primary human nasal epithelial cells that are cultured in an air-liquid interface. Taken together, our work identifies a promising, potent, and new use of the old drug via a distinct mode of action for inhibiting SARS-CoV-2.

    View details for DOI 10.1371/journal.ppat.1009898

    View details for PubMedID 34478458

  • Discovery of ciliary G protein-coupled receptors regulating pancreatic islet insulin and glucagon secretion. Genes & development Wu, C., Hilgendorf, K. I., Bevacqua, R. J., Hang, Y., Demeter, J., Kim, S. K., Jackson, P. K. 2021

    Abstract

    Multiple G protein-coupled receptors (GPCRs) are expressed in pancreatic islet cells, but the majority have unknown functions. We observed specific GPCRs localized to primary cilia, a prominent signaling organelle, in pancreatic alpha and beta cells. Loss of cilia disrupts beta-cell endocrine function, but the molecular drivers are unknown. Using functional expression, we identified multiple GPCRs localized to cilia in mouse and human islet alpha and beta cells, including FFAR4, PTGER4, ADRB2, KISS1R, and P2RY14. Free fatty acid receptor 4 (FFAR4) and prostaglandin E receptor 4 (PTGER4) agonists stimulate ciliary cAMP signaling and promote glucagon and insulin secretion by alpha- and beta-cell lines and by mouse and human islets. Transport of GPCRs to primary cilia requires TULP3, whose knockdown in primary human and mouse islets relocalized ciliary FFAR4 and PTGER4 and impaired regulated glucagon or insulin secretion, without affecting ciliary structure. Our findings provide index evidence that regulated hormone secretion by islet alpha and beta cells is controlled by ciliary GPCRs providing new targets for diabetes.

    View details for DOI 10.1101/gad.348261.121

    View details for PubMedID 34385262

  • SARS-CoV-2 infects human pancreatic beta cells and elicits beta cell impairment. Cell metabolism Wu, C., Lidsky, P. V., Xiao, Y., Lee, I. T., Cheng, R., Nakayama, T., Jiang, S., Demeter, J., Bevacqua, R. J., Chang, C. A., Whitener, R. L., Stalder, A. K., Zhu, B., Chen, H., Goltsev, Y., Tzankov, A., Nayak, J. V., Nolan, G. P., Matter, M. S., Andino, R., Jackson, P. K. 2021

    Abstract

    Emerging evidence points toward an intricate relationship between the pandemic of coronavirus disease 2019 (COVID-19) and diabetes. While preexisting diabetes is associated with severe COVID-19, it is unclear whether COVID-19 severity is a cause or consequence of diabetes. To mechanistically link COVID-19 to diabetes, we tested whether insulin-producing pancreatic beta cells can be infected by SARS-CoV-2 and cause beta cell depletion. We found that the SARS-CoV-2 receptor, ACE2, and related entry factors (TMPRSS2, NRP1, and TRFC) are expressed in beta cells, with selectively high expression of NRP1. We discovered that SARS-CoV-2 infects human pancreatic beta cells in patients who succumbed to COVID-19 and selectively infects human islet beta cells invitro. We demonstrated that SARS-CoV-2 infection attenuates pancreatic insulin levels and secretion and induces beta cell apoptosis, each rescued by NRP1 inhibition. Phosphoproteomic pathway analysis of infected islets indicates apoptotic beta cell signaling, similar to that observed in type 1 diabetes (T1D). In summary, our study shows SARS-CoV-2 can directly induce beta cell killing.

    View details for DOI 10.1016/j.cmet.2021.05.013

    View details for PubMedID 34081912

  • The AMBRA1 E3 ligase adaptor regulates the stability of cyclinD. Nature Chaikovsky, A. C., Li, C., Jeng, E. E., Loebell, S., Lee, M. C., Murray, C. W., Cheng, R., Demeter, J., Swaney, D. L., Chen, S., Newton, B. W., Johnson, J. R., Drainas, A. P., Shue, Y. T., Seoane, J. A., Srinivasan, P., He, A., Yoshida, A., Hipkins, S. Q., McCrea, E., Poltorack, C. D., Krogan, N. J., Diehl, J. A., Kong, C., Jackson, P. K., Curtis, C., Petrov, D. A., Bassik, M. C., Winslow, M. M., Sage, J. 2021

    Abstract

    The initiation of cell division integrates a large number of intra- and extracellular inputs. D-type cyclins (hereafter, cyclinD) couple these inputs to the initiation of DNA replication1. Increased levels of cyclinD promote cell division by activating cyclin-dependent kinases4 and 6 (hereafter, CDK4/6), which in turn phosphorylate and inactivate the retinoblastoma tumour suppressor. Accordingly, increased levels and activity of cyclinD-CDK4/6 complexes are strongly linked to unchecked cell proliferation and cancer2,3. However, the mechanisms that regulate levels of cyclinD are incompletely understood4,5. Here we show that autophagy and beclin1 regulator1 (AMBRA1) is the main regulator of the degradation of cyclinD. We identified AMBRA1 in a genome-wide screen to investigate the genetic basis of the response to CDK4/6 inhibition. Loss of AMBRA1 results in high levels of cyclinD in cells and in mice, which promotes proliferation and decreases sensitivity to CDK4/6 inhibition. Mechanistically, AMBRA1 mediates ubiquitylation and proteasomal degradation of cyclinD as a substrate receptor for the cullin4 E3 ligase complex. Loss of AMBRA1 enhances the growth of lung adenocarcinoma in a mouse model, and low levels of AMBRA1 correlate with worse survival in patients with lung adenocarcinoma. Thus, AMBRA1 regulates cellular levels of cyclinD, and contributes to cancer development and the response of cancer cells to CDK4/6 inhibitors.

    View details for DOI 10.1038/s41586-021-03474-7

    View details for PubMedID 33854239

  • Identifying cancer drivers. Science (New York, N.Y.) Cheng, R., Jackson, P. K. 2021; 374 (6563): 38-39

    Abstract

    [Figure: see text].

    View details for DOI 10.1126/science.abl9080

    View details for PubMedID 34591644

  • Structured elements drive extensive circular RNA translation. Molecular cell Chen, C. K., Cheng, R., Demeter, J., Chen, J., Weingarten-Gabbay, S., Jiang, L., Snyder, M. P., Weissman, J. S., Segal, E., Jackson, P. K., Chang, H. Y. 2021

    Abstract

    The human genome encodes tens of thousands circular RNAs (circRNAs) with mostly unknown functions. Circular RNAs require internal ribosome entry sites (IRES) if they are to undergo translation without a 5' cap. Here, we develop a high-throughput screen to systematically discover RNA sequences that can direct circRNA translation in human cells. We identify more than 17,000 endogenous and synthetic sequences as candidate circRNA IRES. 18S rRNA complementarity and a structured RNA element positioned on the IRES are important for driving circRNA translation. Ribosome profiling and peptidomic analyses show extensive IRES-ribosome association, hundreds of circRNA-encoded proteins with tissue-specific distribution, and antigen presentation. We find that circFGFR1p, a protein encoded by circFGFR1 that is downregulated in cancer, functions as a negative regulator of FGFR1 oncoprotein to suppress cell growth during stress. Systematic identification of circRNA IRES elements may provide important links among circRNA regulation, biological function, and disease.

    View details for DOI 10.1016/j.molcel.2021.07.042

    View details for PubMedID 34437836

  • Structure-activity mapping of ARHGAP36 reveals regulatory roles for its GAP homology and C-terminal domains. PloS one Nano, P. R., Johnson, T. K., Kudo, T., Mooney, N. A., Ni, J., Demeter, J., Jackson, P. K., Chen, J. K. 2021; 16 (5): e0251684

    Abstract

    ARHGAP36 is an atypical Rho GTPase-activating protein (GAP) family member that drives both spinal cord development and tumorigenesis, acting in part through an N-terminal motif that suppresses protein kinase A and activates Gli transcription factors. ARHGAP36 also contains isoform-specific N-terminal sequences, a central GAP-like module, and a unique C-terminal domain, and the functions of these regions remain unknown. Here we have mapped the ARHGAP36 structure-activity landscape using a deep sequencing-based mutagenesis screen and truncation mutant analyses. Using this approach, we have discovered several residues in the GAP homology domain that are essential for Gli activation and a role for the C-terminal domain in counteracting an N-terminal autoinhibitory motif that is present in certain ARHGAP36 isoforms. In addition, each of these sites modulates ARHGAP36 recruitment to the plasma membrane or primary cilium. Through comparative proteomics, we also have identified proteins that preferentially interact with active ARHGAP36, and we demonstrate that one binding partner, prolyl oligopeptidase-like protein, is a novel ARHGAP36 antagonist. Our work reveals multiple modes of ARHGAP36 regulation and establishes an experimental framework that can be applied towards other signaling proteins.

    View details for DOI 10.1371/journal.pone.0251684

    View details for PubMedID 33999959

  • Proteomic analysis of young and old mouse hematopoietic stem cells and their progenitors reveals post-transcriptional regulation in stem cells. eLife Zaro, B. W., Noh, J. J., Mascetti, V. L., Demeter, J., George, B., Zukowska, M., Gulati, G. S., Sinha, R., Flynn, R. A., Banuelos, A., Zhang, A., Wilkinson, A. C., Jackson, P., Weissman, I. L. 2020; 9

    Abstract

    The balance of hematopoietic stem cell (HSC) self-renewal and differentiation is critical for a healthy blood supply; imbalances underlie hematological diseases. The importance of HSCs and their progenitors have led to their extensive characterization at genomic and transcriptomic levels. However, the proteomics of hematopoiesis remains incompletely understood. Here we report a proteomics resource from mass spectrometry of mouse young adult and old adult mouse HSCs, multipotent progenitors and oligopotent progenitors; 12 cell types in total. We validated differential protein levels, including confirmation that Dnmt3a protein levels are undetected in young adult mouse HSCs until forced into cycle. Additionally, through integrating proteomics and RNA-sequencing datasets, we identified a subset of genes with apparent post-transcriptional repression in young adult mouse HSCs. In summary, we report proteomic coverage of young and old mouse HSCs and progenitors, with broader implications for understanding mechanisms for stem cell maintenance, niche interactions and fate determination.

    View details for DOI 10.7554/eLife.62210

    View details for PubMedID 33236985

  • ACE2 localizes to the respiratory cilia and is not increased by ACE inhibitors or ARBs. Nature communications Lee, I. T., Nakayama, T., Wu, C., Goltsev, Y., Jiang, S., Gall, P. A., Liao, C., Shih, L., Schurch, C. M., McIlwain, D. R., Chu, P., Borchard, N. A., Zarabanda, D., Dholakia, S. S., Yang, A., Kim, D., Chen, H., Kanie, T., Lin, C., Tsai, M., Phillips, K. M., Kim, R., Overdevest, J. B., Tyler, M. A., Yan, C. H., Lin, C., Lin, Y., Bau, D., Tsay, G. J., Patel, Z. M., Tsou, Y., Tzankov, A., Matter, M. S., Tai, C., Yeh, T., Hwang, P. H., Nolan, G. P., Nayak, J. V., Jackson, P. K. 2020; 11 (1): 5453

    Abstract

    The coronavirus SARS-CoV-2 is the causative agent of the ongoing severe acute respiratory disease pandemic COVID-19. Tissue and cellular tropism is one key to understanding the pathogenesis of SARS-CoV-2. We investigate the expression and subcellular localization of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), within the upper (nasal) and lower (pulmonary) respiratory tracts of human donors using a diverse panel of banked tissues. Here, we report our discovery that the ACE2 receptor protein robustly localizes within the motile cilia of airway epithelial cells, which likely represents the initial or early subcellular site of SARS-CoV-2 viral entry during host respiratory transmission. We further determine whether ciliary ACE2 expression in the upper airway is influenced by patient demographics, clinical characteristics, comorbidities, or medication use, and show the first mechanistic evidence that the use of angiotensin-converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARBs) does not increase susceptibility to SARS-CoV-2 infection through enhancing the expression of ciliary ACE2 receptor. These findings are crucial to our understanding of the transmission of SARS-CoV-2 for prevention and control of this virulent pathogen.

    View details for DOI 10.1038/s41467-020-19145-6

    View details for PubMedID 33116139

  • Ethacridine inhibits SARS-CoV-2 by inactivating viral particles in cellular models. bioRxiv : the preprint server for biology Li, X., Lidsky, P., Xiao, Y., Wu, C. T., GarciaKnight, M., Yang, J., Nakayama, T., Nayak, J. V., Jackson, P. K., Andino, R., Shu, X. 2020

    Abstract

    More than a million people have now died from COVID-19, because of infection with the SARS-CoV-2 coronavirus. Currently, the FDA has approved remdesivir, an inhibitor of SARS-CoV-2 replication, to treat COVID-19, though very recent data from WHO showed little if any COVID19 protective effect. Here we report that ethacridine, a safe and potent antiseptic use in humans, effectively inhibits SARS-CoV-2, at very low concentrations (EC 50 ~ 0.08 μ M). Ethacridine was identified through a high-throughput screening of an FDA-approved drug library in living cells using a fluorescent assay. Interestingly, the main mode of action of ethacridine is to inactivate virus particles, preventing binding to the host cells. Thus, our work has identified a potent drug with a distinct mode of action against SARS-CoV-2.

    View details for DOI 10.1101/2020.10.28.359042

    View details for PubMedID 33140048

    View details for PubMedCentralID PMC7605555

  • Oncoprotein-specific molecular interaction maps (SigMaps) for cancer network analyses. Nature biotechnology Broyde, J., Simpson, D. R., Murray, D., Paull, E. O., Chu, B. W., Tagore, S., Jones, S. J., Griffin, A. T., Giorgi, F. M., Lachmann, A., Jackson, P., Sweet-Cordero, E. A., Honig, B., Califano, A. 2020

    Abstract

    Tumor-specific elucidation of physical and functional oncoprotein interactions could improve tumorigenic mechanism characterization and therapeutic response prediction. Current interaction models and pathways, however, lack context specificity and are not oncoprotein specific. We introduce SigMaps as context-specific networks, comprising modulators, effectors and cognate binding-partners of a specific oncoprotein. SigMaps are reconstructed de novo by integrating diverse evidence sources-including protein structure, gene expression and mutational profiles-via the OncoSig machine learning framework. We first generated a KRAS-specific SigMap for lung adenocarcinoma, which recapitulated published KRAS biology, identified novel synthetic lethal proteins that were experimentally validated in three-dimensional spheroid models and established uncharacterized crosstalk with RAB/RHO. To show that OncoSig is generalizable, we first inferred SigMaps for the ten most mutated human oncoproteins and then for the full repertoire of 715 proteins in the COSMIC Cancer Gene Census. Taken together, these SigMaps show that the cell's regulatory and signaling architecture is highly tissue specific.

    View details for DOI 10.1038/s41587-020-0652-7

    View details for PubMedID 32929263

  • Combined Proteomic and Genetic Interaction Mapping Reveals New RAS Effector Pathways and Susceptibilities. Cancer discovery Kelly, M. R., Kostyrko, K., Han, K., Mooney, N. A., Jeng, E. E., Spees, K., Dinh, P. T., Abbott, K. L., Gwinn, D. M., Sweet-Cordero, E. A., Bassik, M. C., Jackson, P. K. 2020

    Abstract

    Activating mutations in RAS GTPases drive many cancers, but limited understanding of less-studied RAS interactors, and of the specific roles of different RAS interactor paralogs, continues to limit target discovery. We developed a multistage discovery and screening process to systematically identify genes conferring RAS-related susceptibilities in lung adenocarcinoma. Using affinity purification mass spectrometry, we generated a protein-protein interaction map of RAS interactors and pathway components containing hundreds of interactions. From this network, we constructed a CRISPR dual knockout library targeting 119 RAS-related genes that we screened for KRAS-dependent genetic interactions (GIs). This approach identified new RAS effectors, including the adhesion controller RADIL and the endocytosis regulator RIN1, and >250 synthetic lethal GIs, including a potent KRAS-dependent interaction between RAP1GDS1 and RHOA. Many GIs link specific paralogs within and between gene families. These findings illustrate the power of multiomic approaches to uncover synthetic lethal combinations specific for hitherto untreatable cancer genotypes.

    View details for DOI 10.1158/2159-8290.CD-19-1274

    View details for PubMedID 32727735

  • CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature Han, K., Pierce, S. E., Li, A., Spees, K., Anderson, G. R., Seoane, J. A., Lo, Y. H., Dubreuil, M., Olivas, M., Kamber, R. A., Wainberg, M., Kostyrko, K., Kelly, M. R., Yousefi, M., Simpkins, S. W., Yao, D., Lee, K., Kuo, C. J., Jackson, P. K., Sweet-Cordero, A., Kundaje, A., Gentles, A. J., Curtis, C., Winslow, M. M., Bassik, M. C. 2020; 580 (7801): 136-141

    Abstract

    Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.

    View details for DOI 10.1038/s41586-020-2099-x

    View details for PubMedID 32238925

  • Novel fibrillar structure in the inversin compartment of primary cilia revealed by 3D single-molecule super-resolution microscopy. Molecular biology of the cell Bennett, H. W., Gustavsson, A., Bayas, C. A., Petrov, P. N., Mooney, N., Moerner, W. E., Jackson, P. K. 2020: mbcE19090499

    Abstract

    Primary cilia in many cell types contain a peri-axonemal sub-compartment called the inversin compartment. Four proteins have been found to assemble within the inversin compartment: INVS, ANKS6, NEK8, and NPHP3. The function of the inversin compartment is unknown, but it appears to be critical for normal development including left-right asymmetry and renal tissue homeostasis. Here we combine super-resolution imaging of human RPE1 cells, a classic model for studying primary cilia in vitro, with a genetic dissection of the protein-protein binding relationships that organize compartment assembly to develop a new structural model. We observe that INVS is the core structural determinant of a compartment composed of novel fibril-like substructures, which we identify here by 3D single-molecule super-resolution imaging. We find that NEK8 and ANKS6 depend on INVS for localization to these fibrillar assemblies and that ANKS6-NEK8 density within the compartment is regulated by NEK8. Together, NEK8 and ANKS6 are required downstream of INVS to localize and concentrate NPHP3 within the compartment. In the absence of these upstream components, NPHP3 is redistributed within cilia. These results provide a more detailed structure for the inversin compartment and introduce a new example of a membraneless compartment organized by protein-protein interactions. [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text].

    View details for DOI 10.1091/mbc.E19-09-0499

    View details for PubMedID 31895004

  • Unbiased Proteomic Profiling Uncovers a Targetable GNAS/PKA/PP2A Axis in Small Cell Lung Cancer Stem Cells. Cancer cell Coles, G. L., Cristea, S. n., Webber, J. T., Levin, R. S., Moss, S. M., He, A. n., Sangodkar, J. n., Hwang, Y. C., Arand, J. n., Drainas, A. P., Mooney, N. A., Demeter, J. n., Spradlin, J. N., Mauch, B. n., Le, V. n., Shue, Y. T., Ko, J. H., Lee, M. C., Kong, C. n., Nomura, D. K., Ohlmeyer, M. n., Swaney, D. L., Krogan, N. J., Jackson, P. K., Narla, G. n., Gordan, J. D., Shokat, K. M., Sage, J. n. 2020

    Abstract

    Using unbiased kinase profiling, we identified protein kinase A (PKA) as an active kinase in small cell lung cancer (SCLC). Inhibition of PKA activity genetically, or pharmacologically by activation of the PP2A phosphatase, suppresses SCLC expansion in culture and in vivo. Conversely, GNAS (G-protein α subunit), a PKA activator that is genetically activated in a small subset of human SCLC, promotes SCLC development. Phosphoproteomic analyses identified many PKA substrates and mechanisms of action. In particular, PKA activity is required for the propagation of SCLC stem cells in transplantation studies. Broad proteomic analysis of recalcitrant cancers has the potential to uncover targetable signaling networks, such as the GNAS/PKA/PP2A axis in SCLC.

    View details for DOI 10.1016/j.ccell.2020.05.003

    View details for PubMedID 32531271

  • cAMP Signaling in Nanodomains. Cell Jackson, P. K. 2020; 182 (6): 1379–81

    Abstract

    Cyclic-3',5'-adenosine monophosphate (cAMP) is an ancient second messenger but organizing signaling selectivity on the nanoscale is poorly understood. Examining transport of a new fluorescent cAMP probe, Bock and coworkers observe "buffered diffusion" and establish phosphodiesterase activity can organize cAMP nanodomains, while Zhao and coworkers find that protein kinase A regulatory subunits assemble liquid droplets to further localize cAMP signaling.

    View details for DOI 10.1016/j.cell.2020.08.041

    View details for PubMedID 32946779

  • Robust ACE2 protein expression localizes to the motile cilia of the respiratory tract epithelia and is not increased by ACE inhibitors or angiotensin receptor blockers. medRxiv : the preprint server for health sciences Lee, I. T., Nakayama, T. n., Wu, C. T., Goltsev, Y. n., Jiang, S. n., Gall, P. A., Liao, C. K., Shih, L. C., Schürch, C. M., McIlwain, D. R., Chu, P. n., Borchard, N. A., Zarabanda, D. n., Dholakia, S. S., Yang, A. n., Kim, D. n., Kanie, T. n., Lin, C. D., Tsai, M. H., Phillips, K. M., Kim, R. n., Overdevest, J. B., Tyler, M. A., Yan, C. H., Lin, C. F., Lin, Y. T., Bau, D. T., Tsay, G. J., Patel, Z. M., Tsou, Y. A., Tai, C. J., Yeh, T. H., Hwang, P. H., Nolan, G. P., Nayak, J. V., Jackson, P. K. 2020

    Abstract

    We investigated the expression and subcellular localization of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), within the upper (nasal) and lower (pulmonary) respiratory tracts of healthy human donors. We detected ACE2 protein expression within the cilia organelle of ciliated airway epithelial cells, which likely represents the initial or early subcellular site of SARS-CoV-2 viral entry during respiratory transmission. We further determined whether ACE2 expression in the cilia of upper respiratory cells was influenced by patient demographics, clinical characteristics, co-morbidities, or medication use, and found no evidence that the use of angiotensin-converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARBs) increases ACE2 protein expression.

    View details for DOI 10.1101/2020.05.08.20092866

    View details for PubMedID 32511516

    View details for PubMedCentralID PMC7273284

  • Omega-3 Fatty Acids Activate Ciliary FFAR4 to Control Adipogenesis. Cell Hilgendorf, K. I., Johnson, C. T., Mezger, A., Rice, S. L., Norris, A. M., Demeter, J., Greenleaf, W. J., Reiter, J. F., Kopinke, D., Jackson, P. K. 2019

    Abstract

    Adult mesenchymal stem cells, including preadipocytes, possess a cellular sensory organelle called the primary cilium. Ciliated preadipocytes abundantly populate perivascular compartments in fat and are activated by a high-fat diet. Here, we sought to understand whether preadipocytes use their cilia to sense and respond to external cues to remodel white adipose tissue. Abolishing preadipocyte cilia in mice severely impairs white adipose tissue expansion. We discover that TULP3-dependent ciliary localization of the omega-3 fatty acid receptor FFAR4/GPR120 promotes adipogenesis. FFAR4 agonists and omega-3 fatty acids, but not saturated fatty acids, trigger mitosis and adipogenesis by rapidly activating cAMP production inside cilia. Ciliary cAMP activates EPAC signaling, CTCF-dependent chromatin remodeling, and transcriptional activation of PPARgamma and CEBPalpha to initiate adipogenesis. We propose that dietary omega-3 fatty acids selectively drive expansion of adipocyte numbers to produce new fatcells and store saturated fatty acids, enabling homeostasis of healthy fat tissue.

    View details for DOI 10.1016/j.cell.2019.11.005

    View details for PubMedID 31761534

  • Oligomeric self-association contributes to E2A-PBX1-mediated oncogenesis. Scientific reports Lin, C., Wang, Z., Duque-Afonso, J., Wong, S. H., Demeter, J., Loktev, A. V., Somervaille, T. C., Jackson, P. K., Cleary, M. L. 2019; 9 (1): 4915

    Abstract

    The PBX1 homeodomain transcription factor is converted by t(1;19) chromosomal translocations in acute leukemia into the chimeric E2A-PBX1 oncoprotein. Fusion with E2A confers potent transcriptional activation and constitutive nuclear localization, bypassing the need for dimerization with protein partners that normally stabilize and regulate import of PBX1 into the nucleus, but the mechanisms underlying its oncogenic activation are incompletely defined. We demonstrate here that E2A-PBX1 self-associates through the PBX1 PBC-B domain of the chimeric protein to form higher-order oligomers in t(1;19) human leukemia cells, and that this property is required for oncogenic activity. Structural and functional studies indicate that self-association facilitates the binding of E2A-PBX1 to DNA. Mutants unable to self-associate are transformation defective, however their oncogenic activity is rescued by the synthetic oligomerization domain of FKBP, which confers conditional transformation properties on E2A-PBX1. In contrast to self-association, PBX1 protein domains that mediate interactions with HOX DNA-binding partners are dispensable. These studies suggest that oligomeric self-association may compensate for the inability of monomeric E2A-PBX1 to stably bind DNA and circumvents protein interactions that otherwise modulate PBX1 stability, nuclear localization, DNA binding, and transcriptional activity. The unique dependence on self-association for E2A-PBX1 oncogenic activity suggests potential approaches for mechanism-based targeted therapies.

