Bio


Claude M. Nagamine, DVM, PhD Associate Professor received his D.V.M. from the University of Tennessee in 2004 and completed his residency training in Laboratory Animal Medicine at the Massachusetts Institute of Technology in 2007. He joined the Department of Comparative Medicine at Stanford in 2008. Prior to entering veterinary school, Dr. Nagamine obtained a Ph.D. in Ecology from the University of California, Davis (1979), obtained postdoctoral training in endocrinology, developmental genetics, immunology, and molecular biology of the mouse at the Memorial Sloan-Kettering Cancer Center (NYC), Institut Pasteur (France), and the Howard Hughes Medical Institute at the University of California, San Francisco and was an Assistant Professor of Cell Biology at the Vanderbilt University School of Medicine. His research focuses on using mouse models to study murine and human infectious diseases. These colloborative studies include dengue virus, zika virus, adeno-associated virus, coxsackie virus, enterovirus 71, enterohepatic helicobacters, campylobacters, and anaplasma.

Academic Appointments


Professional Education


  • D.V.M., University of Tennessee, Veterinary Medicine (2004)
  • Ph.D., University of California, Davis, Ecology (1979)
  • M.A., University of California, Davis, Zoology (1975)
  • B.S., University of Hawaii, Manoa, Biology (1973)

Current Research and Scholarly Interests


Mouse models to study murine and human infectious diseases. These colloborative studies include dengue virus, zika virus, adeno-associated virus, coxsackie virus, enterovirus 71, enterohepatic helicobacters, campylobacters, and anaplasma.

2023-24 Courses


Graduate and Fellowship Programs


All Publications


  • Hardwiring tissue-specific AAV transduction in mice through engineered receptor expression. Nature methods Zengel, J., Wang, Y. X., Seo, J. W., Ning, K., Hamilton, J. N., Wu, B., Raie, M., Holbrook, C., Su, S., Clements, D. R., Pillay, S., Puschnik, A. S., Winslow, M. M., Idoyaga, J., Nagamine, C. M., Sun, Y., Mahajan, V. B., Ferrara, K. W., Blau, H. M., Carette, J. E. 2023

    Abstract

    The development of transgenic mouse models that express genes of interest in specific cell types has transformed our understanding of basic biology and disease. However, generating these models is time- and resource-intensive. Here we describe a model system, SELective Expression and Controlled Transduction In Vivo (SELECTIV), that enables efficient and specific expression of transgenes by coupling adeno-associated virus (AAV) vectors with Cre-inducible overexpression of the multi-serotype AAV receptor, AAVR. We demonstrate that transgenic AAVR overexpression greatly increases the efficiency of transduction of many diverse cell types, including muscle stem cells, which are normally refractory to AAV transduction. Superior specificity is achieved by combining Cre-mediated AAVR overexpression with whole-body knockout of endogenous Aavr, which is demonstrated in heart cardiomyocytes, liver hepatocytes and cholinergic neurons. The enhanced efficacy and exquisite specificity of SELECTIV has broad utility in development of new mouse model systems and expands the use of AAV for gene delivery in vivo.

    View details for DOI 10.1038/s41592-023-01896-x

    View details for PubMedID 37291262

    View details for PubMedCentralID 3337962

  • Variant enterovirus A71 found in immune-suppressed patient binds to heparan sulfate and exhibits neurotropism in B-cell-depleted mice. Cell reports Weng, K., Tee, H. K., Tseligka, E. D., Cagno, V., Mathez, G., Rosset, S., Nagamine, C. M., Sarnow, P., Kirkegaard, K., Tapparel, C. 2023; 42 (4): 112389

    Abstract

    Enterovirus A71 (EV-A71) causes hand, foot, and mouth disease outbreaks with neurological complications and deaths. We previously isolated an EV-A71 variant in the stool, cerebrospinal fluid, and blood of an immunocompromised patient who had a leucine-to-arginine substitution on the VP1 capsid protein, resulting in increased heparin sulfate binding. We show here that this mutation increases the virus's pathogenicity in orally infected mice with depleted B cells, which mimics the patient's immune status, and increases susceptibility to neutralizing antibodies. However, a double mutant with even greater heparin sulfate affinity is not pathogenic, suggesting that increased heparin sulfate affinity may trap virions in peripheral tissues and reduce neurovirulence. This research sheds light on the increased pathogenicity of variant with heparin sulfate (HS)-binding ability in individuals with decreased B cell immunity.

    View details for DOI 10.1016/j.celrep.2023.112389

    View details for PubMedID 37058406

  • The human disease gene LYSET is essential for lysosomal enzyme transport and viral infection. Science (New York, N.Y.) Richards, C. M., Jabs, S., Qiao, W., Varanese, L. D., Schweizer, M., Mosen, P. R., Riley, N. M., Klüssendorf, M., Zengel, J. R., Flynn, R. A., Rustagi, A., Widen, J. C., Peters, C. E., Ooi, Y. S., Xie, X., Shi, P. Y., Bartenschlager, R., Puschnik, A. S., Bogyo, M., Bertozzi, C. R., Blish, C. A., Winter, D., Nagamine, C. M., Braulke, T., Carette, J. E. 2022: eabn5648

    Abstract

    Lysosomes are key degradative compartments of the cell. Transport to lysosomes relies on GlcNAc-1-phosphotransferase-mediated tagging of soluble enzymes with mannose 6-phosphate (M6P). GlcNAc-1-phosphotransferase deficiency leads to the severe lysosomal storage disorder mucolipidosis II (MLII). Several viruses require lysosomal cathepsins to cleave structural proteins and thus depend on functional GlcNAc-1-phosphotransferase. Here, we used genome-scale CRISPR screens to identify Lysosomal Enzyme Trafficking factor (LYSET) as essential for infection by cathepsin-dependent viruses including SARS-CoV-2. LYSET deficiency resulted in global loss of M6P tagging and mislocalization of GlcNAc-1-phosphotransferase from the Golgi complex to lysosomes. Lyset knockout mice exhibited MLII-like phenotypes and human pathogenic LYSET alleles failed to restore lysosomal sorting defects. Thus, LYSET is required for correct functioning of the M6P trafficking machinery, and mutations in LYSET can explain the phenotype of the associated disorder.

