Professional Education


  • Doctor of Philosophy, University of Chicago (2021)
  • Master of Science, University of Chicago (2016)
  • PhD, University of Chicago, Cancer Biology (2021)
  • Master of Science, University of Chicago, Translational Research (2016)
  • Bachelor of Art, Vanderbilt University, Molecular and Cellular Biology Economics (2013)

Stanford Advisors


Lab Affiliations


All Publications


  • BET inhibitors enhance embryonic and fetal globin expression in erythroleukemia cell lines HAEMATOLOGICA Cao, J. Z., Bigelow, K., Wickrema, A., Godley, L. A. 2021; 106 (12): 3223-3227

    View details for DOI 10.3324/haematol.2021.278791

    View details for Web of Science ID 000732456400027

    View details for PubMedID 34435481

    View details for PubMedCentralID PMC8634195

  • RBL2 bi-allelic truncating variants cause severe motor and cognitive impairment without evidence for abnormalities in DNA methylation or telomeric function JOURNAL OF HUMAN GENETICS Samra, N., Toubiana, S., Yttervik, H., Tzur-Gilat, A., Morani, I., Itzkovich, C., Giladi, L., Abu Jabal, K., Cao, J. Z., Godley, L. A., Mory, A., Baris Feldman, H., Tveten, K., Selig, S., Weiss, K. 2021; 66 (11): 1101-1112

    Abstract

    RBL2/p130, a member of the retinoblastoma family of proteins, is a key regulator of cell division and propagates irreversible senescence. RBL2/p130 is also involved in neuronal differentiation and survival, and eliminating Rbl2 in certain mouse strains leads to embryonic lethality accompanied by an abnormal central nervous system (CNS) phenotype. Conflicting reports exist regarding a role of RBL2/p130 in transcriptional regulation of DNA methyltransferases (DNMTs), as well as the control of telomere length. Here we describe the phenotype of three patients carrying bi-allelic RBL2-truncating variants. All presented with infantile hypotonia, severe developmental delay and microcephaly. Malignancies were not reported in carriers or patients. Previous studies carried out on mice and human cultured cells, associated RBL2 loss to DNA methylation and telomere length dysregulation. Here, we investigated whether patient cells lacking RBL2 display related abnormalities. The study of primary patient fibroblasts did not detect abnormalities in expression of DNMTs. Furthermore, methylation levels of whole genome DNA, and specifically of pericentromeric repeats and subtelomeric regions, were unperturbed. RBL2-null fibroblasts show no evidence for abnormal elongation by telomeric recombination. Finally, gradual telomere shortening, and normal onset of senescence were observed following continuous culturing of RBL2-mutated fibroblasts. Thus, this study resolves uncertainties regarding a potential non-redundant role for RBL2 in DNA methylation and telomere length regulation, and indicates that loss of function variants in RBL2 cause a severe autosomal recessive neurodevelopmental disorder in humans.

    View details for DOI 10.1038/s10038-021-00931-z

    View details for Web of Science ID 000650066100001

    View details for PubMedID 33980986

  • HIF-1 directly induces TET3 expression to enhance 5-hmC density and induce erythroid gene expression in hypoxia BLOOD ADVANCES Cao, J. Z., Liu, H., Wickrema, A., Godley, L. A. 2020; 4 (13): 3053-3062

    Abstract

    In mammalian cells, cytosines found within cytosine guanine dinucleotides can be methylated to 5-methylcytosine (5-mC) by DNA methyltransferases and further oxidized by the Ten-eleven translocation dioxygenase (TET) enzymes to 5-hydroxymethylcytosine (5-hmC). We have previously shown that hematopoietic stem and progenitor cells (HSPCs) with TET2 mutations have aberrant 5-hmC distribution and less erythroid differentiation potential. However, these experiments were performed under standard tissue culture conditions with 21% oxygen (O2), whereas HSPCs in human bone marrow reside in ∼1% O2. Therefore, to model human erythropoiesis more accurately, we compared 5-hmC distribution and gene expression in hypoxic vs normoxic conditions. Despite TET enzymes having limited O2 as a substrate in hypoxia, 5-hmC peaks were more numerous and pronounced than in normoxia. Among the TET genes, TET3 was upregulated specifically in hypoxia. We identified 2 HIF-1 binding sites in TET3 by chromatin immunoprecipitation of HIF-1α followed by sequencing, and TET3 upregulation was abrogated with deletion of both sites, indicating that TET3 is a direct HIF-1 target. Finally, we showed that loss of one or both of these HIF-1 binding sites in K562 cells disrupted erythroid differentiation in hypoxia and lowered cell viability. This work provides a molecular link between O2 availability, epigenetic modification of chromatin, and erythroid differentiation.

