Administrative Appointments


  • Director of Basic Research, Division of Nephrology (2015 - Present)

Boards, Advisory Committees, Professional Organizations


  • American Society of Transplantation, Board of Directors (2018 - 2021)
  • Federation of Clinical Immunology Societies, Secretary / Treasurer (2017 - Present)

Professional Education


  • BS, MIT, Biology (1989)
  • PhD, University of Pennsylvania School of Medicine, Immunology (1997)
  • MD, University of Pennsylvania School of Medicine, Medicine (1997)
  • Internship/Residency, University of Chicago Hospitals, Internal Medicine (2000)
  • Fellowship, University of Pennsylvania School of Medicine, Nephrology (2003)

2023-24 Courses


All Publications


  • Advancing Mouse Models for Transplantation Research. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons Cravedi, P., Riella, L. V., Ford, M., Valujskikh, A., Menon, M. C., Kirk, A. D., Alegre, M. L., Feng, S., Kehn, P., Najafian, N., Hancock, W., Heeger, P. S., Maltzman, J., Mannon, R. B., Nadig, S., Odim, J., Turnquist, H., Shaw, J., West, L., Luo, X., Chong, A., Bromberg, J. 2024

    Abstract

    Mouse models have been instrumental for understanding mechanisms of transplant rejection and tolerance, but cross-study reproducibility and translation of experimental findings into effective clinical therapies are issues of concern. The MOuse MOdels in Transplantation (MOMOT) symposium gathered scientists and physician-scientists involved in basic and clinical research in transplantation to discuss the strengths and limitations of mouse transplant models and strategies to enhance their utility. Participants recognized that increased procedure standardization, and including use of prespecified, defined endpoints and statistical power analyses, would benefit the field. They also discussed the generation of new models that incorporate environmental and genetic variables affecting clinical outcomes as potentially important. If implemented, these strategies are expected to improve the reproducibility of mouse studies and increase their translation to clinical trials and, ideally, new FDA-approved drugs.

    View details for DOI 10.1016/j.ajt.2024.01.006

    View details for PubMedID 38219866

  • Prior viral infection primes cross-reactive CD8+ T cells that respond to mouse heart allografts. Frontiers in immunology Khorki, M. E., Shi, T., Cianciolo, E. E., Burg, A. R., Chukwuma, P. C., Picarsic, J. L., Morrice, M. K., Woodle, E. S., Maltzman, J. S., Ferguson, A., Katz, J. D., Baker, B. M., Hildeman, D. A. 2023; 14: 1287546

    Abstract

    Significant evidence suggests a connection between transplant rejection and the presence of high levels of pre-existing memory T cells. Viral infection can elicit viral-specific memory T cells that cross-react with allo-MHC capable of driving allograft rejection in mice. Despite these advances, and despite their critical role in transplant rejection, a systematic study of allo-reactive memory T cells, their specificities, and the role of cross-reactivity with viral antigens has not been performed.Here, we established a model to identify, isolate, and characterize cross-reactive T cells using Nur77 reporter mice (C57BL/6 background), which transiently express GFP exclusively upon TCR engagement. We infected Nur77 mice with lymphocytic choriomeningitis virus (LCMV-Armstrong) to generate a robust memory compartment, where quiescent LCMV-specific memory CD8+ T cells could be readily tracked with MHC tetramer staining. Then, we transplanted LCMV immune mice with allogeneic hearts and monitored expression of GFP within MHC-tetramer defined viral-specific T cells as an indicator of their ability to cross-react with alloantigens.Strikingly, prior LCMV infection significantly increased the kinetics and magnitude of rejection as well as CD8+ T cell recruitment into allogeneic, but not syngeneic, transplanted hearts, relative to non-infected controls. Interestingly, as early as day 1 after allogeneic heart transplant an average of ~8% of MHC-tetramer+ CD8+ T cells expressed GFP, in contrast to syngeneic heart transplants, where the frequency of viral-specific CD8+ T cells that were GFP+ was <1%. These data show that a significant percentage of viral-specific memory CD8+ T cells expressed T cell receptors that also recognized alloantigens in vivo. Notably, the frequency of cross-reactive CD8+ T cells differed depending upon the viral epitope. Further, TCR sequences derived from cross-reactive T cells harbored distinctive motifs that may provide insight into cross-reactivity and allo-specificity.In sum, we have established a mouse model to track viral-specific, allo-specific, and cross-reactive T cells; revealing that prior infection elicits substantial numbers of viral-specific T cells that cross-react to alloantigen, respond very early after transplant, and may promote rapid rejection.

    View details for DOI 10.3389/fimmu.2023.1287546

    View details for PubMedID 38143762

    View details for PubMedCentralID PMC10748599

  • CD8 Tregs SQa-1shing transplant rejection. Science immunology Bracey, N. A., Maltzman, J. S. 2023; 8 (90): eadn0644

    Abstract

    MHC-E restricted CD8+ regulatory T cells have a restricted TCR repertoire and eliminate pathogenic CD4 T cells to mediate immune responses.

    View details for DOI 10.1126/sciimmunol.adn0644

    View details for PubMedID 38039378

  • The Effects of Aging on Solid Organ Transplantation-Characteristics and Consequences of Immunosenescence CURRENT TRANSPLANTATION REPORTS Rollenhagen, C., Maltzman, J. S. 2023
  • TCRward personalized transplant therapies. Science immunology Chadha, A., Maltzman, J. S. 2023; 8 (85): eadj4910

    Abstract

    scRNA/TCRseq of kidney biopsies from acute cellular rejection reveals limited clonality and diverse T cell phenotypes.

    View details for DOI 10.1126/sciimmunol.adj4910

    View details for PubMedID 37418546

  • Age-related decline in anti-HBV antibodies in vaccinated kidney transplant recipients. Transplant infectious disease : an official journal of the Transplantation Society Dadhania, D. M., Cravedi, P., Blumberg, E., Stryniak, G., Montez-Rath, M. E., Maltzman, J. S. 2023: e14090

    Abstract

    Hepatitis B virus (HBV) vaccination is indicated for all end stage kidney disease patients, including all solid organ transplant candidates. Maintenance of adequate immunity is especially important for immunosuppressed solid organ recipients who are at increased risk for donor or community acquired HBV. The impact of age and immunosuppression on long-term maintenance of HBV immunity postvaccination has not been fully investigated.We performed a single-center retrospective study of 96 kidney transplant recipients, transplanted between July 2012 and December 2020, who had Hepatitis B surface antibody (HBsAb) levels measured pretransplantation and 1-year posttransplantation. We compared the change in HBsAb levels stratified by patient's age (<45, 45-60, and >60) and by whether or not the patient received lymphocyte depleting induction therapy.Our results demonstrate that HBsAb IgG levels vary by age group, decreased significantly at 1-year posttransplant (p < .0001) and were significantly lower in the older cohort (p = .03). Among recipients who received rabbit anti-thymocyte globulin induction (rATG), the log HbsAb levels were significantly lower in the older age group (2.15 in age <45, 1.75 in age 45-60 and 1.47 in age >60, p = .01). Age group (p = .004), recipient HBcAb status (p = .002), and rATG (p = .048) were independently associated with >20% reduction in log HBsAb levels posttransplant.Significant declines in HBsAb levels occur postkidney transplantation, especially in older individuals, thus placing exposed older kidney transplant recipients at greater risk of HBV infection and associated complications.

    View details for DOI 10.1111/tid.14090

    View details for PubMedID 37377328

  • Humoral and T cell Responses to SARS-CoV-2 Vaccine Booster and Anti-SARS-CoV-2 Monoclonal Antibodies in Patients With End-Stage Kidney Disease. Kidney international reports Zaza, G., Stallone, G., Granata, S., Gentile, M., Panico, M., Bin, S., Wang, L., Rollenhagen, C., Maltzman, J. S., Cravedi, P. 2023

    View details for DOI 10.1016/j.ekir.2023.04.026

    View details for PubMedID 37360818

  • CD8 T cells are forever. Science immunology Maltzman, J. S. 2023; 8 (80): eadg8279

    Abstract

    Iterative acute stimulations can maintain CD8 T cells for longer than their organismal lifespan.

    View details for DOI 10.1126/sciimmunol.adg8279

    View details for PubMedID 36735775

  • CD8 T cells are forever SCIENCE IMMUNOLOGY Maltzman, J. S. 2023; 8 (80)
  • Endothelial Cell-Specific Molecule-1 Inhibits Albuminuria in Diabetic Mice. Kidney360 Zheng, X., Higdon, L., Gaudet, A., Shah, M., Balistieri, A., Li, C., Nadai, P., Palaniappan, L., Yang, X., Santo, B., Ginley, B., Wang, X. X., Myakala, K., Nallagatla, P., Levi, M., Sarder, P., Rosenberg, A., Maltzman, J. S., de Freitas Caires, N., Bhalla, V. 2022; 3 (12): 2059-2076

    Abstract

    Background: Diabetic kidney disease (DKD) is the most common cause of kidney failure in the world, and novel predictive biomarkers and molecular mechanisms of disease are needed. Endothelial cell-specific molecule-1 (Esm-1) is a secreted proteoglycan that attenuates inflammation. We previously identified that a glomerular deficiency of Esm-1 associates with more pronounced albuminuria and glomerular inflammation in DKD-susceptible relative to DKD-resistant mice, but its contribution to DKD remains unexplored.Methods: Using hydrodynamic tail-vein injection, we overexpress Esm-1 in DKD-susceptible DBA/2 mice and delete Esm-1 in DKD-resistant C57BL/6 mice to study the contribution of Esm-1 to DKD. We analyze clinical indices of DKD, leukocyte infiltration, podocytopenia, and extracellular matrix production. We also study transcriptomic changes to assess potential mechanisms of Esm-1 in glomeruli.Results: In DKD-susceptible mice, Esm-1 inversely correlates with albuminuria and glomerular leukocyte infiltration. We show that overexpression of Esm-1 reduces albuminuria and diabetes-induced podocyte injury, independent of changes in leukocyte infiltration. Using a complementary approach, we find that constitutive deletion of Esm-1 in DKD-resistant mice modestly increases the degree of diabetes-induced albuminuria versus wild-type controls. By glomerular RNAseq, we identify that Esm-1 attenuates expression of kidney disease-promoting and interferon (IFN)-related genes, including Ackr2 and Cxcl11.Conclusions: We demonstrate that, in DKD-susceptible mice, Esm-1 protects against diabetes-induced albuminuria and podocytopathy, possibly through select IFN signaling. Companion studies in patients with diabetes suggest a role of Esm-1 in human DKD.