    View details for PubMedID 30894657

  • E2F4 regulates transcriptional activation in mouse embryonic stem cells independently of the RB family. Nature communications Hsu, J. n., Arand, J. n., Chaikovsky, A. n., Mooney, N. A., Demeter, J. n., Brison, C. M., Oliverio, R. n., Vogel, H. n., Rubin, S. M., Jackson, P. K., Sage, J. n. 2019; 10 (1): 2939

    Abstract

    E2F transcription factors are central regulators of cell division and cell fate decisions. E2F4 often represents the predominant E2F activity in cells. E2F4 is a transcriptional repressor implicated in cell cycle arrest and whose repressive activity depends on its interaction with members of the RB family. Here we show that E2F4 is important for the proliferation and the survival of mouse embryonic stem cells. In these cells, E2F4 acts in part as a transcriptional activator that promotes the expression of cell cycle genes. This role for E2F4 is independent of the RB family. Furthermore, E2F4 functionally interacts with chromatin regulators associated with gene activation and we observed decreased histone acetylation at the promoters of cell cycle genes and E2F targets upon loss of E2F4 in RB family-mutant cells. Taken together, our findings uncover a non-canonical role for E2F4 that provide insights into the biology of rapidly dividing cells.

    View details for DOI 10.1038/s41467-019-10901-x

    View details for PubMedID 31270324

  • EZH2 Inactivates Primary Cilia to Activate Wnt and Drive Melanoma. Cancer cell Jackson, P. K. 2018; 34 (1): 3–5

    Abstract

    EZH2 is frequently amplified in human melanomas. In this issue of Cancer Cell, Zingg etal. find that EZH2 overexpression silences genes for the primary cilium, causing deciliation, Wnt pathway activation, and progression of BrafV600E- or NrasQ61N-driven melanomas, thus defining a tumor-suppressor role for cilia in cancer.

    View details for PubMedID 29990499

  • Drebrin restricts rotavirus entry by inhibiting dynamin-mediated endocytosis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Li, B., Ding, S., Feng, N., Mooney, N., Ooi, Y. S., Ren, L., Diep, J., Kelly, M. R., Yasukawa, L. L., Patton, J. T., Yamazaki, H., Shirao, T., Jackson, P. K., Greenberg, H. B. 2017; 114 (18): E3642-E3651

    Abstract

    Despite the wide administration of several effective vaccines, rotavirus (RV) remains the single most important etiological agent of severe diarrhea in infants and young children worldwide, with an annual mortality of over 200,000 people. RV attachment and internalization into target cells is mediated by its outer capsid protein VP4. To better understand the molecular details of RV entry, we performed tandem affinity purification coupled with high-resolution mass spectrometry to map the host proteins that interact with VP4. We identified an actin-binding protein, drebrin (DBN1), that coprecipitates and colocalizes with VP4 during RV infection. Importantly, blocking DBN1 function by siRNA silencing, CRISPR knockout (KO), or chemical inhibition significantly increased host cell susceptibility to RV infection. Dbn1 KO mice exhibited higher incidence of diarrhea and more viral antigen shedding in their stool samples compared with the wild-type littermates. In addition, we found that uptake of other dynamin-dependent cargos, including transferrin, cholera toxin, and multiple viruses, was also enhanced in DBN1-deficient cells. Inhibition of cortactin or dynamin-2 abrogated the increased virus entry observed in DBN1-deficient cells, suggesting that DBN1 suppresses dynamin-mediated endocytosis via interaction with cortactin. Our study unveiled an unexpected role of DBN1 in restricting the entry of RV and other viruses into host cells and more broadly to function as a crucial negative regulator of diverse dynamin-dependent endocytic pathways.

    View details for DOI 10.1073/pnas.1619266114

    View details for Web of Science ID 000400358000009

    View details for PubMedID 28416666

    View details for PubMedCentralID PMC5422808

  • Neural Precursor-Derived Pleiotrophin Mediates Subventricular Zone Invasion by Glioma. Cell Qin, E. Y., Cooper, D. D., Abbott, K. L., Lennon, J. n., Nagaraja, S. n., Mackay, A. n., Jones, C. n., Vogel, H. n., Jackson, P. K., Monje, M. n. 2017; 170 (5): 845–59.e19

    Abstract

    The lateral ventricle subventricular zone (SVZ) is a frequent and consequential site of pediatric and adult glioma spread, but the cellular and molecular mechanisms mediating this are poorly understood. We demonstrate that neural precursor cell (NPC):glioma cell communication underpins this propensity of glioma to colonize the SVZ through secretion of chemoattractant signals toward which glioma cells home. Biochemical, proteomic, and functional analyses of SVZ NPC-secreted factors revealed the neurite outgrowth-promoting factor pleiotrophin, along with required binding partners SPARC/SPARCL1 and HSP90B, as key mediators of this chemoattractant effect. Pleiotrophin expression is strongly enriched in the SVZ, and pleiotrophin knock down starkly reduced glioma invasion of the SVZ in the murine brain. Pleiotrophin, in complex with the binding partners, activated glioma Rho/ROCK signaling, and ROCK inhibition decreased invasion toward SVZ NPC-secreted factors. These findings demonstrate a pathogenic role for NPC:glioma interactions and potential therapeutic targets to limit glioma invasion. PAPERCLIP.

    View details for PubMedID 28823557

  • The CEP19-RABL2 GTPase Complex Binds IFT-B to Initiate Intraflagellar Transport at the Ciliary Base. Developmental cell Kanie, T. n., Abbott, K. L., Mooney, N. A., Plowey, E. D., Demeter, J. n., Jackson, P. K. 2017

    Abstract

    Highly conserved intraflagellar transport (IFT) protein complexes direct both the assembly of primary cilia and the trafficking of signaling molecules. IFT complexes initially accumulate at the base of the cilium and periodically enter the cilium, suggesting an as-yet-unidentified mechanism that triggers ciliary entry of IFT complexes. Using affinity-purification and mass spectrometry of interactors of the centrosomal and ciliopathy protein, CEP19, we identify CEP350, FOP, and the RABL2B GTPase as proteins organizing the first known mechanism directing ciliary entry of IFT complexes. We discover that CEP19 is recruited to the ciliary base by the centriolar CEP350/FOP complex and then specifically captures GTP-bound RABL2B, which is activated via its intrinsic nucleotide exchange. Activated RABL2B then captures and releases its single effector, the intraflagellar transport B holocomplex, from the large pool of pre-docked IFT-B complexes, and thus initiates ciliary entry of IFT.

    View details for PubMedID 28625565

  • Comparative Proteomics Reveals Strain-Specific ß-TrCP Degradation via Rotavirus NSP1 Hijacking a Host Cullin-3-Rbx1 Complex. PLoS pathogens Ding, S., Mooney, N., Li, B., Kelly, M. R., Feng, N., Loktev, A. V., Sen, A., Patton, J. T., Jackson, P. K., Greenberg, H. B. 2016; 12 (10)

    Abstract

    Rotaviruses (RVs) are the leading cause of severe gastroenteritis in young children, accounting for half a million deaths annually worldwide. RV encodes non-structural protein 1 (NSP1), a well-characterized interferon (IFN) antagonist, which facilitates virus replication by mediating the degradation of host antiviral factors including IRF3 and β-TrCP. Here, we utilized six human and animal RV NSP1s as baits and performed tandem-affinity purification coupled with high-resolution mass spectrometry to comprehensively characterize NSP1-host protein interaction network. Multiple Cullin-RING ubiquitin ligase (CRL) complexes were identified. Importantly, inhibition of cullin-3 (Cul3) or RING-box protein 1 (Rbx1), by siRNA silencing or chemical perturbation, significantly impairs strain-specific NSP1-mediated β-TrCP degradation. Mechanistically, we demonstrate that NSP1 localizes to the Golgi with the host Cul3-Rbx1 CRL complex, which targets β-TrCP and NSP1 for co-destruction at the proteasome. Our study uncovers a novel mechanism that RV employs to promote β-TrCP turnover and provides molecular insights into virus-mediated innate immunity inhibition.

    View details for DOI 10.1371/journal.ppat.1005929

    View details for PubMedID 27706223

    View details for PubMedCentralID PMC5051689

  • Metabolic plasticity underpins innate and acquired resistance to LDHA inhibition NATURE CHEMICAL BIOLOGY Boudreau, A., Purkey, H. E., Hitz, A., Robarge, K., Peterson, D., Labadie, S., Kwong, M., Hong, R., Gao, M., Del Nagro, C., Pusapati, R., Ma, S., Salphati, L., Pang, J., Zhou, A., Lai, T., Li, Y., Chen, Z., Wei, B., Yen, I., Sideris, S., McCleland, M., Firestein, R., Corson, L., Vanderbilt, A., Williams, S., Daemen, A., Belvin, M., Eigenbrot, C., Jackson, P. K., Malek, S., Hatzivassiliou, G., Sampath, D., Evangelista, M., O'Brien, T. 2016; 12 (10): 779-?

    Abstract

    Metabolic reprogramming in tumors represents a potential therapeutic target. Herein we used shRNA depletion and a novel lactate dehydrogenase (LDHA) inhibitor, GNE-140, to probe the role of LDHA in tumor growth in vitro and in vivo. In MIA PaCa-2 human pancreatic cells, LDHA inhibition rapidly affected global metabolism, although cell death only occurred after 2 d of continuous LDHA inhibition. Pancreatic cell lines that utilize oxidative phosphorylation (OXPHOS) rather than glycolysis were inherently resistant to GNE-140, but could be resensitized to GNE-140 with the OXPHOS inhibitor phenformin. Acquired resistance to GNE-140 was driven by activation of the AMPK-mTOR-S6K signaling pathway, which led to increased OXPHOS, and inhibitors targeting this pathway could prevent resistance. Thus, combining an LDHA inhibitor with compounds targeting the mitochondrial or AMPK-S6K signaling axis may not only broaden the clinical utility of LDHA inhibitors beyond glycolytically dependent tumors but also reduce the emergence of resistance to LDHA inhibition.

    View details for DOI 10.1038/nchembio.2143

    View details for Web of Science ID 000384248600008

    View details for PubMedID 27479743

  • Membrane trafficking regulation of intracellular ciliogenesis initiation and progression in RPE-1 cells and photoreceptors Insinna, C., Lu, Q., Ott, C., Lippincott-Schwartz, J., Jackson, P., Westlake, C. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2016
  • The ciliopathy-associated CPLANE proteins direct basal body recruitment of intraflagellar transport machinery NATURE GENETICS Toriyama, M., Lee, C., Taylor, S. P., Duran, I., Cohn, D. H., Bruel, A., Tabler, J. M., Drew, K., Kelly, M. R., Kim, S., Park, T. J., Braun, D. A., Pierquin, G., Biver, A., Wagner, K., Malfroot, A., Panigrahi, I., Franco, B., Al-Lami, H. A., Yeung, Y., Choi, Y. J., Duffourd, Y., Faivre, L., Riviere, J., Chen, J., Liu, K. J., Marcotte, E. M., Hildebrandt, F., Thauvin-Robinet, C., Krakow, D., Jackson, P. K., Wallingford, J. B. 2016; 48 (6): 648-?

    Abstract

    Cilia use microtubule-based intraflagellar transport (IFT) to organize intercellular signaling. Ciliopathies are a spectrum of human diseases resulting from defects in cilia structure or function. The mechanisms regulating the assembly of ciliary multiprotein complexes and the transport of these complexes to the base of cilia remain largely unknown. Combining proteomics, in vivo imaging and genetic analysis of proteins linked to planar cell polarity (Inturned, Fuzzy and Wdpcp), we identified and characterized a new genetic module, which we term CPLANE (ciliogenesis and planar polarity effector), and an extensive associated protein network. CPLANE proteins physically and functionally interact with the poorly understood ciliopathy-associated protein Jbts17 at basal bodies, where they act to recruit a specific subset of IFT-A proteins. In the absence of CPLANE, defective IFT-A particles enter the axoneme and IFT-B trafficking is severely perturbed. Accordingly, mutation of CPLANE genes elicits specific ciliopathy phenotypes in mouse models and is associated with ciliopathies in human patients.

    View details for DOI 10.1038/ng.3558

    View details for PubMedID 27158779

  • p73 anc FoxJ1: Programming Multiciliated Epithelia TRENDS IN CELL BIOLOGY Jackson, P. K., Attardi, L. D. 2016; 26 (4): 239–40

    Abstract

    The mysteriously diverse phenotypes in mice lacking the p53 homolog p73 are recently unified by new analysis showing p73 is required for formation of multiciliated epithelia. p73 directly activates FoxJ1, the central transcriptional driver for multiciliation, and induces a host of genes critical for ciliogenesis.

    View details for PubMedID 26988441

  • The primary cilium as a cellular receiver: organizing ciliary GPCR signaling. Current opinion in cell biology Hilgendorf, K. I., Johnson, C. T., Jackson, P. K. 2016; 39: 84-92

    Abstract

    The primary cilium is an antenna-like cellular protrusion mediating sensory and neuroendocrine signaling. Its localization within tissue architecture and a growing list of cilia-localized receptors, in particular G-protein-coupled receptors, determine a host of crucial physiologies, which are disrupted in human ciliopathies. Here, we discuss recent advances in the identification and characterization of ciliary signaling components and pathways. Recent studies have highlighted the unique signaling environment of the primary cilium and we are just beginning to understand how this design allows for highly amplified and regulated signaling.

    View details for DOI 10.1016/j.ceb.2016.02.008

    View details for PubMedID 26926036

    View details for PubMedCentralID PMC4828300

  • Signals straightened out NATURE Jackson, P. K. 2016; 531 (7596): 582–83
  • Smoothened determines beta-arrestin-mediated removal of the G protein-coupled receptor Gpr161 from the primary cilium JOURNAL OF CELL BIOLOGY Pal, K., Hwang, S., Somatilaka, B., Badgandi, H., Jackson, P. K., DeFea, K., Mukhopadhyay, S. 2016; 212 (7): 861-875

    Abstract

    Dynamic changes in membrane protein composition of the primary cilium are central to development and homeostasis, but we know little about mechanisms regulating membrane protein flux. Stimulation of the sonic hedgehog (Shh) pathway in vertebrates results in accumulation and activation of the effector Smoothened within cilia and concomitant disappearance of a negative regulator, the orphan G protein-coupled receptor (GPCR), Gpr161. Here, we describe a two-step process determining removal of Gpr161 from cilia. The first step involves β-arrestin recruitment by the signaling competent receptor, which is facilitated by the GPCR kinase Grk2. An essential factor here is the ciliary trafficking and activation of Smoothened, which by increasing Gpr161-β-arrestin binding promotes Gpr161 removal, both during resting conditions and upon Shh pathway activation. The second step involves clathrin-mediated endocytosis, which functions outside of the ciliary compartment in coordinating Gpr161 removal. Mechanisms determining dynamic compartmentalization of Gpr161 in cilia define a new paradigm for down-regulation of GPCRs during developmental signaling from a specialized subcellular compartment.

    View details for DOI 10.1083/jcb.201506132

    View details for Web of Science ID 000373523200015

    View details for PubMedID 27002170

    View details for PubMedCentralID PMC4810300

  • Systems and structural biology approaches to elucidate new effectors in KRAS mutant tumors Broyde, J., Simpson, D., Wah, D. A., Giorgi, F. M., Petrey, D., Alvarez, M. J., Silkov, A., Lachmann, A., Hill, D. E., Vidal, M., Jackson, P., Honig, B., Sweet-Cordero, A., Califano, A. AMER ASSOC CANCER RESEARCH. 2015
  • Tctex1d2 associates with short-rib polydactyly syndrome proteins and is required for ciliogenesis CELL CYCLE Gholkar, A. A., Senese, S., Lo, Y., Capri, J., Deardorff, W. J., Dharmarajan, H., Contreras, E., Hodara, E., Whitelegge, J. P., Jackson, P. K., Torres, J. Z. 2015; 14 (7): 1116-1125

    Abstract

    Short-rib polydactyly syndromes (SRPS) arise from mutations in genes involved in retrograde intraflagellar transport (IFT) and basal body homeostasis, which are critical for cilia assembly and function. Recently, mutations in WDR34 or WDR60 (candidate dynein intermediate chains) were identified in SRPS. We have identified and characterized Tctex1d2, which associates with Wdr34, Wdr60 and other dynein complex 1 and 2 subunits. Tctex1d2 and Wdr60 localize to the base of the cilium and their depletion causes defects in ciliogenesis. We propose that Tctex1d2 is a novel dynein light chain important for trafficking to the cilium and potentially retrograde IFT and is a new molecular link to understanding SRPS pathology.

    View details for DOI 10.4161/15384101.2014.985066

    View details for Web of Science ID 000352606600028

    View details for PubMedID 25830415

    View details for PubMedCentralID PMC4614626

  • Early steps in primary cilium assembly require EHD1/EHD3-dependent ciliary vesicle formation NATURE CELL BIOLOGY Lu, Q., Insinna, C., Ott, C., Stauffer, J., Pintado, P. A., Rahajeng, J., Baxa, U., Walia, V., Cuenca, A., Hwang, Y., Daar, I. O., Lopes, S., Lippincott-Schwartz, J., Jackson, P. K., Caplan, S., Westlake, C. J. 2015; 17 (3): 228-?

    Abstract

    Membrane association with mother centriole (M-centriole) distal appendages is critical for ciliogenesis initiation. How the Rab GTPase Rab11-Rab8 cascade functions in early ciliary membrane assembly is unknown. Here, we show that the membrane shaping proteins EHD1 and EHD3, in association with the Rab11-Rab8 cascade, function in early ciliogenesis. EHD1 and EHD3 localize to preciliary membranes and the ciliary pocket. EHD-dependent membrane tubulation is essential for ciliary vesicle formation from smaller distal appendage vesicles (DAVs). Importantly, this step functions in M-centriole to basal body transformation and recruitment of transition zone proteins and IFT20. SNAP29, a SNARE membrane fusion regulator and EHD1-binding protein, is also required for DAV-mediated ciliary vesicle assembly. Interestingly, only after ciliary vesicle assembly is Rab8 activated for ciliary growth. Our studies uncover molecular mechanisms informing a previously uncharacterized ciliogenesis step, whereby EHD1 and EHD3 reorganize the M-centriole and associated DAVs before coordinated ciliary membrane and axoneme growth.

    View details for DOI 10.1038/ncb3109

    View details for Web of Science ID 000350770200008

    View details for PubMedID 25686250

    View details for PubMedCentralID PMC4344897

  • 3D spheroid model of mIMCD3 cells for studying ciliopathies and renal epithelial disorders NATURE PROTOCOLS Giles, R. H., Ajzenberg, H., Jackson, P. K. 2014; 9 (12): 2725-2731

    Abstract

    We have developed a novel 3D cell culture model that uses mouse inner-medullary collecting duct (mIMCD3) cells to generate epithelial spheroids. This model is amenable to efficient siRNA knockdown and subsequent rescue with human patient-derived alleles. Spheroids develop apicobasal polarity and complete lumens, and they are consequently an ideal model for polarity defects seen in renal ciliopathies such as nephronophthisis. Briefly, mIMCD3 cells are transfected and subsequently passaged to a Matrigel mixture, which is seeded in chamber slides and covered in growth medium. Once the spheroids are formed, Matrigel is dissolved and immunocytochemistry is performed in the chamber slides. The technique is amenable to semiautomatic imaging analysis, and it can test multiple genes simultaneously, gene-dosing effects and a variety of therapeutic interventions. The spheroid technique is a unique and simple 6-d in vitro method of interrogating ex vivo tissue organization.

    View details for DOI 10.1038/nprot.2014.181

    View details for Web of Science ID 000344249300002

    View details for PubMedID 25356583

  • Regulating Microtubules and Genome Stability via the CUL7/3M Syndrome Complex and CUL9 MOLECULAR CELL Jackson, P. K. 2014; 54 (5): 713–15

    Abstract

    In this issue, two studies from Yue Xiong and colleagues (Li et al., 2014; Yan et al., 2014) show that CUL7, OBSL1, and CCDC8, all mutated in 3M short stature syndrome, form a centrosomal complex that regulates CUL9 and its substrate survivin to link mitosis to cell survival.

    View details for PubMedID 24905004

  • Chk1 inhibition in p53-deficient cell lines drives rapid chromosome fragmentation followed by caspase-independent cell death CELL CYCLE Del Nagro, C. J., Choi, J., Xiao, Y., Rangell, L., Mohan, S., Pandita, A., Zha, J., Jackson, P. K., O'Brien, T. 2014; 13 (2): 303-314

    Abstract

    Activation of Checkpoint kinase 1 (Chk1) following DNA damage mediates cell cycle arrest to prevent cells with damaged DNA from entering mitosis. Here we provide a high-resolution analysis of cells as they undergo S- and G₂-checkpoint bypass in response to Chk1 inhibition with the selective Chk1 inhibitor GNE-783. Within 4-8 h of Chk1 inhibition following gemcitabine induced DNA damage, cells with both sub-4N and 4N DNA content prematurely enter mitosis. Coincident with premature transition into mitosis, levels of DNA damage dramatically increase and chromosomes condense and attempt to align along the metaphase plate. Despite an attempt to congress at the metaphase plate, chromosomes rapidly fragment and lose connection to the spindle microtubules. Gemcitabine mediated DNA damage promotes the formation of Rad51 foci; however, while Chk1 inhibition does not disrupt Rad51 foci that are formed in response to gemcitabine, these foci are lost as cells progress into mitosis. Premature entry into mitosis requires the Aurora, Cdk1/2 and Plk1 kinases and even though caspase-2 and -3 are activated upon mitotic exit, they are not required for cell death. Interestingly, p53, but not p21, deficiency enables checkpoint bypass and chemo-potentiation. Finally, we uncover a differential role for the Wee-1 checkpoint kinase in response to DNA damage, as Wee-1, but not Chk1, plays a more prominent role in the maintenance of S- and G₂-checkpoints in p53 proficient cells.

    View details for DOI 10.4161/cc.27055

    View details for Web of Science ID 000336658200021

    View details for PubMedID 24247149

  • Neuropeptide Y Family Receptors Traffic via the Bardet-Biedl Syndrome Pathway to Signal in Neuronal Primary Cilia CELL REPORTS Loktev, A. V., Jackson, P. K. 2013; 5 (5): 1316-1329

    Abstract

    Human monogenic obesity syndromes, including Bardet-Biedl syndrome (BBS), implicate neuronal primary cilia in regulation of energy homeostasis. Cilia in hypothalamic neurons have been hypothesized to sense and regulate systemic energy status, but the molecular mechanism of this signaling remains unknown. Here, we report a comprehensive localization screen of 42 G-protein-coupled receptors (GPCR) revealing seven ciliary GPCRs, including the neuropeptide Y (NPY) receptors NPY2R and NPY5R. We show that mice modeling BBS disease or obese tubby mice fail to localize NPY2R to cilia in the hypothalamus and that BBS mutant mice fail to activate c-fos or decrease food intake in response to the NPY2R ligand PYY3-36. We find that cells with ciliary NPY2R show augmented PYY3-36-dependent cAMP signaling. Our data demonstrate that ciliary targeting of NPY receptors is important for controlling energy balance in mammals, revealing a physiologically defined ligand-receptor pathway signaling within neuronal cilia.

    View details for DOI 10.1016/j.celrep.2013.11.011

    View details for Web of Science ID 000328266400015

    View details for PubMedID 24316073

  • Supplementation of Nicotinic Acid with NAMPT Inhibitors Results in Loss of In Vivo Efficacy in NAPRT1-Deficient Tumor Models NEOPLASIA O'Brien, T., Oeh, J., Xiao, Y., Liang, X., Vanderbilt, A., Qin, A., Yang, L., Lee, L. B., Ly, J., Cosino, E., Lacap, J. A., Ogasawara, A., Williams, S., Nannini, M., Liederer, B. M., Jackson, P., Dragovich, P. S., Sampath, D. 2013; 15 (12): 1314-?