    View details for DOI 10.1182/bloodadvances.2020001535

    View details for Web of Science ID 000551200600023

    View details for PubMedID 32634239

    View details for PubMedCentralID PMC7362358

  • MYC Regulation of D2HGDH and L2HGDH Influences the Epigenome and Epitranscriptome CELL CHEMICAL BIOLOGY Qiu, Z., Lin, A., Jiang, S., Elkashef, S. M., Myers, J., Srikantan, S., Sasi, B., Cao, J. Z., Godley, L. A., Rakheja, D., Lyu, Y., Zheng, S., Madesh, M., Shiio, Y., Dahia, P. M., Aguiar, R. T. 2020; 27 (5): 538-+

    Abstract

    Mitochondrial D2HGDH and L2HGDH catalyze the oxidation of D-2-HG and L-2-HG, respectively, into αKG. This contributes to cellular homeostasis in part by modulating the activity of αKG-dependent dioxygenases. Signals that control the expression/activity of D2HGDH/L2HGDH are presumed to broadly influence physiology and pathology. Using cell and mouse models, we discovered that MYC directly induces D2HGDH and L2HGDH transcription. Furthermore, in a manner suggestive of D2HGDH, L2HGDH, and αKG dependency, MYC activates TET enzymes and RNA demethylases, and promotes their nuclear localization. Consistent with these observations, in primary B cell lymphomas MYC expression positively correlated with enhancer hypomethylation and overexpression of lymphomagenic genes. Together, these data provide additional evidence for the role of mitochondria metabolism in influencing the epigenome and epitranscriptome, and imply that in specific contexts wild-type TET enzymes could demethylate and activate oncogenic enhancers.

    View details for DOI 10.1016/j.chembiol.2020.02.002

    View details for Web of Science ID 000536032400007

    View details for PubMedID 32101699

    View details for PubMedCentralID PMC7714266

  • Mitochondrial superoxide disrupts the metabolic and epigenetic landscape of CD4+ and CD8+ T-lymphocytes REDOX BIOLOGY Moshfegh, C. M., Collins, C. W., Gunda, V., Vasanthakumar, A., Cao, J. Z., Singh, P. K., Godley, L. A., Case, A. J. 2019; 27: 101141

    Abstract

    While the role of mitochondrial metabolism in controlling T-lymphocyte activation and function is becoming more clear, the specifics of how mitochondrial redox signaling contributes to T-lymphocyte regulation remains elusive. Here, we examined the global effects of elevated mitochondrial superoxide (O2-) on T-lymphocyte activation using a novel model of inducible manganese superoxide dismutase (MnSOD) knock-out. Loss of MnSOD led to specific increases in mitochondrial O2- with no evident changes in hydrogen peroxide (H2O2), peroxynitrite (ONOO-), or copper/zinc superoxide dismutase (CuZnSOD) levels. Unexpectedly, both mitochondrial and glycolytic metabolism showed significant reductions in baseline, maximal capacities, and ATP production with increased mitochondrial O2- levels. MnSOD knock-out T-lymphocytes demonstrated aberrant activation including widespread dysregulation in cytokine production and increased cellular apoptosis. Interestingly, an elevated proliferative signature defined by significant upregulation of cell cycle regulatory genes was also evident in MnSOD knock-out T-lymphocytes, but these cells did not show accelerated proliferative rates. Global disruption in T-lymphocyte DNA methylation and hydroxymethylation was also observed with increased mitochondrial O2-, which was correlated to alterations in intracellular metabolite pools linked to the methionine cycle. Together, these results demonstrate a mitochondrial redox and metabolic couple that when disrupted may alter cellular processes necessary for proper T-lymphocyte activation.