    View details for DOI 10.1126/science.abn5648

    View details for PubMedID 36074821

  • Efficacy of 3 Buprenorphine Formulations for the Attenuation of Hypersensitivity after Plantar Incision in Immunodeficient NSG Mice. Journal of the American Association for Laboratory Animal Science : JAALAS Arthur, J. D., Alamaw, E. D., Jampachairsri, K., Sharp, P., Nagamine, C. M., Huss, M. K., Pacharinsak, C. 2022

    Abstract

    Buprenorphine is perhaps the most prescribed analgesic for management of postoperative pain in mice. Although various buprenorphine formulations are effective in commonly used immunocompetent mouse strains, a knowledge gap exists regarding its efficacy in immunodeficient mice. Here we used a plantar incision to evaluate the efficacy of 3 buprenorphine formulations for attenuating postoperative mechanical and thermal hypersensitivity in the immunodeficient NSG mouse strain. We also characterized the pharmacokinetics of these formulations over a 72-h period. We hypothesized that all 3 buprenorphine formulations evaluated-the standard preparation and 2 extended-release products (Bup-HCl, Bup-ER, and Bup-XR, respectively)-would attenuate postoperative mechanical and thermal hypersensitivity resulting from a plantar incision in NSG mice. Male and female NSG mice (n = 48) were allocated to 4 treatment groups: saline (0.9% NaCl, 5 mL/kg SC once); Bup-HCl (0.1 mg/kg SC, BID for 2 d); Bup-ER (1.0 mg/kg SC once); and Bup-XR (3.25 mg/kg SC once). Mechanical and thermal hypersensitivity assessments were conducted 24 h before surgery and at 4, 8, 24, 48, and 72 h afterward. All groups of mice showed mechanical and thermal hypersensitivity within the first 24 h after surgery. Behavioral pain indicators (guarding, toe-touching [intermittent partial weight bearing], licking the incision, vocalizations) were observed in some mice from each group at every postoperative time point. Plasma buprenorphine was measured in a separate group of mice and concentrations surpassed the suggested therapeutic level (1.0 ng/mL) for less than 4 h for Bup-HCl, for at least 24 h for Bup-ER, and for 72 h for Bup-XR. Our results indicate that at the dosages studied, these buprenorphine formulations do not adequately attenuate postoperative mechanical and thermal hypersensitivity in the plantar incisional model in NSG mice. These findings support the need for strain-specific analgesic protocols for mice used in research.

    View details for DOI 10.30802/AALAS-JAALAS-22-000058

    View details for PubMedID 36068076

  • A Targeted Computational Screen of the SWEETLEAD Database Reveals FDA-Approved Compounds with Anti-Dengue Viral Activity. mBio Moshiri, J., Constant, D. A., Liu, B., Mateo, R., Kearnes, S., Novick, P., Prasad, R., Nagamine, C., Pande, V., Kirkegaard, K. 2020; 11 (6)

    Abstract

    Affordable and effective antiviral therapies are needed worldwide, especially against agents such as dengue virus that are endemic in underserved regions. Many antiviral compounds have been studied in cultured cells but are unsuitable for clinical applications due to pharmacokinetic profiles, side effects, or inconsistent efficacy across dengue serotypes. Such tool compounds can, however, aid in identifying clinically useful treatments. Here, computational screening (Rapid Overlay of Chemical Structures) was used to identify entries in an in silico database of safe-in-human compounds (SWEETLEAD) that display high chemical similarities to known inhibitors of dengue virus. Inhibitors of the dengue proteinase NS2B/3, the dengue capsid, and the host autophagy pathway were used as query compounds. Three FDA-approved compounds that resemble the tool molecules structurally, cause little toxicity, and display strong antiviral activity in cultured cells were selected for further analysis. Pyrimethamine (50% inhibitory concentration [IC50] = 1.2muM), like the dengue proteinase inhibitor ARDP0006 to which it shows structural similarity, inhibited intramolecular NS2B/3 cleavage. Lack of toxicity early in infection allowed testing in mice, in which pyrimethamine also reduced viral loads. Niclosamide (IC50 = 0.28muM), like dengue core inhibitor ST-148, affected structural components of the virion and inhibited early processes during infection. Vandetanib (IC50 = 1.6muM), like cellular autophagy inhibitor spautin-1, blocked viral exit from cells and could be shown to extend survival in vivo Thus, three FDA-approved compounds with promising utility for repurposing to treat dengue virus infections and their potential mechanisms were identified using computational tools and minimal phenotypic screening.IMPORTANCE No antiviral therapeutics are currently available for dengue virus infections. By computationally overlaying the three-dimensional (3D) chemical structures of compounds known to inhibit dengue virus over those of compounds known to be safe in humans, we identified three FDA-approved compounds that are attractive candidates for repurposing as antivirals. We identified targets for two previously identified antiviral compounds and revealed a previously unknown potential anti-dengue compound, vandetanib. This computational approach to analyze a highly curated library of structures has the benefits of speed and cost efficiency. It also leverages mechanistic work with query compounds used in biomedical research to provide strong hypotheses for the antiviral mechanisms of the safer hit compounds. This workflow to identify compounds with known safety profiles can be expanded to any biological activity for which a small-molecule query compound has been identified, potentially expediting the translation of basic research to clinical interventions.

    View details for DOI 10.1128/mBio.02839-20

    View details for PubMedID 33173007

  • Hardwiring Tissue-Specific AAV Transduction in Mice Through Engineered AAVR Expression Zengel, J., Puschnik, A. S., Pillay, S., Nagamine, C. M., Carette, J. E. CELL PRESS. 2020: 253
  • Hamsters and Gerbils EXOTIC ANIMAL LABORATORY DIAGNOSIS McKeon, G. P., Nagamine, C. M., Felt, S. A., Heatley, J. J., Russell, K. E. 2020: 113-128
  • Enterovirus pathogenesis requires the host methyltransferase SETD3. Nature microbiology Diep, J. n., Ooi, Y. S., Wilkinson, A. W., Peters, C. E., Foy, E. n., Johnson, J. R., Zengel, J. n., Ding, S. n., Weng, K. F., Laufman, O. n., Jang, G. n., Xu, J. n., Young, T. n., Verschueren, E. n., Kobluk, K. J., Elias, J. E., Sarnow, P. n., Greenberg, H. B., Hüttenhain, R. n., Nagamine, C. M., Andino, R. n., Krogan, N. J., Gozani, O. n., Carette, J. E. 2019

    Abstract

    Enteroviruses (EVs) comprise a large genus of positive-sense, single-stranded RNA viruses whose members cause a number of important and widespread human diseases, including poliomyelitis, myocarditis, acute flaccid myelitis and the common cold. How EVs co-opt cellular functions to promote replication and spread is incompletely understood. Here, using genome-scale CRISPR screens, we identify the actin histidine methyltransferase SET domain containing 3 (SETD3) as critically important for viral infection by a broad panel of EVs, including rhinoviruses and non-polio EVs increasingly linked to severe neurological disease such as acute flaccid myelitis (EV-D68) and viral encephalitis (EV-A71). We show that cytosolic SETD3, independent of its methylation activity, is required for the RNA replication step in the viral life cycle. Using quantitative affinity purification-mass spectrometry, we show that SETD3 specifically interacts with the viral 2A protease of multiple enteroviral species, and we map the residues in 2A that mediate this interaction. 2A mutants that retain protease activity but are unable to interact with SETD3 are severely compromised in RNA replication. These data suggest a role of the viral 2A protein in RNA replication beyond facilitating proteolytic cleavage. Finally, we show that SETD3 is essential for in vivo replication and pathogenesis in multiple mouse models for EV infection, including CV-A10, EV-A71 and EV-D68. Our results reveal a crucial role of a host protein in viral pathogenesis, and suggest targeting SETD3 as a potential mechanism for controlling viral infections.