    Abstract

    Nicotinamide adenine dinucleotide (NAD) is a metabolite essential for cell survival and generated de novo from tryptophan or recycled from nicotinamide (NAM) through the nicotinamide phosphoribosyltransferase (NAMPT)-dependent salvage pathway. Alternatively, nicotinic acid (NA) is metabolized to NAD through the nicotinic acid phosphoribosyltransferase domain containing 1 (NAPRT1)-dependent salvage pathway. Tumor cells are more reliant on the NAMPT salvage pathway making this enzyme an attractive therapeutic target. Moreover, the therapeutic index of NAMPT inhibitors may be increased by in NAPRT-deficient tumors by NA supplementation as normal tissues may regenerate NAD through NAPRT1. To confirm the latter, we tested novel NAMPT inhibitors, GNE-617 and GNE-618, in cell culture- and patient-derived tumor models. While NA did not protect NAPRT1-deficient tumor cell lines from NAMPT inhibition in vitro, it rescued efficacy of GNE-617 and GNE-618 in cell culture- and patient-derived tumor xenografts in vivo. NA co-treatment increased NAD and NAM levels in NAPRT1-deficient tumors to levels that sustained growth in vivo. Furthermore, NAM co-administration with GNE-617 led to increased tumor NAD levels and rescued in vivo efficacy as well. Importantly, tumor xenografts remained NAPRT1-deficient in the presence of NA, indicating that the NAPRT1-dependent pathway is not reactivated. Protection of NAPRT1-deficient tumors in vivo may be due to increased circulating levels of metabolites generated by mouse liver, in response to NA or through competitive reactivation of NAMPT by NAM. Our results have important implications for the development of NAMPT inhibitors when considering NA co-treatment as a rescue strategy.

    View details for DOI 10.1593/neo.131718

    View details for Web of Science ID 000330128200001

    View details for PubMedID 24403854

  • Identification of Preferred Chemotherapeutics for Combining with a CHK1 Inhibitor MOLECULAR CANCER THERAPEUTICS Xiao, Y., Ramiscal, J., Kowanetz, K., Del Nagro, C., Malek, S., Evangelista, M., Blackwood, E., Jackson, P. K., O'Brien, T. 2013; 12 (11): 2285-2295

    Abstract

    Here we report that GNE-783, a novel checkpoint kinase-1 (CHK1) inhibitor, enhances the activity of gemcitabine by disabling the S- and G2 cell-cycle checkpoints following DNA damage. Using a focused library of 51 DNA-damaging agents, we undertook a systematic screen using three different cell lines to determine which chemotherapeutics have their activity enhanced when combined with GNE-783. We found that GNE-783 was most effective at enhancing activity of antimetabolite-based DNA-damaging agents; however, there was a surprisingly wide range of activity within each class of agents. We, next, selected six different therapeutic agents and screened these in combination with GNE-783 across a panel of cell lines. This revealed a preference for enhanced chemopotentiation of select agents within tumor types, as, for instance, GNE-783 preferentially enhanced the activity of temozolomide only in melanoma cell lines. Additionally, although p53 mutant status was important for the overall response to combinations with some agents; our data indicate that this alone was insufficient to predict synergy. We finally compared the ability of a structurally related CHK1 inhibitor, GNE-900, to enhance the in vivo activity of gemcitabine, CPT-11, and temozolomide in xenograft models. GNE-900 significantly enhanced activity of only gemcitabine in vivo, suggesting that strong chemopotentiation in vitro can translate into chemopotentiation in vivo. In conclusion, our results show that selection of an appropriate agent to combine with a CHK1 inhibitor needs to be carefully evaluated in the context of the genetic background and tumor type in which it will be used.

    View details for DOI 10.1158/1535-7163.MCT-13-0404

    View details for Web of Science ID 000326886000001

    View details for PubMedID 24038068

  • Dependence of Tumor Cell Lines and Patient-Derived Tumors on the NAD Salvage Pathway Renders Them Sensitive to NAMPT Inhibition with GNE-618 NEOPLASIA Xiao, Y., Elkins, K., Durieux, J. K., Lee, L., Oeh, J., Yang, L. X., Liang, X., Delnagro, C., Tremayne, J., Kwong, M., Liederer, B. M., Jackson, P. K., Belmont, L. D., Sampath, D., O'Brien, T. 2013; 15 (10): 1137-1146

    View details for DOI 10.1593/neo.131304

    View details for Web of Science ID 000328136300003

  • Combination Drug Scheduling Defines a "Window of Opportunity" for Chemopotentiation of Gemcitabine by an Orally Bioavailable, Selective ChK1 Inhibitor, GNE-900 MOLECULAR CANCER THERAPEUTICS Blackwood, E., Epler, J., Yen, I., Flagella, M., O'Brien, T., Evangelista, M., Schmidt, S., Xiao, Y., Choi, J., Kowanetz, K., Ramiscal, J., Wong, K., Jakubiak, D., Yee, S., Cain, G., Gazzard, L., Williams, K., Halladay, J., Jackson, P. K., Malek, S. 2013; 12 (10): 1968-1980

    Abstract

    Checkpoint kinase 1 (ChK1) is a serine/threonine kinase that functions as a central mediator of the intra-S and G2-M cell-cycle checkpoints. Following DNA damage or replication stress, ChK1-mediated phosphorylation of downstream effectors delays cell-cycle progression so that the damaged genome can be repaired. As a therapeutic strategy, inhibition of ChK1 should potentiate the antitumor effect of chemotherapeutic agents by inactivating the postreplication checkpoint, causing premature entry into mitosis with damaged DNA resulting in mitotic catastrophe. Here, we describe the characterization of GNE-900, an ATP-competitive, selective, and orally bioavailable ChK1 inhibitor. In combination with chemotherapeutic agents, GNE-900 sustains ATR/ATM signaling, enhances DNA damage, and induces apoptotic cell death. The kinetics of checkpoint abrogation seems to be more rapid in p53-mutant cells, resulting in premature mitotic entry and/or accelerated cell death. Importantly, we show that GNE-900 has little single-agent activity in the absence of chemotherapy and does not grossly potentiate the cytotoxicity of gemcitabine in normal bone marrow cells. In vivo scheduling studies show that optimal administration of the ChK1 inhibitor requires a defined lag between gemcitabine and GNE-900 administration. On the refined combination treatment schedule, gemcitabine's antitumor activity against chemotolerant xenografts is significantly enhanced and dose-dependent exacerbation of DNA damage correlates with extent of tumor growth inhibition. In summary, we show that in vivo potentiation of gemcitabine activity is mechanism based, with optimal efficacy observed when S-phase arrest and release is followed by checkpoint abrogation with a ChK1 inhibitor.

    View details for DOI 10.1158/1535-7163.MCT-12-1218

    View details for Web of Science ID 000325548400005

    View details for PubMedID 23873850

  • Covalent and allosteric inhibitors of the ATPase VCP/p97 induce cancer cell death. Nature chemical biology Magnaghi, P., D'alessio, R., Valsasina, B., Avanzi, N., Rizzi, S., Asa, D., Gasparri, F., Cozzi, L., Cucchi, U., Orrenius, C., Polucci, P., Ballinari, D., Perrera, C., Leone, A., Cervi, G., Casale, E., Xiao, Y., Wong, C., Anderson, D. J., Galvani, A., Donati, D., O'Brien, T., Jackson, P. K., Isacchi, A. 2013; 9 (9): 548-556

    Abstract

    VCP (also known as p97 or Cdc48p in yeast) is an AAA(+) ATPase regulating endoplasmic reticulum-associated degradation. After high-throughput screening, we developed compounds that inhibit VCP via different mechanisms, including covalent modification of an active site cysteine and a new allosteric mechanism. Using photoaffinity labeling, structural analysis and mutagenesis, we mapped the binding site of allosteric inhibitors to a region spanning the D1 and D2 domains of adjacent protomers encompassing elements important for nucleotide-state sensing and ATP hydrolysis. These compounds induced an increased affinity for nucleotides. Interference with nucleotide turnover in individual subunits and distortion of interprotomer communication cooperated to impair VCP enzymatic activity. Chemical expansion of this allosteric class identified NMS-873, the most potent and specific VCP inhibitor described to date, which activated the unfolded protein response, interfered with autophagy and induced cancer cell death. The consistent pattern of cancer cell killing by covalent and allosteric inhibitors provided critical validation of VCP as a cancer target.

    View details for DOI 10.1038/nchembio.1313

    View details for PubMedID 23892893

  • Nek8 Couples Renal Ciliopathies to DNA Damage and Checkpoint Control MOLECULAR CELL Jackson, P. K. 2013; 51 (4): 407–8

    Abstract

    In this issue, Choi et al. (2013) discover a molecular link between the Nek8 kinase, mutated in the renal ciliopathy nephronophthisis, and DNA damage control by cyclin A/Cdk2 and ATR-Chk1, providing new ideas for targeted therapies limiting tissue degeneration.

    View details for PubMedID 23973371

  • The Ciliary G-Protein-Coupled Receptor Gpr161 Negatively Regulates the Sonic Hedgehog Pathway via cAMP Signaling CELL Mukhopadhyay, S., Wen, X., Ratti, N., Loktev, A., Rangell, L., Scales, S. J., Jackson, P. K. 2013; 152 (1-2): 210-223

    Abstract

    The primary cilium is required for Sonic hedgehog (Shh) signaling in vertebrates. In contrast to mutants affecting ciliary assembly, mutations in the intraflagellar transport complex A (IFT-A) paradoxically cause increased Shh signaling. We previously showed that the IFT-A complex, in addition to its canonical role in retrograde IFT, binds to the tubby-like protein, Tulp3, and recruits it to cilia. Here, we describe a conserved vertebrate G-protein-coupled receptor, Gpr161, which localizes to primary cilia in a Tulp3/IFT-A-dependent manner. Complete loss of Gpr161 in mouse causes midgestation lethality and increased Shh signaling in the neural tube, phenocopying Tulp3/IFT-A mutants. Constitutive Gpr161 activity increases cAMP levels and represses Shh signaling by determining the processing of Gli3 to its repressor form. Conversely, Shh signaling directs Gpr161 to be internalized from cilia, preventing its activity. Thus, Gpr161 defines a morphogenetic pathway coupling protein kinase A activation to Shh signaling during neural tube development.

    View details for DOI 10.1016/j.cell.2012.12.026

    View details for Web of Science ID 000313719800018

    View details for PubMedID 23332756

  • Our thanks to Cilia's reviewers. Cilia Beales, P., Jackson, P. K. 2013; 2 (1): 4-?

    Abstract

    CONTRIBUTING REVIEWERS: We would like to thank all our reviewers for their contribution to the success of Cilia. Their voluntary participation has led to the launch of a new journal consistently publishing high quality articles and an intelligent peer review process for all our authors.

    View details for DOI 10.1186/2046-2530-2-4

    View details for PubMedID 23497485

  • TTBK2 Kinase: Linking Primary Cilia and Cerebellar Ataxias CELL Jackson, P. K. 2012; 151 (4): 697-699

    Abstract

    Mutations disrupting primary cilia cause retinal, renal, and cerebellar defects, and misregulated Sonic hedgehog signaling. A new mouse mutant in the TTBK2 kinase fails to make cilia, and shows neural tube and Sonic hedgehog signaling defects. Ciliary targeting mutations in human TTBK2 are linked to spinocerebellar ataxia, suggesting cilia protect from neurodegeneration.

    View details for DOI 10.1016/j.cell.2012.10.027

    View details for Web of Science ID 000310921200002

    View details for PubMedID 23141531

  • Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Maurer, T., Garrenton, L. S., Oha, A., Pitts, K., Anderson, D. J., Skelton, N. J., Fauber, B. P., Pan, B., Malek, S., Stokoe, D., Ludlam, M. J., Bowman, K. K., Wu, J., Giannetti, A. M., Starovasnik, M. A., Mellman, I., Jackson, P. K., Rudolph, J., Wang, W., Fang, G. 2012; 109 (14): 5299-5304

    Abstract

    The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.

    View details for DOI 10.1073/pnas.1116510109

    View details for Web of Science ID 000302294700043

    View details for PubMedID 22431598

  • Cilia develop long-lasting contacts, with other cilia. Cilia Jackson, P. K. 2012; 1 (1): 5-?

    View details for DOI 10.1186/2046-2530-1-5

    View details for PubMedID 23351843

  • Cilia - the prodigal organelle. Cilia Beales, P., Jackson, P. K. 2012; 1 (1): 1-?

    View details for DOI 10.1186/2046-2530-1-1

    View details for PubMedID 23351984

  • The STARD9/Kif16a Kinesin Associates with Mitotic Microtubules and Regulates Spindle Pole Assembly CELL Torres, J. Z., Summers, M. K., Peterson, D., Brauer, M. J., Lee, J., Senese, S., Gholkar, A. A., Lo, Y., Lei, X., Jung, K., Anderson, D. C., Davis, D. P., Belmont, L., Jackson, P. K. 2011; 147 (6): 1309-1323

    Abstract

    During cell division, cells form the microtubule-based mitotic spindle, a highly specialized and dynamic structure that mediates proper chromosome transmission to daughter cells. Cancer cells can show perturbed mitotic spindles and an approach in cancer treatment has been to trigger cell killing by targeting microtubule dynamics or spindle assembly. To identify and characterize proteins necessary for spindle assembly, and potential antimitotic targets, we performed a proteomic and genetic analysis of 592 mitotic microtubule copurifying proteins (MMCPs). Screening for regulators that affect both mitosis and apoptosis, we report the identification and characterization of STARD9, a kinesin-3 family member, which localizes to centrosomes and stabilizes the pericentriolar material (PCM). STARD9-depleted cells have fragmented PCM, form multipolar spindles, activate the spindle assembly checkpoint (SAC), arrest in mitosis, and undergo apoptosis. Interestingly, STARD9-depletion synergizes with the chemotherapeutic agent taxol to increase mitotic death, demonstrating that STARD9 is a mitotic kinesin and a potential antimitotic target.

    View details for DOI 10.1016/j.cell.2011.11.020

    View details for PubMedID 22153075

  • An ARL3-UNC119-RP2 GTPase cycle targets myristoylated NPHP3 to the primary cilium GENES & DEVELOPMENT Wright, K. J., Baye, L. M., Olivier-Mason, A., Mukhopadhyay, S., Sang, L., Kwong, M., Wang, W., Pretorius, P. R., Sheffield, V. C., Sengupta, P., Slusarski, D. C., Jackson, P. K. 2011; 25 (22): 2347-2360

    Abstract

    The membrane of the primary cilium is a highly specialized compartment that organizes proteins to achieve spatially ordered signaling. Disrupting ciliary organization leads to diseases called ciliopathies, with phenotypes ranging from retinal degeneration and cystic kidneys to neural tube defects. How proteins are selectively transported to and organized in the primary cilium remains unclear. Using a proteomic approach, we identified the ARL3 effector UNC119 as a binding partner of the myristoylated ciliopathy protein nephrocystin-3 (NPHP3). We mapped UNC119 binding to the N-terminal 200 residues of NPHP3 and found the interaction requires myristoylation. Creating directed mutants predicted from a structural model of the UNC119-myristate complex, we identified highly conserved phenylalanines within a hydrophobic β sandwich to be essential for myristate binding. Furthermore, we found that binding of ARL3-GTP serves to release myristoylated cargo from UNC119. Finally, we showed that ARL3, UNC119b (but not UNC119a), and the ARL3 GAP Retinitis Pigmentosa 2 (RP2) are required for NPHP3 ciliary targeting and that targeting requires UNC119b myristoyl-binding activity. Our results uncover a selective, membrane targeting GTPase cycle that delivers myristoylated proteins to the ciliary membrane and suggest that other myristoylated proteins may be similarly targeted to specialized membrane domains.

    View details for DOI 10.1101/gad.173443.111

    View details for Web of Science ID 000297154700004

    View details for PubMedID 22085962

  • Live-Cell Microscopy Reveals Small Molecule Inhibitor Effects on MAPK Pathway Dynamics PLOS ONE Anderson, D. J., Durieux, J. K., Song, K., Alvarado, R., Jackson, P. K., Hatzivassiliou, G., Ludlam, M. J. 2011; 6 (8)

    Abstract

    Oncogenic mutations in the mitogen activated protein kinase (MAPK) pathway are prevalent in human tumors, making this pathway a target of drug development efforts. Recently, ATP-competitive Raf inhibitors were shown to cause MAPK pathway activation via Raf kinase priming in wild-type BRaf cells and tumors, highlighting the need for a thorough understanding of signaling in the context of small molecule kinase inhibitors. Here, we present critical improvements in cell-line engineering and image analysis coupled with automated image acquisition that allow for the simultaneous identification of cellular localization of multiple MAPK pathway components (KRas, CRaf, Mek1 and Erk2). We use these assays in a systematic study of the effect of small molecule inhibitors across the MAPK cascade either as single agents or in combination. Both Raf inhibitor priming as well as the release from negative feedback induced by Mek and Erk inhibitors cause translocation of CRaf to the plasma membrane via mechanisms that are additive in pathway activation. Analysis of Erk activation and sub-cellular localization upon inhibitor treatments reveals differential inhibition and activation with the Raf inhibitors AZD628 and GDC0879 respectively. Since both single agent and combination studies of Raf and Mek inhibitors are currently in the clinic, our assays provide valuable insight into their effects on MAPK signaling in live cells.

    View details for DOI 10.1371/journal.pone.0022607

    View details for Web of Science ID 000293561200025

    View details for PubMedID 21829637

  • Deubiquitinase USP37 Is Activated by CDK2 to Antagonize APC(CDH1) and Promote S Phase Entry MOLECULAR CELL Huang, X., Summers, M. K., Pham, V., Lill, J. R., Liu, J., Lee, G., Kirkpatrick, D. S., Jackson, P. K., Fang, G., Dixit, V. M. 2011; 42 (4): 511-523

    Abstract

    Cell cycle progression requires the E3 ubiquitin ligase anaphase-promoting complex (APC/C), which uses the substrate adaptors CDC20 and CDH1 to target proteins for proteasomal degradation. The APC(CDH1) substrate cyclin A is critical for the G1/S transition and, paradoxically, accumulates even when APC(CDH1) is active. We show that the deubiquitinase USP37 binds CDH1 and removes degradative polyubiquitin from cyclin A. USP37 was induced by E2F transcription factors in G1, peaked at G1/S, and was degraded in late mitosis. Phosphorylation of USP37 by CDK2 stimulated its full activity. USP37 overexpression caused premature cyclin A accumulation in G1 and accelerated S phase entry, whereas USP37 knockdown delayed these events. USP37 was inactive in mitosis because it was no longer phosphorylated by CDK2. Indeed, it switched from an antagonist to a substrate of APC(CDH1) and was modified with degradative K11-linked polyubiquitin.

    View details for DOI 10.1016/j.molcel.2011.03.027

    View details for Web of Science ID 000291113800013

    View details for PubMedID 21596315

  • Mapping the NPHP-JBTS-MKS Protein Network Reveals Ciliopathy Disease Genes and Pathways CELL Sang, L., Miller, J. J., Corbit, K. C., Giles, R. H., Brauer, M. J., Otto, E. A., Baye, L. M., Wen, X., Scales, S. J., Kwong, M., Huntzicker, E. G., Sfakianos, M. K., Sandoval, W., Bazan, J. F., Kulkarni, P., Garcia-Gonzalo, F. R., Seol, A. D., O'Toole, J. F., Held, S., Reutter, H. M., Lane, W. S., Rafiq, M. A., Noor, A., Ansar, M., Devi, A. R., Sheffield, V. C., Slusarski, D. C., Vincent, J. B., Doherty, D. A., Hildebrandt, F., Reiter, J. F., Jackson, P. K. 2011; 145 (4): 513-528

    Abstract

    Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.

    View details for DOI 10.1016/j.cell.2011.04.019

    View details for Web of Science ID 000290560800006

    View details for PubMedID 21565611

    View details for PubMedCentralID PMC3383065

  • Do cilia put brakes on the cell cycle? NATURE CELL BIOLOGY Jackson, P. K. 2011; 13 (4): 340-342

    Abstract

    Two papers in this issue show that dynein-binding proteins may regulate the G1-S transition through an effect on cilia. Nde1, a known partner of dynein light chain LC8, controls ciliary length in vitro and in zebrafish, and influences the G1-S progression. The phosphorylation of Tctex1, a dynein light chain, modulates cilia length and accelerates G1-S, thereby regulating proliferation-differentiation decisions in the developing mouse neocortex.

    View details for DOI 10.1038/ncb0411-340

    View details for Web of Science ID 000289070900003

    View details for PubMedID 21460803

  • Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7 NATURE Wertz, I. E., Kusam, S., Lam, C., Okamoto, T., Sandoval, W., Anderson, D. J., Helgason, E., Ernst, J. A., Eby, M., Liu, J., Belmont, L. D., Kaminker, J. S., O'Rourke, K. M., Pujara, K., Kohli, P. B., Johnson, A. R., Chiu, M. L., Lill, J. R., Jackson, P. K., Fairbrother, W. J., Seshagiri, S., Ludlam, M. J., Leong, K. G., Dueber, E. C., Maecker, H., Huang, D. C., Dixit, V. M. 2011; 471 (7336): 110-114

    Abstract

    Microtubules have pivotal roles in fundamental cellular processes and are targets of antitubulin chemotherapeutics. Microtubule-targeted agents such as Taxol and vincristine are prescribed widely for various malignancies, including ovarian and breast adenocarcinomas, non-small-cell lung cancer, leukaemias and lymphomas. These agents arrest cells in mitosis and subsequently induce cell death through poorly defined mechanisms. The strategies that resistant tumour cells use to evade death induced by antitubulin agents are also unclear. Here we show that the pro-survival protein MCL1 (ref. 3) is a crucial regulator of apoptosis triggered by antitubulin chemotherapeutics. During mitotic arrest, MCL1 protein levels decline markedly, through a post-translational mechanism, potentiating cell death. Phosphorylation of MCL1 directs its interaction with the tumour-suppressor protein FBW7, which is the substrate-binding component of a ubiquitin ligase complex. The polyubiquitylation of MCL1 then targets it for proteasomal degradation. The degradation of MCL1 was blocked in patient-derived tumour cells that lacked FBW7 or had loss-of-function mutations in FBW7, conferring resistance to antitubulin agents and promoting chemotherapeutic-induced polyploidy. Additionally, primary tumour samples were enriched for FBW7 inactivation and elevated MCL1 levels, underscoring the prominent roles of these proteins in oncogenesis. Our findings suggest that profiling the FBW7 and MCL1 status of tumours, in terms of protein levels, messenger RNA levels and genetic status, could be useful to predict the response of patients to antitubulin chemotherapeutics.

    View details for DOI 10.1038/nature09779

    View details for Web of Science ID 000287924100044

    View details for PubMedID 21368834

  • A novel acetylation of beta-tubulin by San modulates microtubule polymerization via down-regulating tubulin incorporation MOLECULAR BIOLOGY OF THE CELL Chu, C., Hou, F., Zhang, J., Phu, L., Loktev, A. V., Kirkpatrick, D. S., Jackson, P. K., Zhao, Y., Zou, H. 2011; 22 (4): 448-456

    Abstract

    Dynamic instability is a critical property of microtubules (MTs). By regulating the rate of tubulin polymerization and depolymerization, cells organize the MT cytoskeleton to accommodate their specific functions. Among many processes, posttranslational modifications of tubulin are implicated in regulating MT functions. Here we report a novel tubulin acetylation catalyzed by acetyltransferase San at lysine 252 (K252) of β-tubulin. This acetylation, which is also detected in vivo, is added to soluble tubulin heterodimers but not tubulins in MTs. The acetylation-mimicking K252A/Q mutants were incorporated into the MT cytoskeleton in HeLa cells without causing any obvious MT defect. However, after cold-induced catastrophe, MT regrowth is accelerated in San-siRNA cells while the incorporation of acetylation-mimicking mutant tubulins is severely impeded. K252 of β-tubulin localizes at the interface of α-/β-tubulins and interacts with the phosphate group of the α-tubulin-bound GTP. We propose that the acetylation slows down tubulin incorporation into MTs by neutralizing the positive charge on K252 and allowing tubulin heterodimers to adopt a conformation that disfavors tubulin incorporation.

    View details for DOI 10.1091/mbc.E10-03-0203

    View details for Web of Science ID 000287305700002

    View details for PubMedID 21177827

  • Primary cilia membrane assembly is initiated by Rab11 and transport protein particle II (TRAPPII) complex-dependent trafficking of Rabin8 to the centrosome PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Westlake, C. J., Baye, L. M., Nachury, M. V., Wright, K. J., Ervin, K. E., Phu, L., Chalouni, C., Beck, J. S., Kirkpatrick, D. S., Slusarski, D. C., Sheffield, V. C., Scheller, R. H., Jackson, P. K. 2011; 108 (7): 2759-2764

    Abstract

    Sensory and signaling pathways are exquisitely organized in primary cilia. Bardet-Biedl syndrome (BBS) patients have compromised cilia and signaling. BBS proteins form the BBSome, which binds Rabin8, a guanine nucleotide exchange factor (GEF) activating the Rab8 GTPase, required for ciliary assembly. We now describe serum-regulated upstream vesicular transport events leading to centrosomal Rab8 activation and ciliary membrane formation. Using live microscopy imaging, we show that upon serum withdrawal Rab8 is observed to assemble the ciliary membrane in ∼100 min. Rab8-dependent ciliary assembly is initiated by the relocalization of Rabin8 to Rab11-positive vesicles that are transported to the centrosome. After ciliogenesis, Rab8 ciliary transport is strongly reduced, and this reduction appears to be associated with decreased Rabin8 centrosomal accumulation. Rab11-GTP associates with the Rabin8 COOH-terminal region and is required for Rabin8 preciliary membrane trafficking to the centrosome and for ciliogenesis. Using zebrafish as a model organism, we show that Rabin8 and Rab11 are associated with the BBS pathway. Finally, using tandem affinity purification and mass spectrometry, we determined that the transport protein particle (TRAPP) II complex associates with the Rabin8 NH(2)-terminal domain and show that TRAPP II subunits colocalize with centrosomal Rabin8 and are required for Rabin8 preciliary targeting and ciliogenesis.