    View details for DOI 10.34067/KID.0001712022

    View details for PubMedID 36591362

  • Analysis of Cross-sectional and Longitudinal HLA and Anti-viral Responses After COVID Infection in Renal Allograft Recipients: Differences and Correlates. Transplantation Girnita, A. L., Wang, L., Colovai, A. I., Ahearn, P., Azzi, Y., Menon, M. C., Fernandez-Vina, M., Gebel, H. M., Steve Woodle, E., Cravedi, P., Maltzman, J. S., Akalin, E. 2022

    Abstract

    BACKGROUND: Characterization of anti-HLA versus anti-severe acute respiratory syndrome coronavirus 2 (anti-SARS-CoV-2) immune globulin isotypes in organ transplant recipients after coronavirus disease 2019 (COVID-19) infection has not been reported. We aimed to determine changes in anti-HLA antibodies in renal transplant patients with COVID-19 and compare the immunoglobulin and epitope-binding pattern versus anti-SARS-CoV-2 antibodies.METHODS: This is a cross-sectional study of 46 kidney transplant recipients including 21 with longitudinal sampling. Using a semi-quantitative multiplex assay, we determined immunoglobulin (Ig) M, IgA, IgG, and IgG1-2-3-4 antibodies against Class I and Class II HLA, and 5 SARS-CoV-2 epitopes including the nucleocapsid protein and multiple regions of the spike protein.RESULTS: Fourteen of 46 (30%) patients had donor-specific anti-HLA antibodies (donor-specific antibody [DSA]), 12 (26%) had non-DSA anti-HLA antibodies and 45 (98%) had anti-SARS-CoV-2 antibodies. Most DSAs targeted HLA-DQ (71%), with a dominant IgG isotype and IgG1 subtype prevalence (93%), and/or IgG3 (64%), followed by IgG2 (36%). Comparatively, there was a higher prevalence of IgA (85% versus 14%, P = 0.0001) and IgM (87%, versus 36%, P = 0.001) in the anti-SARS-CoV-2 antibody profile, when compared to DSAs, respectively. Anti-SARS-CoV-2 antibody profile was characterized by increased prevalence of IgM and IgA, when compared to DSAs. The median calculated panel reactive antibody before COVID-19 diagnosis (24%) tended to decrease after COVID-19 diagnosis (10%) but it was not statistically significant (P = 0.1).CONCLUSIONS: Anti-HLA antibody strength and calculated panel reactive antibody in kidney transplant recipients after COVID-19 do not significantly increase after infection. Although the IgG isotype was the dominant form in both HLA and SARS-CoV-2 antigens, the alloimmune response had a low IgA pattern, whereas anti-SARS-CoV-2 antibodies were high IgA/IgM.

    View details for DOI 10.1097/TP.0000000000004277

    View details for PubMedID 36070571

  • A SPARC-ling link to inflammaging. Science immunology Maltzman, J. S. 2022; 7 (75): eade5698

    Abstract

    Adipocyte derived SPARC induces pro-inflammatory changes to macrophages, leading to aging that can be reduced by caloric restriction.

    View details for DOI 10.1126/sciimmunol.ade5698

    View details for PubMedID 36054338

  • Blood Transcriptomes of SARS-CoV-2-Infected Kidney Transplant Recipients Associated with Immune Insufficiency Proportionate to Severity. Journal of the American Society of Nephrology : JASN Sun, Z., Zhang, Z., Banu, K., Azzi, Y. A., Reghuvaran, A., Fredericks, S., Planoutene, M., Hartzell, S., Kim, Y., Pell, J., Tietjen, G., Asch, W., Kulkarni, S., Formica, R., Rana, M., Maltzman, J. S., Zhang, W., Akalin, E., Heeger, P. S., Cravedi, P., Menon, M. C. 2022

    Abstract

    Among patients with COVID-19, kidney transplant recipients (KTRs) have poor outcomes compared with non-KTRs. To provide insight into management of immunosuppression during acute illness, we studied immune signatures from the peripheral blood during and after COVID-19 infection from a multicenter KTR cohort.We ascertained clinical data by chart review. A single sample of blood was collected for transcriptome analysis. Total RNA was poly-A selected and RNA was sequenced to evaluate transcriptome changes. We also measured cytokines and chemokines of serum samples collected during acute infection.A total of 64 patients with COVID-19 in KTRs were enrolled, including 31 with acute COVID-19 (<4 weeks from diagnosis) and 33 with post-acute COVID-19 (>4 weeks postdiagnosis). In the blood transcriptome of acute cases, we identified genes in positive or negative association with COVID-19 severity scores. Functional enrichment analyses showed upregulation of neutrophil and innate immune pathways but downregulation of T cell and adaptive immune activation pathways. This finding was independent of lymphocyte count, despite reduced immunosuppressant use in most KTRs. Compared with acute cases, post-acute cases showed "normalization" of these enriched pathways after 4 weeks, suggesting recovery of adaptive immune system activation despite reinstitution of immunosuppression. Analysis of the non-KTR cohort with COVID-19 showed significant overlap with KTRs in these functions. Serum inflammatory cytokines followed an opposite trend (i.e., increased with disease severity), indicating that blood lymphocytes are not the primary source.The blood transcriptome of KTRs affected by COVID-19 shows decreases in T cell and adaptive immune activation pathways during acute disease that, despite reduced immunosuppressant use, associate with severity. These pathways show recovery after acute illness.

    View details for DOI 10.1681/ASN.2022010125

    View details for PubMedID 36041788

  • Infection, Rejection, and the Connection. Transplantation Higdon, L. E., Tan, J. C., Maltzman, J. S. 2022

    Abstract

    Solid organ transplantation is a life-saving treatment for people with end-stage organ disease. Immune-mediated transplant rejection is a common complication that decreases allograft survival. Although immunosuppression is required to prevent rejection, it also increases the risk of infection. Some infections, such as cytomegalovirus and BK virus, can promote inflammatory gene expression that can further tip the balance toward rejection. BK virus and other infections can induce damage that resembles the clinical pathology of rejection, and this complicates accurate diagnosis. Moreover, T cells specific for viral infection can lead to rejection through heterologous immunity to donor antigen directly mediated by antiviral cells. Thus, viral infections and allograft rejection interact in multiple ways that are important to maintain immunologic homeostasis in solid organ transplant recipients. Better insight into this dynamic interplay will help promote long-term transplant survival.

    View details for DOI 10.1097/TP.0000000000004297

    View details for PubMedID 36017937

  • Chimeric Antigen Receptor Regulatory T Cell in Transplantation: TheFuture of Cell Therapy? Kidney international reports Lamarche, C., Maltzman, J. S. 2022; 7 (6): 1149-1152

    View details for DOI 10.1016/j.ekir.2022.04.003

    View details for PubMedID 35694556

  • The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science (New York, N.Y.) Jones, R. C., Karkanias, J., Krasnow, M. A., Pisco, A. O., Quake, S. R., Salzman, J., Yosef, N., Bulthaup, B., Brown, P., Harper, W., Hemenez, M., Ponnusamy, R., Salehi, A., Sanagavarapu, B. A., Spallino, E., Aaron, K. A., Concepcion, W., Gardner, J. M., Kelly, B., Neidlinger, N., Wang, Z., Crasta, S., Kolluru, S., Morri, M., Pisco, A. O., Tan, S. Y., Travaglini, K. J., Xu, C., Alcántara-Hernández, M., Almanzar, N., Antony, J., Beyersdorf, B., Burhan, D., Calcuttawala, K., Carter, M. M., Chan, C. K., Chang, C. A., Chang, S., Colville, A., Crasta, S., Culver, R. N., Cvijović, I., D'Amato, G., Ezran, C., Galdos, F. X., Gillich, A., Goodyer, W. R., Hang, Y., Hayashi, A., Houshdaran, S., Huang, X., Irwin, J. C., Jang, S., Juanico, J. V., Kershner, A. M., Kim, S., Kiss, B., Kolluru, S., Kong, W., Kumar, M. E., Kuo, A. H., Leylek, R., Li, B., Loeb, G. B., Lu, W. J., Mantri, S., Markovic, M., McAlpine, P. L., de Morree, A., Morri, M., Mrouj, K., Mukherjee, S., Muser, T., Neuhöfer, P., Nguyen, T. D., Perez, K., Phansalkar, R., Pisco, A. O., Puluca, N., Qi, Z., Rao, P., Raquer-McKay, H., Schaum, N., Scott, B., Seddighzadeh, B., Segal, J., Sen, S., Sikandar, S., Spencer, S. P., Steffes, L. C., Subramaniam, V. R., Swarup, A., Swift, M., Travaglini, K. J., Van Treuren, W., Trimm, E., Veizades, S., Vijayakumar, S., Vo, K. C., Vorperian, S. K., Wang, W., Weinstein, H. N., Winkler, J., Wu, T. T., Xie, J., Yung, A. R., Zhang, Y., Detweiler, A. M., Mekonen, H., Neff, N. F., Sit, R. V., Tan, M., Yan, J., Bean, G. R., Charu, V., Forgó, E., Martin, B. A., Ozawa, M. G., Silva, O., Tan, S. Y., Toland, A., Vemuri, V. N., Afik, S., Awayan, K., Botvinnik, O. B., Byrne, A., Chen, M., Dehghannasiri, R., Detweiler, A. M., Gayoso, A., Granados, A. A., Li, Q., Mahmoudabadi, G., McGeever, A., de Morree, A., Olivieri, J. E., Park, M., Pisco, A. O., Ravikumar, N., Salzman, J., Stanley, G., Swift, M., Tan, M., Tan, W., Tarashansky, A. J., Vanheusden, R., Vorperian, S. K., Wang, P., Wang, S., Xing, G., Xu, C., Yosef, N., Alcántara-Hernández, M., Antony, J., Chan, C. K., Chang, C. A., Colville, A., Crasta, S., Culver, R., Dethlefsen, L., Ezran, C., Gillich, A., Hang, Y., Ho, P. Y., Irwin, J. C., Jang, S., Kershner, A. M., Kong, W., Kumar, M. E., Kuo, A. H., Leylek, R., Liu, S., Loeb, G. B., Lu, W. J., Maltzman, J. S., Metzger, R. J., de Morree, A., Neuhöfer, P., Perez, K., Phansalkar, R., Qi, Z., Rao, P., Raquer-McKay, H., Sasagawa, K., Scott, B., Sinha, R., Song, H., Spencer, S. P., Swarup, A., Swift, M., Travaglini, K. J., Trimm, E., Veizades, S., Vijayakumar, S., Wang, B., Wang, W., Winkler, J., Xie, J., Yung, A. R., Artandi, S. E., Beachy, P. A., Clarke, M. F., Giudice, L. C., Huang, F. W., Huang, K. C., Idoyaga, J., Kim, S. K., Krasnow, M., Kuo, C. S., Nguyen, P., Quake, S. R., Rando, T. A., Red-Horse, K., Reiter, J., Relman, D. A., Sonnenburg, J. L., Wang, B., Wu, A., Wu, S. M., Wyss-Coray, T. 2022; 376 (6594): eabl4896

    Abstract

    Molecular characterization of cell types using single-cell transcriptome sequencing is revolutionizing cell biology and enabling new insights into the physiology of human organs. We created a human reference atlas comprising nearly 500,000 cells from 24 different tissues and organs, many from the same donor. This atlas enabled molecular characterization of more than 400 cell types, their distribution across tissues, and tissue-specific variation in gene expression. Using multiple tissues from a single donor enabled identification of the clonal distribution of T cells between tissues, identification of the tissue-specific mutation rate in B cells, and analysis of the cell cycle state and proliferative potential of shared cell types across tissues. Cell type-specific RNA splicing was discovered and analyzed across tissues within an individual.