    View details for DOI 10.1038/s41564-019-0551-1

    View details for PubMedID 31527793

  • Targeting intramolecular proteinase NS2B/3 cleavages for trans-dominant inhibition of dengue virus PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Constant, D. A., Mateo, R., Nagamine, C. M., Kirkegaard, K. 2018; 115 (40): 10136–41
  • Delayed Onset and Altered Biodistribution of a Non-Canonical AAV Entry Pathway Dudek, A. M., Pillay, S., Puschnik, A. S., Nagamine, C. M., Carette, J. E., Vandenberghe, L. H. CELL PRESS. 2018: 188
  • Identification and Characterization of an Alternate, AAVR Independent, AAV Entry Mechanism Using a Genome-Wide CRISPR/Cas9 Knock-Out Screen Dudek, A. M., Zinn, E. M., Pillay, S., Puschnik, A. S., Nagamine, C. M., Cheng, F., Qiu, J., Carette, J. E., Vandenberghe, L. H. CELL PRESS. 2018: 323
  • SETD3 is an actin histidine methyltransferase that prevents primary dystocia. Nature Wilkinson, A. W., Diep, J. n., Dai, S. n., Liu, S. n., Ooi, Y. S., Song, D. n., Li, T. M., Horton, J. R., Zhang, X. n., Liu, C. n., Trivedi, D. V., Ruppel, K. M., Vilches-Moure, J. G., Casey, K. M., Mak, J. n., Cowan, T. n., Elias, J. E., Nagamine, C. M., Spudich, J. A., Cheng, X. n., Carette, J. E., Gozani, O. n. 2018

    Abstract

    For more than 50 years, the methylation of mammalian actin at histidine 73 has been known to occur1. Despite the pervasiveness of His73 methylation, which we find is conserved in several model animals and plants, its function remains unclear and the enzyme that generates this modification is unknown. Here we identify SET domain protein 3 (SETD3) as the physiological actin His73 methyltransferase. Structural studies reveal that an extensive network of interactions clamps the actin peptide onto the surface of SETD3 to orient His73 correctly within the catalytic pocket and to facilitate methyl transfer. His73 methylation reduces the nucleotide-exchange rate on actin monomers and modestly accelerates the assembly of actin filaments. Mice that lack SETD3 show complete loss of actin His73 methylation in several tissues, and quantitative proteomics analysis shows that actin His73 methylation is the only detectable physiological substrate of SETD3. SETD3-deficient female mice have severely decreased litter sizes owing to primary maternal dystocia that is refractory to ecbolic induction agents. Furthermore, depletion of SETD3 impairs signal-induced contraction in primary human uterine smooth muscle cells. Together, our results identify a mammalian histidine methyltransferase and uncover a pivotal role for SETD3 and actin His73 methylation in the regulation of smooth muscle contractility. Our data also support the broader hypothesis that protein histidine methylation acts as a common regulatory mechanism.

    View details for PubMedID 30626964

  • An alternate route for adeno-associated virus entry independent of AAVR. Journal of virology Dudek, A. M., Pillay, S. n., Puschnik, A. S., Nagamine, C. M., Cheng, F. n., Qiu, J. n., Carette, J. E., Vandenberghe, L. H. 2018

    Abstract

    Determinants and mechanisms of cell attachment and entry steer the Adeno-Associated Virus (AAV) in its utility as a gene therapy vector. Thus far a systematic assessment of how diverse AAV serotypes engage their proteinaceous receptor AAVR (KIAA0319L) to establish transduction has been lacking, despite potential implications for cell and tissue tropism. Here, a large set of human and simian AAVs as well as in silico reconstructed ancestral AAV capsids were interrogated for AAVR usage. We identified a distinct AAV capsid lineage comprised of AAV4 and AAVrh32.33 that can bind and transduce cells in the absence of AAVR, independent of multiplicity of infection. Viral overlay assays and rescue experiments in non-permisive cells demonstrate that these AAVs are unable to bind to or use the AAVR protein for entry. Further evidence for a distinct entry pathway was observed in vivo, as AAVR knock out mice were equally permissive to transduction by AAVrh32.33 compared to wild type mice upon systemic injection. We interestingly observe that some AAV capsids undergo a low level of transduction in the absence of AAVR, both in vitro and in vivo, suggesting that some capsids may have a multi-modal entry pathway. In aggregate, our results demonstrate that AAVR usage is conserved amongst all primate AAVs except for those in the AAV4 lineage, and a non-AAVR pathway may be available to other serotypes. This work furthers our understanding of entry of AAV, a vector system of broad utility in gene therapy.Importance: Adeno-Associated Virus (AAV) is a non-pathogenic virus that is used as a vehicle for gene delivery. Here, we have identified several situations in which transduction is retained in both cell lines and a mouse model in the absence of a previously defined entry receptor, AAVR. Defining the molecular determinants of the infectious pathway of this highly relevant viral vector system can help refine future applications and therapies of this vector.

    View details for PubMedID 29343568

  • Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antiviral research Pu, S. Y., Xiao, F. n., Schor, S. n., Bekerman, E. n., Zanini, F. n., Barouch-Bentov, R. n., Nagamine, C. M., Einav, S. n. 2018; 155: 67–75

    Abstract

    There is an urgent need for strategies to combat dengue virus (DENV) infection; a major global threat. We reported that the cellular kinases AAK1 and GAK regulate intracellular trafficking of multiple viruses and that sunitinib and erlotinib, approved anticancer drugs with potent activity against these kinases, protect DENV-infected mice from mortality. Nevertheless, further characterization of the therapeutic potential and underlying mechanism of this approach is required prior to clinical evaluation. Here, we demonstrate that sunitinib/erlotinib combination achieves sustained suppression of systemic infection at approved dose in DENV-infected IFN-α/β and IFN-γ receptor-deficient mice. Nevertheless, treatment with these blood-brain barrier impermeable drugs delays, yet does not prevent, late-onset paralysis; a common manifestation in this immunodeficient mouse model but not in humans. Sunitinib and erlotinib treatment also demonstrates efficacy in human primary monocyte-derived dendritic cells. Additionally, DENV infection induces expression of AAK1 transcripts, but not GAK, via single-cell transcriptomics, and these kinases are important molecular targets underlying the anti-DENV effect of sunitinib and erlotinib. Lastly, sunitinib/erlotinib combination alters inflammatory cytokine responses in DENV-infected mice. These findings support feasibility of repurposing sunitinib/erlotinib combination as a host-targeted antiviral approach and contribute to understanding its mechanism of antiviral action.