    View details for DOI 10.1073/pnas.1018823108

    View details for Web of Science ID 000287377000029

    View details for PubMedID 21273506

    View details for PubMedCentralID PMC3041065

  • The tubby family proteins GENOME BIOLOGY Mukhopadhyay, S., Jackson, P. K. 2011; 12 (6)

    Abstract

    The tubby mouse shows a tripartite syndrome characterized by maturity-onset obesity, blindness and deafness. The causative gene Tub is the founding member of a family of related proteins present throughout the animal and plant kingdoms, each characterized by a signature carboxy-terminal tubby domain. This domain consists of a β barrel enclosing a central α helix and binds selectively to specific membrane phosphoinositides. The vertebrate family of tubby-like proteins (TULPs) includes the founding member TUB and the related TULPs, TULP1 to TULP4. Tulp1 is expressed in the retina and mutations in TULP1 cause retinitis pigmentosa in humans; Tulp3 is expressed ubiquitously in the mouse embryo and is important in sonic hedgehog (Shh)-mediated dorso-ventral patterning of the spinal cord. The amino terminus of these proteins is diverse and directs distinct functions. In the best-characterized example, the TULP3 amino terminus binds to the IFT-A complex, a complex important in intraflagellar transport in the primary cilia, through a short conserved domain. Thus, the tubby family proteins seem to serve as bipartite bridges through their phosphoinositide-binding tubby and unique amino-terminal functional domains, coordinating multiple signaling pathways, including ciliary G-protein-coupled receptor trafficking and Shh signaling. Molecular studies on this functionally diverse protein family are beginning to provide us with remarkable insights into the tubby-mouse syndrome and other related diseases.

    View details for DOI 10.1186/gb-2011-12-6-225

    View details for Web of Science ID 000296646600015

    View details for PubMedID 21722349

  • TULP3 bridges the IFT-A complex and membrane phosphoinositides to promote trafficking of G protein-coupled receptors into primary cilia GENES & DEVELOPMENT Mukhopadhyay, S., Wen, X., Chih, B., Nelson, C. D., Lane, W. S., Scales, S. J., Jackson, P. K. 2010; 24 (19): 2180-2193

    Abstract

    Primary cilia function as a sensory signaling compartment in processes ranging from mammalian Hedgehog signaling to neuronal control of obesity. Intraflagellar transport (IFT) is an ancient, conserved mechanism required to assemble cilia and for trafficking within cilia. The link between IFT, sensory signaling, and obesity is not clearly defined, but some novel monogenic obesity disorders may be linked to ciliary defects. The tubby mouse, which presents with adult-onset obesity, arises from mutation in the Tub gene. The tubby-like proteins comprise a related family of poorly understood proteins with roles in neural development and function. We find that specific Tubby family proteins, notably Tubby-like protein 3 (TULP3), bind to the IFT-A complex. IFT-A is linked to retrograde ciliary transport, but, surprisingly, we find that the IFT-A complex has a second role directing ciliary entry of TULP3. TULP3 and IFT-A, in turn, promote trafficking of a subset of G protein-coupled receptors (GPCRs), but not Smoothened, to cilia. Both IFT-A and membrane phosphoinositide-binding properties of TULP3 are required for ciliary GPCR localization. TULP3 and IFT-A proteins both negatively regulate Hedgehog signaling in the mouse embryo, and the TULP3-IFT-A interaction suggests how these proteins cooperate during neural tube patterning.

    View details for DOI 10.1101/gad.1966210

    View details for Web of Science ID 000282376200008

    View details for PubMedID 20889716

  • Candidate exome capture identifies mutation of SDCCAG8 as the cause of a retinal-renal ciliopathy NATURE GENETICS Otto, E. A., Hurd, T. W., Airik, R., Chaki, M., Zhou, W., Stoetzel, C., Patil, S. B., Levy, S., Ghosh, A. K., Murga-Zamalloa, C. A., van Reeuwijk, J., Letteboer, S. J., Sang, L., Giles, R. H., Liu, Q., Coene, K. L., Estrada-Cuzcano, A., Collin, R. W., McLaughlin, H. M., Held, S., Kasanuki, J. M., Ramaswami, G., Conte, J., Lopez, I., Washburn, J., MacDonald, J., Hu, J., Yamashita, Y., Maher, E. R., Guay-Woodford, L. M., Neumann, H. P., Obermueller, N., Koenekoop, R. K., Bergmann, C., Bei, X., Lewis, R. A., Katsanis, N., Lopes, V., Williams, D. S., Lyons, R. H., Dang, C. V., Brito, D. A., Dias, M. B., Zhang, X., Cavalcoli, J. D., Nuernberg, G., Nuernberg, P., Pierce, E. A., Jackson, P. K., Antignac, C., Saunier, S., Roepman, R., Dollfus, H., Khanna, H., Hildebrandt, F. 2010; 42 (10): 840-?

    Abstract

    Nephronophthisis-related ciliopathies (NPHP-RC) are recessive disorders that feature dysplasia or degeneration occurring preferentially in the kidney, retina and cerebellum. Here we combined homozygosity mapping with candidate gene analysis by performing 'ciliopathy candidate exome capture' followed by massively parallel sequencing. We identified 12 different truncating mutations of SDCCAG8 (serologically defined colon cancer antigen 8, also known as CCCAP) in 10 families affected by NPHP-RC. We show that SDCCAG8 is localized at both centrioles and interacts directly with OFD1 (oral-facial-digital syndrome 1), which is associated with NPHP-RC. Depletion of sdccag8 causes kidney cysts and a body axis defect in zebrafish and induces cell polarity defects in three-dimensional renal cell cultures. This work identifies loss of SDCCAG8 function as a cause of a retinal-renal ciliopathy and validates exome capture analysis for broadly heterogeneous single-gene disorders.

    View details for DOI 10.1038/ng.662

    View details for Web of Science ID 000282276600013

    View details for PubMedID 20835237

  • APC/C-Cdc20 targets E2F1 for degradation in prometaphase CELL CYCLE Peart, M. J., Poyurovsky, M. V., Kass, E. M., Urist, M., Verschuren, E. W., Summers, M. K., Jackson, P. K., Prives, C. 2010; 9 (19): 3956-3964

    Abstract

    The mechanisms that control E2F-1 activity are complex. We previously showed that Chk1 and Chk2 are required for E2F1 stabilization and p73 target gene induction following DNA damage. To gain further insight into the processes regulating E2F1 protein stability, we focused our investigation on the mechanisms responsible for regulating E2F1 turnover. Here we show that E2F1 is a substrate of the anaphase promoting complex or cyclosome (APC/C), a ubiquitin ligase that plays an important role in cell cycle progression. Ectopic expression of the APC/C activators Cdh1 and Cdc20 reduced the levels of co-expressed E2F-1 protein. Co-expression of DP1 with E2F1 blocked APC/C-induced E2F1 degradation, suggesting that the E2F1/DP1 heterodimer is protected from APC/C regulation. Following Cdc20 knockdown, E2F1 levels increased and remained stable in extracts over a time course, indicating that APC/C(Cdc20) is a primary regulator of E2F1 stability in vivo. Moreover, cell synchronization experiments showed that siRNA directed against Cdc20 induced an accumulation of E2F1 protein in prometaphase cells. These data suggest that APC/C(Cdc20) specifically targets E2F1 for degradation in early mitosis and reveal a novel mechanism for limiting free E2F1 levels in cells, failure of which may compromise cell survival and/or homeostasis.

    View details for DOI 10.4161/cc.9.19.13162

    View details for Web of Science ID 000282703600026

    View details for PubMedID 20948288

  • A Chemosensitization Screen Identifies TP53RK, a Kinase that Restrains Apoptosis after Mitotic Stress CANCER RESEARCH Peterson, D., Lee, J., Lei, X. C., Forrest, W. F., Davis, D. P., Jackson, P. K., Belmont, L. D. 2010; 70 (15): 6325-6335

    Abstract

    Taxanes are very effective at causing mitotic arrest; however, there is variability among cancer cells in the apoptotic response to mitotic arrest. The variability in clinical efficacy of taxane-based therapy is likely a reflection of this variability in apoptotic response, thus elucidation of the molecular mechanism of the apoptotic response to mitotic stress could lead to improved clinical strategies. To identify genes whose expression influences the rate and extent of apoptosis after mitotic arrest, we screened a kinase-enriched small interfering RNA library for effects on caspase activation in response to maximally effective doses of paclitaxel, a PLK1 inhibitor, or cisplatin. Small interfering RNA oligonucleotides directed against an atypical protein kinase, TP53RK, caused the greatest increase in caspase-3/7 activation in response to antimitotic agents. Time-lapse microscopy revealed that cells entered mitosis with normal kinetics, but died after entry into mitosis in the presence of paclitaxel more rapidly when TP53RK was depleted. Because expression levels of TP53RK vary in cancers, TP53RK levels could provide a molecular marker to predict response to antimitotic agents. TP53RK inhibition may also sensitize cancers to taxanes.

    View details for DOI 10.1158/0008-5472.CAN-10-0015

    View details for Web of Science ID 000280557500023

    View details for PubMedID 20647325

  • A Specific Form of Phospho Protein Phosphatase 2 Regulates Anaphase-promoting Complex/Cyclosome Association with Spindle Poles MOLECULAR BIOLOGY OF THE CELL Torres, J. Z., Ban, K. H., Jackson, P. K. 2010; 21 (6): 897-904

    Abstract

    In early mitosis, the END (Emi1/NuMA/Dynein-dynactin) network anchors the anaphase-promoting complex/cyclosome (APC/C) to the mitotic spindle and poles. Spindle anchoring restricts APC/C activity, thereby limiting the destruction of spindle-associated cyclin B and ensuring maintenance of spindle integrity. Emi1 binds directly to hypophosphorylated APC/C, linking the APC/C to the spindle via NuMA. However, whether the phosphorylation state of the APC/C is important for its association with the spindle and what kinases and phosphatases are necessary for regulating this event remain unknown. Here, we describe the regulation of APC/C-mitotic spindle pole association by phosphorylation. We find that only hypophosphorylated APC/C associates with microtubule asters, suggesting that phosphatases are important. Indeed, a specific form of PPP2 (CA/R1A/R2B) binds APC/C, and PPP2 activity is necessary for Cdc27 dephosphorylation. Screening by RNA interference, we find that inactivation of CA, R1A, or R2B leads to delocalization of APC/C from spindle poles, early mitotic spindle defects, a failure to congress chromosomes, and decreased levels of cyclin B on the spindle. Consistently, inhibition of cyclin B/Cdk1 activity increased APC/C binding to microtubules. Thus, cyclin B/Cdk1 and PPP2 regulate the dynamic association of APC/C with spindle poles in early mitosis, a step necessary for proper spindle formation.

    View details for DOI 10.1091/mbc.E09-07-0598

    View details for Web of Science ID 000275433500005

    View details for PubMedID 20089842

    View details for PubMedCentralID PMC2836970

  • Individuals with mutations in XPNPEP3, which encodes a mitochondrial protein, develop a nephronophthisis-like nephropathy JOURNAL OF CLINICAL INVESTIGATION O'Toole, J. F., Liu, Y., Davis, E. E., Westlake, C. J., Attanasio, M., Otto, E. A., Seelow, D., Nurnberg, G., Becker, C., Nuutinen, M., Karppa, M., Ignatius, J., Uusimaa, J., Pakanen, S., Jaakkola, E., Van den Heuvel, L. P., Fehrenbach, H., Wiggins, R., Goyal, M., Zhou, W., Wolf, M. T., Wise, E., Helou, J., Allen, S. J., Murga-Zamalloa, C. A., Ashraf, S., Chaki, M., Heeringa, S., Chernin, G., Hoskins, B. E., Chaib, H., Gleeson, J., Kusakabe, T., Suzuki, T., Isaac, R. E., Quarmby, L. M., Tennant, B., Fujioka, H., Tuominen, H., Hassinen, I., Lohi, H., Van Houten, J. L., Rotig, A., Sayer, J. A., Rolinski, B., Freisinger, P., Madhavan, S. M., Herzer, M., Madignier, F., Prokisch, H., Nurnberg, P., Jackson, P., Khanna, H., Katsanis, N., Hildebrandt, F. 2010; 120 (3): 791-802

    Abstract

    The autosomal recessive kidney disease nephronophthisis (NPHP) constitutes the most frequent genetic cause of terminal renal failure in the first 3 decades of life. Ten causative genes (NPHP1-NPHP9 and NPHP11), whose products localize to the primary cilia-centrosome complex, support the unifying concept that cystic kidney diseases are "ciliopathies". Using genome-wide homozygosity mapping, we report here what we believe to be a new locus (NPHP-like 1 [NPHPL1]) for an NPHP-like nephropathy. In 2 families with an NPHP-like phenotype, we detected homozygous frameshift and splice-site mutations, respectively, in the X-prolyl aminopeptidase 3 (XPNPEP3) gene. In contrast to all known NPHP proteins, XPNPEP3 localizes to mitochondria of renal cells. However, in vivo analyses also revealed a likely cilia-related function; suppression of zebrafish xpnpep3 phenocopied the developmental phenotypes of ciliopathy morphants, and this effect was rescued by human XPNPEP3 that was devoid of a mitochondrial localization signal. Consistent with a role for XPNPEP3 in ciliary function, several ciliary cystogenic proteins were found to be XPNPEP3 substrates, for which resistance to N-terminal proline cleavage resulted in attenuated protein function in vivo in zebrafish. Our data highlight an emerging link between mitochondria and ciliary dysfunction, and suggest that further understanding the enzymatic activity and substrates of XPNPEP3 will illuminate novel cystogenic pathways.

    View details for DOI 10.1172/JCI40076

    View details for Web of Science ID 000275272700017

    View details for PubMedID 20179356

  • Navigating the Deubiquitinating Proteome with a CompPASS CELL Jackson, P. K. 2009; 138 (2): 222-224

    Abstract

    Deubiquitinating enzymes (DUBs) act on ubiquitinated substrates to regulate their modification and stability. In this issue, Sowa et al. (2009) present a comprehensive proteomic analysis of DUB interacting proteins in humans and a new quantitative scoring system for hits (CompPASS), providing a resource that links DUBs to biological pathways.

    View details for DOI 10.1016/j.cell.2009.07.008

    View details for Web of Science ID 000268277000003

    View details for PubMedID 19632171

  • High-throughput generation of tagged stable cell lines for proteomic analysis PROTEOMICS Torres, J. Z., Miller, J. J., Jackson, P. K. 2009; 9 (10): 2888-2891

    Abstract

    We present an optimized system for rapid generation of localization and affinity purification-tagged mammalian stable cell lines that facilitates complex purification and interacting protein identification. The improved components of this method, including the flexibility of inducible expression, circumvent issues associated with toxicity, clonal selection, sample yields and time to data acquisition. We have applied this method to the study of cell-cycle regulators and novel microtubule-associated proteins.

    View details for DOI 10.1002/pmic.200800873

    View details for Web of Science ID 000266707800024

    View details for PubMedID 19405035

    View details for PubMedCentralID PMC4785821

  • Biochemical analysis of the Anaphase Promoting Complex: activities of E2 enzymes and substrate competitive (pseudosubstrate) inhibitors. Methods in molecular biology (Clifton, N.J.) Summers, M. K., Jackson, P. K. 2009; 545: 313-330

    Abstract

    The Anaphase Promoting Complex (APC) ubiquitin ligase is critical for multiple processes including cell cycle, development, meiosis, and senescence. The importance of regulation of the APC by substrate competitive (pseudosubstrate) inhibitors, such as Emi1 and BubR1, has recently been demonstrated. Substrate competitive inhibitors typically bind to enzymes via the same site as substrates, but by having any combination of increased enzyme affinity and low turnover numbers, are able to "clog" the ability of the enzyme to bind and turnover substrates. For the APC, these pseudosubstrates can both position and block the APC and have been well validated as critical regulators for the APC enzymes.We have found that the substrate competitive mechanism of inhibition is sensitive to the E2 activity driving APC catalyzed ubiquitination events. This chapter provides detailed protocols for multiple in vitro ubiquitination assays of increasing complexity and the use of pseudosubstrate inhibitors in these assays. These assays are instrumental in examining the use of E2 enzymes by the APC and the intimate relationship this has with pseudosubstrate inhibition.

    View details for DOI 10.1007/978-1-60327-993-2_20

    View details for PubMedID 19475398

  • A BBSome Subunit Links Ciliogenesis, Microtubule Stability, and Acetylation DEVELOPMENTAL CELL Loktev, A. V., Zhang, Q., Beck, J. S., Searby, C. C., Scheetz, T. E., Bazan, J. F., Slusarski, D. C., Sheffield, V. C., Jackson, P. K., Nachury, M. V. 2008; 15 (6): 854-865

    Abstract

    Primary cilium dysfunction affects the development and homeostasis of many organs in Bardet-Biedl syndrome (BBS). We recently showed that seven highly conserved BBS proteins form a stable complex, the BBSome, that functions in membrane trafficking to and inside the primary cilium. We have now discovered a BBSome subunit that we named BBIP10. Similar to other BBSome subunits, BBIP10 localizes to the primary cilium, BBIP10 is present exclusively in ciliated organisms, and depletion of BBIP10 yields characteristic BBS phenotypes in zebrafish. Unexpectedly, BBIP10 is required for cytoplasmic microtubule polymerization and acetylation, two functions not shared with any other BBSome subunits. Strikingly, inhibition of the tubulin deacetylase HDAC6 restores microtubule acetylation in BBIP10-depleted cells, and BBIP10 physically interacts with HDAC6. BBSome-bound BBIP10 may therefore function to couple acetylation of axonemal microtubules and ciliary membrane growth.

    View details for DOI 10.1016/j.devcel.2008.11.001

    View details for Web of Science ID 000261631500009

    View details for PubMedID 19081074

  • New ligands for melanocortin receptors 20th Conference of the Pennington-Biomedical-Research-Center Kaelin, C. B., Candille, S. I., Yu, B., Jackson, P., Thompson, D. A., Nix, M. A., Binkley, J., Millhauser, G. L., Barsh, G. S. NATURE PUBLISHING GROUP. 2008: S19–S27

    Abstract

    Named originally for their effects on peripheral end organs, the melanocortin system controls a diverse set of physiological processes through a series of five G-protein-coupled receptors and several sets of small peptide ligands. The central melanocortin system plays an essential role in homeostatic regulation of body weight, in which two alternative ligands, alpha-melanocyte-stimulating hormone and agouti-related protein, stimulate and inhibit receptor signaling in several key brain regions that ultimately affect food intake and energy expenditure. Much of what we know about the relationship between central melanocortin signaling and body weight regulation stems from genetic studies. Comparative genomic studies indicate that melanocortin receptors used for controlling pigmentation and body weight regulation existed more than 500 million years ago in primitive vertebrates, but that fine-grained control of melanocortin receptors through neuropeptides and endogenous antagonists developed more recently. Recent studies based on dog coat-color genetics revealed a new class of melanocortin ligands, the beta-defensins, which reveal the potential for cross talk between the melanocortin and the immune systems.

    View details for DOI 10.1038/ijo.2008.234

    View details for Web of Science ID 000262376700005

    View details for PubMedID 19136986

  • The unique N terminus of the UbcH10 E2 enzyme controls the threshold for APC activation and enhances checkpoint regulation of the APC MOLECULAR CELL Summers, M. K., Pan, B., Mukhyala, K., Jackson, P. K. 2008; 31 (4): 544-556

    Abstract

    In vitro, the anaphase-promoting complex (APC) E3 ligase functions with E2 ubiquitin-conjugating enzymes of the E2-C and Ubc4/5 families to ubiquitinate substrates. However, only the use of the E2-C family, notably UbcH10, is genetically well validated. Here, we biochemically demonstrate preferential use of UbcH10 by the APC, specified by the E2 core domain. Importantly, an additional E2-E3 interaction mediated by the N-terminal extension of UbcH10 regulates APC activity. Mutating the highly conserved N terminus increases substrate ubiquitination and the number of substrate lysines targeted, allows ubiquitination of APC substrates lacking their destruction boxes, increases resistance to the APC inhibitors Emi1 and BubR1 in vitro, and bypasses the spindle checkpoint in vivo. Fusion of the UbcH10 N terminus to UbcH5 restricts ubiquitination activity but does not direct specific interactions with the APC. Thus, UbcH10 combines a specific E2-E3 interface and regulation via its N-terminal extension to limit APC activity for substrate selection and checkpoint control.

    View details for DOI 10.1016/j.molcel.2008.07.014

    View details for Web of Science ID 000258744100009

    View details for PubMedID 18722180

    View details for PubMedCentralID PMC2813190

  • Cdc2 and Mos regulate Emi2 stability to promote the meiosis I-meiosis II transition MOLECULAR BIOLOGY OF THE CELL Tang, W., Wu, J. Q., Guo, Y., Hansen, D. V., Perry, J. A., Freel, C. D., Nutt, L., Jackson, P. K., Kornbluth, S. 2008; 19 (8): 3536-3543

    Abstract

    The transition of oocytes from meiosis I (MI) to meiosis II (MII) requires partial cyclin B degradation to allow MI exit without S phase entry. Rapid reaccumulation of cyclin B allows direct progression into MII, producing a cytostatic factor (CSF)-arrested egg. It has been reported that dampened translation of the anaphase-promoting complex (APC) inhibitor Emi2 at MI allows partial APC activation and MI exit. We have detected active Emi2 translation at MI and show that Emi2 levels in MI are mainly controlled by regulated degradation. Emi2 degradation in MI depends not on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), but on Cdc2-mediated phosphorylation of multiple sites within Emi2. As in MII, this phosphorylation is antagonized by Mos-mediated recruitment of PP2A to Emi2. Higher Cdc2 kinase activity in MI than MII allows sufficient Emi2 phosphorylation to destabilize Emi2 in MI. At MI anaphase, APC-mediated degradation of cyclin B decreases Cdc2 activity, enabling Cdc2-mediated Emi2 phosphorylation to be successfully antagonized by Mos-mediated PP2A recruitment. These data suggest a model of APC autoinhibition mediated by stabilization of Emi2; Emi2 proteins accumulate at MI exit and inhibit APC activity sufficiently to prevent complete degradation of cyclin B, allowing MI exit while preventing interphase before MII entry.

    View details for DOI 10.1091/mbc.E08-04-0417

    View details for Web of Science ID 000259160400033

    View details for PubMedID 18550795

    View details for PubMedCentralID PMC2488281

  • The hunt for cyclin CELL Jackson, P. K. 2008; 134 (2): 199-202

    Abstract

    It is 25 years since Tim Hunt discovered cyclin, the oscillating protein that drives activation of cyclin-dependent kinases and entry into mitosis (Evans et al., 1983).

    View details for DOI 10.1016/j.cell.2008.07.011

    View details for Web of Science ID 000257891700006

    View details for PubMedID 18662532

  • Stopping replication, at the beginning NATURE CHEMICAL BIOLOGY Jackson, P. K. 2008; 4 (6): 331-332

    View details for DOI 10.1038/nchembio0608-331

    View details for Web of Science ID 000256068600004

    View details for PubMedID 18488006

  • The nucleolar phosphatase Cdc14B is dispensable for chromosome segregation and mitotic exit in human cells CELL CYCLE Berdougo, E., Nachury, M. V., Jackson, P. K., Jallepalli, P. V. 2008; 7 (9): 1184-1190

    Abstract

    In yeast, the protein phosphatase Cdc14 promotes chromosome segregation, mitotic exit, and cytokinesis by reversing M-phase phosphorylations catalyzed by Cdk1. A key feature of Cdc14 regulation is its sequestration within the nucleolus, which restricts its access to potential substrates for much of the cell cycle. Mammals also possess a nucleolar Cdc14 homolog, termed Cdc14B, but its roles during mitosis and cell division remain speculative. Here we analyze Cdc14B's subcellular dynamics during mitosis and rigorously test its functional contributions to cell division through homozygous disruption of the Cdc14B locus in human somatic cells. While Cdc14B is initially released from nucleoli at the start of mitosis, the phosphatase quickly redistributes onto segregating sister chromatids during anaphase. This relocalization is mainly driven by Cdk1 inactivation, as pharmacologic inhibition of Cdk1 in prometaphase cells redirects Cdc14B onto chromosomes. However, in sharp contrast to yeast cdc14 mutants, human Cdc14B(Delta/Delta) cells were viable and lacked defects in spindle assembly, anaphase progression, mitotic exit, and cytokinesis, and continued to segregate ribosomal DNA repeats with near-normal proficiency. Our findings reveal substantial divergence in mitotic regulation between yeast and mammalian cells, as the latter possess efficient mechanisms for completing late M-phase events in the absence of a nucleolar Cdc14-related phosphatase.