    View details for DOI 10.1126/science.abl4896

    View details for PubMedID 35549404

  • Cell types of origin of the cell-free transcriptome. Nature biotechnology Vorperian, S. K., Moufarrej, M. N., Tabula Sapiens Consortium, Quake, S. R., Jones, R. C., Karkanias, J., Krasnow, M., Pisco, A. O., Quake, S. R., Salzman, J., Yosef, N., Bulthaup, B., Brown, P., Harper, W., Hemenez, M., Ponnusamy, R., Salehi, A., Sanagavarapu, B. A., Spallino, E., Aaron, K. A., Concepcion, W., Gardner, J. M., Kelly, B., Neidlinger, N., Wang, Z., Crasta, S., Kolluru, S., Morri, M., Tan, S. Y., Travaglini, K. J., Xu, C., Alcantara-Hernandez, M., Almanzar, N., Antony, J., Beyersdorf, B., Burhan, D., Calcuttawala, K., Carter, M. M., Chan, C. K., Chang, C. A., Chang, S., Colville, A., Culver, R. N., Cvijovic, I., D'Amato, G., Ezran, C., Galdos, F. X., Gillich, A., Goodyer, W. R., Hang, Y., Hayashi, A., Houshdaran, S., Huang, X., Irwin, J. C., Jang, S., Juanico, J. V., Kershner, A. M., Kim, S., Kiss, B., Kong, W., Kumar, M. E., Kuo, A. H., Leylek, R., Li, B., Loeb, G. B., Lu, W., Mantri, S., Markovic, M., McAlpine, P. L., de Morree, A., Mrouj, K., Mukherjee, S., Muser, T., Neuhofer, P., Nguyen, T. D., Perez, K., Phansalkar, R., Puluca, N., Qi, Z., Rao, P., Raquer-McKay, H., Schaum, N., Scott, B., Seddighzadeh, B., Segal, J., Sen, S., Sikandar, S., Spencer, S. P., Steffes, L., Subramaniam, V. R., Swarup, A., Swift, M., Van Treuren, W., Trimm, E., Veizades, S., Vijayakumar, S., Vo, K. C., Vorperian, S. K., Wang, W., Weinstein, H. N., Winkler, J., Wu, T. T., Xie, J., Yung, A. R., Zhang, Y., Detweiler, A. M., Mekonen, H., Neff, N. F., Sit, R. V., Tan, M., Yan, J., Bean, G. R., Charu, V., Forgo, E., Martin, B. A., Ozawa, M. G., Silva, O., Toland, A., Vemuri, V. N., Afik, S., Awayan, K., Bierman, R., Botvinnik, O. B., Byrne, A., Chen, M., Dehghannasiri, R., Gayoso, A., Granados, A. A., Li, Q., Mahmoudabadi, G., McGeever, A., Olivieri, J. E., Park, M., Ravikumar, N., Stanley, G., Tan, W., Tarashansky, A. J., Vanheusden, R., Wang, P., Wang, S., Xing, G., Xu, C., Yosef, N., Culver, R., Dethlefsen, L., Ho, P., Liu, S., Maltzman, J. S., Metzger, R. J., Sasagawa, K., Sinha, R., Song, H., Wang, B., Artandi, S. E., Beachy, P. A., Clarke, M. F., Giudice, L. C., Huang, F. W., Huang, K. C., Idoyaga, J., Kim, S. K., Kuo, C. S., Nguyen, P., Rando, T. A., Red-Horse, K., Reiter, J., Relman, D. A., Sonnenburg, J. L., Wu, A., Wu, S. M., Wyss-Coray, T. 2022

    Abstract

    Cell-free RNA from liquid biopsies can be analyzed to determine disease tissue of origin. We extend this concept to identify cell types of origin using the Tabula Sapiens transcriptomic cell atlas as well as individual tissue transcriptomic cell atlases in combination with the Human Protein Atlas RNA consensus dataset. We define cell type signature scores, which allow the inference of cell types that contribute to cell-free RNA for a variety of diseases.

    View details for DOI 10.1038/s41587-021-01188-9

    View details for PubMedID 35132263

  • CMV-Responsive CD4 T Cells Have a Stable Cytotoxic Phenotype Over the First Year Post-Transplant in Patients Without Evidence of CMV Viremia. Frontiers in immunology Higdon, L. E., Ahmad, A. A., Schaffert, S., Margulies, K. B., Maltzman, J. S. 2022; 13: 904705

    Abstract

    Cytomegalovirus (CMV) infection is a known cause of morbidity and mortality in solid organ transplant recipients. While primary infection is controlled by a healthy immune system, CMV is never eradicated due to viral latency and periodic reactivation. Transplantation and associated therapies hinder immune surveillance of CMV. CD4 T cells are an important part of control of CMV reactivation. We therefore investigated how CMV impacts differentiation, functionality, and expansion of protective CD4 T cells from recipients of heart or kidney transplant in the first year post-transplant without evidence of CMV viremia. We analyzed longitudinal peripheral blood samples by flow cytometry and targeted single cell RNA sequencing coupled to T cell receptor (TCR) sequencing. At the time of transplant, CD4 T cells from CMV seropositive transplant recipients had a higher degree of immune aging than the seronegative recipients. The phenotype of CD4 T cells was stable over time. CMV-responsive CD4 T cells in our transplant cohort included a large proportion with cytotoxic potential. We used sequence analysis of TCRalphabeta to identify clonal expansion and found that clonally expanded CMV-responsive CD4 T cells were of a predominantly aged cytotoxic phenotype. Overall, our analyses suggest that the CD4 response to CMV is dominated by cytotoxicity and not impacted by transplantation in the first year. Our findings indicate that CMV-responsive CD4 T cells are homeostatically stable in the first year after transplantation and identify subpopulations relevant to study the role of this CD4 T cell population in post-transplant health.

    View details for DOI 10.3389/fimmu.2022.904705

    View details for PubMedID 35837398

  • Differences in Humoral and Cellular Vaccine Responses to SARS-CoV-2 in Kidney and Liver Transplant Recipients. Frontiers in immunology Furian, L., Russo, F. P., Zaza, G., Burra, P., Hartzell, S., Bizzaro, D., Di Bello, M., Di Bella, C., Nuzzolese, E., Agnolon, C., Florman, S., Rana, M., Lee, J., Kim, Y., Maggiore, U., Maltzman, J. S., Cravedi, P. 2022; 13: 853682

    Abstract

    The antibody and T cell responses after SARS-CoV-2 vaccination have not been formally compared between kidney and liver transplant recipients. Using a multiplex assay, we measured IgG levels against 4 epitopes of SARS-CoV-2 spike protein and nucleocapsid (NC) antigen, SARS-CoV-2 variants, and common coronaviruses in serial blood samples from 52 kidney and 50 liver transplant recipients undergoing mRNA SARS-CoV-2 vaccination. We quantified IFN-gamma/IL-2 T cells reactive against SARS-CoV-2 spike protein by FluoroSpot. We used multivariable generalized linear models to adjust for the differences in immunosuppression between groups. In liver transplant recipients, IgG levels against every SARS-CoV-2 spike epitope increased significantly more than in kidney transplant recipients (MFI: 19,617 vs 6,056; P<0.001), a difference that remained significant after adjustments. Vaccine did not affect IgG levels against NC nor common coronaviruses. Elicited antibodies recognized all variants tested but at significantly lower strength than the original Wuhan strain. Anti-spike IFN-gamma-producing T cells increased significantly more in liver than in kidney transplant recipients (IFN-gamma-producing T cells 28 vs 11 spots/5x105 cells), but this difference lost statistical significance after adjustments. SARS-CoV-2 vaccine elicits a stronger antibody response in liver than in kidney transplant recipients, a phenomenon that is not entirely explained by the different immunosuppression.

    View details for DOI 10.3389/fimmu.2022.853682

    View details for PubMedID 35493446

  • Mitochondria-More than just ATP for CTLs. Science immunology Maltzman, J. S. 2021; 6 (65): eabn0249

    Abstract

    [Figure: see text].

    View details for DOI 10.1126/sciimmunol.eabn0249

    View details for PubMedID 34739341

  • Functional Consequences of Memory Inflation after Solid Organ Transplantation. Journal of immunology (Baltimore, Md. : 1950) Higdon, L. E., Schaffert, S., Cohen, R. H., Montez-Rath, M. E., Lucia, M., Saligrama, N., Margulies, K. B., Martinez, O. M., Tan, J. C., Davis, M. M., Khatri, P., Maltzman, J. S. 2021

    Abstract

    CMV is a major infectious complication following solid organ transplantation. Reactivation of CMV leads to memory inflation, a process in which CD8 T cells expand over time. Memory inflation is associated with specific changes in T cell function, including increased oligoclonality, decreased cytokine production, and terminal differentiation. To address whether memory inflation during the first year after transplantation in human subjects alters T cell differentiation and function, we employed single-cell-matched TCRalphabeta and targeted gene expression sequencing. Expanded T cell clones exhibited a terminally differentiated, immunosenescent, and polyfunctional phenotype whereas rare clones were less differentiated. Clonal expansion occurring between pre- and 3 mo posttransplant was accompanied by enhancement of polyfunctionality. In contrast, polyfunctionality and differentiation state were largely maintained between 3 and 12 mo posttransplant. Highly expanded clones had a higher degree of polyfunctionality than rare clones. Thus, CMV-responsive CD8 T cells differentiated during the pre- to posttransplant period then maintained their differentiation state and functional capacity despite posttransplant clonal expansion.