    View details for PubMedID 29753658

  • Animal Models of Zika Virus COMPARATIVE MEDICINE Bradley, M. P., Nagamine, C. M. 2017; 67 (3): 242–52

    Abstract

    Zika virus has garnered great attention over the last several years, as outbreaks of the disease have emerged throughout the Western Hemisphere. Until quite recently Zika virus was considered a fairly benign virus, with limited clinical severity in both people and animals. The size and scope of the outbreak in the Western Hemisphere has allowed for the identification of severe clinical disease that is associated with Zika virus infection, most notably microcephaly among newborns, and an association with Guillian-Barré syndrome in adults. This recent association with severe clinical disease, of which further analysis strongly suggested causation by Zika virus, has resulted in a massive increase in the amount of both basic and applied research of this virus. Both small and large animal models are being used to uncover the pathogenesis of this emerging disease and to develop vaccine and therapeutic strategies. Here we review the animal-model-based Zika virus research that has been performed to date.

    View details for PubMedID 28662753

    View details for PubMedCentralID PMC5482516

  • Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects JOURNAL OF CLINICAL INVESTIGATION Bekerman, E., Neveu, G., Shulla, A., Brannan, J., Pu, S., Wang, S., Xiao, F., Barouch-Bentov, R., Bakken, R. R., Mateo, R., Govero, J., Nagamine, C. M., Diamond, M. S., De Jonghe, S., Herdewijn, P., Dye, J. M., Randall, G., Einav, S. 2017; 127 (4): 1338-1352

    Abstract

    Global health is threatened by emerging viral infections, which largely lack effective vaccines or therapies. Targeting host pathways that are exploited by multiple viruses could offer broad-spectrum solutions. We previously reported that AAK1 and GAK, kinase regulators of the host adaptor proteins AP1 and AP2, are essential for hepatitis C virus (HCV) infection, but the underlying mechanism and relevance to other viruses or in vivo infections remained unknown. Here, we have discovered that AP1 and AP2 cotraffic with HCV particles in live cells. Moreover, we found that multiple viruses, including dengue and Ebola, exploit AAK1 and GAK during entry and infectious virus production. In cultured cells, treatment with sunitinib and erlotinib, approved anticancer drugs that inhibit AAK1 or GAK activity, or with more selective compounds inhibited intracellular trafficking of HCV and multiple unrelated RNA viruses with a high barrier to resistance. In murine models of dengue and Ebola infection, sunitinib/erlotinib combination protected against morbidity and mortality. We validated sunitinib- and erlotinib-mediated inhibition of AAK1 and GAK activity as an important mechanism of antiviral action. Additionally, we revealed potential roles for additional kinase targets. These findings advance our understanding of virus-host interactions and establish a proof of principle for a repurposed, host-targeted approach to combat emerging viruses.

    View details for DOI 10.1172/JCI89857

    View details for Web of Science ID 000398183300024

    View details for PubMedID 28240606

  • Influence of Genetic Background on Hematologic and Histopathologic Alterations during Acute Granulocytic Anaplasmosis in 129/SvEv and C57BL/6J Mice Lacking Type I and Type II Interferon Signaling COMPARATIVE MEDICINE Johns, J. L., Discipulo, M. L., Koehne, A. L., Moorhead, K. A., Nagamine, C. M. 2017; 67 (2): 127-137

    Abstract

    The role of host type I IFN signaling and its interaction with other immune pathways during bacterial infections is incompletely understood. Type II IFN signaling plays a key role during numerous bacterial infections including granulocytic anaplasmosis (GA) caused by Anaplasma phagocytophilum infection. The function of combined type I and type II IFN signaling and their potential synergism during GA and similar tick-borne diseases is a topic of current research investigation. The goal of this study was to evaluate 2 mouse models of absent type I/type II IFN signaling in experimental A. phagocytophilum infection to determine the effects of background strain. Mice lacking both type I and type II IFN receptor signaling (IFNAR-/-/IFNGR-/-) on either the 129/SvEv or C57BL/6J genetic background were evaluated at days 0, 6, 8, and 12 of infection. Pathogen burden in multiple organs was largely similar between strains of infected mice, with few significant differences. Background strain influenced the immune response to infection. Mice of the 129/SvEv strain developed more severe hematologic abnormalities, particularly more severe leukocytosis with marked neutrophilia and lymphocytosis, throughout acute infection. Histopathologic changes occurred in infected mice of both strains and varied in severity by organ. 129/SvEv mice developed more severe pathologic changes in spleen and bone marrow, whereas C57BL/6J mice developed more severe renal pathology. This work highlights the importance of mouse background strain in dictating pathophysiologic response to infection and informs future work regarding the loss of type I and type II IFN signaling on the immune response during GA.

    View details for Web of Science ID 000398880200006

    View details for PubMedID 28381313

  • Development of Novel ImmunoPET Tracers to Image Human PD-1 Checkpoint Expression on Tumor-Infiltrating Lymphocytes in a Humanized Mouse Model. Molecular imaging and biology Natarajan, A., Mayer, A. T., Reeves, R. E., Nagamine, C. M., Gambhir, S. S. 2017

    Abstract

    It is well known that cancers exploit immune checkpoints (programmed death 1 receptor (PD-1) and its ligand (PD-L1)) to evade anti-tumor immune responses. Although immune checkpoint (IC) blockade is a promising approach, not all patients respond. Hence, imaging of tumor-infiltrating lymphocytes (TILs) is of high specific interest, as they are known to express PD-1 during activation and subsequent exhaustion in the tumor microenvironment and are thought to be potentially predictive of therapeutic responses to IC blockade.We developed immune-tracers for positron emission tomography (PET) to image hPD-1 status of human peripheral blood mononuclear cells (hPBMCs) adoptively transferred to NOD-scid IL-2Rγ(null) (NSG) mice (hNSG) bearing A375 human skin melanoma tumors. The anti-PD-1 human antibody (IgG; keytruda) was labeled with either Zr-89 or Cu-64 radiometals to image PD-1-expressing human TILs in vivo.[(89)Zr] Keytruda (groups = 2; NSG-ctl (control) and hNSG-nblk (non-blocking), n = 3-5, 3.2 ± 0.4 MBq/15-16 μg/200 μl) and [(64)Cu] Keytruda (groups = 3; NSG-ctl, NSG-blk (blocking), and hNSG-nblk; n = 4, 7.4 ± 0.4 MBq /20-25 μg/200 μl) were administered in mice. PET-CT scans were performed over 1-144 h ([(89)Zr] Keytruda) and 1-48 h ([(64)Cu] Keytruda) on mice. hNSG mice exhibited a high tracer uptake in the spleen, lymphoid organs and tumors. At 24 h, human TILs homing into melanoma of hNSG-nblk mice exhibited high signal (mean %ID/g ± SD) of 3.8 ± 0.4 ([(89)Zr] Keytruda), and 6.4 ± 0.7 ([(64)Cu] Keytruda), which was 1.5- and 3-fold higher uptake compared to NSG-ctl mice (p = 0.01), respectively. Biodistribution measurements of hNSG-nblk mice performed at 144 h ([(89)Zr] Keytruda) and 48 h ([(64)Cu] Keytruda) p.i. revealed tumor to muscle ratios as high as 45- and 12-fold, respectively.Our immunoPET study clearly demonstrates specific imaging of human PD-1-expressing TILs within the tumor and lymphoid tissues. This suggests these anti-human-PD-1 tracers could be clinically translatable to monitor cancer treatment response to IC blockade therapy.