    View details for Web of Science ID 000256102700012

    View details for PubMedID 18418058

  • Emi1 protein accumulation implicates misregulation of the anaphase promoting complex/cyclosome pathway in ovarian clear cell carcinoma MODERN PATHOLOGY Guetgemann, I., Lehman, N. L., Jackson, P. K., Longacre, T. A. 2008; 21 (4): 445-454

    Abstract

    Clear cell carcinoma is a clinically and pathologically distinct entity among surface epithelial ovarian neoplasms, recognized for its resistance to standard platinum-based chemotherapy at advanced stage disease and poor prognosis. Despite advances in our understanding of the biology of other surface epithelial ovarian neoplasms, very little is known about the molecular genetic mechanisms that are involved in clear cell tumorigenesis. Early mitotic inhibitor-1 (Emi1) protein is a key cell cycle regulator, that promotes S-phase and mitotic entry by inhibiting the anaphase promoting complex. In cell culture systems, overexpression of Emi1 leads to tetraploidy and genomic instability, especially in the absence of normal p53 function. We investigated Emi1 protein expression in ovarian neoplasms using a tissue microarray constructed from 339 primary ovarian surface epithelial (serous, endometrioid, clear cell, and mucinous) and peritoneal (serous) neoplasms, stromal and mesenchymal tumors, germ cell tumors, and normal ovarian tissue. Significant overexpression of Emi1 protein was present in 82% (27/33) clear cell carcinoma, including one borderline tumor in a diffuse, granular cytoplasmic and perinuclear staining pattern, independent of patient age, presence of ovarian and/or pelvic endometriosis, and FIGO stage. In contrast, only 10% (17/177) primary ovarian and primary peritoneal serous carcinomas, 0% (0/10) mucinous carcinomas, and 19% (6/32) endometrioid carcinomas exhibited significant Emi1 protein overexpression. Accumulation of Emi1 protein was not linked to Ki-67 labeling index, but was directly correlated with cyclin E and inversely correlated with ER in clear cell carcinoma (P<0.001). Emi1 protein expression was present in mixed endometrioid/clear cell tumors but absent in tumors with mixed serous/clear cell histology. These findings represent a potentially important insight into the molecular pathway underlying ovarian carcinogenesis and provide a possible cell cycle model for the development and progression of ovarian clear cell carcinoma.

    View details for DOI 10.1038/modpathol.3801022

    View details for Web of Science ID 000254288500011

    View details for PubMedID 18204430

  • Cell division, growth and death CURRENT OPINION IN CELL BIOLOGY Jackson, P. K., Peters, J. 2007; 19 (6): 646–48
  • Loss of Emi1-dependent anaphase-promoting complex/cyclosome inhibition deregulates E2F target expression and elicits DNA damage-induced senescence MOLECULAR AND CELLULAR BIOLOGY Verschuren, E. W., Ban, K. H., Masek, M. A., Lehman, N. L., Jackson, P. K. 2007; 27 (22): 7955-7965

    Abstract

    Expression of the anaphase-promoting complex/cyclosome (APC/C) inhibitor Emi1 is required for the accumulation of APC/C substrates crucial for DNA synthesis and mitotic entry. We show that in vivo Emi1 expression correlates with the proliferative status of the cellular compartment and that cells lacking Emi1 undergo cellular senescence. Emi1 depletion leads to strong decreases in E2F target mRNA and APC/C substrate protein abundances. However, cyclin E mRNA and cyclin E protein levels and associated kinase activities are increased. Cells lacking Emi1 undergo DNA damage, likely explained by replication stress upon deregulated cyclin E- and A-associated kinase activities. Inhibition of ATM kinase prevents induction of senescence, implying that senescence is a consequence of DNA damage. Surprisingly, no senescence or no extensive amount of senescence is evident upon depletion of the Emi1-stabilizing factor Evi5 or Pin1, respectively. Our data suggest that maintenance of a protein stabilization/mRNA expression positive-feedback circuit fueled by Emi1 is required for accurate cell cycle progression, maintenance of DNA integrity, and prevention of cellular senescence.

    View details for DOI 10.1128/MCB.00908-07

    View details for Web of Science ID 000250800900019

    View details for PubMedID 17875940

    View details for PubMedCentralID PMC2169152

  • Control of Emi2 activity and stability through Mos-mediated recruitment of PP2A PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wu, J. Q., Hansen, D. V., Guo, Y., Wang, M. Z., Tang, W., Freel, C. D., Tung, J. J., Jackson, P. K., Kornbluth, S. 2007; 104 (42): 16564-16569

    Abstract

    Before fertilization, vertebrate eggs are arrested in meiosis II by cytostatic factor (CSF), which holds the anaphase-promoting complex (APC) in an inactive state. It was recently reported that Mos, an integral component of CSF, acts in part by promoting the Rsk-mediated phosphorylation of the APC inhibitor Emi2/Erp1. We report here that Rsk phosphorylation of Emi2 promotes its interaction with the protein phosphatase PP2A. Emi2 residues adjacent to the Rsk phosphorylation site were important for PP2A binding. An Emi2 mutant that retained Rsk phosphorylation but lacked PP2A binding could not be modulated by Mos. PP2A bound to Emi2 acted on two distinct clusters of sites phosphorylated by Cdc2, one responsible for modulating its stability during CSF arrest and one that controls binding to the APC. These findings provide a molecular mechanism for Mos action in promoting CSF arrest and also define an unusual mechanism, whereby protein phosphorylation recruits a phosphatase for dephosphorylation of distinct sites phosphorylated by another kinase.

    View details for DOI 10.1073/pnas.0707537104

    View details for Web of Science ID 000250373400033

    View details for PubMedID 17881560

    View details for PubMedCentralID PMC2034268

  • A bacterial effector targets Mad2L2, an APC inhibitor, to modulate host cell cycling CELL Iwai, H., Kim, M., Yoshikawa, Y., Ashida, H., Ogawa, M., Fujita, Y., Muller, D., Kirikae, T., Jackson, P. K., Kotani, S., Sasakawa, C. 2007; 130 (4): 611-623

    Abstract

    The gut epithelium self-renews every several days, providing an important innate defense system that limits bacterial colonization. Nevertheless, many bacterial pathogens, including Shigella, efficiently colonize the intestinal epithelium. Here, we show that the Shigella effector IpaB, when delivered into epithelial cells, causes cell-cycle arrest by targeting Mad2L2, an anaphase-promoting complex/cyclosome (APC) inhibitor. Cyclin B1 ubiquitination assays revealed that APC undergoes unscheduled activation due to IpaB interaction with the APC inhibitor Mad2L2. Synchronized HeLa cells infected with Shigella failed to accumulate Cyclin B1, Cdc20, and Plk1, causing cell-cycle arrest at the G2/M phase in an IpaB/Mad2L2-dependent manner. IpaB/Mad2L2-dependent cell-cycle arrest by Shigella infection was also demonstrated in rabbit intestinal crypt progenitors, and the IpaB-mediated arrest contributed to efficient colonization of the host cells. These results strongly indicate that Shigella employ special tactics to influence epithelial renewal in order to promote bacterial colonization of intestinal epithelium.

    View details for DOI 10.1016/j.cell.2007.06.043

    View details for Web of Science ID 000249319400013

    View details for PubMedID 17719540

  • Cyclin E overexpression impairs progression through mitosis by inhibiting APC(Cdh1) JOURNAL OF CELL BIOLOGY Keck, J. M., Summers, M. K., Tedesco, D., Ekholm-Reed, S., Chuang, L., Jackson, P. K., Reed, S. I. 2007; 178 (3): 371-385

    Abstract

    Overexpression of cyclin E, an activator of cyclin-dependent kinase 2, has been linked to human cancer. In cell culture models, the forced expression of cyclin E leads to aneuploidy and polyploidy, which is consistent with a direct role of cyclin E overexpression in tumorigenesis. In this study, we show that the overexpression of cyclin E has a direct effect on progression through the latter stages of mitotic prometaphase before the complete alignment of chromosomes at the metaphase plate. In some cases, such cells fail to divide chromosomes, resulting in polyploidy. In others, cells proceed to anaphase without the complete alignment of chromosomes. These phenotypes can be explained by an ability of overexpressed cyclin E to inhibit residual anaphase-promoting complex (APC(Cdh1)) activity that persists as cells progress up to and through the early stages of mitosis, resulting in the abnormal accumulation of APC(Cdh1) substrates as cells enter mitosis. We further show that the accumulation of securin and cyclin B1 can account for the cyclin E-mediated mitotic phenotype.

    View details for DOI 10.1083/jcb.200703202

    View details for Web of Science ID 000248544500007

    View details for PubMedID 17664332

    View details for PubMedCentralID PMC2064850

  • The END network couples spindle pole assembly to inhibition of the anaphase-promoting Complex/Cyclosome in early mitosis DEVELOPMENTAL CELL Ban, K. H., Torres, J. Z., Miller, J. J., Mikhailov, A., Nachury, M. V., Tung, J. J., Rieder, C. L., Jackson, P. K. 2007; 13 (1): 29-42

    Abstract

    Cyclin-dependent kinase 1 (Cdk1) initiates mitosis and later activates the anaphase-promoting complex/cyclosome (APC/C) to destroy cyclins. Kinetochore-derived checkpoint signaling delays APC/C-dependent cyclin B destruction, and checkpoint-independent mechanisms cooperate to limit APC/C activity when kinetochores lack checkpoint components in early mitosis. The APC/C and cyclin B localize to the spindle and poles, but the significance and regulation of these populations remain unclear. Here we describe a critical spindle pole-associated mechanism, called the END (Emi1/NuMA/dynein-dynactin) network, that spatially restricts APC/C activity in early mitosis. The APC/C inhibitor Emi1 binds the spindle-organizing NuMA/dynein-dynactin complex to anchor and inhibit the APC/C at spindle poles, and thereby limits destruction of spindle-associated cyclin B. Cyclin B/Cdk1 activity recruits the END network and establishes a positive feedback loop to stabilize spindle-associated cyclin B critical for spindle assembly. The organization of the APC/C on the spindle also provides a framework for understanding microtubule-dependent organization of protein destruction.

    View details for DOI 10.1016/j.devcel.2007.04.017

    View details for Web of Science ID 000247837800008

    View details for PubMedID 17609108

  • A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis CELL Nachury, M. V., Loktev, A. V., Zhang, Q., Westlake, C. J., Peranen, J., Merdes, A., Slusarski, D. C., Scheller, R. H., Bazan, J. F., Sheffield, V. C., Jackson, P. K. 2007; 129 (6): 1201-1213

    Abstract

    Primary cilium dysfunction underlies the pathogenesis of Bardet-Biedl syndrome (BBS), a genetic disorder whose symptoms include obesity, retinal degeneration, and nephropathy. However, despite the identification of 12 BBS genes, the molecular basis of BBS remains elusive. Here we identify a complex composed of seven highly conserved BBS proteins. This complex, the BBSome, localizes to nonmembranous centriolar satellites in the cytoplasm but also to the membrane of the cilium. Interestingly, the BBSome is required for ciliogenesis but is dispensable for centriolar satellite function. This ciliogenic function is mediated in part by the Rab8 GDP/GTP exchange factor, which localizes to the basal body and contacts the BBSome. Strikingly, Rab8(GTP) enters the primary cilium and promotes extension of the ciliary membrane. Conversely, preventing Rab8(GTP) production blocks ciliation in cells and yields characteristic BBS phenotypes in zebrafish. Our data reveal that BBS may be caused by defects in vesicular transport to the cilium.

    View details for DOI 10.1016/j.cell.2007.03.053

    View details for Web of Science ID 000247390400022

    View details for PubMedID 17574030

  • Oncogenic regulators and substrates of the anaphase promoting complex/cyclosome are frequently overexpressed in malignant tumors AMERICAN JOURNAL OF PATHOLOGY Lehman, N. L., Tibshirani, R., Hsu, J. Y., Natkunam, Y., Harris, B. T., West, R. B., Masek, M. A., Montgomery, K., van de Rijn, M., Jackson, P. K. 2007; 170 (5): 1793-1805

    Abstract

    The fidelity of cell division is dependent on the accumulation and ordered destruction of critical protein regulators. By triggering the appropriately timed, ubiquitin-dependent proteolysis of the mitotic regulatory proteins securin, cyclin B, aurora A kinase, and polo-like kinase 1, the anaphase promoting complex/cyclosome (APC/C) ubiquitin ligase plays an essential role in maintaining genomic stability. Misexpression of these APC/C substrates, individually, has been implicated in genomic instability and cancer. However, no comprehensive survey of the extent of their misregulation in tumors has been performed. Here, we analyzed more than 1600 benign and malignant tumors by immunohistochemical staining of tissue microarrays and found frequent overexpression of securin, polo-like kinase 1, aurora A, and Skp2 in malignant tumors. Positive and negative APC/C regulators, Cdh1 and Emi1, respectively, were also more strongly expressed in malignant versus benign tumors. Clustering and statistical analysis supports the finding that malignant tumors generally show broad misregulation of mitotic APC/C substrates not seen in benign tumors, suggesting that a "mitotic profile" in tumors may result from misregulation of the APC/C destruction pathway. This profile of misregulated mitotic APC/C substrates and regulators in malignant tumors suggests that analysis of this pathway may be diagnostically useful and represent a potentially important therapeutic target.

    View details for DOI 10.2353/ajpath.2007.060767

    View details for PubMedID 17456782

  • Emi2 at the crossroads - Where CSF meets MPF CELL CYCLE Hansen, D. V., Pomerening, J. R., Summers, M. K., Miller, J. J., Ferrell, J. E., Jackson, P. K. 2007; 6 (6): 732-738

    Abstract

    Vertebrate eggs arrest at metaphase of meiosis II due to an activity known as cytostatic factor (CSF). CSF antagonizes the ubiquitin ligase activity of the anaphase-promoting complex/cyclosome (APC/C), preventing cyclin B destruction and meiotic exit until fertilization occurs. A puzzling feature of CSF arrest is that APC/C inhibition is leaky. Ongoing cyclin B synthesis is counterbalanced by a limited amount of APC/C-mediated cyclin B destruction; thus, cyclin B/Cdc2 activity remains at steady state. How the APC/C can be slightly active toward cyclin B, and yet restrained from ubiquitinating cyclin B altogether, is unknown. Emi2/XErp1 is the critical CSF component directly responsible for APC/C inhibition during CSF arrest. Fertilization triggers the Ca2+-dependent destruction of Emi2, releasing the APC/C to ubiquitinate the full pool of cyclin B and initiate completion of meiosis. Previously, we showed that a phosphatase maintains Emi2's APC/C-inhibitory activity in CSF-arrested Xenopus egg extracts. Here, we demonstrate that phosphatase inhibition permits Emi2 phosphorylation at thr-545 and -551, which inactivates Emi2. Furthermore, we provide evidence that adding excess cyclin B to CSF extracts stimulates Cdc2 phosphorylation of these same residues, antagonizing Emi2-APC/C association. Our findings suggest a model wherein the pool of Emi2 acts analogously to a rheostat by integrating Cdc2 and phosphatase activities to prevent cyclin B overaccumulation and Cdc2 hyperactivity during the indefinite period of time between arrival at metaphase II and eventual fertilization. Finally, we propose that inactivation of Emi2 by Cdc2 permits mitotic progression during early embryonic cleavage cycles.

    View details for Web of Science ID 000245496500017

    View details for PubMedID 17361104

  • Translational unmasking of Emi2 directs cytostatic factor arrest in meiosis II CELL CYCLE Tung, J. J., Padmanabhan, K., Hansen, D. V., Richter, J. D., Jackson, P. K. 2007; 6 (6): 725-731

    Abstract

    Cytostatic factor (CSF) arrests unfertilized vertebrate eggs in metaphase of meiosis II by inhibiting the anaphase-promoting complex/cyclosome (APC/C) from mediating cyclin destruction. The APC/C inhibitor Emi2/XErp1 satisfies a number of historical criteria for the molecular identification of CSF, but the mechanism by which CSF is activated selectively in meiosis II is the remaining unexplained criterion. Here we provide an explanation by showing that Emi2 is expressed specifically in meiosis II through translational de-repression or "unmasking" of its mRNA. We find that Emi2 protein is undetectable in immature, G2/prophase-arrested Xenopus oocytes and accumulates approximately 90 minutes after germinal vesicle breakdown. The 3' untranslated region of Emi2 mRNA contains cytoplasmic polyadenylation elements that directly bind the CPEB protein and confer temporal regulation of Emi2 polyadenylation and translation. Our results demonstrate that cytoplasmic polyadenylation and translational unmasking of Emi2 directs meiosis II-specific CSF arrest.

    View details for Web of Science ID 000245496500016

    View details for PubMedID 17361107

  • A role for Cdc2- and PP2A-mediated regulation of Emi2 in the maintenance of CSF arrest CURRENT BIOLOGY Wu, Q., Guo, Y., Yamada, A., Perry, J. A., Wang, M. Z., Araki, M., Freel, C. D., Tung, J. J., Tang, W., Margolis, S. S., Jackson, P. K., Yamano, H., Asano, M., Kornbluth, S. 2007; 17 (3): 213-224

    Abstract

    Vertebrate oocytes are arrested in metaphase II of meiosis prior to fertilization by cytostatic factor (CSF). CSF enforces a cell-cycle arrest by inhibiting the anaphase-promoting complex (APC), an E3 ubiquitin ligase that targets Cyclin B for degradation. Although Cyclin B synthesis is ongoing during CSF arrest, constant Cyclin B levels are maintained. To achieve this, oocytes allow continuous slow Cyclin B degradation, without eliminating the bulk of Cyclin B, which would induce release from CSF arrest. However, the mechanism that controls this continuous degradation is not understood.We report here the molecular details of a negative feedback loop wherein Cyclin B promotes its own destruction through Cdc2/Cyclin B-mediated phosphorylation and inhibition of the APC inhibitor Emi2. Emi2 bound to the core APC, and this binding was disrupted by Cdc2/Cyclin B, without affecting Emi2 protein stability. Cdc2-mediated phosphorylation of Emi2 was antagonized by PP2A, which could bind to Emi2 and promote Emi2-APC interactions.Constant Cyclin B levels are maintained during a CSF arrest through the regulation of Emi2 activity. A balance between Cdc2 and PP2A controls Emi2 phosphorylation, which in turn controls the ability of Emi2 to bind to and inhibit the APC. This balance allows proper maintenance of Cyclin B levels and Cdc2 kinase activity during CSF arrest.

    View details for DOI 10.1016/j.cub.2006.12.045

    View details for Web of Science ID 000244164700020

    View details for PubMedID 17276914

  • Putting transcription repression and protein destruction in pRb's pocket DEVELOPMENTAL CELL Verschuren, E. W., Jackson, P. K. 2007; 12 (2): 169-170

    Abstract

    The tumor suppressor function of the retinoblastoma protein (pRb) is historically attributed to inhibition of E2F gene transcription. In a recent issue of Nature Cell Biology, Binné and colleagues show that pRB is physically linked to the active anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase, suggesting that pRB-mediated tumor suppression may also function by directing the local degradation of E2F targets, including Skp2 (Binné et al., 2006).

    View details for DOI 10.1016/j.devcel.2007.01.015

    View details for Web of Science ID 000245087200002

    View details for PubMedID 17276331

  • Identification of Rab11 as a small GTPase binding protein for the Evi5 oncogene PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Westlake, C. J., Junutula, J. R., Simon, G. C., Pilli, M., Prekeris, R., Scheller, R. H., Jackson, P. K., Eldridge, A. G. 2007; 104 (4): 1236-1241

    Abstract

    The Evi5 oncogene has recently been shown to regulate the stability and accumulation of critical G(1) cell cycle factors including Emi1, an inhibitor of the anaphase-promoting complex/cyclosome, and cyclin A. Sequence analysis of the amino terminus of Evi5 reveals a Tre-2, Bub2, Cdc16 domain, which has been shown to be a binding partner and GTPase-activating protein domain for the Rab family of small Ras-like GTPases. Here we describe the identification of Evi5 as a candidate binding protein for Rab11, a GTPase that regulates intracellular transport and has specific roles in endosome recycling and cytokinesis. By yeast two-hybrid analysis, immunoprecipitation, and Biacore analysis, we demonstrate that Evi5 binds Rab11a and Rab11b in a GTP-dependent manner. However, Evi5 displays no activation of Rab11 GTPase activity in vitro. Evi5 colocalizes with Rab11 in vivo, and overexpression of Rab11 perturbs the localization of Evi5, redistributing it into Rab11-positive recycling endosomes. Interestingly, in vitro binding studies show that Rab11 effector proteins including FIP3 compete with Evi5 for binding to Rab11, suggesting a partitioning between Rab11-Evi5 and Rab11 effector complexes. Indeed, ablation of Evi5 by RNA interference causes a mislocalization of FIP3 at the abscission site during cytokinesis. These data demonstrate that Evi5 is a Rab11 binding protein and that Evi5 may cooperate with Rab11 to coordinate vesicular trafficking, cytokinesis, and cell cycle control independent of GTPase-activating protein function.

    View details for DOI 10.1073/pnas.0610500104

    View details for Web of Science ID 000243849900024

    View details for PubMedID 17229837

  • Prophase I arrest and progression to metaphase I in mouse oocytes are controlled by Emi1-dependent regulation of APC(Cdh1) JOURNAL OF CELL BIOLOGY Marangos, P., Verschuren, E. W., Chen, R., Jackson, P. K., Carroll, J. 2007; 176 (1): 65-75

    Abstract

    Mammalian oocytes are arrested in prophase of the first meiotic division. Progression into the first meiotic division is driven by an increase in the activity of maturation-promoting factor (MPF). In mouse oocytes, we find that early mitotic inhibitor 1 (Emi1), an inhibitor of the anaphase-promoting complex (APC) that is responsible for cyclin B destruction and inactivation of MPF, is present at prophase I and undergoes Skp1-Cul1-F-box/betaTrCP-mediated destruction immediately after germinal vesicle breakdown (GVBD). Exogenous Emi1 or the inhibition of Emi1 destruction in prophase-arrested oocytes leads to a stabilization of cyclin B1-GFP that is sufficient to trigger GVBD. In contrast, the depletion of Emi1 using morpholino oligonucleotides increases cyclin B1-GFP destruction, resulting in an attenuation of MPF activation and a delay of entry into the first meiotic division. Finally, we show that Emi1-dependent effects on meiosis I require the presence of Cdh1. These observations reveal a novel mechanism for the control of entry into the first meiotic division: an Emi1-dependent inhibition of APC(Cdh1).

    View details for DOI 10.1083/jcb.200607070

    View details for Web of Science ID 000243581600008

    View details for PubMedID 17190794

    View details for PubMedCentralID PMC2063628

  • Mouse Emi2 is required to enter meiosis II by reestablishing cyclin B1 during interkinesis JOURNAL OF CELL BIOLOGY Madgwick, S., Hansen, D. V., Levasseur, M., Jackson, P. K., Jones, K. T. 2006; 174 (6): 791-801

    Abstract

    During interkinesis, a metaphase II (MetII) spindle is built immediately after the completion of meiosis I. Oocytes then remain MetII arrested until fertilization. In mouse, we find that early mitotic inhibitor 2 (Emi2), which is an anaphase-promoting complex inhibitor, is involved in both the establishment and the maintenance of MetII arrest. In MetII oocytes, Emi2 needs to be degraded for oocytes to exit meiosis, and such degradation, as visualized by fluorescent protein tagging, occurred tens of minutes ahead of cyclin B1. Emi2 antisense morpholino knockdown during oocyte maturation did not affect polar body (PB) extrusion. However, in interkinesis the central spindle microtubules from meiosis I persisted for a short time, and a MetII spindle failed to assemble. The chromatin in the oocyte quickly decondensed and a nucleus formed. All of these effects were caused by the essential role of Emi2 in stabilizing cyclin B1 after the first PB extrusion because in Emi2 knockdown oocytes a MetII spindle was recovered by Emi2 rescue or by expression of nondegradable cyclin B1 after meiosis I.