    View details for DOI 10.4049/jimmunol.2100405

    View details for PubMedID 34551963

  • Evolution of Cytomegalovirus-Responsive T Cell Clonality following Solid Organ Transplantation. Journal of immunology (Baltimore, Md. : 1950) Higdon, L. E., Schaffert, S., Huang, H., Montez-Rath, M. E., Lucia, M., Jha, A., Saligrama, N., Margulies, K. B., Martinez, O. M., Davis, M. M., Khatri, P., Maltzman, J. S. 2021

    Abstract

    CMV infection is a significant complication after solid organ transplantation. We used single cell TCR alphabeta sequencing to determine how memory inflation impacts clonality and diversity of the CMV-responsive CD8 and CD4 T cell repertoire in the first year after transplantation in human subjects. We observed CD8 T cell inflation but no changes in clonal diversity, indicating homeostatic stability in clones. In contrast, the CD4 repertoire was diverse and stable over time, with no evidence of CMV-responsive CD4 T cell expansion. We identified shared CDR3 TCR motifs among patients but no public CMV-specific TCRs. Temporal changes in clonality in response to transplantation and in the absence of detectable viral reactivation suggest changes in the repertoire immediately after transplantation followed by an expansion with stable clonal competition that may mediate protection.

    View details for DOI 10.4049/jimmunol.2100404

    View details for PubMedID 34551964

  • RNA splicing programs define tissue compartments and cell types at single-cell resolution ELIFE Olivieri, J., Dehghannasiri, R., Wang, P. L., Jang, S., de Morree, A., Tan, S. Y., Ming, J., Wu, A., Consortium, T., Quake, S. R., Krasnow, M. A., Salzman, J. 2021; 10
  • SARS-CoV-2 Vaccination and Antibody Testing in Immunosuppressed Populations: You Can't Tell the Players Without a Scorecard. Transplantation Woodle, E. S., Gebel, H. M., Montgomery, R. A., Maltzman, J. S. 2021

    View details for DOI 10.1097/TP.0000000000003882

    View details for PubMedID 34224542

  • SARS-CoV-2 Vaccination, Immune Responses, and Antibody Testing in Immunosuppressed Populations: Tip of the Iceberg. Transplantation Woodle, E. S., Gebel, H. M., Montgomery, R. A., Maltzman, J. S. 2021

    View details for DOI 10.1097/TP.0000000000003859

    View details for PubMedID 34144554

  • It's good to have Gremlins in the zone. Science immunology Gustafson, C. E., Maltzman, J. S. 2021; 6 (60)

    Abstract

    A unique subset of Gremlin1-expressing fibroblastic reticular cells mediate cDC homeostasis and functionality within lymph nodes.

    View details for DOI 10.1126/sciimmunol.abj7174

    View details for PubMedID 34088748

  • Association of Premature Immune Aging and Cytomegalovirus After Solid Organ Transplant FRONTIERS IN IMMUNOLOGY Higdon, L. E., Gustafson, C. E., Ji, X., Sahoo, M. K., Pinsky, B. A., Margulies, K. B., Maecker, H. T., Goronzy, J., Maltzman, J. S. 2021; 12
  • Association of Premature Immune Aging and Cytomegalovirus After Solid Organ Transplant. Frontiers in immunology Higdon, L. E., Gustafson, C. E., Ji, X., Sahoo, M. K., Pinsky, B. A., Margulies, K. B., Maecker, H. T., Goronzy, J., Maltzman, J. S. 2021; 12: 661551

    Abstract

    Immune function is altered with increasing age. Infection with cytomegalovirus (CMV) accelerates age-related immunological changes resulting in expanded oligoclonal memory CD8 T cell populations with impaired proliferation, signaling, and cytokine production. As a consequence, elderly CMV seropositive (CMV+) individuals have increased mortality and impaired responses to other infections in comparison to seronegative (CMV-) individuals of the same age. CMV is also a significant complication after organ transplantation, and recent studies have shown that CMV-associated expansion of memory T cells is accelerated after transplantation. Thus, we investigated whether immune aging is accelerated post-transplant, using a combination of telomere length, flow cytometry phenotyping, and single cell RNA sequencing. Telomere length decreased slightly in the first year after transplantation in a subset of both CMV+ and CMV- recipients with a strong concordance between CD57+ cells and short telomeres. Phenotypically aged cells increased post-transplant specifically in CMV+ recipients, and clonally expanded T cells were enriched for terminally differentiated cells post-transplant. Overall, these findings demonstrate a pattern of accelerated aging of the CD8 T cell compartment in CMV+ transplant recipients.

    View details for DOI 10.3389/fimmu.2021.661551

    View details for PubMedID 34122420

    View details for PubMedCentralID PMC8190404

  • Immunological yin-yang after pregnancy. Science immunology Maltzman, J. S. 2021; 6 (57)

    Abstract

    Pregnancy induces both humoral sensitization and T cell tolerance to paternal tissues.

    View details for DOI 10.1126/sciimmunol.abh3186

    View details for PubMedID 33674324

  • The ONE Study: One Small Step for Patient Care, a Giant Leap for Cell Therapy AMERICAN JOURNAL OF KIDNEY DISEASES Lamarche, C., Maltzman, J. S. 2021; 77 (2): 297–99
  • Anti-HLA and anti-SARS-CoV2 Antibodies in Kidney Transplant Recipient with COVID-19. Transplant international : official journal of the European Society for Organ Transplantation Gandolfini, I. n., Zanelli, P. n., Palmisano, A. n., Salvetti, D. n., Parmigiani, A. n., Maltzman, J. S., Labate, C. n., Fiaccadori, E. n., Cravedi, P. n., Maggiore, U. n. 2021

    Abstract

    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and its associated illness coronavirus disease 2019 (COVID-19) have severely affected organ transplant recipients, with all-cause mortality rates exceeding 20% (1, 2). Although no clear guidelines exist on how to adjust immunosuppression, most centers reduce anti-rejection drugs to facilitate the T and B cell response against the virus.

    View details for DOI 10.1111/tri.13829

    View details for PubMedID 33481306

  • Delayed Kinetics of IgG, but not IgA, Anti-spike Antibodies in Transplant Recipients following SARS-CoV-2 Infection. J Am Soc Nephrol. Cravedi, P. 2021

    View details for DOI 10.1681/ASN.2021040573

  • Aging Tregs need DCAFinating. Science immunology Dong, D., Maltzman, J. S. 2020; 5 (52)

    Abstract

    Loss of DCAF1 and resulting ROS leads to Treg aging and inflammation.

    View details for DOI 10.1126/sciimmunol.abe9581

    View details for PubMedID 33008917

  • Sestrins teach old T cells new tricks. Science immunology Maltzman, J. S. 2020; 5 (47)

    Abstract

    Sestrin proteins dampen TCR signaling and induce antigen-independent natural killer-like cytotoxicity in highly differentiated CD8 T cells.

    View details for DOI 10.1126/sciimmunol.abc4460

    View details for PubMedID 32358171

  • Development and Validation of a Multiplex, Bead-Based Assay to Detect Antibodies Directed Against SARS-CoV-2 Proteins. Transplantation Bray, R. A., Lee, J. H., Brescia, P. n., Kumar, D. n., Nong, T. n., Shih, R. n., Woodle, E. S., Maltzman, J. S., Gebel, H. M. 2020

    Abstract

    Transplant recipients who develop COVID-19 may be at increased risk for morbidity and mortality. Determining the status of antibodies against SARS-CoV-2 in both candidates and recipients will be important to understand the epidemiology and clinical course of COVID-19 in this population. While there are multiple tests to detect antibodies to SARS-CoV-2, their performance is variable. Tests vary according to their platforms and the antigenic targets which make interpretation of the results challenging. Furthermore, for some assays, sensitivity and specificity is less than optimal. Additionally, currently available serological tests do not exclude the possibility that positive responses are due to cross reactive antibodies to community coronaviruses rather than SARS-CoV-2.This study describes the development and validation of a high throughput multiplex antibody detection assay.The multiplex assay has the capacity to identify, simultaneously, patient responses to 5 SARS-CoV-2 proteins, namely, the full spike protein, 3 individual domains of the spike protein (S1, S2 and receptor binding domain) and the nucleocapsid protein. The antibody response to the above proteins are SARS-CoV-2 specific, as antibodies against 4 common coronaviruses do not cross-react.This new assay provides a novel tool to interrogate the spectrum of immune responses to SAR-CoV-2 and is uniquely suitable for use in the transplant setting. Test configuration is essentially identical to the single antigen bead assays used in the majority of histocompatibility laboratories around the world and could easily be implemented into routine screening of transplant candidates and recipients.

    View details for DOI 10.1097/TP.0000000000003524

    View details for PubMedID 33273320

  • The ONE Study: One Small Step for Patient Care, a Giant Leap for Cell Therapy. American journal of kidney diseases : the official journal of the National Kidney Foundation Lamarche, C. n., Maltzman, J. S. 2020

    View details for DOI 10.1053/j.ajkd.2020.07.006

    View details for PubMedID 32763258

  • To debug or not to debug, a question worth asking. Science immunology Higdon, L. E., Maltzman, J. S. 2019; 4 (41)

    Abstract

    Recipient antibiotic pre-treatment protects both mice and humans from ischemia-reperfusion injury after liver transplantation.

    View details for DOI 10.1126/sciimmunol.aaz9474

    View details for PubMedID 33681670

    View details for PubMedCentralID PMC7932049

  • To debug or not to debug, a question worth asking. Science immunology Higdon, L. E., Maltzman, J. S. 2019; 4 (41)

    Abstract

    Recipient antibiotic pretreatment protects both mice and humans from ischemia-reperfusion injury after liver transplantation.

    View details for DOI 10.1126/sciimmunol.aaz9474

    View details for PubMedID 31676498

  • Transplantation Alters Function and Clonality of Cytomegalovirus-Responsive T Cells Higdon, L. E., Schaffert, S., Saligrama, N., Davis, M. M., Khatri, P., Maltzman, J. S. LIPPINCOTT WILLIAMS & WILKINS. 2019
  • Duel(ling) precursors in thymic Treg development. Science immunology Maltzman, J. S. 2019; 4 (36)

    Abstract

    Functional thymus derived regulatory T cells can be generated by two distinct pathways.