    View details for DOI 10.1007/s11307-017-1060-3

    View details for PubMedID 28247187

  • An Essential and Ubiquitous Protein Receptor for AAV; Glycans as Attachment Receptors Meyer, N. L., Pillay, S., Xie, Q., Davulcu, O., Puschnik, A., Diep, J., Ishikawa, Y., Jae, L., Wosen, J., Nagamine, C., Noble, A., Stagg, S., Carette, J. E., Chapman, M. S. NATURE PUBLISHING GROUP. 2016: S189
  • Discovery of an Essential Receptor for Adeno-Associated Virus Infection Pillay, S., Meyer, N. L., Puschnik, A. S., Davulco, O., Diep, J., Ishikawa, Y., Jae, L. T., Wosen, J. E., Nagamine, C. M., Chapman, M. S., Carette, J. E. NATURE PUBLISHING GROUP. 2016: S118
  • Evaluation of Isoflurane Overdose for Euthanasia of Neonatal Mice JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE Seymour, T. L., Nagamine, C. M. 2016; 55 (3): 321-323

    Abstract

    Neonatal mice (that is, pups younger than 6 d) must be exposed to CO2 for as long as 50 min to achieve euthanasia. Alternatively, other inhalant anesthetic agents have been used to euthanize laboratory rodent species. We investigated the efficacy of isoflurane at saturated vapor pressure to euthanize neonatal mice. Neonatal mice (n = 76; age, 1 or 2 d) were exposed to isoflurane in a sealed, quart-size (0.95-L) plastic bag at room temperature. Righting and withdrawal reflexes were absent in less than 2 min. After 30 min of exposure to isoflurane, pups were removed and monitored for recovery. All pups were cyanotic and showed no detectable signs of life when they were removed from the bag. However, after 30 to 120 min after removal from the bag, 24% of isoflurane-overexposed pups began gasping and then resumed normal respiration and regained a normal pink coloration. These results demonstrate that isoflurane overexposure at saturated vapor pressure for 30 min is insufficient to euthanize neonatal mice and that isoflurane overexposure must be followed by a secondary means of euthanasia.

    View details for Web of Science ID 000375510400012

    View details for PubMedID 27177567

  • An essential receptor for adeno-associated virus infection. Nature Pillay, S., Meyer, N. L., Puschnik, A. S., Davulcu, O., Diep, J., Ishikawa, Y., Jae, L. T., Wosen, J. E., Nagamine, C. M., Chapman, M. S., Carette, J. E. 2016; 530 (7588): 108-112

    Abstract

    Adeno-associated virus (AAV) vectors are currently the leading candidates for virus-based gene therapies because of their broad tissue tropism, non-pathogenic nature and low immunogenicity. They have been successfully used in clinical trials to treat hereditary diseases such as haemophilia B (ref. 2), and have been approved for treatment of lipoprotein lipase deficiency in Europe. Considerable efforts have been made to engineer AAV variants with novel and biomedically valuable cell tropisms to allow efficacious systemic administration, yet basic aspects of AAV cellular entry are still poorly understood. In particular, the protein receptor(s) required for AAV entry after cell attachment remains unknown. Here we use an unbiased genetic screen to identify proteins essential for AAV serotype 2 (AAV2) infection in a haploid human cell line. The most significantly enriched gene of the screen encodes a previously uncharacterized type I transmembrane protein, KIAA0319L (denoted hereafter as AAV receptor (AAVR)). We characterize AAVR as a protein capable of rapid endocytosis from the plasma membrane and trafficking to the trans-Golgi network. We show that AAVR directly binds to AAV2 particles, and that anti-AAVR antibodies efficiently block AAV2 infection. Moreover, genetic ablation of AAVR renders a wide range of mammalian cell types highly resistant to AAV2 infection. Notably, AAVR serves as a critical host factor for all tested AAV serotypes. The importance of AAVR for in vivo gene delivery is further highlighted by the robust resistance of Aavr(-/-) (also known as Au040320(-/-) and Kiaa0319l(-/-)) mice to AAV infection. Collectively, our data indicate that AAVR is a universal receptor involved in AAV infection.

    View details for DOI 10.1038/nature16465

    View details for PubMedID 26814968

  • Suppression of Drug Resistance in Dengue Virus. mBio Mateo, R., Nagamine, C. M., Kirkegaard, K. 2015; 6 (6): e01960-15

    Abstract

    Dengue virus is a major human pathogen responsible for 400 million infections yearly. As with other RNA viruses, daunting challenges to antiviral design exist due to the high error rates of RNA-dependent RNA synthesis. Indeed, treatment of dengue virus infection with a nucleoside analog resulted in the expected genetic selection of resistant viruses in tissue culture and in mice. However, when the function of the oligomeric core protein was inhibited, no detectable selection of drug resistance in tissue culture or in mice was detected, despite the presence of drug-resistant variants in the population. Suppressed selection of drug-resistant virus correlated with cooligomerization of the targeted drug-susceptible and drug-resistant core proteins. The concept of "dominant drug targets," in which inhibition of oligomeric viral assemblages leads to the formation of drug-susceptible chimeras, can therefore be used to prevent the outgrowth of drug resistance during dengue virus infection.Drug resistance is a major hurdle in the development of effective antivirals, especially those directed at RNA viruses. We have found that one can use the concept of the genetic dominance of defective subunits to "turn cousins into enemies," i.e., to thwart the outgrowth of drug-resistant viral genomes as soon as they are generated. This requires deliberate targeting of larger assemblages, which would otherwise rarely be considered by antiviral researchers.

    View details for DOI 10.1128/mBio.01960-15

    View details for PubMedID 26670386

    View details for PubMedCentralID PMC4701834

  • Suppression of Drug Resistance in Dengue Virus MBIO Mateo, R., Nagamine, C. M., Kirkegaard, K. 2015; 6 (6)

    Abstract

    Dengue virus is a major human pathogen responsible for 400 million infections yearly. As with other RNA viruses, daunting challenges to antiviral design exist due to the high error rates of RNA-dependent RNA synthesis. Indeed, treatment of dengue virus infection with a nucleoside analog resulted in the expected genetic selection of resistant viruses in tissue culture and in mice. However, when the function of the oligomeric core protein was inhibited, no detectable selection of drug resistance in tissue culture or in mice was detected, despite the presence of drug-resistant variants in the population. Suppressed selection of drug-resistant virus correlated with cooligomerization of the targeted drug-susceptible and drug-resistant core proteins. The concept of "dominant drug targets," in which inhibition of oligomeric viral assemblages leads to the formation of drug-susceptible chimeras, can therefore be used to prevent the outgrowth of drug resistance during dengue virus infection.Drug resistance is a major hurdle in the development of effective antivirals, especially those directed at RNA viruses. We have found that one can use the concept of the genetic dominance of defective subunits to "turn cousins into enemies," i.e., to thwart the outgrowth of drug-resistant viral genomes as soon as they are generated. This requires deliberate targeting of larger assemblages, which would otherwise rarely be considered by antiviral researchers.