    View details for Web of Science ID 000240697100009

    View details for PubMedID 16966421

    View details for PubMedCentralID PMC2064334

  • Emi1 stably binds and inhibits the anaphase-promoting complex/cyclosome as a pseudosubstrate inhibitor GENES & DEVELOPMENT Miller, J. J., Summers, M. K., Hansen, D. V., Nachury, M. V., Lehman, N. L., Loktev, A., Jackson, P. K. 2006; 20 (17): 2410-2420

    Abstract

    The periodic destruction of mitotic cyclins is triggered by the activation of the anaphase-promoting complex/cyclosome (APC/C) in mitosis. Although the ability of the APC/C to recognize destruction box (D-box) substrates oscillates throughout the cell cycle, the mechanism regulating APC/C binding to D-box substrates remains unclear. Here, we show that the APC/C inhibitor Emi1 tightly binds both the APC/C and its Cdh1 activator, binds to the D-box receptor site on the APC/C(Cdh1), and competes with APC/C substrates for D-box binding. Emi1 itself contains a conserved C-terminal D-box, which provides APC/C-binding affinity, and a conserved zinc-binding region (ZBR), which antagonizes APC/C E3 ligase activity independent of tight APC binding. Mutation of the ZBR converts Emi1 into a D-box-dependent APC/C substrate. The identification of a direct Emi1-APC/C complex further explains how Emi1 functions as a stabilizing factor for cyclin accumulation and the need to destroy Emi1 for APC/C activation in mitosis. The combination of a degron/E3 recognition site and an anti-ligase function in Emi1 suggests a general model for how E3 substrates evolve to become pseudosubstrate inhibitors.

    View details for DOI 10.1101/gad.1454006

    View details for Web of Science ID 000240303400010

    View details for PubMedID 16921029

    View details for PubMedCentralID PMC1560415

  • Developmental neurobiology - A destructive switch for neurons NATURE Jackson, P. K. 2006; 442 (7101): 365-366

    View details for DOI 10.1038/442365a

    View details for Web of Science ID 000239278900026

    View details for PubMedID 16871201

  • Overexpression of the anaphase promoting complex/cyclosome inhibitor Emi1 leads to tetraploidy and genomic instability of p53-deficient cells CELL CYCLE Lehman, N. L., Verschuren, E. W., Hsu, J. Y., Cherry, A. M., Jackson, P. K. 2006; 5 (14): 1569-1573

    Abstract

    The anaphase promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that controls the cell cycle by directing the ubiquitin-dependent proteolysis of S-phase and mitosis promoting factors. Emi1 is an E2F transcriptional target that drives cell cycle progression from G1/S through early mitosis by inhibiting the APC/C's ubiquitin ligase activity, and thus facilitates accumulation of APC/C substrates. Using cell culture model systems, we found that Emi1 overexpression leads to proliferation, tetraploidy and genome instability of cells deficient for p53. We propose that loss of pRb repression of E2F-mediated transcription causing misregulation of Emi1 and APC/C substrates results in the generation of tetraploidy and proliferation of genomically unstable cells in the absence of normal p53 function. This represents a potentially important mechanism by which pRb and p53 dysfunction may contribute to tumorigenesis through the generation of genomic instability.

    View details for Web of Science ID 000240697800017

    View details for PubMedID 16861914

  • Climbing the Greatwall to mitosis MOLECULAR CELL Jackson, P. K. 2006; 22 (2): 156-157

    Abstract

    Greatwall kinase is a conserved regulator of mitotic entry, and new work in Xenopus egg extracts (Yu et al., 2006) shows that Greatwall is required for the positive feedback loop that removes inhibitory tyrosine phosphate from the central mitotic regulatory kinase Cdc2.

    View details for DOI 10.1016/j.molcel.2006.04.005

    View details for Web of Science ID 000237150400005

    View details for PubMedID 16630886

  • The Evi5 oncogene regulates cyclin accumulation by stabilizing the anaphase-promoting complex inhibitor Emi1 (vol 124, pg 367, 2006) CELL Eldridge, A. G., Loktev, A. V., Hansen, D. V., Verschuren, E. W., Reimann, J. D., Jackson, P. K. 2006; 124 (6): 1301–2
  • Dual degradation signals control Gli protein stability and tumor formation GENES & DEVELOPMENT Huntzicker, E. G., Estay, I. S., Zhen, H., Lokteva, L. A., Jackson, P. K., Oro, A. E. 2006; 20 (3): 276-281

    Abstract

    Regulated protein destruction controls many key cellular processes with aberrant regulation increasingly found during carcinogenesis. Gli proteins mediate the transcriptional effects of the Sonic hedgehog pathway, which is implicated in up to 25% of human tumors. Here we show that Gli is rapidly destroyed by the proteasome and that mouse basal cell carcinoma induction correlates with Gli protein accumulation. We identify two independent destruction signals in Gli1, D(N) and D(C), and show that removal of these signals stabilizes Gli1 protein and rapidly accelerates tumor formation in transgenic animals. These data argue that control of Gli protein accumulation underlies tumorigenesis and suggest a new avenue for antitumor therapy.

    View details for DOI 10.1101/gad.1380906

    View details for Web of Science ID 000235267000002

    View details for PubMedID 16421275

    View details for PubMedCentralID PMC1361699

  • The Evi5 oncogene regulates cyclin accumulation by stabilizing the anaphase-promoting complex inhibitor Emi1 CELL Eldridge, A. G., Loktev, A. V., Hansen, D. V., Verschuren, E. W., Reimann, J. D., Jackson, P. K. 2006; 124 (2): 367-380

    Abstract

    The anaphase-promoting complex/cyclosome (APC/C) inhibitor Emi1 controls progression to S phase and mitosis by stabilizing key APC/C ubiquitination substrates, including cyclin A. Examining Emi1 binding proteins, we identified the Evi5 oncogene as a regulator of Emi1 accumulation. Evi5 antagonizes SCF(betaTrCP)-dependent Emi1 ubiquitination and destruction by binding to a site adjacent to Emi1's DSGxxS degron and blocking both degron phosphorylation by Polo-like kinases and subsequent betaTrCP binding. Thus, Evi5 functions as a stabilizing factor maintaining Emi1 levels in S/G2 phase. Evi5 protein accumulates in early G1 following Plk1 destruction and is degraded in a Plk1- and ubiquitin-dependent manner in early mitosis. Ablation of Evi5 induces precocious degradation of Emi1 by the Plk/SCF(betaTrCP) pathway, causing premature APC/C activation; cyclin destruction; cell-cycle arrest; centrosome overduplication; and, finally, mitotic catastrophe. We propose that the balance of Evi5 and Polo-like kinase activities determines the timely accumulation of Emi1 and cyclin, ensuring mitotic fidelity.

    View details for DOI 10.1016/j.cell.2005.10.038

    View details for Web of Science ID 000235068500024

    View details for PubMedID 16439210

  • CaMKII and Polo-like kinase 1 sequentially phosphorylate the cytostatic factor Emi2/XErp1 to trigger its destruction and meiotic exit PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Hansen, D. V., Tung, J. J., Jackson, P. K. 2006; 103 (3): 608-613

    Abstract

    In vertebrate meiosis, unfertilized eggs are arrested in metaphase II by cytostatic factor (CSF), which is required to maintain mitotic cyclin-dependent kinase activity. Fertilization triggers a transient increase in cytosolic free Ca(2+), which leads to CSF inactivation and ubiquitin-dependent cyclin destruction through the anaphase promoting complex or cyclosome (APC/C). The Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and the Polo-like kinase Plx1 are essential factors for Ca(2+)-induced meiotic exit, but the critical targets of these kinases were unknown. The APC/C inhibitor Emi2 or XErp1 has recently been characterized as a pivotal CSF component, required to maintain metaphase II arrest and rapidly destroyed in response to Ca(2+) signaling through phosphorylation by Plx1 and ubiquitination by the SCF(betaTrCP) complex. An important question is how the increase in free Ca(2+) targets Plx1 activity toward Emi2. Here, we demonstrate that CaMKII is required for Ca(2+)-induced Emi2 destruction, and that CaMKII functions as a "priming kinase," directly phosphorylating Emi2 at a specific motif to induce a strong interaction with the Polo Box domain of Plx1. We show that the strict requirement for CaMKII to phosphorylate Emi2 is a specific feature of CSF arrest, and we also use phosphatase inhibitors to demonstrate an additional mode of Emi2 inactivation independent of its destruction. We firmly establish the CSF component Emi2 as the first-known critical and direct target of CaMKII in CSF release, providing a detailed molecular mechanism explaining how CaMKII and Plx1 coordinately direct APC/C activation and meiotic exit upon fertilization.

    View details for DOI 10.1073/pnas.0509549102

    View details for Web of Science ID 000234727800021

    View details for PubMedID 16407128

    View details for PubMedCentralID PMC1325965

  • Inhibition of the anaphase-promoting complex by the Xnf7 ubiquitin ligase JOURNAL OF CELL BIOLOGY Casaletto, J. B., Nutt, L. K., Wu, Q. J., Moore, J. D., ETKIN, L. D., Jackson, P. K., Hunt, T., Kornbluth, S. 2005; 169 (1): 61-71

    Abstract

    Degradation of specific protein substrates by the anaphase-promoting complex/cyclosome (APC) is critical for mitotic exit. We have identified the protein Xenopus nuclear factor 7 (Xnf7) as a novel APC inhibitor able to regulate the timing of exit from mitosis. Immunodepletion of Xnf7 from Xenopus laevis egg extracts accelerated the degradation of APC substrates cyclin B1, cyclin B2, and securin upon release from cytostatic factor arrest, whereas excess Xnf7 inhibited APC activity. Interestingly, Xnf7 exhibited intrinsic ubiquitin ligase activity, and this activity was required for APC inhibition. Unlike other reported APC inhibitors, Xnf7 did not associate with Cdc20, but rather bound directly to core subunits of the APC. Furthermore, Xnf7 was required for spindle assembly checkpoint function in egg extracts. These data suggest that Xnf7 is an APC inhibitor able to link spindle status to the APC through direct association with APC core components.

    View details for DOI 10.1083/jcb.200411056

    View details for Web of Science ID 000228327500006

    View details for PubMedID 15824132

    View details for PubMedCentralID PMC2171901

  • A role for the anaphase-promoting complex inhibitor Emi2/XErp1, a homolog of early mitotic inhibitor 1, in cytostatic factor arrest of Xenopus eggs PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Tung, J. J., Hansen, D. V., Ban, K. H., Loktev, A. V., Summers, M. K., Adler, J. R., Jackson, P. K. 2005; 102 (12): 4318-4323

    Abstract

    Unfertilized vertebrate eggs are arrested in metaphase of meiosis II with high cyclin B/Cdc2 activity to prevent parthenogenesis. Until fertilization, exit from metaphase is blocked by an activity called cytostatic factor (CSF), which stabilizes cyclin B by inhibiting the anaphase-promoting complex (APC) ubiquitin ligase. The APC inhibitor early mitotic inhibitor 1 (Emi1) was recently found to be required for maintenance of CSF arrest. We show here that exogenous Emi1 is unstable in CSF-arrested Xenopus eggs and is destroyed by the SCF(betaTrCP) ubiquitin ligase, suggesting that endogenous Emi1, an apparent 44-kDa protein, requires a stabilizing factor. However, anti-Emi1 antibodies crossreact with native Emi2/Erp1/FBXO43, a homolog of Emi1 and conserved APC inhibitor. Emi2 is stable in CSF-arrested eggs, is sufficient to prevent CSF release, and is rapidly degraded in a Polo-like kinase 1-dependent manner in response to calcium-mediated egg activation. These results identify Emi2 as a candidate CSF maintenance protein.

    View details for DOI 10.1073/pnas.0501108102

    View details for Web of Science ID 000227854800019

    View details for PubMedID 15753281

    View details for PubMedCentralID PMC552977

  • Structures of the agouti signaling protein JOURNAL OF MOLECULAR BIOLOGY McNulty, J. C., Jackson, P. J., Thompson, D. A., Chai, B. X., Gantz, I., Barsh, G. S., Dawson, P. E., Millhauser, G. L. 2005; 346 (4): 1059-1070

    Abstract

    Expression of the agouti signaling protein (ASIP) during hair growth produces the red/yellow pigment pheomelanin. ASIP, and its neuropeptide homolog the agouti-related protein (AgRP) involved in energy balance, are novel, paracrine signaling molecules that act as inverse agonists at distinct subsets of melanocortin receptors. Ubiquitous ASIP expression in mice gives rise to a pleiotropic phenotype characterized by a uniform yellow coat color, obesity, overgrowth, and metabolic derangements similar to type II diabetes in humans. Here we report the synthesis and NMR structure of ASIP's active, cysteine-rich, C-terminal domain. ASIP adopts the inhibitor cystine knot fold and, along with AgRP, are the only known mammalian proteins in this structure class. Moreover, ASIP populates two distinct conformers resulting from a cis peptide bond at Pro102-Pro103 and a coexistence of cis/trans isomers of Ala104-Pro105. Pharmacologic studies of Pro-->Ala mutants demonstrate that the minor conformation with two cis peptide bonds is responsible for activity at all MCRs. The loop containing the heterogeneous Ala-Pro peptide bond is conserved in mammals, and suggests that ASIP is either trapped by evolution in this unusual configuration or possesses function outside of strict MCR antagonism.

    View details for DOI 10.1016/j.jmb.2004.12.030

    View details for Web of Science ID 000227187800010

    View details for PubMedID 15701517

  • Emi1 class of proteins regulate entry into meiosis and the meiosis I to meiosis II transition in Xenopus oocytes CELL CYCLE Tung, J. J., Jackson, P. K. 2005; 4 (3): 478-482

    Abstract

    Xenopus oocytes are arrested at the G2/prophase boundary of meiosis I and enter meiosis in response to progesterone. A hallmark of meiosis is the absence of DNA replication between the successive cell division phases meiosis I (MI) and meiosis II (MII). After the MI-MII transition, Xenopus eggs are locked in metaphase II by the cytostatic factor (CSF) arrest to prevent parthenogenesis. Early Mitotic Inhibitor 1 (Emi1) maintains CSF arrest by inhibiting the ability of the Anaphase Promoting Complex (APC) to direct the destruction of cyclin B. To investigate whether Emi1 has an earlier role in meiosis, we injected Xenopus oocytes with neutralizing antibodies against Emi1 at G2/prophase and during the MI-MII transition. Progesterone-treated G2/prophase oocytes injected with anti-Emi1 antibody fail to activate Maturation Promoting Factor (MPF), a complex of cdc2/cyclin B, and the MAPK pathway, and do not undergo germinal vesicle breakdown (GVBD). Injection of purified Delta90 cyclin B protein or blocking anti-Emi1 antibody with purified Emi1 protein rescues these meiotic processes in Emi1-neutralized oocytes. Acute inhibition of Emi1 in progesterone treated oocytes immediately after GVBD causes rapid loss of cdc2 activity with simultaneous loss of cyclin B levels and inactivation of the MAPK pathway. These oocytes decondense their chromosomes and enter a DNA replication phase instead of progressing to MII. Prior ablation of Cdc20, addition of methyl-ubiquitin, or addition of nondestructible Delta90 cyclin B rescues the MI-MII transition in Emi1-inhibited oocytes.

    View details for Web of Science ID 000229538500030

    View details for PubMedID 15701974

  • Screening of tissue microarrays for ubiquitin proteasome system components in tumors UBIQUITIN AND PROTEIN DEGRADATION, PT B Lehman, N. L., van de Rijn, M., Jackson, P. K. 2005; 399: 334-?

    Abstract

    The turnover of key proteins that mediate development, cellular proliferation, and a host of essential biological processes is controlled by the ubiquitin proteasome system (UPS). In several well-studied examples, notably in the cell cycle, regulatory proteins that control ubiquitin-dependent destruction are themselves substrates of the UPS, creating a multilayered system to ensure precise and dynamic control of protein stability. UPS regulators controlled at the level of protein stability--including the F-box protein Skp2 and the VHL protein (substrate adapter proteins for multicomponent E3 ubiquitin ligases)-- seem to be misregulated in tumors. In these cases, especially, measuring levels of critical regulatory and target proteins will often present a more biologically meaningful picture than examining relative mRNA levels, which do not always reflect corresponding protein levels. Tissue microarrays (TMAs) allow simultaneous screening of large numbers of tumors for expression of specific proteins by immunohistochemical staining of a single microscope slide prepared from a TMA paraffin block. Replicate slides prepared from the same block can be immunostained for multiple proteins functioning in a related pathway, and a semiquantitative protein expression profile for a given subset of UPS pathway components, or other subsets of proteins of interest, can be assembled. Protein expression profiles of individual tumors or tissue types can be compared and visualized by hierarchical clustering methods. These expression profiles may be used as screening tools to investigate the relative abundance of components of a biochemical pathway in tumors or other tissues. TMAs have an exciting future as tools for basic research, diagnostic pathology, and drug targeting. In this article, we provide an introduction to the use of TMAs to study the expression of UPS component proteins and substrates in tumors by immunohistochemistry.

    View details for DOI 10.1016/S0076-6879(05)99023-X

    View details for PubMedID 16338367

  • Plk1 regulates activation of the anaphase promoting complex by phosphorylating and triggering SCF beta TrCP-dependent destruction of the APC inhibitor Emi1 MOLECULAR BIOLOGY OF THE CELL Hansen, D. V., Loktev, A. V., Ban, K. H., Jackson, P. K. 2004; 15 (12): 5623-5634

    Abstract

    Progression through mitosis requires activation of cyclin B/Cdk1 and its downstream targets, including Polo-like kinase and the anaphase-promoting complex (APC), the ubiquitin ligase directing degradation of cyclins A and B. Recent evidence shows that APC activation requires destruction of the APC inhibitor Emi1. In prophase, phosphorylation of Emi1 generates a D-pS-G-X-X-pS degron to recruit the SCF(betaTrCP) ubiquitin ligase, causing Emi1 destruction and allowing progression beyond prometaphase, but the kinases directing this phosphorylation remain undefined. We show here that the polo-like kinase Plk1 is strictly required for Emi1 destruction and that overexpression of Plk1 is sufficient to trigger Emi1 destruction. Plk1 stimulates Emi1 phosphorylation, betaTrCP binding, and ubiquitination in vitro and cyclin B/Cdk1 enhances these effects. Plk1 binds to Emi1 in mitosis and the two proteins colocalize on the mitotic spindle poles, suggesting that Plk1 may spatially control Emi1 destruction. These data support the hypothesis that Plk1 activates the APC by directing the SCF-dependent destruction of Emi1 in prophase.

    View details for DOI 10.1091/mbc.E04-07-0598

    View details for Web of Science ID 000225372800037

    View details for PubMedID 15469984

    View details for PubMedCentralID PMC532041

  • Wagging the dogma: Tissue-specific cell cycle control in the mouse embryo CELL Pagano, M., Jackson, P. K. 2004; 118 (5): 535-538

    Abstract

    The family of cyclin-dependent kinases (Cdks) lies at the core of the machinery that drives the cell division cycle. Studies in cultured mammalian cells have provided insight into the cellular functions of many Cdks. Recent Cdk and cyclin knockouts in the mouse show that the functions of G1 cell cycle regulatory genes are often essential only in specific cell types, pointing to our limited understanding of tissue-specific expression, redundancy, and compensating mechanisms in the Cdk network.

    View details for Web of Science ID 000223730300004

    View details for PubMedID 15339658

  • Xenopus Cdc14 alpha/beta are localized to the nucleolus and centrosome and are required for embryonic cell division BMC CELL BIOLOGY Kaiser, B. K., Nachury, M. V., Gardner, B. E., Jackson, P. K. 2004; 5

    Abstract

    The dual specificity phosphatase Cdc14 has been shown to be a critical regulator of late mitotic events in several eukaryotes, including S. cerevisiae, S. pombe. C. elegans and H. sapiens. However, Cdc14 homologs have clearly evolved to regulate distinct cellular processes and to respond to regulatory signals important for these processes. The human paralogs hCdc14A and B are the only vertebrate Cdc14 homologues studied to date, but their functions are not well understood. Therefore, it is of great interest to examine the function Cdc14 homologs in other vertebrate species.We identified two open reading frames from Xenopus laevis closely related to human Cdc14A, called XCdc14alpha and XCdc14beta, although no obvious paralog of the hCdc14B was found. To begin a functional characterization of Xcdc14alpha and XCdc14beta, we raised polyclonal antibodies against a conserved region. These antibodies stained both the nucleolus and centrosome in interphase Xenopus tissue culture cells, and the mitotic centrosomes. GFP-tagged version of XCdc14alpha localized to the nucleulus and GFP-XCdc14beta localized to the centrosome, although not exclusively. XCdc14alpha was also both meiotically and mitotically phosphorylated. Injection of antibodies raised against a conserved region of XCdc14/beta into Xenopus embryos at the two-cell stage blocked division of the injected blastomeres, suggesting that activities of XCdc14alpha/beta are required for normal cell division.These results provide evidence that XCdc14alpha/beta are required for normal cellular division and are regulated by at least two mechanisms, subcellular localization and possibly phosphorylation. Due to the high sequence conservation between Xcdc14alpha and hCdc14A, it seems likely that both mechanisms will contribute to regulation of Cdc14 homologs in vertebrates.

    View details for DOI 10.1186/1471-2121-5-27

    View details for Web of Science ID 000222850900001

    View details for PubMedID 15251038

    View details for PubMedCentralID PMC481057

  • Linking tumor suppression, DNA damage and the anaphase-promoting complex TRENDS IN CELL BIOLOGY Jackson, P. K. 2004; 14 (7): 331-334

    Abstract

    A recent study shows that the RASSF1A tumor suppressor functions as a regulator of the ordered proteolytic steps that organize mitosis. By controlling the stability of microtubules and the activity of the anaphase-promoting complex (APC), RASSF1A might provide a crucial link between mechanisms of tumor suppression and mitotic cell division. Furthermore, another recent study shows that protein kinase A, which is a key growth regulator, inhibits the APC during mitosis in yeast.

    View details for DOI 10.1016/j.tcb.2004.05.005

    View details for Web of Science ID 000223010200001

    View details for PubMedID 15246424

  • Can Fizzy fly solo? NATURE CELL BIOLOGY Jackson, P. K. 2003; 5 (10): 864-865

    View details for DOI 10.1038/ncb1003-864

    View details for Web of Science ID 000185647400010

    View details for PubMedID 14523400

  • Prophase destruction of Emi1 by the SCF beta TrCP/Slimb ubiquitin ligase activates the anaphase promoting complex to allow progression beyond prometaphase DEVELOPMENTAL CELL Margottin-Goguet, F., Hsu, J. Y., Loktev, A., Hsieh, H. M., Reimann, J. D., Jackson, P. K. 2003; 4 (6): 813-826

    Abstract

    Progression through mitosis occurs because cyclin B/Cdc2 activation induces the anaphase promoting complex (APC) to cause cyclin B destruction and mitotic exit. To ensure that cyclin B/Cdc2 does not prematurely activate the APC in early mitosis, there must be a mechanism delaying APC activation. Emi1 is a protein capable of inhibiting the APC in S and G2. We show here that Emi1 is phosphorylated by Cdc2, and on a DSGxxS consensus site, is subsequently recognized by the SCF(betaTrCP/Slimb) ubiquitin ligase and destroyed, thus providing a delay for APC activation. Failure of betaTrCP-dependent Emi1 destruction stabilizes APC substrates and results in mitotic catastrophe including centrosome overduplication, potentially explaining mitotic deficiencies in Drosophila Slimb/betaTrCP mutants. We hypothesize that Emi1 destruction relieves a late prophase checkpoint for APC activation.

    View details for Web of Science ID 000183383100007

    View details for PubMedID 12791267

  • Control of meiotic and mitotic progression by the F box protein beta-Trcp1 in vivo DEVELOPMENTAL CELL Guardavaccaro, D., Kudo, Y., Boulaire, J., Barchi, M., Busino, L., Donzelli, M., Margottin-Goguet, F., Jackson, P. K., Yamasaki, L., Pagano, M. 2003; 4 (6): 799-812

    Abstract

    SCF ubiquitin ligases, composed of three major subunits, Skp1, Cul1, and one of many F box proteins (Fbps), control the proteolysis of important cellular regulators. We have inactivated the gene encoding the Fbp beta-Trcp1 in mice. beta-Trcp1(-/-) males show reduced fertility correlating with an accumulation of methaphase I spermatocytes. beta-Trcp1(-/-) MEFs display a lengthened mitosis, centrosome overduplication, multipolar metaphase spindles, and misaligned chromosomes. Furthermore, cyclin A, cyclin B, and Emi1, an inhibitor of the anaphase promoting complex, are stabilized in mitotic beta-Trcp1(-/-) MEFs. Indeed, we demonstrate that Emi1 is a bona fide substrate of beta-Trcp1. In contrast, stabilization of beta-catenin and IkappaBalpha, two previously reported beta-Trcp1 substrates, does not occur in the absence of beta-Trcp1 and instead requires the additional silencing of beta-Trcp2 by siRNA. Thus, beta-Trcp1 regulates the timely order of meiotic and mitotic events.