    View details for DOI 10.1126/sciimmunol.aax9509

    View details for PubMedID 32619185

  • Single cell immune profiling in transplantation research AMERICAN JOURNAL OF TRANSPLANTATION Higdon, L. E., Schaffert, S., Khatri, P., Maltzman, J. S. 2019; 19 (5): 1278–87

    View details for DOI 10.1111/ajt.15316

    View details for Web of Science ID 000471342300007

  • Optimization of single-cell plate sorting for high throughput sequencing applications JOURNAL OF IMMUNOLOGICAL METHODS Higdon, L. E., Cain, C. J., Colden, M. A., Maltzman, J. S. 2019; 466: 17–23
  • Single cell immune profiling in transplantation research. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons Higdon, L. E., Schaffert, S., Khatri, P., Maltzman, J. S. 2019

    Abstract

    Recently developed single-cell profiling technologies hold promise to provide new insights including analysis of population heterogeneity and linkage of antigen receptors with gene expression. These technologies produce complex data sets that require knowledge of bioinformatics for appropriate analysis. In this minireview, we discuss several single-cell immune profiling technologies for gene and protein expression, including cytometry by time-of-flight, RNA sequencing, and antigen receptor sequencing, as well as key considerations for analysis that apply to each. Because of the critical importance of data analysis for high parameter single cell analysis, we discuss essential factors in analysis of these data, including quality control, quantification, examples of methods for high dimensional analysis, immune repertoire analysis, and preparation of analysis pipelines. We provide examples of, and suggestions for, application of these innovative methods to transplantation research. This article is protected by copyright. All rights reserved.

    View details for PubMedID 30768832

  • Duel(ling) precursors in thymic Treg development. Science immunology Maltzman, J. S. 2019; 4 (36)

    Abstract

    Functional thymus derived regulatory T cells can be generated by two distinct pathways.

    View details for DOI 10.1126/sciimmunol.aax9509

    View details for PubMedID 31175179

  • Optimization of single-cell plate sorting for high throughput sequencing applications. Journal of immunological methods Higdon, L. E., Cain, C. J., Colden, M. A., Maltzman, J. S. 2018

    Abstract

    Single cell sequencing has recently been applied to many immunological studies. Flow cytometric index sorting isolates cells for single cell sequencing with protein level data linked to sequences. However, successful sequencing of index sorted samples requires careful optimization of several sort parameters, including nozzle size, flow rate, threshold rate, and yield calculations. In this study, considerations and optimization data for each of these variables are presented. Our analysis focused on index sorting, but the findings can be applied to any plate sorting protocol. Minimization of flow rates and use of the 70 mum nozzle improved cell yields. Improvements in total read counts after sequencing were obtained by decreasing the threshold rate, or the number of cells processed per second. In addition, this technique provided linked protein and gene expression analysis of the cytokine interferon (IFN)gamma, demonstrating that on a single cell basis IFNgamma+ cells tend to express IFNG mRNA, and IFNgamma- cells do not. Through rigorous optimization and quality control, we have identified parameters important to plate sorting and recommend the use of the 70 mum nozzle and low flow and threshold rates for analysis of rare populations of human lymphocytes.

    View details for PubMedID 30590019

  • Inflammatory macrophage-associated 3-gene signature predicts subclinical allograft injury and graft survival. JCI insight Azad, T. D., Donato, M. n., Heylen, L. n., Liu, A. B., Shen-Orr, S. S., Sweeney, T. E., Maltzman, J. S., Naesens, M. n., Khatri, P. n. 2018; 3 (2)

    Abstract

    Late allograft failure is characterized by cumulative subclinical insults manifesting over many years. Although immunomodulatory therapies targeting host T cells have improved short-term survival rates, rates of chronic allograft loss remain high. We hypothesized that other immune cell types may drive subclinical injury, ultimately leading to graft failure. We collected whole-genome transcriptome profiles from 15 independent cohorts composed of 1,697 biopsy samples to assess the association of an inflammatory macrophage polarization-specific gene signature with subclinical injury. We applied penalized regression to a subset of the data sets and identified a 3-gene inflammatory macrophage-derived signature. We validated discriminatory power of the 3-gene signature in 3 independent renal transplant data sets with mean AUC of 0.91. In a longitudinal cohort, the 3-gene signature strongly correlated with extent of injury and accurately predicted progression of subclinical injury 18 months before clinical manifestation. The 3-gene signature also stratified patients at high risk of graft failure as soon as 15 days after biopsy. We found that the 3-gene signature also distinguished acute rejection (AR) accurately in 3 heart transplant data sets but not in lung transplant. Overall, we identified a parsimonious signature capable of diagnosing AR, recognizing subclinical injury, and risk-stratifying renal transplant patients. Our results strongly suggest that inflammatory macrophages may be a viable therapeutic target to improve long-term outcomes for organ transplantation patients.

    View details for PubMedID 29367465

  • The outstanding questions in transplantation: It's about time... AMERICAN JOURNAL OF TRANSPLANTATION Azzi, J., Raimondi, G., Mas, V., Riella, L. V., Elfadawy, N., Safa, K., Wojciechowski, D., Kanak, M., Nog, R., Maltzman, J. S., Ford, M. L., Pober, J. S., Luo, X., Rothstein, D., Miller, M. L., Matthews, D., Burlingham, W., Levings, M., Heeger, P., Higdon, L., Gill, J., Gill, R. G., Alegre, M. 2018; 18 (1): 271–72

    View details for PubMedID 28758364

  • Expanding the Toolkit for the Study of Allospecific B and T Cell Responses TRANSPLANTATION Higdon, L. E., Maltzman, J. S. 2017; 101 (11): 2661–62

    View details for PubMedID 29059129

    View details for PubMedCentralID PMC5724565

  • T cells expand after solid organ transplantation in the absence of CMV disease. American journal of transplantation Higdon, L. E., Trofe-Clark, J., Liu, S., Margulies, K. B., Sahoo, M. K., Blumberg, E., Pinsky, B. A., Maltzman, J. S. 2017

    Abstract

    Cytomegalovirus (CMV) is a major cause of morbidity and mortality in solid-organ transplant recipients. Approximately 60% of adults are CMV seropositive indicating previous exposure. Following resolution of primary infection, CMV remains in a latent state. Reactivation is controlled by memory T cells in healthy individuals; transplant recipients have reduced memory T cell function due to chronic immunosuppressive therapies. In this study, CD8(+) T cell responses to CMV polypeptides IE-1 and pp65 were analyzed in sixteen CMV seropositive renal and cardiac transplant recipients longitudinally pre- and post-transplant. All patients received standard of care maintenance immunosuppression, antiviral prophylaxis and CMV viral load monitoring, with approximately half receiving T cell depleting induction therapy. The frequency of CMV-responsive CD8(+) T cells, defined by production of effector molecules in response to CMV peptides, increased during the course of a year post-transplant. The increase commenced after the completion of antiviral prophylaxis, and these T cells tended to be terminally differentiated effector cells. Based on this small cohort, these data suggest that even in the absence of disease, antigenic exposure may continually shape the CMV-responsive T cell population post-transplant. This article is protected by copyright. All rights reserved.

    View details for DOI 10.1111/ajt.14227

    View details for PubMedID 28199780

  • Virtual Global Transplant Laboratory Standard Operating Procedures for Blood Collection, PBMC Isolation, and Storage. Transplantation direct Higdon, L. E., Lee, K., Tang, Q., Maltzman, J. S. 2016; 2 (9)

    Abstract

    Research on human immune responses frequently involves the use of peripheral blood mononuclear cells (PBMC) immediately, or at significantly delayed timepoints, after collection. This requires PBMC isolation from whole blood and cryopreservation for some applications. It is important to standardize protocols for blood collection, PBMC isolation, cryopreservation, and thawing that maximize survival and functionality of PBMC at the time of analysis. This resource includes detailed protocols describing blood collection tubes, isolation of PBMC using a density gradient, cryopreservation of PBMC, and thawing of cells as well as preparation for functional assays. For each protocol, we include important considerations, such as timing, storage temperatures, and freezing rate. In addition, we provide alternatives so that researchers can make informed decisions in determining the optimal protocol for their application.

    View details for PubMedID 27795993

  • Differentially Expressed Gene Transcripts Using RNA Sequencing from the Blood of Immunosuppressed Kidney Allograft Recipients PLOS ONE Dorr, C., Wu, B., Guan, W., Muthusamy, A., Sanghavi, K., Schladt, D. P., Maltzman, J. S., Scherer, S. E., Brott, M. J., Matas, A. J., Jacobson, P. A., Oetting, W. S., Israni, A. K. 2015; 10 (5)

    Abstract

    We performed RNA sequencing (RNAseq) on peripheral blood mononuclear cells (PBMCs) to identify differentially expressed gene transcripts (DEGs) after kidney transplantation and after the start of immunosuppressive drugs. RNAseq is superior to microarray to determine DEGs because it's not limited to available probes, has increased sensitivity, and detects alternative and previously unknown transcripts. DEGs were determined in 32 adult kidney recipients, without clinical acute rejection (AR), treated with antibody induction, calcineurin inhibitor, mycophenolate, with and without steroids. Blood was obtained pre-transplant (baseline), week 1, months 3 and 6 post-transplant. PBMCs were isolated, RNA extracted and gene expression measured using RNAseq. Principal components (PCs) were computed using a surrogate variable approach. DEGs post-transplant were identified by controlling false discovery rate (FDR) at < 0.01 with at least a 2 fold change in expression from pre-transplant. The top 5 DEGs with higher levels of transcripts in blood at week 1 were TOMM40L, TMEM205, OLFM4, MMP8, and OSBPL9 compared to baseline. The top 5 DEGs with lower levels at week 1 post-transplant were IL7R, KLRC3, CD3E, CD3D, and KLRC2 (Striking Image) compared to baseline. The top pathways from genes with lower levels at 1 week post-transplant compared to baseline, were T cell receptor signaling and iCOS-iCOSL signaling while the top pathways from genes with higher levels than baseline were axonal guidance signaling and LXR/RXR activation. Gene expression signatures at month 3 were similar to week 1. DEGs at 6 months post-transplant create a different gene signature than week 1 or month 3 post-transplant. RNAseq analysis identified more DEGs with lower than higher levels in blood compared to baseline at week 1 and month 3. The number of DEGs decreased with time post-transplant. Further investigations to determine the specific lymphocyte(s) responsible for differential gene expression may be important in selecting and personalizing immune suppressant drugs and may lead to targeted therapies.

    View details for DOI 10.1371/journal.pone.0125045

    View details for PubMedID 25946140

  • Regulatory T Cells Require TCR Signaling for Their Suppressive Function JOURNAL OF IMMUNOLOGY Schmidt, A. M., Lu, W., Sindhava, V. J., Huang, Y., Burkhardt, J. K., Yang, E., Riese, M. J., Maltzman, J. S., Jordan, M. S., Kambayashi, T. 2015; 194 (9): 4362-4370

    Abstract

    Regulatory T cells (Tregs) are a subset of CD4(+) T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4(+) T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase-like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.