    View details for DOI 10.1128/mBio.01960-15

    View details for Web of Science ID 000367524700060

    View details for PubMedCentralID PMC4701834

  • Co-infection of the Siberian hamster (Phodopus sungorus) with a novel Helicobacter sp and Campylobacter sp. JOURNAL OF MEDICAL MICROBIOLOGY Nagamine, C. M., Shen, Z., Luong, R. H., McKeon, G. P., Ruby, N. F., Fox, J. G. 2015; 64: 575-581

    Abstract

    We report the isolation of a novel helicobacter isolated from the caecum of the Siberian hamster (Phodopus sungorus). Sequence analysis showed 97% sequence similarity to Helicobacter ganmani. In addition, we report the co-infection of these Siberian hamsters with a Campylobacter sp. and a second Helicobacter sp. with 99% sequence similarity to Helicobacter sp. flexispira taxon 8 (Helicobacter bilis), a species isolated previously from patients with bacteraemia. Gross necropsy and histopathology did not reveal any overt pathological lesions of the liver and gastrointestinal tract that could be attributed to the Helicobacter or Campylobacter spp. infections. This is the first helicobacter to be identified in the Siberian hamster and the first report of co-infection of Helicobacter spp. and Campylobacter sp. in asymptomatic Siberian hamsters.

    View details for DOI 10.1099/jmm.0.000051

    View details for Web of Science ID 000358189300012

  • Co-infection of the Siberian hamster (Phodopus sungorus) with a novel Helicobacter sp. and Campylobacter sp. Journal of medical microbiology Nagamine, C. M., Shen, Z., Luong, R. H., McKeon, G. P., Ruby, N. F., Fox, J. G. 2015; 64 (Pt 5): 575-81

    Abstract

    We report the isolation of a novel helicobacter isolated from the caecum of the Siberian hamster (Phodopus sungorus). Sequence analysis showed 97% sequence similarity to Helicobacter ganmani. In addition, we report the co-infection of these Siberian hamsters with a Campylobacter sp. and a second Helicobacter sp. with 99% sequence similarity to Helicobacter sp. flexispira taxon 8 (Helicobacter bilis), a species isolated previously from patients with bacteraemia. Gross necropsy and histopathology did not reveal any overt pathological lesions of the liver and gastrointestinal tract that could be attributed to the Helicobacter or Campylobacter spp. infections. This is the first helicobacter to be identified in the Siberian hamster and the first report of co-infection of Helicobacter spp. and Campylobacter sp. in asymptomatic Siberian hamsters.

    View details for DOI 10.1099/jmm.0.000051

    View details for PubMedID 25752854

  • Evaluation of Zr-89-rituximab Tracer by Cerenkov Luminescence Imaging and Correlation with PET in a Humanized Transgenic Mouse Model to Image NHL MOLECULAR IMAGING AND BIOLOGY Natarajan, A., Habte, F., Liu, H., Sathirachinda, A., Hu, X., Cheng, Z., Nagamine, C. M., Gambhir, S. S. 2013; 15 (4): 468-475

    Abstract

    PURPOSE: This research aimed to study the use of Cerenkov luminescence imaging (CLI) for non-Hodgkin's lymphoma (NHL) using (89)Zr-rituximab positron emission tomography (PET) tracer with a humanized transgenic mouse model that expresses human CD20 and the correlation of CLI with PET. PROCEDURES: Zr-rituximab (2.6 MBq) was tail vein-injected into transgenic mice that express the human CD20 on their B cells (huCD20TM). One group (n = 3) received 2 mg/kg pre-dose (blocking) of cold rituximab 2 h prior to tracer; a second group (n = 3) had no pre-dose (non-blocking). CLI was performed using a cooled charge-coupled device optical imager. We also performed PET imaging and ex vivo studies in order to confirm the in vivo CLI results. At each time point (4, 24, 48, 72, and 96 h), two groups of mice were imaged in vivo and ex vivo with CLI and PET, and at 96 h, organs were measured by gamma counter. RESULTS: huCD20 transgenic mice injected with (89)Zr-rituximab demonstrated a high-contrast CLI image compared to mice blocked with a cold dose. At various time points of 4-96 h post-radiotracer injection, the in vivo CLI signal intensity showed specific uptake in the spleen where B cells reside and, hence, the huCD20 biomarker is present at very high levels. The time-activity curve of dose decay-corrected CLI intensity and percent injected dose per gram of tissue of PET uptake in the spleen were increased over the time period (4-96 h). At 96 h, the (89)Zr-rituximab uptake ratio (non-blocking vs blocking) counted (mean ± standard deviation) for the spleen was 1.5 ± 0.6 for CLI and 1.9 ± 0.3 for PET. Furthermore, spleen uptake measurements (non-blocking and blocking of all time points) of CLI vs PET showed good correlation (R (2) = 0.85 and slope = 0.576), which also confirmed the corresponding correlations parameter value (R (2) = 0.834 and slope = 0.47) obtained for ex vivo measurements. CONCLUSIONS: CLI and PET of huCD20 transgenic mice injected with (89)Zr-rituximab demonstrated that the tracer was able to target huCD20-expressing B cells. The in vivo and ex vivo tracer uptake corresponding to the CLI radiance intensity from the spleen is in good agreement with PET. In this report, we have validated the use of CLI with PET for NHL imaging in huCD20TM.

    View details for DOI 10.1007/s11307-013-0624-0

    View details for Web of Science ID 000321972500014

    View details for PubMedID 23471750

  • Inhibition of Cellular Autophagy Deranges Dengue Virion Maturation JOURNAL OF VIROLOGY Mateo, R., Nagamine, C. M., Spagnolo, J., Mendez, E., Rahe, M., Gale, M., Yuan, J., Kirkegaard, K. 2013; 87 (3): 1312-1321

    Abstract

    Autophagy is an important component of the innate immune response, directly destroying many intracellular pathogens. However, some pathogens, including several RNA viruses, subvert the autophagy pathway, or components of the pathway, to facilitate their replication. In the present study, the effect of inhibiting autophagy on the growth of dengue virus was tested using a novel inhibitor, spautin-1 (specific and potent autophagy inhibitor 1). Inhibition of autophagy by spautin-1 generated heat-sensitive, noninfectious dengue virus particles, revealing a large effect of components of the autophagy pathway on viral maturation. A smaller effect on viral RNA accumulation was also observed. Conversely, stimulation of autophagy resulted in increased viral titers and pathogenicity in the mouse. We conclude that the presence of functional autophagy components facilitates viral RNA replication and, more importantly, is required for infectious dengue virus production. Pharmacological inhibition of host processes is an attractive antiviral strategy to avoid selection of treatment-resistant variants, and inhibitors of autophagy may prove to be valuable therapeutics against dengue virus infection and pathogenesis.