    View details for Web of Science ID 000183383100006

    View details for PubMedID 12791266

  • The activation of the Anaphase Promoting Complex in mitosis requires prior destruction of Emi1 by the SCFbetaTrCP/Slimb ubiquitin ligase Experimental Biology 2003 Annual Meeting Jackson, P. K., Margottin-Goguet, F., Loktev, A., Hsu, J., Regan-Reimann, J. FEDERATION AMER SOC EXP BIOL. 2003: A1182–A1182
  • Spongiform degeneration in mahoganoid mutant mice SCIENCE He, L., Lu, X. Y., Jolly, A. F., Eldridge, A. G., Watson, S. J., Jackson, P. K., Barsh, G. S., Gunn, T. M. 2003; 299 (5607): 710-712

    Abstract

    mahoganoid is a mouse coat-color mutation whose pigmentary phenotype and genetic interactions resemble those of Attractin (Atrn). Atrn mutations also cause spongiform neurodegeneration. Here, we show that a null mutation for mahoganoid causes a similar age-dependent neuropathology that includes many features of prion diseases but without accumulation of protease-resistant prion protein. The gene mutated in mahoganoid encodes a RING-containing protein with E3 ubiquitin ligase activity in vitro. Similarities in phenotype, expression, and genetic interactions suggest that mahoganoid and Atrn genes are part of a conserved pathway for regulated protein turnover whose function is essential for neuronal viability.

    View details for Web of Science ID 000180687700048

    View details for PubMedID 12560552

  • Ubiquitinating a phosphorylated Cdk inhibitor on the blades of the Cdc4 beta-propeller CELL Jackson, P. K. 2003; 112 (2): 142-144

    Abstract

    Substrate binding by the SCFCdc4 ubiquitin ligase is regulated by phosphorylation. In this issue of Cell, Orlicky et al. describe the crystal structure of the Cdc4 subunit bound to a high-affinity substrate phosphopeptide. This structure provides insights into the binding interaction and how a precise mechanism involving multiple regulatory phosphorylations may be mediated by a single binding site.

    View details for Web of Science ID 000181191600002

    View details for PubMedID 12553901

  • Accessory proteins for melanocortin signaling - Attractin and mahogunin 5th International Melanocortin Meeting He, L., Eldridge, A. G., Jackson, P. K., Gunn, T. M., Barsh, G. S. NEW YORK ACAD SCIENCES. 2003: 288–298

    Abstract

    Switching from eumelanin to pheomelanin synthesis during hair growth is accomplished by transient synthesis of Agouti protein, an inverse agonist for the melanocortin-1 receptor (Mc1r). The coat color mutations mahogany and mahoganoid prevent hair follicle melanocytes from responding to Agouti protein. The gene mutated in mahogany, which is also known as Attractin (Atrn), encodes a type I transmembrane protein that functions as an accessory receptor for Agouti protein. We have recently determined that the gene mutated in mahoganoid, which is also known as Mahogunin (Mgrn1), encodes an E3 ubiquitin ligase. Like Attractin, Mahogunin is conserved in invertebrate genomes, and its absence causes a pleiotropic phenotype that includes spongiform neurodegeneration.

    View details for Web of Science ID 000184303800037

    View details for PubMedID 12851328

  • Emi-1 is upregulated in clear cell carcinomas of the ovary: A preliminary comparative analysis of protein expression using ovarian tissue arrays 92nd Annual Meeting of the United-States-and-Canadian-Academy-of-Pathology Gutgemann, I., Hsiu, I., Lehman, N., Montgomery, K., Masek, M., Jackson, P., Longacre, T. NATURE PUBLISHING GROUP. 2003: 191A–191A
  • Loops and links: Structural insights into the remarkable function of the agouti-related protein 5th International Melanocortin Meeting Millhauser, G. L., McNulty, J. C., Jackson, P. J., Thompson, D. A., Barsh, G. S., Gantz, I. NEW YORK ACAD SCIENCES. 2003: 27–35

    Abstract

    The agouti-related protein (AGRP) is an endogenous antagonist of the melanocortin receptors MC3R and MC4R found in the hypothalamus and exhibits potent orexigenic activity. The cysteine-rich C-terminal domain of this protein, corresponding to AGRP(87-132), exhibits receptor binding affinity and antagonism equivalent to that of the full-length protein. We recently determined the NMR structure of AGRP(87-132) and demonstrated that a portion of the domain adopts the inhibitor cystine-knot fold. Remarkably, this is the first identification of a mammalian protein with this specific architecture. Further analysis of the structure suggests that melanocortin receptor contacts are made primarily by two loops presented within the cystine knot. (10) To test this hypothesis we designed a 34-residue AGRP analogue corresponding to only the cystine knot. We found that this designed miniprotein folds to a homogeneous product, retains the desired cystine-knot architecture, functions as a potent antagonist, and maintains the melanocortin receptor pharmacological profile of AGRP(87-132). (26) The AGRP-like activity of this molecule supports the hypothesis that indeed the cystine-knot region possesses the melanocortin receptor contacts. Based on these design and structure studies, we propose that the N-terminal loop of AGRP(87-132) makes contact with a receptor exoloop and helps confer AGRP's selectivity for the central MCRs.

    View details for Web of Science ID 000184303800004

    View details for PubMedID 12851295

  • The E3 ubiquitin ligase GREUL1 anteriorizes ectoderm during Xenopus development DEVELOPMENTAL BIOLOGY Borchers, A. G., Hufton, A. L., Eldridge, A. G., Jackson, P. K., Harland, R. M., Baker, J. C. 2002; 251 (2): 395-408

    Abstract

    We have identified a family of RING finger proteins that are orthologous to Drosophila Goliath (G1, Gol). One of the members, GREUL1 (Goliath Related E3 Ubiquitin Ligase 1), can convert Xenopus ectoderm into XAG-1- and Otx2-expressing cells in the absence of both neural tissue and muscle. This activity, combined with the finding that XGREUL1 is expressed within the cement gland, suggests a role for GREUL1 in the generation of anterior ectoderm. Although GREUL1 is not a direct inducer of neural tissue, it can activate the formation of ectopic neural cells within the epidermis of intact embryos. This suggests that GREUL1 can sensitize ectoderm to neuralizing signals. In this paper, we provide evidence that GREUL1 is an E3 ubiquitin ligase. Using a biochemical assay, we show that GREUL1 catalyzes the addition of polyubiquitin chains. These events are mediated by the RING domain since a mutation in two of the cysteines abolishes ligase activity. Mutation of these cysteines also compromises GREUL1's ability to induce cement gland. Thus, GREUL1's RING domain is necessary for both the ubiquitination of substrates and for the conversion of ectoderm to an anterior fate.

    View details for DOI 10.1006/dbio.2002.0814

    View details for Web of Science ID 000179377900015

    View details for PubMedID 12435366

  • hEmi1 links the Rb/E2F pathway to spindle abnormalities and chromosome missegregation 42nd Annual Meeting of the American-Society-for-Cell-Biology Hsu, J. Y., Reimann, J. R., Sorensen, C. S., Lukas, J., Jackson, P. K. AMER SOC CELL BIOLOGY. 2002: 299A–299A
  • Control of the centriole and centrosome cycles by ubiquitination enzymes ONCOGENE Hansen, D. V., Hsu, J. Y., Kaiser, B. K., Jackson, P. K., Eldridge, A. G. 2002; 21 (40): 6209-6221

    View details for Web of Science ID 000177840500010

    View details for PubMedID 12214251

  • Disruption of centrosome structure, chromosome segregation, and cytokinesis by misexpression of human Cdc14A phosphatase MOLECULAR BIOLOGY OF THE CELL Kaiser, B. K., Zimmerman, Z. A., Charbonneau, H., Jackson, P. K. 2002; 13 (7): 2289-2300

    Abstract

    In budding yeast, the Cdc14p phosphatase activates mitotic exit by dephosphorylation of specific cyclin-dependent kinase (Cdk) substrates and seems to be regulated by sequestration in the nucleolus until its release in mitosis. Herein, we have analyzed the two human homologs of Cdc14p, hCdc14A and hCdc14B. We demonstrate that the human Cdc14A phosphatase is selective for Cdk substrates in vitro and that although the protein abundance and intrinsic phosphatase activity of hCdc14A and B vary modestly during the cell cycle, their localization is cell cycle regulated. hCdc14A dynamically localizes to interphase but not mitotic centrosomes, and hCdc14B localizes to the interphase nucleolus. These distinct patterns of localization suggest that each isoform of human Cdc14 likely regulates separate cell cycle events. In addition, hCdc14A overexpression induces the loss of the pericentriolar markers pericentrin and gamma-tubulin from centrosomes. Overproduction of hCdc14A also causes mitotic spindle and chromosome segregation defects, defective karyokinesis, and a failure to complete cytokinesis. Thus, the hCdc14A phosphatase appears to play a role in the regulation of the centrosome cycle, mitosis, and cytokinesis, thereby influencing chromosome partitioning and genomic stability in human cells.

    View details for DOI 10.1091/mbc.01-11-0535

    View details for Web of Science ID 000177192600009

    View details for PubMedID 12134069

    View details for PubMedCentralID PMC117313

  • The SCF ubiquitin ligase: An extended look MOLECULAR CELL Jackson, P. K., Eldridge, A. G. 2002; 9 (5): 923-925

    Abstract

    The SCF E3 ubiquitin ligases select specific proteins for ubiquitination (and typically destruction) by coupling variable adaptor (F box) proteins that bind protein substrates to a conserved catalytic engine containing a cullin, Cul1, and the Rbx1/Roc1 RING finger protein. A new crystal structure of the SCF(Skp2) ubiquitin ligase shows the molecular organization of this complex and raises important questions as to how substrate ubiquitination is accomplished.

    View details for Web of Science ID 000175967100002

    View details for PubMedID 12049727

  • E2F-dependent accumulation of hEmi1 regulates S phase entry by inhibiting APC(Cdh1) NATURE CELL BIOLOGY Hsu, J. Y., Reimann, J. D., Sorensen, C. S., Lukas, J., Jackson, P. K. 2002; 4 (5): 358-366

    Abstract

    Emi1 promotes mitotic entry in Xenopus laevis embryos by inhibiting the APC(Cdc20) ubiquitination complex to allow accumulation of cyclin B. We show here that human Emi1 (hEmi1) functions to promote cyclin A accumulation and S phase entry in somatic cells by inhibiting the APC(Cdh1) complex. At the G1-S transition, hEmi1 is transcriptionally induced by the E2F transcription factor, much like cyclin A. hEmi1 overexpression accelerates S phase entry and can override a G1 block caused by overexpression of Cdh1 or the E2F-inhibitor p105 retinoblastoma protein (pRb). Depleting cells of hEmi1 through RNA interference prevents accumulation of cyclin A and inhibits S phase entry. These data suggest that E2F can activate both transcription of cyclin A and the hEmi1-dependent stabilization of APC(Cdh1) targets, such as cyclin A, to promote S phase entry.

    View details for DOI 10.1038/ncb785

    View details for Web of Science ID 000175325100017

    View details for PubMedID 11988738

  • Emi1 is required for cytostatic factor arrest in vertebrate eggs NATURE Reimann, J. D., Jackson, P. K. 2002; 416 (6883): 850-854

    Abstract

    Vertebrate eggs are arrested at metaphase of meiosis II with stable cyclin B and high cyclin B/Cdc2 kinase activity. The ability of the anaphase-promoting complex/cyclosome (APC), an E3 ubiquitin ligase, to trigger cyclin B destruction and metaphase exit is blocked in eggs by the activity of cytostatic factor (CSF) (reviewed in ref. 1). CSF was defined as an activity in mature oocytes that caused mitotic arrest when injected into dividing embryos. Fertilization causes a transient increase in cytoplasmic calcium concentration leading to CSF inactivation, APC activation, cyclin B destruction and mitotic exit. The APC activator Cdc20 is required for APC activation after fertilization. We show here that the APC(cdc20) inhibitor Emi1 (ref. 6) is necessary and sufficient to inhibit the APC and to prevent mitotic exit in CSF-arrested eggs. CSF extracts immunodepleted of Emi1 degrade cyclin B, and exit from mitosis prematurely in the absence of calcium. Addition of Emi1 to these Emi1-depleted extracts blocks premature inactivation of the CSF-arrested state. Emi1 is required to arrest unfertilized eggs at metaphase of meiosis II and seems to be the long-sought mediator of CSF activity.

    View details for Web of Science ID 000175163800052

    View details for PubMedID 11976684

  • Deregulated human Cdc14A phosphatase disrupts centrosome separation and chromosome segregation NATURE CELL BIOLOGY Mailand, N., Lukas, C., Kaiser, B. K., Jackson, P. K., Bartek, J., Lukas, J. 2002; 4 (4): 317-322

    Abstract

    We show that human Cdc14A phosphatase interacts with interphase centrosomes, and that this interaction is independent of microtubules and Cdc14A phosphatase activity, but requires active nuclear export. Disrupting the nuclear export signal (NES) led to Cdc14A being localized in nucleoli, which in unperturbed cells selectively contain Cdc14B (ref. 1). Conditional overproduction of Cdc14A, but not its phosphatase-dead or NES-deficient mutants, or Cdc14B, resulted in premature centrosome splitting and formation of supernumerary mitotic spindles. In contrast, downregulation of endogenous Cdc14A by short inhibitory RNA duplexes (siRNA) induced mitotic defects including impaired centrosome separation and failure to undergo productive cytokinesis. Consequently, both overexpression and downregulation of Cdc14A caused aberrant chromosome partitioning into daughter cells. These results indicate that Cdc14A is a physiological regulator of the centrosome duplication cycle, which, when disrupted, can lead to genomic instability in mammalian cells.

    View details for DOI 10.1038/ncb777

    View details for Web of Science ID 000174994000021

    View details for PubMedID 11901424

  • Emi1 regulates the anaphase-promoting complex by a different mechanism than Mad2 proteins GENES & DEVELOPMENT Reimann, J. D., Gardner, B. E., Margottin-Goguet, F., Jackson, P. K. 2001; 15 (24): 3278-3285

    Abstract

    The anaphase-promoting complex/cyclosome (APC) ubiquitin ligase is activated by Cdc20 and Cdh1 and inhibited by Mad2 and the spindle assembly checkpoint complex, Mad2B, and the early mitotic inhibitor Emi1. Mad2 inhibits APC(Cdc20), whereas Mad2B preferentially inhibits APC(Cdh1). We have examined the mechanism of APC inhibition by Emi1 and find that unlike Mad2 proteins, Emi1 binds and inhibits both APC(Cdh1) and APC(Cdc20). Also unlike Mad2, Emi1 stabilizes cyclin A in the embryo and requires zinc for its APC inhibitory activity. We find that Emi1 binds the substrate-binding region of Cdc20 and prevents substrate binding to the APC, illustrating a novel mechanism of APC inhibition.

    View details for Web of Science ID 000172879200006

    View details for PubMedID 11751633

    View details for PubMedCentralID PMC312853

  • A new RING for SUMO: wrestling transcriptional responses into nuclear bodies with PIAS family E3 SUMO ligases GENES & DEVELOPMENT Jackson, P. K. 2001; 15 (23): 3053-3058

    View details for Web of Science ID 000172598800001

    View details for PubMedID 11731472

  • Emi1 controls S phase and genome stability by regulating the Cdh1-APC complex Jackson, P. K., Reimann, J., Hsu, J. AMER SOC CELL BIOLOGY. 2001: 265A
  • Triggering ubiquitination of a CDK inhibitor at origins of DNA replication NATURE CELL BIOLOGY Furstenthal, L., Swanson, C., Kaiser, B. K., Eldridge, A. G., Jackson, P. K. 2001; 3 (8): 715-722

    Abstract

    To ensure proper timing of the G1-S transition in the cell cycle, the cyclin E-Cdk2 complex, which is responsible for the initiation of DNA replication, is restrained by the p21(Cip1)/p27(Kip1)/p57(Kip2) family of CDK (cyclin-dependent kinase) inhibitors in humans and by the related p27(Xic1) protein in Xenopus. Activation of cyclin E-Cdk2 is linked to the ubiquitination of human p27(Kip1) or Xenopus p27(Xic1) by SCF (for Skp1-Cullin-F-box protein) ubiquitin ligases. For human p27(Kip1), ubiquitination requires direct phosphorylation by cyclin E-Cdk2. We show here that Xic1 ubiquitination does not require phosphorylation by cyclin E-Cdk2, but it does require nuclear accumulation of the Xic1-cyclin E-Cdk2 complex and recruitment of this complex to chromatin by the origin-recognition complex together with Cdc6 replication preinitiation factors; it also requires an activation step necessitating cyclin E-Cdk2-kinase and SCF ubiquitin-ligase activity, and additional factors associated with mini-chromosome maintenance proteins, including the inactivation of geminin. Components of the SCF ubiquitin-ligase complex, including Skp1 and Cul1, are also recruited to chromatin through cyclin E-Cdk2 and the preinitiation complex. Thus, activation of the cyclin E-Cdk2 kinase and ubiquitin-dependent destruction of its inhibitor are spatially constrained to the site of a properly assembled preinitiation complex.

    View details for Web of Science ID 000170393200016

    View details for PubMedID 11483956

  • Emi1 is a mitotic regulator that interacts with Cdc20 and inhibits the anaphase promoting complex CELL Reimann, J. D., Freed, E., Hsu, J. Y., Kramer, E. R., Peters, J. M., Jackson, P. K. 2001; 105 (5): 645-655

    Abstract

    We have discovered an early mitotic inhibitor, Emi1, which regulates mitosis by inhibiting the anaphase promoting complex/cyclosome (APC). Emi1 is a conserved F box protein containing a zinc binding region essential for APC inhibition. Emi1 accumulates before mitosis and is ubiquitylated and destroyed in mitosis, independent of the APC. Emi1 immunodepletion from cycling Xenopus extracts strongly delays cyclin B accumulation and mitotic entry, whereas nondestructible Emi1 stabilizes APC substrates and causes a mitotic block. Emi1 binds the APC activator Cdc20, and Cdc20 can rescue an Emi1-induced block to cyclin B destruction. Our results suggest that Emi1 regulates progression through early mitosis by preventing premature APC activation, and may help explain the well-known delay between cyclin B/Cdc2 activation and cyclin B destruction.

    View details for Web of Science ID 000169123200011

    View details for PubMedID 11389834

  • Cyclin E uses Cdc6 as a chromatin-associated receptor required for DNA replication JOURNAL OF CELL BIOLOGY Furstenthal, L., Kaiser, B. K., Swanson, C., Jackson, P. K. 2001; 152 (6): 1267-1278

    Abstract

    Using an in vitro chromatin assembly assay in Xenopus egg extract, we show that cyclin E binds specifically and saturably to chromatin in three phases. In the first phase, the origin recognition complex and Cdc6 prereplication proteins, but not the minichromosome maintenance complex, are necessary and biochemically sufficient for ATP-dependent binding of cyclin E--Cdk2 to DNA. We find that cyclin E binds the NH(2)-terminal region of Cdc6 containing Cy--Arg-X-Leu (RXL) motifs. Cyclin E proteins with mutated substrate selection (Met-Arg-Ala-Ile-Leu; MRAIL) motifs fail to bind Cdc6, fail to compete with endogenous cyclin E--Cdk2 for chromatin binding, and fail to rescue replication in cyclin E--depleted extracts. Cdc6 proteins with mutations in the three consensus RXL motifs are quantitatively deficient for cyclin E binding and for rescuing replication in Cdc6-depleted extracts. Thus, the cyclin E--Cdc6 interaction that localizes the Cdk2 complex to chromatin is important for DNA replication. During the second phase, cyclin E--Cdk2 accumulates on chromatin, dependent on polymerase activity. In the third phase, cyclin E is phosphorylated, and the cyclin E--Cdk2 complex is displaced from chromatin in mitosis. In vitro, mitogen-activated protein kinase and especially cyclin B--Cdc2, but not the polo-like kinase 1, remove cyclin E--Cdk2 from chromatin. Rebinding of hyperphosphorylated cyclin E--Cdk2 to interphase chromatin requires dephosphorylation, and the Cdk kinase-directed Cdc14 phosphatase is sufficient for this dephosphorylation in vitro. These three phases of cyclin E association with chromatin may facilitate the diverse activities of cyclin E--Cdk2 in initiating replication, blocking rereplication, and allowing resetting of origins after mitosis.

    View details for Web of Science ID 000167715600014

    View details for PubMedID 11257126

    View details for PubMedCentralID PMC2199215

  • Characterization of a putative human homolog of Emi1, a mitotic regulator in Xenopus laevis Hsu, J. Y., Reimann, J. D., Jackson, P. K. AMER SOC CELL BIOLOGY. 2000: 448A
  • Novel roles for substrate phosphorylation in SCF ubiquitin ligase function Eldridge, A. G., Jackson, P. K. AMER SOC CELL BIOLOGY. 2000: 341A–342A
  • Cell cycle regulation by the Cdc14 phosphatase Kaiser, B. K., Furstenthal, L., Swanson, C., Eldridge, A. G., Jackson, P. K. AMER SOC CELL BIOLOGY. 2000: 342A
  • Cdc6 function as a substrate and chromatin-associated receptor for cyclin E/Cdk2 is required for DNA replication Furstenthal, L. E., Kaiser, B. K., Swanson, C. A., Jackson, P. K. AMER SOC CELL BIOLOGY. 2000: 345A
  • Emi1, a novel F-box, zinc finger protein, regulates mitosis by inhibiting the APC regulator Cdc20 Reimann, J. D., Freed, E., Kramer, E. R., Hsu, J. Y., Peters, J. M., Jackson, P. K. AMER SOC CELL BIOLOGY. 2000: 7A–7A
  • The lore of the RINGs: substrate recognition and catalysis by ubiquitin ligases TRENDS IN CELL BIOLOGY Jackson, P. K., Eldridge, A. G., Freed, E., Furstenthal, L., Hsu, J. Y., Kaiser, B. K., Reimann, J. D. 2000; 10 (10): 429-439

    Abstract

    Recently, many new examples of E3 ubiquitin ligases or E3 enzymes have been found to regulate a host of cellular processes. These E3 enzymes direct the formation of multiubiquitin chains on specific protein substrates, and - typically - the subsequent destruction of those proteins. We discuss how the modular architecture of E3 enzymes connects one of two distinct classes of catalytic domains to a wide range of substrate-binding domains. In one catalytic class, a HECT domain transfers ubiquitin directly to substrate bound to a non-catalytic domain. Members of the other catalytic class, found in the SCF, VBC and APC complexes, use a RING finger domain to facilitate ubiquitylation. The separable substrate-recognition domains of E3 enzymes provides a flexible means of linking a conserved ubiquitylation function to potentially thousands of ubiquitylated substrates in eukaryotic cells.

    View details for Web of Science ID 000089588600006

    View details for PubMedID 10998601

  • Nuclear accumulation of cyclin E/Cdk2 triggers a concentration-dependent switch for the destruction of p27 (Xic1) PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Swanson, C., Ross, J., Jackson, P. K. 2000; 97 (14): 7796-7801

    Abstract

    The action of cyclin-dependent kinases (CDKs) is regulated by phosphorylation, cyclin levels, the abundance of CDK inhibitors, and, as recently has been shown for cyclin B/cdc2, their localization. It is unclear how localization regulates the action of cyclin E/Cdk2 and its inhibitors. Here, we show that the closest known Xenopus laevis homolog of mammalian Cdk2 inhibitors p27(Kip1) and p21(CIP1), Xic1, is concentrated, ubiquitinated, and destroyed in the nucleus. Furthermore, Xic1 destruction requires nuclear import, but not nuclear export, and requires the formation of a transport-competent nuclear envelope, but not interactions between the lamina and chromatin. We show that (i) cyclin E/Cdk2 and Xic1 are transported into the nucleus as a complex and that Xic1 destruction requires the activity of cyclin E, (ii) that phosphorylation of Xic1 by cyclin E/Cdk2 bypasses the requirement for nuclear formation, and (iii) that the phosphorylation of Xic1 by cyclin E/Cdk2 is concentration dependent and likely realized through second-order interactions between stable cyclin E/Cdk2/Xic1 ternary complexes. Based on these results we propose a model wherein nuclear accumulation of the cyclin E/Cdk2/Xic1 complex triggers a concentration-dependent switch that promotes the phosphorylation of Xic1 and, consequently, its ubiquitination and destruction, thus allowing subsequent activation of cyclin E/Cdk2.

    View details for Web of Science ID 000088048400027

    View details for PubMedID 10884410

  • Identification of novel F-box proteins in Xenopus laevis CURRENT BIOLOGY Regan-Reimann, J. D., Duong, Q. V., Jackson, P. K. 1999; 9 (20): R762-R763

    View details for Web of Science ID 000083289200005

    View details for PubMedID 10531041

  • Budding yeast Cdc6p induces re-replication in fission yeast by inhibition at SCFPop-mediated proteolysis MOLECULAR AND GENERAL GENETICS Wolf, D. A., McKeon, F., Jackson, P. K. 1999; 262 (3): 473-480

    Abstract

    In fission yeast, overexpression of the replication initiator protein Cdc18p induces re-replication, a phenotype characterized by continuous DNA synthesis in the absence of cell division. In contrast, overexpression of Cdc6p, the budding yeast homolog of Cdc18p, does not cause re-replication in S. cerevisiae. However, we have found that Cdc6p has the ability to induce rereplication in fission yeast. Cdc6p cannot functionally replace Cdc18p, but instead interferes with the proteolysis of both Cdc18p and Rum1p, the inhibitor of the protein kinase Cdc2p. This activity of Cdc6p is entirely contained within a short N-terminal peptide, which forms a tight complex with Cdc2p and the F-box/WD-repeat protein Sud1p/Pop2p, a component of the SCF(Pop) ubiquitin ligase in fission yeast. These interactions are mediated by two distinct regions within the N-terminal region of Cdc6p and depend on the integrity of its Cdc2p phosphorylation sites. The data suggest that disruption of re-replication control by overexpression of Cdc6p in fission yeast is a consequence of sequestration of Cdc2p and Pop2p, two factors involved in the negative regulation of Rum1p, Cdc18p and potentially other replication proteins.