    View details for DOI 10.4049/jimmunol.1402384

    View details for Web of Science ID 000353727400034

    View details for PubMedID 25821220

  • AST Cutting Edge of Transplantation 2013 Meeting Report: A Comprehensive Look at B Cells and Antibodies in Transplantation AMERICAN JOURNAL OF TRANSPLANTATION Mengel, M., Chong, A., Rothstein, D. M., Zorn, E., Maltzman, J. S. 2014; 14 (3): 524-530

    Abstract

    Antibody-mediated rejection (ABMR) represents a significant clinical challenge for solid organ transplantation. Mechanistic understanding of ABMR is incomplete and diagnostic accuracy for ABMR is limited, and as a result, targeted treatment remains elusive and new treatment modalities are difficult to validate. Three hundred twenty-six participants from 15 countries met for the first Cutting Edge of Transplantation (CEOT) symposium organized by the American Society of Transplantation (AST) in Chandler, Arizona, February 14-16, 2013. During the 3-day interactive symposium, presentations, moderated poster sessions and round table discussions addressed cutting edge knowledge of B and plasma cell biology, mechanisms of antibody-mediated tissue injury, advances and limitations in ABMR diagnostics, as well as current and potential new treatment options for ABMR. The outcome of the meeting identified the following unmet needs for: (a) improved understanding of the regulation of B cell maturation and antibody response to enable targeted therapies; (b) more precise diagnostics of ABMR, including molecular pathology, risk stratification by sensitive antibody testing and monitoring of treatment effects; and (c) innovative multicenter trial designs that enhance observational power, in particular, in assessing synergistic multimodality therapies with reduced toxicities.

    View details for DOI 10.1111/ajt.12593

    View details for Web of Science ID 000331783200007

    View details for PubMedID 24674597

  • T-Cell Costimulatory Blockade in Organ Transplantation COLD SPRING HARBOR PERSPECTIVES IN MEDICINE Maltzman, J. S., Turka, L. A. 2013; 3 (12)

    Abstract

    Before it became possible to derive T-cell lines and clones, initial experimentation on the activation requirements of T lymphocytes was performed on transformed cell lines, such as Jurkat. These studies, although technically correct, proved misleading as most transformed T cells can be activated by stimulation of the clonotypic T-cell receptor (TCR) alone. In contrast, once it became possible to study nontransformed T cells, it quickly became clear that TCR stimulation by itself is insufficient for optimal activation of naïve T cells, but in fact, induces a state of anergy. It then became clear that functional activation of T cells requires not only recognition of major histocompatibility complex (MHC) and peptide by the TCR, but also requires ligation of costimulatory receptors expressed on the cell surface.

    View details for DOI 10.1101/cshperspect.a015537

    View details for Web of Science ID 000328277800002

    View details for PubMedID 24296352

  • An obligate cell-intrinsic function for CD28 in Tregs JOURNAL OF CLINICAL INVESTIGATION Zhang, R., Huynh, A., Whitcher, G., Chang, J., Maltzman, J. S., Turka, L. A. 2013; 123 (2): 580-593

    Abstract

    Tregs expressing the transcription factor FOXP3 are critical for immune homeostasis. The costimulatory molecule CD28 is required for optimal activation and function of naive T cells; however, its role in Treg function has been difficult to dissect, as CD28 is required for thymic Treg development, and blockade of CD28-ligand interactions has confounding effects in trans on nonregulatory cells. To address this question, we created Treg-specific Cd28 conditional knockout mice. Despite the presence of normal numbers of FOXP3+ cells, these animals accumulated large numbers of activated T cells, developed severe autoimmunity that primarily affected the skin and lungs, and failed to appropriately resolve induced experimental allergic encephalomyelitis. This in vivo functional impairment was accompanied by dampened expression of CTLA-4, PD-1, and CCR6. Disease occurrence was not due to subversion of Cd28-deficient Tregs into pathogenic cells, as complementation with normal Tregs prevented disease occurrence. Interestingly, in these "competitive" environments, Cd28-deficient Tregs exhibited a pronounced proliferative/survival disadvantage. These data demonstrate clear postmaturational roles for CD28 in FOXP3+ Tregs and provide mechanisms which we believe to be novel to explain how interruption of CD28-ligand interactions may enhance immune responses independent of effects on thymic development or on other cell types.

    View details for DOI 10.1172/JCI65013

    View details for Web of Science ID 000314553600015

    View details for PubMedID 23281398

  • Homeostatic Division Is Not Necessary for Antigen-Specific CD4(+) Memory T Cell Persistence JOURNAL OF IMMUNOLOGY Corbo-Rodgers, E., Wiehagen, K. R., Staub, E. S., Maltzman, J. S. 2012; 189 (7): 3378-3385

    Abstract

    CD4(+) memory T cells are generated in response to infection or vaccination, provide protection to the host against reinfection, and persist through a combination of enhanced survival and slow homeostatic turnover. We used timed deletion of the TCR-signaling adaptor molecule Src homology 2 domain-containing phosphoprotein of 76 kDa (SLP-76) with MHC:peptide tetramers to study the requirements for tonic TCR signals in the maintenance of polyclonal Ag-specific CD4(+) memory T cells. SLP-76-deficient I-A(b):gp61 cells are unable to rapidly generate effector cytokines or proliferate in response to secondary infection. In mice infected with lymphocytic choriomeningitis virus (LCMV) or Listeria monocytogenes expressing the LCMV gp61-80 peptide, SLP-76-deficient I-A(b):gp61(+) cells exhibit reduced division, similar to that seen in in vitro-generated CD44(hi) and endogenous CD4(+)CD44(hi) cells. Competitive bone marrow chimera experiments demonstrated that the decrease in homeostatic turnover in the absence of SLP-76 is a cell-intrinsic process. Surprisingly, despite the reduction in turnover, I-A(b):gp61(+) Ag-specific memory cells persist in normal numbers for >30 wk after LCMV infection in the absence of SLP-76. These data suggest the independent maintenance of a population of Ag-specific CD4(+) memory T cells in the absence of SLP-76 and normal levels of homeostatic division.

    View details for DOI 10.4049/jimmunol.1201583

    View details for Web of Science ID 000309164300013

    View details for PubMedID 22956580

  • Foxp4 Is Dispensable for T Cell Development, but Required for Robust Recall Responses PLOS ONE Wiehagen, K. R., Corbo-Rodgers, E., Li, S., Staub, E. S., Hunter, C. A., Morrisey, E. E., Maltzman, J. S. 2012; 7 (8)

    Abstract

    Transcription factors regulate T cell fates at every stage of development and differentiation. Members of the Foxp family of forkhead transcription factors are essential for normal T lineage development; Foxp3 is required for T regulatory cell generation and function, and Foxp1 is necessary for generation and maintenance of naïve T cells. Foxp4, an additional member of the Foxp family, is highly homologous to Foxp1 and has been shown to dimerize with other Foxp proteins. We report the initial characterization of Foxp4 in T lymphocytes. Foxp4 is expressed in both thymocytes and peripheral CD4(+) and CD8(+) T cells. We used a CD4Cre mediated approach to evaluate the cell autonomous role for Foxp4 in murine T lymphocytes. T cell development, peripheral cellularity and cell surface phenotype are normal in the absence of Foxp4. Furthermore, Foxp3(+) T regulatory cells develop normally in Foxp4 deficient animals and naïve Foxp4 deficient CD4 T cells can differentiate to inducible T regulatory cells in vitro. In wild-type T cells, expression of Foxp4 increases following activation, but deletion of Foxp4 does not affect T cell proliferative responses or in vitro effector T cell differentiation. In vivo, despite effective control of Toxoplasma gondii and acute lymphocytic choriomeningitis virus infections, effector cytokine production during antigen specific recall responses are reduced in the absence of Foxp4. We conclude that Foxp4 is dispensable for T cell development, but necessary for normal T cell cytokine recall responses to antigen following pathogenic infection.

    View details for DOI 10.1371/journal.pone.0042273

    View details for Web of Science ID 000307500100010

    View details for PubMedID 22912696

  • Cutting Edge: IL-2 Signals Determine the Degree of TCR Signaling Necessary To Support Regulatory T Cell Proliferation In Vivo JOURNAL OF IMMUNOLOGY Zou, T., Satake, A., Corbo-Rodgers, E., Schmidt, A. M., Farrar, M. A., Maltzman, J. S., Kambayashi, T. 2012; 189 (1): 28-32

    Abstract

    To ensure immune tolerance, regulatory T cell (Treg) numbers must be maintained by cell division. This process has been thought to be strictly dependent on the Treg TCR interacting with MHC class II. In this study, we report that Treg division does not absolutely require cell-autonomous TCR signaling in vivo, depending on the degree of IL-2-mediated stimulation provided. At steady state IL-2 levels, Tregs require cell-autonomous TCR signaling to divide. However, when given exogenous IL-2 or when STAT5 is selectively activated in Tregs, Treg division can occur independently of MHC class II and TCR signaling. Thus, depending on the amount of IL-2R stimulation, a wide range of TCR signals supports Treg division, which may contribute to preservation of a diverse repertoire of Treg TCR specificities. These findings also have therapeutic implications, as TCR signaling by Tregs may not be required when using IL-2 to increase Treg numbers for treatment of inflammatory disorders.

    View details for DOI 10.4049/jimmunol.1200507

    View details for Web of Science ID 000305636000005

    View details for PubMedID 22623329

  • Site-specific accumulation of recently activated CD4+Foxp3+ regulatory T cells following adoptive transfer EUROPEAN JOURNAL OF IMMUNOLOGY Lieberman, S. M., Kim, J. S., Corbo-Rodgers, E., Kambayashi, T., Maltzman, J. S., Behrens, E. M., Turka, L. A. 2012; 42 (6): 1429-1435

    Abstract

    CD4(+) Foxp3(+) regulatory T (Treg) cells are required for the maintenance of self-tolerance, as demonstrated by profound autoimmunity in mice and humans with inactivating Foxp3 mutations. Recent studies demonstrate that Treg cells are anatomically compartmentalized within secondary lymphoid organs based on their TCR repertoire and specific organ-protective function; however, whether this reflects differential homing or in situ selection is not known. Here, using Foxp3-GFP reporter mice, we have examined the ability of polyclonal Treg cells from cervical LNs to return to their site-of-origin following adoptive transfer to nonlymphopenic congenic recipients. We find that bulk cervical LN Treg cells do not home directly to cervical LNs but rather accumulate site specifically over time following transfer. Site-specific enrichment is both more rapid and more pronounced among a population of recently activated (CD69(+) ) Treg cells. These data suggest that compartmentalization of Treg cells within secondary lymphoid organs may be governed by antigen recognition and implicate CD69 as a potential marker of recently activated Treg cells recognizing locally expressed antigens.