    View details for DOI 10.1016/j.redox.2019.101141

    View details for Web of Science ID 000496261300003

    View details for PubMedID 30819616

    View details for PubMedCentralID PMC6859572

  • Cytokine-Regulated Phosphorylation and Activation of TET2 by JAK2 in Hematopoiesis. Cancer discovery Jeong, J. J., Gu, X., Nie, J., Sundaravel, S., Liu, H., Kuo, W. L., Bhagat, T. D., Pradhan, K., Cao, J., Nischal, S., McGraw, K. L., Bhattacharyya, S., Bishop, M. R., Artz, A., Thirman, M. J., Moliterno, A., Ji, P., Levine, R. L., Godley, L. A., Steidl, U., Bieker, J. J., List, A. F., Saunthararajah, Y., He, C., Verma, A., Wickrema, A. 2019; 9 (6): 778-795

    Abstract

    Even though the Ten-eleven translocation (TET) enzymes catalyze the generation of 5-hydroxymethylcytosines required for lineage commitment and subsequent differentiation of stem cells into erythroid cells, the mechanisms that link extracellular signals to TET activation and DNA hydroxymethylation are unknown. We demonstrate that hematopoietic cytokines phosphorylate TET2, leading to its activation in erythroid progenitors. Specifically, cytokine receptor-associated JAK2 phosphorylates TET2 at tyrosines 1939 and 1964. Phosphorylated TET2 interacts with the erythroid transcription factor KLF1, and this interaction with TET2 is increased upon exposure to erythropoietin. The activating JAK2V617F mutation seen in myeloproliferative disease patient samples and in mouse models is associated with increased TET activity and cytosine hydroxymethylation as well as genome-wide loss of cytosine methylation. These epigenetic and functional changes are also associated with increased expression of several oncogenic transcripts. Thus, we demonstrate that JAK2-mediated TET2 phosphorylation provides a mechanistic link between extracellular signals and epigenetic changes during hematopoiesis. SIGNIFICANCE: Identification of TET2 phosphorylation and activation by cytokine-stimulated JAK2 links extracellular signals to chromatin remodeling during hematopoietic differentiation. This provides potential avenues to regulate TET2 function in the context of myeloproliferative disorders and myelodysplastic syndromes associated with the JAK2V617F-activating mutation.This article is highlighted in the In This Issue feature, p. 681.

    View details for DOI 10.1158/2159-8290.CD-18-1138

    View details for PubMedID 30944118

    View details for PubMedCentralID PMC6697164

  • Regulation of 5-Hydroxymethylcytosine Distribution by the TET Enzymes DNA, RNA, AND HISTONE METHYLOMES Cao, J. Z., Hains, A. E., Godley, L. A., Jurga, S., Barciszewski, J. 2019: 229-263
  • Fumarate and Succinate Regulate Expression of Hypoxia-inducible Genes via TET Enzymes JOURNAL OF BIOLOGICAL CHEMISTRY Laukka, T., Mariani, C. J., Ihantola, T., Cao, J. Z., Hokkanen, J., Kaelin, W. G., Godley, L. A., Koivunen, P. 2016; 291 (8): 4256-4265

    Abstract

    The TET enzymes are members of the 2-oxoglutarate-dependent dioxygenase family and comprise three isoenzymes in humans: TETs 1-3. These TETs convert 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC) in DNA, and high 5-hmC levels are associated with active transcription. The importance of the balance in these modified cytosines is emphasized by the fact that TET2 is mutated in several human cancers, including myeloid malignancies such as acute myeloid leukemia (AML). We characterize here the kinetic and inhibitory properties of Tets and show that the Km value of Tets 1 and 2 for O2 is 30 μm, indicating that they retain high activity even under hypoxic conditions. The AML-associated mutations in the Fe(2+) and 2-oxoglutarate-binding residues increased the Km values for these factors 30-80-fold and reduced the Vmax values. Fumarate and succinate, which can accumulate to millimolar levels in succinate dehydrogenase and fumarate hydratase-mutant tumors, were identified as potent Tet inhibitors in vitro, with IC50 values ∼400-500 μm. Fumarate and succinate also down-regulated global 5-hmC levels in neuroblastoma cells and the expression levels of some hypoxia-inducible factor (HIF) target genes via TET inhibition, despite simultaneous HIFα stabilization. The combination of fumarate or succinate treatment with TET1 or TET3 silencing caused differential effects on the expression of specific HIF target genes. Altogether these data show that hypoxia-inducible genes are regulated in a multilayered manner that includes epigenetic regulation via TETs and 5-hmC levels in addition to HIF stabilization.

    View details for DOI 10.1074/jbc.M115.688762

    View details for Web of Science ID 000371338400048

    View details for PubMedID 26703470

    View details for PubMedCentralID PMC4759199