    View details for Web of Science ID 000083839000010

    View details for PubMedID 10589835

  • Components of an SCE ubiquitin ligase localize to the centrosome and regulate the centrosome duplication cycle GENES & DEVELOPMENT Freed, E., Lacey, K. R., Huie, P., Lyapina, S. A., Deshaies, R. J., Stearns, T., Jackson, P. K. 1999; 13 (17): 2242-2257

    Abstract

    Centrosomes organize the mitotic spindle to ensure accurate segregation of the chromosomes in mitosis. The mechanism that ensures accurate duplication and separation of the centrosomes underlies the fidelity of chromosome segregation, but remains unknown. In Saccharomyces cerevisiae, entry into S phase and separation of spindle pole bodies each require CDC4 and CDC34, which encode components of an SCF (Skp1-cullin-F-box) ubiquitin ligase, but a direct (SCF) connection to the spindle pole body is unknown. Using immunofluorescence microscopy, we show that in mammalian cells the Skp1 protein and the cullin Cul1 are localized to interphase and mitotic centrosomes and to the cytoplasm and nucleus. Deconvolution and immunoelectron microscopy suggest that Skp1 forms an extended pericentriolar structure that may function to organize the centrosome. Purified centrosomes also contain Skp1, and Cul1 modified by the ubiquitin-like molecule NEDD8, suggesting a role for NEDD8 in targeting. Using an in vitro assay for centriole separation in Xenopus extracts, antibodies to Skp1 or Cul1 block separation. Proteasome inhibitors block both centriole separation in vitro and centrosome duplication in Xenopus embryos. We identify candidate centrosomal F-box proteins, suggesting that distinct SCF complexes may direct proteolysis of factors mediating multiple steps in the centrosome cycle.

    View details for Web of Science ID 000082647200006

    View details for PubMedID 10485847

    View details for PubMedCentralID PMC316987

  • F-box/WD-repeat proteins Pop1p and Sud1p/Pop2p form complexes that bind and direct the proteolysis of Cdc18p CURRENT BIOLOGY Wolf, D. A., McKeon, F., Jackson, P. K. 1999; 9 (7): 373-376

    Abstract

    Ubiquitin-dependent proteolysis plays an important role in cell-cycle control [1] [2]. In budding yeast, the protein Skp1p, the cullin-family member Cdc53p, and the F-box/WD-repeat protein Cdc4p form the SCFCdc4p ubiquitin ligase complex, which targets the cyclin-dependent kinase (Cdk) inhibitor Sic1p for proteolysis [3] [4] [5] [6] [7] [8]. Sic1p is recruited to the SCFCdc4p complex by binding to the WD-repeat region of Cdc4p [5] [6], while Skp1p binds to the F-box of Cdc4p [9]. In fission yeast, two distinct Cdc4p-related proteins, Pop1p/Ste16p [10] [11] and the recently identified Sud1p/Pop2p [12], regulate the stability of the replication initiator Cdc18p and the Cdk inhibitor Rum1p. We show here that, despite their structural and functional similarities, the pop1 and pop2 genes fail to complement each other's deletion phenotypes, indicating that they perform non-redundant, but potentially interdependent, functions in proteolysis. Consistent with this hypothesis, Pop1p and Pop2p formed heterooligomeric complexes when overexpressed, and binding of Cdc18p to Pop2p was dependent on Pop1p. The Pop1p-Pop2p interaction was mediated by the amino-terminal domain of Pop2p which, when fused to full-length Pop1p, rescued the phenotype of a Deltapop1Deltapop2 double mutant. Thus, close physical proximity of two distinct F-box/WD-repeat proteins directs proteolysis mediated by the SCFPop ubiquitin ligase complex.

    View details for Web of Science ID 000079732800019

    View details for PubMedID 10209119

  • Cyclin-dependent kinase control of centrosome duplication PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lacey, K. R., Jackson, P. K., Stearns, T. 1999; 96 (6): 2817-2822

    Abstract

    Centrosomes nucleate microtubules and duplicate once per cell cycle. This duplication and subsequent segregation in mitosis results in maintenance of the one centrosome/cell ratio. Centrosome duplication occurs during the G1/S transition in somatic cells and must be coupled to the events of the nuclear cell cycle; failure to coordinate duplication and mitosis results in abnormal numbers of centrosomes and aberrant mitoses. Using both in vivo and in vitro assays, we show that centrosome duplication in Xenopus laevis embryos requires cyclin/cdk2 kinase activity. Injection of the cdk (cyclin-dependent kinase) inhibitor p21 into one blastomere of a dividing embryo blocks centrosome duplication in that blastomere; the related cdk inhibitor p27 has a similar effect. An in vitro system using Xenopus extracts carries out separation of the paired centrioles within the centrosome. This centriole separation activity is dependent on cyclin/cdk2 activity; depletion of either cdk2 or of the two activating cyclins, cyclin A and cyclin E, eliminates centriole separation activity. In addition, centriole separation is inhibited by the mitotic state, suggesting a mechanism of linking the cell cycle to periodic duplication of the centrosome.

    View details for Web of Science ID 000079224500048

    View details for PubMedID 10077594

    View details for PubMedCentralID PMC15852

  • Identification of two cyclin A regulators that control DNA replication in Xenopus egg extracts Regan, J. D., Jackson, P. K. AMER SOC CELL BIOLOGY. 1998: 112A
  • Nuclear transport couples the initiation of DNA replication to nuclear formation Swanson, C., Kaiser, B., Jackson, P. AMER SOC CELL BIOLOGY. 1998: 361A
  • Cell cycle: Oiling the gears of anaphase CURRENT BIOLOGY Wolf, D. A., Jackson, P. K. 1998; 8 (18): R636-R639

    Abstract

    The anaphase-promoting complex (APC) or cyclosome directs the ubiquitination and destruction of proteins that control specific steps in mitosis. Recent studies show that APC activity requires WD40 domain proteins, and that one of these proteins is part of the checkpoint control that ensures accurate chromosome segregation.

    View details for Web of Science ID 000075862000007

    View details for PubMedID 9740795

  • Cell cycle: Cull and destroy CURRENT BIOLOGY Jackson, P. K. 1996; 6 (10): 1209-1212

    Abstract

    A newly discovered family of proteins homologous to yeast Cdc53, called cullins, may play a key role in the targeting of cell-cycle regulators, such as cyclins, for destruction by ubiquitin-dependent proteolysis.

    View details for Web of Science ID A1996VN63500007

    View details for PubMedID 8939566

  • The cytostatic function of c-Abl is controlled by multiple nuclear localization signals and requires the p53 and Rb tumor suppressor gene products EMBO JOURNAL Wen, S. T., Jackson, P. K., VANETTEN, R. A. 1996; 15 (7): 1583-1595

    Abstract

    c-Abl is a non-receptor protein-tyrosine kinase lacking a clear physiological role. A clue to its normal function is suggested by overexpression of Abl in fibroblasts, which leads to inhibition of cell growth. This effect requires tyrosine kinase activity and the Abl C-terminus. c-Abl is localized to the cell nucleus, where it can bind DNA, and interacts with the retinoblastoma protein, a potential mediator of the growth-inhibitory effect. Nuclear localization of Abl can be directed by a pentalysine nuclear localization signal in the Abl C-terminus. Here, we have identified two additional basic motifs in the Abl C-terminus, either of which can function independently of the pentalysine signal to localize Abl to the nucleus. Using a quantitative transfection assay, we show that both c-Abl and transforming Abl proteins inhibit entry into S phase and this effect is absolutely dependent on nuclear localization. Further, we demonstrate that the Abl cytostatic effect requires both the Rb and p53 tumor suppressor gene products. These results indicate that Abl inhibits cell proliferation by interacting with central elements of the cell cycle control apparatus in the nucleus, and suggest a direct connection between p53 and Rb in this growth-inhibitory pathway.

    View details for Web of Science ID A1996UF65800013

    View details for PubMedID 8612582

  • EARLY EVENTS IN DNA-REPLICATION REQUIRE CYCLIN-E AND ARE BLOCKED BY P21(CIP1) JOURNAL OF CELL BIOLOGY Jackson, P. K., Chevalier, S., Philippe, M., Kirschner, M. W. 1995; 130 (4): 755-769

    Abstract

    Using immunodepletion of cyclin E and the inhibitor protein p21WAF/CIP1, we demonstrate that the cyclin E protein, in association with Cdk2, is required for the elongation phase of replication on single-stranded substrates. Although cyclin E/Cdk2 is likely to be the major target by which p21 inhibits the initiation of sperm DNA replication, p21 can inhibit single-stranded replication through a mechanism dependent on PCNA. While the cyclin E/Cdk2 complex appears to have a role in the initiation of DNA replication, another Cdk kinase, possibly cyclin A/Cdk, may be involved in a later step controlling the switch from initiation to elongation. The provision of a large maternal pool of cyclin E protein shows that regulators of replication are constitutively present, which explains the lack of a protein synthesis requirement for replication in the early embryonic cell cycle.

    View details for Web of Science ID A1995RQ21900001

    View details for PubMedID 7642695

  • CELL-CYCLE ARREST BY TYROSINE KINASE ABL INVOLVES ALTERED EARLY MITOGENIC RESPONSE ONCOGENE Mattioni, T., Jackson, P. K., VANHUIJSDUIJNEN, O. B., Picard, D. 1995; 10 (7): 1325-1333

    Abstract

    Activated forms of the nuclear and cytoplasmic tyrosine kinase c-Abl are completely cytoplasmic and oncogenic. The overexpression of c-Abl, and in certain fibroblast cell lines even of v-Abl, leads to a cell cycle arrest revealing an alternative Abl function. To facilitate the analysis of this growth inhibitory function we have taken advantage of regulable Abl-estrogen receptor (ABL:ER) fusion proteins. Oncogenic in the presence of estrogen, they are reversibly switched to inhibit cell proliferation upon removal of hormone. Using this system, we demonstrate that inhibition is effected by Abl derivatives which we have previously shown to be hypo-phosphorylated and to have low kinase activity. Since an almost exclusively cytoplasmic ABL:ER protein is fully growth inhibitory, relevant interactions may occur in the cytoplasm. We identify the cell cycle arrest as an early G1 or G0-like block. Interestingly, growth inhibition correlates with an altered expression pattern of early serum response genes; c-Jun mRNA and c-Fos protein levels are elevated in Abl-blocked cells. In view of the two functional modes of overexpressed Abl proteins, one can speculate that normal c-Abl may be involved in relaying growth regulatory signals from the membrane to the nucleus.

    View details for Web of Science ID A1995QR65100009

    View details for PubMedID 7731683

  • SEPARATE DOMAINS OF P21 INVOLVED IN THE INHIBITION OF CDK KINASE AND PCNA NATURE Chen, J. J., Jackson, P. K., Kirschner, M. W., Dutta, A. 1995; 374 (6520): 386-388

    Abstract

    The protein p21 (WAF1, CIP1 or sdi1), induced by the tumour-suppressor protein p53, interacts with and inhibits two different targets essential for cell-cycle progression. One of these is the cyclin-Cdk family of kinases and the other is the essential DNA replication factor, proliferating-cell nuclear antigen (PCNA). We report here that separate domains of p21 are responsible for interacting with and inhibiting the two targets. An amino-terminal domain inhibits cyclin-Cdk kinases and a carboxy-terminal domain inhibits PCNA. Using these separated domains, we have determined that p21 inhibits different biological systems through different targets. The PCNA-binding domain is sufficient for inhibition of DNA replication based on simian virus 40, whereas the Cdk2-binding domain is sufficient for inhibition of DNA replication based on Xenopus egg extract and for growth suppression in transformed human cells.

    View details for Web of Science ID A1995QN63000067

    View details for PubMedID 7885482

  • MITOSIS IN TRANSITION CELL King, R. W., Jackson, P. K., Kirschner, M. W. 1994; 79 (4): 563-571

    View details for Web of Science ID A1994PT48100004

    View details for PubMedID 7954823

  • THE COOH TERMINUS OF THE C-ABL TYROSINE KINASE CONTAINS DISTINCT F-ACTIN AND G-ACTIN BINDING DOMAINS WITH BUNDLING ACTIVITY JOURNAL OF CELL BIOLOGY VANETTEN, R. A., Jackson, P. K., Baltimore, D., Sanders, M. C., Matsudaira, P. T., JANMEY, P. A. 1994; 124 (3): 325-340

    Abstract

    The myristoylated form of c-Abl protein, as well as the P210bcr/abl protein, have been shown by indirect immunofluorescence to associate with F-actin stress fibers in fibroblasts. Analysis of deletion mutants of c-Abl stably expressed in fibroblasts maps the domain responsible for this interaction to the extreme COOH-terminus of Abl. This domain mediates the association of a heterologous protein with F-actin filaments after microinjection into NIH 3T3 cells, and directly binds to F-actin in a cosedimentation assay. Microinjection and cosedimentation assays localize the actin-binding domain to a 58 amino acid region, including a charged motif at the extreme COOH-terminus that is important for efficient binding. F-actin binding by Abl is calcium independent, and Abl competes with gelsolin for binding to F-actin. In addition to the F-actin binding domain, the COOH-terminus of Abl contains a proline-rich region that mediates binding and sequestration of G-actin, and the Abl F- and G-actin binding domains cooperate to bundle F-actin filaments in vitro. The COOH terminus of Abl thus confers several novel localizing functions upon the protein, including actin binding, nuclear localization, and DNA binding. Abl may modify and receive signals from the F-actin cytoskeleton in vivo, and is an ideal candidate to mediate signal transduction from the cell surface and cytoskeleton to the nucleus.

    View details for Web of Science ID A1994MT69900010

    View details for PubMedID 8294516

  • HORMONE-CONDITIONAL TRANSFORMATION BY FUSION PROTEINS OF C-ABL AND ITS TRANSFORMING VARIANTS EMBO JOURNAL Jackson, P., Baltimore, D., Picard, D. 1993; 12 (7): 2809-2819

    Abstract

    Fusion of the hormone binding domain (HBD) of steroid receptors to transcription factors renders them hormone-dependent. We show here that an SH3-deleted, oncogenic variant of the Abl tyrosine kinase becomes hormone-dependent for transformation by fusion to the estrogen receptor (ER) HBD, extending the phenomenon to tyrosine kinases. Surprisingly, fusion of the HBD to the normal, non-transforming c-Abl (IV) protein activated transforming activity in a hormone-dependent fashion. In the presence of hormone, the c-Abl:ER fusion protein was transforming, cytoplasmic and tyrosine phosphorylated, whereas it was non-transforming, nuclear and hypophosphorylated without hormone. We have examined the kinetics of activation of the c-Abl:ER protein and found that protein synthesis is required both for kinase activation and for redistribution of the c-Abl:ER protein from the nucleus to the cytoplasm. We suggest that the activation of c-Abl could be due to HBD-mediated dimerization and/or to the ability to overexpress conditionally the normally toxic c-Abl protein. This novel approach may be applicable to a wide variety of proteins, particularly when activating mutations or physiological inducers are unknown or when the protein is toxic to cells.

    View details for Web of Science ID A1993LK36400025

    View details for PubMedID 8334997

  • MUTATION OF A PHENYLALANINE CONSERVED IN SH3-CONTAINING TYROSINE KINASES ACTIVATES THE TRANSFORMING ABILITY OF C-ABL ONCOGENE Jackson, P. K., Paskind, M., Baltimore, D. 1993; 8 (7): 1943-1956

    Abstract

    c-abl is the normal cellular homolog of the v-abl transforming gene of Abelson murine leukemia virus. By constructing recombinants between c- and v-abl retroviruses, we show that a point mutation in c-Abl is sufficient to change the myristoylated form of c-Abl into a protein able to transform fibroblasts, but not capable of transforming bone marrow or inducing Abelson disease. This activating mutation, which changes the phenylalanine at amino acid 420 to valine (F420V) found in the homologous position of v-Abl, is positioned outside of the SH3 domain, a region typically modified in transforming alleles of abl. Phenylalanine 420 is perfectly conserved among tyrosine kinases with N-terminal SH3 domains (the Src and Abl families). The equivalent position in other protein tyrosine kinases is a conserved hydrophobic residue that predicts the specific family to which that kinase belongs. Mutation of phenylalanine 420 to other hydrophobic residues activates c-Abl. Unlike other transforming variants of Abl, the F420V mutant protein is not highly phosphorylated on tyrosine. Mutation of the nearby proposed autophosphorylation site, tyrosine 412, shows that this tyrosine is not strictly required for fibroblast transformation in either F420V or SH3-deleted variants of c-Abl (IV).

    View details for Web of Science ID A1993LG68200027

    View details for PubMedID 8510937

  • NONMYRISTOYLATED ABL PROTEINS TRANSFORM A FACTOR-DEPENDENT HEMATOPOIETIC-CELL LINE MOLECULAR AND CELLULAR BIOLOGY Daley, G. Q., VANETTEN, R. A., Jackson, P. K., Bernards, A., Baltimore, D. 1992; 12 (4): 1864-1871

    Abstract

    N-terminal myristoylation can promote the association of proteins with the plasma membrane, a property that is required for oncogenic variants of Src and Abl to transform fibroblastic cell types. The P210bcr/abl protein of chronic myelogenous leukemia cells is not myristoylated and does not stably transform NIH 3T3 fibroblasts; however, it will transform lymphoid and myeloid cell types in vitro and in vivo, suggesting that myristoylation is not required for Abl variants to transform hematopoietic cells. To test this hypothesis, we introduced point mutations that disrupt myristoylation into two activated Abl proteins, v-Abl and a deletion mutant of c-Abl (delta XB), and examined their ability to transform an interleukin-3-dependent lymphoblastoid cell line, Ba/F3. Neither of the nonmyristoylated Abl proteins transformed NIH 3T3 fibroblasts, but like P210bcr/abl, both were capable of transforming the Ba/F3 cells to factor independence and tumorigenicity. Nonmyristoylated Abl variants did not associate with the plasma membrane in the transformed Ba/F3 cells. These results demonstrate that Abl proteins can transform hematopoietic cells in the absence of membrane association and suggest that distinct functions of Abl are required for transformation of fibroblast and hematopoietic cell types.

    View details for Web of Science ID A1992HK75100050

    View details for PubMedID 1549131

  • POINT MUTATIONS IN THE ABL SH2 DOMAIN COORDINATELY IMPAIR PHOSPHOTYROSINE BINDING INVITRO AND TRANSFORMING ACTIVITY INVIVO MOLECULAR AND CELLULAR BIOLOGY Mayer, B. J., Jackson, P. K., VANETTEN, R. A., Baltimore, D. 1992; 12 (2): 609-618

    Abstract

    We have constructed a series of point mutations in the highly conserved FLVRES motif of the src homology 2 (SH2) domain of the abl tyrosine kinase. Mutant SH2 domains were expressed in bacteria, and their ability to bind to tyrosine-phosphorylated proteins was examined in vitro. Three mutants were greatly reduced in their ability to bind both phosphotyrosine itself and tyrosine-phosphorylated cellular proteins. All of the mutants that retained activity bound to the same set of tyrosine-phosphorylated proteins as did the wild type, suggesting that binding specificity was unaffected. These results implicate the FLVRES motif in direct binding to phosphotyrosine. When the mutant SH2 domains were inserted into an activated abl kinase and expressed in murine fibroblasts, decreased in vitro phosphotyrosine binding correlated with decreased transforming ability. This finding implies that SH2-phosphotyrosine interactions are involved in transmission of positive growth signals by the nonreceptor tyrosine kinases, most likely via the assembly of multiprotein complexes with other tyrosine-phosphorylated proteins.

    View details for Web of Science ID A1992HB06600017

    View details for PubMedID 1370711

  • NEONATAL LETHALITY AND LYMPHOPENIA IN MICE WITH A HOMOZYGOUS DISRUPTION OF THE C-ABL PROTOONCOGENE CELL Tybulewicz, V. L., Crawford, C. E., Jackson, P. K., Bronson, R. T., Mulligan, R. C. 1991; 65 (7): 1153-1163

    Abstract

    The c-abl proto-oncogene, which encodes a cytoplasmic protein-tyrosine kinase, is expressed throughout murine gestation and ubiquitously in adult mouse tissues. However, its levels are highest in thymus, spleen, and testes. To examine the in vivo role of c-abl, the gene was disrupted in embryonic stem cells, and the resulting genetically modified cells were used to establish a mouse strain carrying the mutation. Most mice homozygous for the c-abl mutation became runted and died 1 to 2 weeks after birth. In addition, many showed thymic and splenic atrophy and a T and B cell lymphopenia.

    View details for Web of Science ID A1991FU89900008

    View details for PubMedID 2065352

  • THE NONCATALYTIC SRC HOMOLOGY REGION-2 SEGMENT OF ABL TYROSINE KINASE BINDS TO TYROSINE-PHOSPHORYLATED CELLULAR PROTEINS WITH HIGH-AFFINITY PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Mayer, B. J., Jackson, P. K., Baltimore, D. 1991; 88 (2): 627-631

    Abstract

    Several proteins implicated in the regulation of cell proliferation contain a common noncatalytic domain, src homology region 2 (SH2). We have used the bacterially expressed SH2 domain of abl protein-tyrosine kinase to evaluate the ability of this domain to bind to cellular proteins. ablSH2 specifically bound to a number of tyrosine-phosphorylated proteins from cells transformed by tyrosine kinase oncogenes in a filter-binding assay and to a subset of those proteins in solution. The SH2 probe bound almost exclusively to tyrosine-phosphorylated proteins, and binding was eliminated by dephosphorylation of cell proteins. Free phosphotyrosine could partially disrupt SH2 binding, suggesting that phosphotyrosine is directly involved in the binding interaction. These results demonstrate that an SH2 domain is sufficient to confer direct, high-affinity phosphotyrosine-dependent binding to proteins and suggest a general role for SH2 domains in cellular signaling pathways.

    View details for Web of Science ID A1991ET52100065

    View details for PubMedID 1703304

  • THE MOUSE TYPE-IV C-ABL GENE-PRODUCT IS A NUCLEAR-PROTEIN, AND ACTIVATION OF TRANSFORMING ABILITY IS ASSOCIATED WITH CYTOPLASMIC LOCALIZATION CELL VANETTEN, R. A., Jackson, P., Baltimore, D. 1989; 58 (4): 669-678

    Abstract

    The subcellular localization of the mouse type IV c-abl protein was determined by indirect immunofluorescence of nontransformed NIH 3T3 fibroblasts that overexpress the protein. Unlike the viral transforming protein p160gag/v-abl, which has cytoplasmic and plasma membrane localization, a large fraction of the c-abl (IV) protein is nuclear, with the remainder in the cytoplasm and plasma membrane. Deletion of a small N-terminal regulatory region of the c-abl (IV) protein, sufficient to activate its transforming potential fully, changes the distribution of the protein from the nucleus to the cytoplasm. Mapping of an amino acid sequence responsible for the nuclear localization of the c-abl (IV) protein reveals a nuclear localization signal similar to that of SV40 large T antigen.

    View details for Web of Science ID A1989AM54700010

    View details for PubMedID 2670246

  • N-TERMINAL MUTATIONS ACTIVATE THE LEUKEMOGENIC POTENTIAL OF THE MYRISTOYLATED FORM OF C-ABL EMBO JOURNAL Jackson, P., Baltimore, D. 1989; 8 (2): 449-456

    Abstract

    The two major forms of the c-abl gene differ from their activated counterpart, the v-abl oncogene of the Abelson murine leukemia virus by the replacement of their N-terminal sequences with viral gag sequences. Overexpression of p150c-abl type IV in a retroviral vector similar to Abelson virus does not transform NIH 3T3 fibroblasts, even though it is expressed and myristoylated at levels comparable to pp160v-abl. Members of a nested set of deletion mutations of the N-terminus of c-abl type IV in this expression system will activate abl to transform murine fibroblasts. The smallest of these deletions, delta XB, efficiently transforms lymphoid cells in vitro and causes leukemia in vivo demonstrating that gag sequences are not necessary for abl-induced leukemogenesis. The delta XB mutation defines an N-terminal regulatory domain, which shares a surprising homology with chicken oncogene v-crk and phospholipase C-II. Although overexpression of the myristoylated form of c-abl does not transform cells, it nonetheless has a profound effect on cell growth.

    View details for Web of Science ID A1989T348700016

    View details for PubMedID 2542016