    View details for DOI 10.1128/JVI.02177-12

    View details for Web of Science ID 000313558100003

    View details for PubMedID 23175363

    View details for PubMedCentralID PMC3554187

  • Maternal antibodies or nonproductive infections confound the need for rederivation. Journal of the American Association for Laboratory Animal Science Nagamine, C. M., Chen, L., Ho, W. Q., Felt, S. A. 2013; 52 (4): 495-498

    Abstract

    After rederivation of a mouse parvovirus (MPV)-contaminated transgenic mouse strain, serology and PCR testing of the surrogate dam showed it to be infected with mouse parvovirus strain 1 (MPV-1). The rederived pups (n = 3) also were MPVpositive, according to serology. Despite MPV seropositivity, fecal PCR tests of the pups were negative, as were serologic results from direct-contact sentinels. Only one rederived pup survived, and this male was bred successfully. None of its mates or progeny seroconverted to MPV. At 14.5 mo of age, the rederived male mouse was euthanized; tissues were collected and submitted for MPV testing; both serologic tests and PCR analysis of mesenteric lymph nodes were MPV-negative. One explanation for the rederived pups' MPV seropostivity is passive transfer of maternal antibodies or a nonproductive MPV infection. This case illustrates that although routine serological testing of surrogate mothers and pups is appropriate, any positive results should be further investigated by using transmissibility testing (fecal PCR or contact sentinels or both) prior to repeat rederivation.

    View details for PubMedID 23849450

  • Carbon Dioxide and Oxygen Levels in Disposable Individually Ventilated Cages after Removal from Mechanical Ventilation JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE Nagamine, C. M., Long, C. T., McKeon, G. P., Felt, S. A. 2012; 51 (2): 155-161

    Abstract

    Disposable individually ventilated cages have lids that restrict air exchange when the cage is not mechanically ventilated. This design feature may cause intracage CO2 to increase and O2 to decrease (hypercapnic and hypoxic conditions, respectively) when the electrical supply to the ventilated rack fails, the ventilated rack malfunctions, cages are docked in the rack incorrectly, or cages are removed from the ventilated rack for extended periods of time. We investigated how quickly hypercapnic and hypoxic conditions developed within disposable individually ventilated cages after removal from mechanical ventilation and compared the data with nondisposable static cages, disposable static cages, and unventilated nondisposable individually ventilated cages. When disposable individually ventilated cages with 5 adult mice per cage were removed from mechanical ventilation, CO2 concentrations increased from less than 1% at 0 h to approximately 5% at 3 h and O2 levels dropped from more than 20% at 0 h to 11.7% at 6 h. The breathing pattern of the mice showed a prominent abdominal component (hyperventilation). Changes were similar for 4 adult mice per cage, reaching at least 5% CO2 at 4 h and 13.0% O2 at 6 h. For 3 or 2 mice per cage, values were 4.6% CO2 and 14.7% O2 and 3.04% CO2 and 17.1% O2, respectively, at 6 h. These results document that within disposable individually ventilated cages, a hypercapnic and hypoxic microenvironment develops within hours in the absence of mechanical ventilation.

    View details for Web of Science ID 000306772200003

    View details for PubMedID 22776114

  • Interleukin-16 deficiency suppresses the development of chronic rejection in murine cardiac transplantation model JOURNAL OF HEART AND LUNG TRANSPLANTATION Kimura, N., Itoh, S., Nakae, S., Axtell, R. C., Velotta, J. B., Bos, E. J., Merk, D. R., Gong, Y., Okamura, H., Nagamine, C. M., Adachi, H., Kornfeld, H., Robbins, R. C., Fischbein, M. P. 2011; 30 (12): 1409-1417

    Abstract

    IL-16 promotes the recruitment of various cells expressing CD4, a receptor for IL-16. The precise role of IL-16 in transplant rejection remains unknown; therefore, the present study investigated the contribution of IL-16 to the development of chronic rejection in heart transplants.C-H-2(bm12)KhEg (H-2(bm12)) donor hearts were transplanted into (1) IL-16-deficient (IL-16(-/-)) C57BL/6J or (b) wild type (WT) control recipients (MHC class II mismatch). Grafts were harvested at 52 days, parenchymal rejection was assessed by the ISHLT grading system, and CAV was examined morphometrically. Graft infiltrating cells were detected 10 and 52 days after transplantation. Intragraft cytokine and chemokine profiles were assessed. To confirm the role of IL-16 in CAV development, C-H-2(bm12)KhEg (H-2(bm12)) donor hearts were transplanted into C57BL/6J WT recipients treated with (1) anti-IL-16-neutralization monoclonal antibody or (b) control immunoglobulin G. Grafts were harvested at 52 days, and CAV was quantified morphometrically. Graft-infiltrating cells were examined histologically.Parenchymal rejection and CAV was significantly attenuated in donor hearts transplanted into IL-16(-/-) recipient mice compared with WT controls. Donor hearts transplanted into IL-16(-/-) recipients had a significant reduction in coronary artery luminal occlusion, intima-to-media ratio, and percentage of diseased vessels. CAV was associated with decreased donor organ inflammation, as well as donor organ cytokine (IL-1β and IL-6) and chemokine (MCP-1 and KC) protein expression. Intimal proliferation and inflammatory cell infiltration were significantly reduced in hearts transplanted into recipients treated with an IL-16-neutralization antibody.IL-16-deficiency reduced graft inflammatory cell recruitment, and allograft inflammatory cytokine and chemokine production. Therefore, IL-16 neutralization may provide a potential target for novel therapeutic treatment for cardiac allograft rejection.

    View details for DOI 10.1016/j.healun.2011.08.017

    View details for Web of Science ID 000297385400016

    View details for PubMedID 22055099

  • Hematologic, Serologic, and Histologic Profile of Aged Siberian Hamsters (Phodopus sungorus) JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE McKeon, G. P., Nagamine, C. M., Ruby, N. F., Luong, R. H. 2011; 50 (3): 308-316

    Abstract

    Biologic samples from 18 (12 female, 6 male) Siberian hamsters (Phodopus sungorus) representing an aged colony (17 to 27 mo) were examined. Values for CBC and serum biochemical parameters were determined, and macroscopic and microscopic pathologic evaluations were performed. Blood urea nitrogen levels were significantly higher in male (54.2 ± 14 mg/dL) compared with female (35.3 ± 22 mg/dL) hamsters and correlated histologically with a higher incidence of chronic glomerulonephropathy in males (5 of 6 males; 0 of 12 females). All 18 hamsters had histologic evidence of follicular mite infestation. Half (6 of 12) of the female hamsters showed cystic rete ovarii. Other histologic findings included thymic or thyroid branchial cysts (3 of 18), focal enteritis (2 of 18), and single cases of hepatic hemangiosarcoma, renal adenoma, subcutaneous mast cell tumor, cutaneous sebaceous adenoma, cutaneous trichofolliculoma, squamous papilloma of the nonglandular stomach, epididymal cholesteatoma, pyometra, and pituitary craniopharyngeal cyst. This study is the first published report of hematologic and serum chemical values for any population of Siberian hamsters and the first published report showing a potential male predisposition for chronic progressive glomerulonephropathy and a potential female predisposition for cystic rete ovarii.

