1
|
He C, Hua G, Liu Y, Li S. Unveiling the hidden role of the interaction between CD36 and FcγRIIb: implications for autoimmune disorders. Cell Mol Biol Lett 2024; 29:76. [PMID: 38762740 PMCID: PMC11102138 DOI: 10.1186/s11658-024-00593-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND The role of the scavenger receptor CD36 in cell metabolism and the immune response has been investigated mainly in macrophages, dendritic cells, and T cells. However, its involvement in B cells has not been comprehensively examined. METHODS To investigate the function of CD36 in B cells, we exposed Cd36fl/flMB1cre mice, which lack CD36 specifically in B cells, to apoptotic cells to trigger an autoimmune response. To validate the proteins that interact with CD36 in primary B cells, we conducted mass spectrometry analysis following anti-CD36 immunoprecipitation. Immunofluorescence and co-immunoprecipitation were used to confirm the protein interactions. RESULTS The data revealed that mice lacking CD36 in B cells exhibited a reduction in germinal center B cells and anti-DNA antibodies in vivo. Mass spectrometry analysis identified 30 potential candidates that potentially interact with CD36. Furthermore, the interaction between CD36 and the inhibitory Fc receptor FcγRIIb was first discovered by mass spectrometry and confirmed through immunofluorescence and co-immunoprecipitation techniques. Finally, deletion of FcγRIIb in mice led to decreased expression of CD36 in marginal zone B cells, germinal center B cells, and plasma cells. CONCLUSIONS Our data indicate that CD36 in B cells is a critical regulator of autoimmunity. The interaction of CD36-FcγRIIb has the potential to serve as a therapeutic target for the treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Chenfei He
- Center for Research in Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Guoying Hua
- Center for Research in Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yong Liu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna Campus, Stockholm, Sweden
| | - Shuijie Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, China.
- Heilongjiang Province Key Laboratory of Research On Molecular Targeted Anti-Tumor Drugs, Harbin, China.
| |
Collapse
|
2
|
Bodansky A, Yu DJ, Rallistan A, Kalaycioglu M, Boonyaratanakornkit J, Green DJ, Gauthier J, Turtle CJ, Zorn K, O’Donovan B, Mandel-Brehm C, Asaki J, Kortbawi H, Kung AF, Rackaityte E, Wang CY, Saxena A, de Dios K, Masi G, Nowak RJ, O’Connor KC, Li H, Diaz VE, Saloner R, Casaletto KB, Gontrum EQ, Chan B, Kramer JH, Wilson MR, Utz PJ, Hill JA, Jackson SW, Anderson MS, DeRisi JL. Unveiling the proteome-wide autoreactome enables enhanced evaluation of emerging CAR T cell therapies in autoimmunity. J Clin Invest 2024; 134:e180012. [PMID: 38753445 PMCID: PMC11213466 DOI: 10.1172/jci180012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
Given the global surge in autoimmune diseases, it is critical to evaluate emerging therapeutic interventions. Despite numerous new targeted immunomodulatory therapies, comprehensive approaches to apply and evaluate the effects of these treatments longitudinally are lacking. Here, we leveraged advances in programmable-phage immunoprecipitation methodology to explore the modulation, or lack thereof, of autoantibody profiles, proteome-wide, in both health and disease. Using a custom set of over 730,000 human-derived peptides, we demonstrated that each individual, regardless of disease state, possesses a distinct and complex constellation of autoreactive antibodies. For each individual, the set of resulting autoreactivites constituted a unique immunological fingerprint, or "autoreactome," that was remarkably stable over years. Using the autoreactome as a primary output, we evaluated the relative effectiveness of various immunomodulatory therapies in altering autoantibody repertoires. We found that therapies targeting B cell maturation antigen (BCMA) profoundly altered an individual's autoreactome, while anti-CD19 and anti-CD20 therapies had minimal effects. These data both confirm that the autoreactome comprises autoantibodies secreted by plasma cells and strongly suggest that BCMA or other plasma cell-targeting therapies may be highly effective in treating currently refractory autoantibody-mediated diseases.