    View details for DOI 10.1002/eji.201142286

    View details for Web of Science ID 000304997800008

    View details for PubMedID 22678899

  • Oral ivermectin as an unexpected initiator of CreT2-mediated deletion in T cells NATURE IMMUNOLOGY Corbo-Rodgers, E., Staub, E. S., Zou, T., Smith, A., Kambayashi, T., Maltzman, J. S. 2012; 13 (3): 197-198

    View details for Web of Science ID 000300510600001

    View details for PubMedID 22344272

  • Probucol ameliorates renal and metabolic sequelae of primary CoQ deficiency in Pdss2 mutant mice EMBO MOLECULAR MEDICINE Falk, M. J., Polyak, E., Zhang, Z., Peng, M., King, R., Maltzman, J. S., Okwuego, E., Horyn, O., Nakamaru-Ogiso, E., Ostrovsky, J., Xie, L. X., Chen, J. Y., Marbois, B., Nissim, I., Clarke, C. F., Gasser, D. L. 2011; 3 (7): 410-427

    Abstract

    Therapy of mitochondrial respiratory chain diseases is complicated by limited understanding of cellular mechanisms that cause the widely variable clinical findings. Here, we show that focal segmental glomerulopathy-like kidney disease in Pdss2 mutant animals with primary coenzyme Q (CoQ) deficiency is significantly ameliorated by oral treatment with probucol (1% w/w). Preventative effects in missense mutant mice are similar whether fed probucol from weaning or for 3 weeks prior to typical nephritis onset. Furthermore, treating symptomatic animals for 2 weeks with probucol significantly reduces albuminuria. Probucol has a more pronounced health benefit than high-dose CoQ(10) supplementation and uniquely restores CoQ(9) content in mutant kidney. Probucol substantially mitigates transcriptional alterations across many intermediary metabolic domains, including peroxisome proliferator-activated receptor (PPAR) pathway signaling. Probucol's beneficial effects on the renal and metabolic manifestations of Pdss2 disease occur despite modest induction of oxidant stress and appear independent of its hypolipidemic effects. Rather, decreased CoQ(9) content and altered PPAR pathway signaling appear, respectively, to orchestrate the glomerular and global metabolic consequences of primary CoQ deficiency, which are both preventable and treatable with oral probucol therapy.

    View details for DOI 10.1002/emmm.201100149

    View details for Web of Science ID 000293277400005

    View details for PubMedID 21567994

  • Conditional deletion of SLP-76 in mature T cells abrogates peripheral immune responses EUROPEAN JOURNAL OF IMMUNOLOGY Wu, G. F., Corbo, E., Schmidt, M., Smith-Garvin, J. E., Riese, M. J., Jordan, M. S., Laufer, T. M., Brown, E. J., Maltzman, J. S. 2011; 41 (7): 2064-2073

    Abstract

    The adaptor protein Src homology 2 domain-containing leukocyte-specific protein of 76 kDa (SLP-76) is central to the organization of intracellular signaling downstream of the T-cell receptor (TCR). Evaluation of its role in mature, primary T cells has been hampered by developmental defects that occur in the absence of WT SLP-76 protein in thymocytes. Here, we show that following tamoxifen-regulated conditional deletion of SLP-76, mature, antigen-inexperienced T cells maintain normal TCR surface expression but fail to transduce TCR-generated signals. Conditionally deficient T cells fail to proliferate in response to antigenic stimulation or a lymphopenic environment. Mice with induced deletion of SLP-76 are resistant to induction of the CD4+ T-cell-mediated autoimmune disease experimental autoimmune encephalomyelitis. Altogether, our findings demonstrate the critical role of SLP-76-mediated signaling in initiating T-cell-directed immune responses both in vitro and in vivo and highlight the ability to analyze signaling processes in mature T cells in the absence of developmental defects.

    View details for DOI 10.1002/eji.201040809

    View details for Web of Science ID 000293131200027

    View details for PubMedID 21469089

  • T cell receptor signal strength in T-reg and iNKT cell development demonstrated by a novel fluorescent reporter mouse JOURNAL OF EXPERIMENTAL MEDICINE Moran, A. E., Holzapfel, K. L., Xing, Y., Cunningham, N. R., Maltzman, J. S., Punt, J., Hogquist, K. A. 2011; 208 (6): 1279-1289

    Abstract

    The ability of antigen receptors to engage self-ligands with varying affinity is crucial for lymphocyte development. To further explore this concept, we generated transgenic mice expressing GFP from the immediate early gene Nr4a1 (Nur77) locus. GFP was up-regulated in lymphocytes by antigen receptor stimulation but not by inflammatory stimuli. In T cells, GFP was induced during positive selection, required major histocompatibility complex for maintenance, and directly correlated with the strength of T cell receptor (TCR) stimulus. Thus, our results define a novel tool for studying antigen receptor activation in vivo. Using this model, we show that regulatory T cells (T(reg) cells) and invariant NKT cells (iNKT cells) perceived stronger TCR signals than conventional T cells during development. However, although T(reg) cells continued to perceive strong TCR signals in the periphery, iNKT cells did not. Finally, we show that T(reg) cell progenitors compete for recognition of rare stimulatory TCR self-ligands.

    View details for DOI 10.1084/jem.20110308

    View details for Web of Science ID 000291424200014

    View details for PubMedID 21606508

  • Antiviral memory CD8 T-cell differentiation, maintenance, and secondary expansion occur independently of MyD88 BLOOD Rahman, A. H., Zhang, R., Blosser, C. D., Hou, B., DeFranco, A. L., Maltzman, J. S., Wherry, E. J., Turka, L. A. 2011; 117 (11): 3123-3130

    Abstract

    Inflammatory signals induced during infection regulate T-cell expansion, differentiation, and memory formation. Toll-like receptors (TLRs) are inflammatory mediators that allow innate immune cells to recognize and respond to invading pathogens. In addition to their role in innate immune cells, we have found that signals delivered through the TLR adapter protein myeloid differentiation protein 88 (MyD88) play a critical, T cell-intrinsic role in supporting the survival and accumulation of antigen-specific effector cells after acute viral infection. However, the importance of MyD88-dependent signals in regulating the generation and maintenance of memory T cells remained unclear. To address this, we used a novel, inducible knockout system to examine whether MyD88 is required for optimal memory CD8 T-cell generation and responses after lymphocytic choriomeningitis virus infection. We show that whereas MyD88 is critical for initial T-cell expansion, it is not required for the subsequent differentiation and stable maintenance of a memory T-cell population. Furthermore, in contrast to naive CD8 T cells, memory CD8 T cells do not depend on MyD88 for their secondary expansion. Our findings clarify the importance of MyD88 during distinct phases of the antiviral T-cell response and establish differential dependence on MyD88 signaling as a novel characteristic that distinguishes naive from memory CD8 T cells.

    View details for DOI 10.1182/blood-2010-11-318485

    View details for Web of Science ID 000288496300022

    View details for PubMedID 21233312

  • Loss of tonic T-cell receptor signals alters the generation but not the persistence of CD8(+) memory T cells BLOOD Wiehagen, K. R., Corbo, E., Schmidt, M., Shin, H., Wherry, E. J., Maltzman, J. S. 2010; 116 (25): 5560-5570

    Abstract

    The requirements for tonic T-cell receptor (TCR) signaling in CD8(+) memory T-cell generation and homeostasis are poorly defined. The SRC homology 2 (SH2)-domain-containing leukocyte protein of 76 kDa (SLP-76) is critical for proximal TCR-generated signaling. We used temporally mediated deletion of SLP-76 to interrupt tonic and activating TCR signals after clearance of the lymphocytic choriomeningitis virus (LCMV). SLP-76-dependent signals are required during the contraction phase of the immune response for the normal generation of CD8 memory precursor cells. Conversely, LCMV-specific memory CD8 T cells generated in the presence of SLP-76 and then acutely deprived of TCR-mediated signals persist in vivo in normal numbers for more than 40 weeks. Tonic TCR signals are not required for the transition of the memory pool toward a central memory phenotype, but the absence of SLP-76 during memory homeostasis substantially alters the kinetics. Our data are consistent with a model in which tonic TCR signals are required at multiple stages of differentiation, but are dispensable for memory CD8 T-cell persistence.

    View details for DOI 10.1182/blood-2010-06-292458

    View details for Web of Science ID 000285383900019

    View details for PubMedID 20884806

  • Ablation of SLP-76 signaling after T cell priming generates memory CD4 T cells impaired in steady-state and cytokine-driven homeostasis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Bushar, N. D., Corbo, E., Schmidt, M., Maltzman, J. S., Farber, D. L. 2010; 107 (2): 827-831

    Abstract

    The intracellular signaling mechanisms regulating the generation and long-term persistence of memory T cells in vivo remain unclear. In this study, we used mouse models with conditional deletion of the key T cell receptor (TCR)-coupled adaptor molecule SH2-domain-containing phosphoprotein of 76 kDa (SLP-76), to analyze signaling mechanisms for memory CD4 T cell generation, maintenance, and homeostasis. We found that ablation of SLP-76 expression after T cell priming did not inhibit generation of phenotypic effector or memory CD4 T cells; however, the resultant SLP-76-deficient memory CD4 T cells could not produce recall cytokines in response to TCR-mediated stimulation and showed decreased persistence in vivo. In addition, SLP-76-deficient memory CD4 T cells exhibited reduced steady-state homeostasis and were impaired in their ability to homeostatically expand in vivo in response to the gamma(c) cytokine IL-7, despite intact proximal signaling through the IL-7R-coupled JAK3/STAT5 pathway. Direct in vivo deletion of SLP-76 in polyclonal memory CD4 T cells likewise led to impaired steady-state homeostasis as well as impaired homeostatic responses to IL-7. Our findings demonstrate a dominant role for SLP-76-dependent TCR signals in regulating turnover and perpetuation of memory CD4 T cells and their responses to homeostatic cytokines, with implications for the selective survival of memory CD4 T cells following pathogen exposure, vaccination, and aging.