    View details for Web of Science ID 000299026400002

    View details for PubMedID 21640024

  • Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/cJ-IL10-deficient mice. Int J Cancer Nagamine CM, Rogers AB, Fox JG, Schauer DB 2008; 122: 832-838
  • Acute paraplegia in a young adult Long-Evans rat resulting from T-cell lymphoma. Contemp Topics Lab Anim Sci Nagamine CM, Jackson C, Beck KA, Marini RP, Fox JG, Nambiar PR 2005; 44: 53-56
  • Proliferative pododermatitis (canker) with intralesional spirochetes in three horses. J Vet Diagn Invest Nagamine CM, Castro F, Buchanan B, Schumacher J, Craig L. 2005; 17: 269-271
  • Sex Reversal Caused by Mus musculus domesticus Y Chromosomes Linked to Variant Expression of the Testis-Determining Gene Sry Dev. Biol. Nagamine CM, Morohashi K, Carlisle C, Chang D 1999; 216: 182-194
  • Ovotestes in B6-XXSxr sex reversed mice. Dev. Biol. Nagamine CM, Capehart J., Carlisle C, Chang D 1998; 196: 24-32
  • Zfy2/1 fusion gene fails to replicate Zfy1 expression pattern in fetal gonads. Genomics Nagamine CM, Carlisle C 1997; 43: 397-398
  • Absence of correlation between Sry polymorphisms and XY sex reversal caused by M. m. domesticus Y Chromosome. Genomics Carlisle C, Winking H, Weichenhan D, Nagamine CM 1996; 33: 32-45
  • The dominant white spotting oncogene allele Kit(W-42J) exacerbates XY(DOM) sex reversal. Development Nagamine CM, Carlisle C 1996; 123: 3597-3605
  • The testis-determining gene, SRY, exists in multiple copies in Old World rodents. Genetical Research Nagamine CM 1994; 64: 151-159
  • Distribution of the molossinus allele of Sry, the testis-determining gene, in wild mice. Molecular Biology and Evolution Nagamine CM, Shiroishi T, Miyashita N, Tsuchiya K, Ikeda H, Takao N, Wu X-L, Jin M-L, Wang F-S, Kryukov AP, Akbar 1994; 11: 864-874
  • Polymorphism of a CAG trinucleotide repeat within Sry correlates with B6.YDom sex reversal. Nature Genetics Coward P, Nagai K, Chen D, Thomas HD, Nagamine CM, Lau Y-FC 1994; 6: 245-250
  • The musculus-type Y chromosome of the laboratory mouse is of Asian origin. Mammalian Genome Nagamine CM, Nishioka Y, Moriwaki K, Boursot P, Bonhomme F, Lau Y-FC 1992; 3: 84-91
  • Hst-3: an X-linked hybrid sterility gene. Genetical Research Guénet J-L, Nagamine CM, Simon-Chazottes D, Montagutelli X, Bonhomme F 1990; 56: 163-165
  • The two candidate testis-determining Y genes (Zfy-1, Zfy-2) are differentially expressed in fetal and adult mouse tissues. Genes Develop Nagamine CM, Chan K, Hake LE, Lau Y-FC 1990; 4: 63-74
  • Development and fertility of ovaries in the B6.YDom sex-reversed female mouse. Development Taketo-Hosotani T, Nishioka Y, Nagamine CM, Villalpando I, Merchant-Larios H 1989; 107: 95-105
  • Chromosome mapping and expression of a putative testis-determining gene in mouse. Science Nagamine CM, Chan K, Kozak CA, Lau Y-F 1989; 243: 80-83
  • A PCR artifact: generation of heteroduplexes. Am J Hum Genet Nagamine CM, Chan K, Lau Y-FC 1989; 45: 337-339
  • Morphological development of the gonad in tda-1 XY sex reversal. Differentiation Nagamine CM, Taketo T, Koo GC 1987; 33: 214-222
  • Linkage of the murine steroid sulfatase locus, Sts, to sex reversal, Sxr: a genetic and molecular analysis. Nucl Acid Res Nagamine CM, Michot J-L, Roberts C, Guénet J-L, Bishop CE 1987; 15: 9227-9238
  • The use of specific DNA probes to analyze the Sxr mutation in the mouse. Development Bishop CE, Roberts C, Michot J-L, Nagamine CM, Winking H, Guénet J-L, Weith A 1987; 101 (S): 165-167
  • Masculinization of female crayfish, Procambarus clarki (Girard), by androgenic gland implantation. Int J Invert Reprod Devel Nagamine, C., Knight AW 1987; 11: 77-87
  • Induction of female breeding characteristics by ovarian tissue implants in androgenic gland ablated male freshwater prawns Macrobrachium rosenbergii (de Man)(Decapoda, Palaemonidae). Int J Invert Reprod Devel Nagamine CM, Knight AW 1987; 11: 225-234
  • Studies on the genetics of tda-1 XY sex reversal. Differentiation Nagamine CM, Taketo T, Koo GC 1987; 33: 223-231
  • A radiobinding assay for H-Y antigen using monoclonal antibodies. Transplantation Nagamine C, Reidy J, Koo GC 1984; 37: 13-17
  • H-Y antigen in XO mice. Immunogenetics Koo G, Reidy J, Nagamine CM 1983; 18: 37-44
  • The development, maturation, and function of the sexually dimorphic characteristics of the Malaysian prawn, Macrobrachium rosenbergii (de Man)(Decapoda, Palaemonidae). Crustaceana Nagamine C, Knight AW 1980; 39: 141-152
  • Effects of androgenic gland ablation on the male primary and secondary sexual characteristics in the Malaysian prawn, Macrobrachium rosenbergii (de Man), with the first evidence of induced feminization in a nonhermaphroditic decapod. Gen Comp Endocrinol Nagamine C, Knight A, Maggenti A , Paxman G 1980; 41: 423-441
  • Masculinization of female Macrobrachium rosenbergii (de Man)(Decapoda, Palaemonidae) by androgenic gland implantation. Gen Comp Endocrinol Nagamine C, Knight A, Maggenti A, Paxman G 1980; 41: 442-457