Collapse
Affiliation(s)
- Aaron Bodansky
- Department of Pediatrics, Division of Critical Care, and
| | - David J.L. Yu
- Diabetes Center, School of Medicine, UCSF, San Francisco, California, USA
| | - Alysa Rallistan
- Department of Medicine, Division of Immunology and Rheumatology, and
| | - Muge Kalaycioglu
- Institute of Immunity, Transplantation, and Infection, Stanford University, Stanford, California, USA
| | - Jim Boonyaratanakornkit
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Damian J. Green
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Jordan Gauthier
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Cameron J. Turtle
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | | - Hannah Kortbawi
- Department of Biochemistry and Biophysics
- Medical Scientist Training Program, and
| | - Andrew F. Kung
- Department of Biochemistry and Biophysics
- Biological and Medical Informatics Program, UCSF, San Francisco, California, USA
| | | | - Chung-Yu Wang
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| | - Aditi Saxena
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| | - Kimberly de Dios
- Diabetes Center, School of Medicine, UCSF, San Francisco, California, USA
| | - Gianvito Masi
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Richard J. Nowak
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kevin C. O’Connor
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Hao Li
- Department of Biochemistry and Biophysics
| | - Valentina E. Diaz
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Eva Q. Gontrum
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Brandon Chan
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences
| | - Michael R. Wilson
- Weill Institute for Neurosciences, and
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, and
| | - Joshua A. Hill
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Shaun W. Jackson
- Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mark S. Anderson
- Diabetes Center, School of Medicine, UCSF, San Francisco, California, USA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| |
Collapse
|
3
|
Dennis E, Murach M, Blackburn CM, Marshall M, Root K, Pattarabanjird T, Deroissart J, Erickson LD, Binder CJ, Bekiranov S, McNamara CA. Loss of TET2 increases B-1 cell number and IgM production while limiting CDR3 diversity. Front Immunol 2024; 15:1380641. [PMID: 38601144 PMCID: PMC11004297 DOI: 10.3389/fimmu.2024.1380641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Recent studies have demonstrated a role for Ten-Eleven Translocation-2 (TET2), an epigenetic modulator, in regulating germinal center formation and plasma cell differentiation in B-2 cells, yet the role of TET2 in regulating B-1 cells is largely unknown. Here, B-1 cell subset numbers, IgM production, and gene expression were analyzed in mice with global knockout of TET2 compared to wildtype (WT) controls. Results revealed that TET2-KO mice had elevated numbers of B-1a and B-1b cells in their primary niche, the peritoneal cavity, as well as in the bone marrow (B-1a) and spleen (B-1b). Consistent with this finding, circulating IgM, but not IgG, was elevated in TET2-KO mice compared to WT. Analysis of bulk RNASeq of sort purified peritoneal B-1a and B-1b cells revealed reduced expression of heavy and light chain immunoglobulin genes, predominantly in B-1a cells from TET2-KO mice compared to WT controls. As expected, the expression of IgM transcripts was the most abundant isotype in B-1 cells. Yet, only in B-1a cells there was a significant increase in the proportion of IgM transcripts in TET2-KO mice compared to WT. Analysis of the CDR3 of the BCR revealed an increased abundance of replicated CDR3 sequences in B-1 cells from TET2-KO mice, which was more clearly pronounced in B-1a compared to B-1b cells. V-D-J usage and circos plot analysis of V-J combinations showed enhanced usage of VH11 and VH12 pairings. Taken together, our study is the first to demonstrate that global loss of TET2 increases B-1 cell number and IgM production and reduces CDR3 diversity, which could impact many biological processes and disease states that are regulated by IgM.
Collapse
Affiliation(s)
- Emily Dennis
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Maria Murach
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Cassidy M.R. Blackburn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Melissa Marshall
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Katherine Root
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Tanyaporn Pattarabanjird
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Justine Deroissart
- Department for Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Loren D. Erickson
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Christoph J. Binder
- Department for Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan Bekiranov
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Coleen A. McNamara
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Bodansky A, Yu DJL, Rallistan A, Kalaycioglu M, Boonyaratanakornkit J, Green DJ, Gauthier J, Turtle CJ, Zorn K, O’Donovan B, Mandel-Brehm C, Asaki J, Kortbawi H, Kung AF, Rackaityte E, Wang CY, Saxena A, de Dios K, Masi G, Nowak RJ, O’Connor KC, Li H, Diaz VE, Casaletto KB, Gontrum EQ, Chan B, Kramer JH, Wilson MR, Utz PJ, Hill JA, Jackson SW, Anderson MS, DeRisi JL. Unveiling the autoreactome: Proteome-wide immunological fingerprints reveal the promise of plasma cell depleting therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.19.23300188. [PMID: 38196603 PMCID: PMC10775319 DOI: 10.1101/2023.12.19.23300188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
The prevalence and burden of autoimmune and autoantibody mediated disease is increasing worldwide, yet most disease etiologies remain unclear. Despite numerous new targeted immunomodulatory therapies, comprehensive approaches to apply and evaluate the effects of these treatments longitudinally are lacking. Here, we leverage advances in programmable-phage immunoprecipitation (PhIP-Seq) methodology to explore the modulation, or lack thereof, of proteome-wide autoantibody profiles in both health and disease. We demonstrate that each individual, regardless of disease state, possesses a distinct set of autoreactivities constituting a unique immunological fingerprint, or "autoreactome", that is remarkably stable over years. In addition to uncovering important new biology, the autoreactome can be used to better evaluate the relative effectiveness of various therapies in altering autoantibody repertoires. We find that therapies targeting B-Cell Maturation Antigen (BCMA) profoundly alter an individual's autoreactome, while anti-CD19 and CD-20 therapies have minimal effects, strongly suggesting a rationale for BCMA or other plasma cell targeted therapies in autoantibody mediated diseases.