    View details for DOI 10.1073/pnas.0908126107

    View details for Web of Science ID 000273559300057

    View details for PubMedID 20080760

  • Conditional gene expression: A new tool for the transplantologist AMERICAN JOURNAL OF TRANSPLANTATION Maltzman, J. S., Turka, L. A. 2007; 7 (4): 733-740

    Abstract

    The ability to generate genetically manipulated mice has revolutionized the study of development, cell biology, immunobiology and transplantation. Conventional gene targeting approaches lead to inactivation of the target gene in all tissues. This approach often has unintended consequences, such as embryonic lethality, which preclude studying the originally intended tissue. Newer approaches allowing conditional gene expression in a tissue-specific or temporally controlled fashion have the advantage of normal development with gene deletion only in the desired tissues. While nuances to these techniques continue to be developed, the underlying concepts remain consistent. This minireview focuses on the use of conditional gene targeting in mice using the Cre-loxP system and drug inducible gene expression using the tetracycline system.

    View details for DOI 10.1111/j.1600-6143.2006.01685.x

    View details for Web of Science ID 000245151500002

    View details for PubMedID 17391118

  • Conditional deletion reveals a cell-autonomous requirement of SLP-76 for thymocyte selection JOURNAL OF EXPERIMENTAL MEDICINE Maltzman, J. S., Kovoor, L., Clements, J. L., Koretzky, G. A. 2005; 202 (7): 893-900

    Abstract

    The SH2 domain containing leukocyte phosphoprotein of 76 kD (SLP-76) is critical for pre-TCR-mediated maturation to the CD4+CD8+ double positive (DP) stage in the thymus. The absolute block in SLP-76null mice at the CD4-CD8-CD44-CD25+ (double-negative 3, DN3) stage has hindered our understanding of the role of this adaptor in alphabeta TCR-mediated signal transduction in primary thymocytes and peripheral T lymphocytes. To evaluate the requirements for SLP-76 in these events, we used a cre-loxP approach to generate mice that conditionally delete SLP-76 after the DN3 checkpoint. These mice develop DP thymocytes that express the alphabeta TCR on the surface, but lack SLP-76 at the genomic DNA and protein levels. The DP compartment has reduced cellularity in young mice and fails to undergo positive selection to CD4+ or CD8+ single positive (SP) cells in vivo or activation-induced cell death in vitro. A small number of CD4+SP thymocytes are generated, but these cells fail to flux calcium in response to an alphabeta TCR-generated signal. Peripheral T cells are reduced in number, lack SLP-76 protein, and have an abnormal surface phenotype. These studies show for the first time that SLP-76 is required for signal transduction through the mature alphabeta TCR in primary cells of the T lineage.

    View details for DOI 10.1084/jem.20051128

    View details for Web of Science ID 000232619000003

    View details for PubMedID 16186188

  • Role of EGR1 in regulation of stimulus-dependent CD44 transcription in B lymphocyte MOLECULAR AND CELLULAR BIOLOGY Maltzman, J. S., Carman, J. A., Monroe, J. G. 1996; 16 (5): 2283-2294

    Abstract

    The immediate-early gene egr-1 encodes a transcription factor (EGR1) that links B-cell antigen receptor (BCR) signals to downstream activation events through the regulation of previously unidentified target genes. Here we identify the gene encoding the lymphocyte homing and migration protein CD44 as a target of EGR1 regulation in B cells. BCR-induced increases in CD44 mRNA expression and transcription levels are shown to occur in EGR1-expressing but not in nonexpressing subclones of the B-cell line WEHI-231. Kinetics of egr-1 transcription and the appearance of nuclear EGR1 protein precede CD44 induction and occur within 30 min after stimulation in the EGR1-expressing subclone. A single EGR1 binding motif is demonstrated at bp -301 of the human CD44 promoter. Cotransfection of a CD44 promoter-chloramphenicol acetyltransferase reporter construct with an egr-1 expression vector resulted in a 6.5- to 8.5-fold induction of transcriptional activity relative to an empty expression vector. The EGR1 binding motif was shown to be necessary for stimulus-induced expression of a CD44 promoter-chloramphenicol acetyltransferase reporter construct in nontransformed B lymphocytes and was required for transactivation by an EGR1 expression vector in a B-cell line. These studies identify EGR1 as an intermediary linking BCR-derived signals to the induction of CD44. The relevance of these molecular events to BCR signal transduction and antigen-stimulated B-cell-mediated immune responses is discussed.

    View details for Web of Science ID A1996UG29900042

    View details for PubMedID 8628295

  • Transcriptional regulation of the Icam-1 gene in antigen receptor- and phorbol ester-stimulated B lymphocytes: Role for transcription factor EGR1 JOURNAL OF EXPERIMENTAL MEDICINE Maltzman, J. S., Carman, J. A., Monroe, J. G. 1996; 183 (4): 1747-1759

    Abstract

    Intercellular adhesion molecule (ICAM) 1/CD54 plays an important role in T cell dependent B cell activation and for function of B lymphocytes as antigen-presenting cells. ICAM-1 expression is upregulated as a consequence of B lymphocyte antigen receptor (BCR) signaling, thereby serving to render antigen-stimulated B cells more receptive to T cell-mediated costimulatory signals. We have investigated BCR-induced expression of the Icam-1 gene in primary B cells and B cell lines and have found it to be dependent on BCR-induced expression of the transcription factor EGR1. Icam-1 transcription, induced by BCR cross-linking or bypassing the BCR with phorbol ester, is absent in a B cell line in which the EGR1-encoding gene (egr-1) is methylated and not expressed. A potential EGR1-binding site was located at -701 bp upstream of the murine Icam-1 gene transcription start site and shown by electrophoretic mobility shift assay to bind to murine EGR1. Mutation of this site in the context of 1.1 kb of the Icam-1 promoter significantly abrogated transcriptional induction by phorbol ester and anti-mu stimulation in primary B cells. A direct effect of EGR1 on the Icam-1 promoter is suggested by the ability of EGR1 expressed from an SV40-driven expression vector transactivate the wild-type Icam-1 promoter, whereas mutation of the EGR1 mutation of the EGR1 binding motif at -701 bp markedly compromises this induction. These data identify EGR1 as a signaling intermediate in BCR-stimulated B cell functional responses, specifically linking BCR signal transduction to induction of the Icam-1 gene. Furthermore, similar findings for BCR-induced CD44 gene induction (Maltzman, J.S., J.A. Carman, and J.G. Monroe. 1996. Role of EGR1 in regulation of stimulus-dependent CD44 transcription in B lymphocytes. Mol. Cell. Biol. In press) suggest that EGR1 may be an important signaling molecule for regulating levels of migration and adhesion molecules during humoral immune responses.

    View details for Web of Science ID A1996UH14400048

    View details for PubMedID 8666932

  • MOLECULAR EVIDENCE FOR THE EXPRESSION OF NICOTINIC ACETYLCHOLINE-RECEPTOR ALPHA-CHAIN IN MOUSE THYMUS JOURNAL OF IMMUNOLOGY Wheatley, L. M., Urso, D., TUMAS, K., Maltzman, J., Loh, E., Levinson, A. I. 1992; 148 (10): 3105-3109

    Abstract

    The presence and structure of nicotinic acetylcholine receptor (nAChR) in the thymus has been a subject of interest for many years because of its possible role in the pathogenesis of the autoimmune disease myasthenia gravis. Using the polymerase chain reaction with primers specific for the alpha-chain of nAChR (nAChR-alpha), an 880-bp homologous band was found after amplification of cDNA prepared from mouse thymus, thymic medullary and cortical epithelial cell lines, but not from thymocytes or kidney. Sequencing of the polymerase chain reaction product from the thymus and thymic medullary and cortical epithelial lines showed identity with skeletal muscle nAChR-alpha over the region examined. This region includes the domains of the molecule on which B cell and T cell autoantigenic targets have been described. No evidence was found in mouse tissue for the exon 3A, which has been described in human muscle and the human rhabdomyosarcoma cell line TE671. Our results provide evidence at the RNA level for the expression of the nAChR-alpha on stromal cells but not on thymocytes in normal murine thymus and are consistent with a role for intrathymic autoantigen expression in the pathogenesis of myasthenia gravis.

    View details for Web of Science ID A1992HU68600019

    View details for PubMedID 1578136

  • EXPRESSION OF THE V-BETA-5.1 GENE BY MURINE PERIPHERAL T-CELLS IS CONTROLLED BY MHC GENES AND SKEWED TO THE CD8+ SUBSET JOURNAL OF IMMUNOLOGY Liao, N. S., Maltzman, J., Raulet, D. H. 1990; 144 (3): 844-848

    Abstract

    We have examined the usage of the V beta 5.1 region in peripheral T cell populations of several mouse strains by measuring V beta 5.1 transcript levels. The results show that V beta 5.1 is expressed predominantly by CD8+ T cells in mice of several MHC haplotypes. This finding suggests that V beta 5.1 may confer restriction to class 1 molecules present in these strains. Interestingly, however, T cells expressing V beta 5.1, like those expressing V beta 17a and V beta 11, are clonally deleted from both CD4 and CD8 subsets in mice that express MHC IE molecules. The latter result implies that V beta 5.1 confers reactivity to a class 2 molecule (IE). These results are discussed in terms of positive selection and clonal deletion. Finally we provide evidence that genes in the K or A regions of the MHC also control V beta 5.1 usage.

    View details for Web of Science ID A1990CL02700011

    View details for PubMedID 1967275

  • POSITIVE SELECTION DETERMINES T-CELL RECEPTOR V-BETA-14 GENE USAGE BY CD8+ T-CELLS JOURNAL OF EXPERIMENTAL MEDICINE Liao, N. S., Maltzman, J., Raulet, D. H. 1989; 170 (1): 135-143

    Abstract

    We report here a mAb, 14-2, reactive with TCRs that include V beta 14. The frequency of V beta 14+ T cells varies with CD4 and CD8 subset and is controlled by the H-2 genes. Thus CD8+ T cells from H-2b mice include approximately 2.3% V beta 14+ T cells while CD8+ T cells from mice expressing K kappa include greater than 8% V beta 14+ T cells. In all strains examined, 7-8% of CD4+ T cells express V beta 14. The frequent usage of V beta 14 in CD8+ T cells of K kappa-expressing mice is a result of preferential positive selection of V beta 14+ CD8+ T cells as demonstrated by analysis of radiation chimeras. These studies demonstrate that H-2-dependent positive selection occurs in unmanipulated mice. Furthermore, the results imply that positive selection, and possibly H-2 restriction, can be strongly influenced by a V beta domain, with some independence from the beta-junctional sequence and alpha chain.

    View details for Web of Science ID A1989AE68500010

    View details for PubMedID 2501444