Collapse
Affiliation(s)
- Aaron Bodansky
- Department of Pediatrics, Division of Critical Care, University of California San Francisco, San Francisco, CA
| | - David JL Yu
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA
| | - Alysa Rallistan
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305
| | - Muge Kalaycioglu
- Institute of Immunity, Transplantation, and Infection (ITI), Stanford University, Stanford, CA 94305
| | - Jim Boonyaratanakornkit
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Damian J. Green
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Jordan Gauthier
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Cameron J. Turtle
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Kelsey Zorn
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | - Brian O’Donovan
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | - Caleigh Mandel-Brehm
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | - James Asaki
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA
| | - Hannah Kortbawi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA
| | - Andrew F. Kung
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
- Biological and Medical Informatics Program, University of California San Francisco, San Francisco, CA
| | - Elze Rackaityte
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | | | | | - Kimberly de Dios
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA
| | - Gianvito Masi
- Department of Neurology, Yale School of Medicine, New Haven, CT
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT
| | | | - Kevin C. O’Connor
- Department of Neurology, Yale School of Medicine, New Haven, CT
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | - Valentina E. Diaz
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Eva Q. Gontrum
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Brandon Chan
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Michael R. Wilson
- Weill Institute for Neurosciences, University of California San Francisco; San Francisco, CA
- Department of Neurology, University of California San Francisco; San Francisco, CA
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305
| | - Joshua A. Hill
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Shaun W. Jackson
- Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
- Seattle Children’s Research Institute, Seattle, WA
- Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Mark S. Anderson
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| |
Collapse
|
5
|
Webster SE, Tsuji NL, Clemente MJ, Holodick NE. Age-related changes in antigen-specific natural antibodies are influenced by sex. Front Immunol 2023; 13:1047297. [PMID: 36713434 PMCID: PMC9878317 DOI: 10.3389/fimmu.2022.1047297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction Natural antibody (NAb) derived from CD5+ B-1 cells maintains tissue homeostasis, controls inflammation, aids in establishing long-term protective responses against pathogens, and provides immediate protection from infection. CD5+ B-1 cell NAbs recognize evolutionarily fixed epitopes, such as phosphatidylcholine (PtC), found on bacteria and senescent red blood cells. Anti-PtC antibodies are essential in protection against bacterial sepsis. CD5+ B-1 cell-derived NAbs have a unique germline-like structure that lacks N-additions, a feature critical for providing protection against infection. Previously, we demonstrated the repertoire and germline status of PtC+CD5+ B-1 cell IgM obtained from male mice changes with age depending on the anatomical location of the B-1 cells. More recently, we demonstrated serum antibody from aged female mice maintains protection against pneumococcal infection, whereas serum antibody from male mice does not provide protection. Results Here, we show that aged female mice have significantly more splenic PtC+CD5+ B-1 cells and more PtC specific serum IgM than aged male mice. Furthermore, we find both age and biological sex related repertoire differences when comparing B cell receptor (BCR) sequencing results of PtC+CD5+ B-1 cells. While BCR germline status of PtC+CD5+ B-1 cells from aged male and female mice is similar in the peritoneal cavity, it differs significantly in the spleen, where aged females retain germline configuration and aged males do not. Nucleic acid sensing toll-like receptors are critical in the maintenance of PtC+ B-1 cells; therefore, to begin to understand the mechanism of differences observed between the male and female PtC+CD5+ B-1 cell repertoire, we analyzed levels of cell-free nucleic acids and found increases in aged females. Conclusion Our results suggest the antigenic milieu differs between aged males and females, leading to differential selection of antigen-specific B-1 cells over time. Further elucidation of how biological sex differences influence the maintenance of B-1 cells within the aging environment will be essential to understand sex and age-related disparities in the susceptibility to bacterial infection and will aid in the development of more effective vaccination and/or therapeutic strategies specific for males and females.
Collapse
Affiliation(s)
- Sarah E. Webster
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Naomi L. Tsuji
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Michael J. Clemente
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
- Flow Cytometry and Imaging Core, Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Nichol E. Holodick
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
- Flow Cytometry and Imaging Core, Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
6
|
Shinton SA, Brill-Dashoff J, Hayakawa K. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase B1a cells. Sci Rep 2022; 12:14899. [PMID: 36050343 PMCID: PMC9437038 DOI: 10.1038/s41598-022-18876-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Newborns require early generation of effective innate immunity as a primary physiological mechanism for survival. The neonatal Lin28+Let7– developmental pathway allows increased generation of Th2-type cells and B1a (B-1 B) cells compared to adult cells and long-term maintenance of these initially generated innate cells. For initial B1a cell growth from the neonatal to adult stage, Th2-type IL-5 production from ILC2s and NKT2 cells is important to increase B1a cells. The Th17 increase is dependent on extracellular bacteria, and increased bacteria leads to lower Th2-type generation. Secreted group IIA-phospholipase A2 (sPLA2-IIA) from the Pla2g2a gene can bind to gram-positive bacteria and degrade bacterial membranes, controlling microbiota in the intestine. BALB/c mice are Pla2g2a+, and express high numbers of Th2-type cells and B1a cells. C57BL/6 mice are Pla2g2a-deficient and distinct from the SLAM family, and exhibit fewer NKT2 cells and fewer B1a cells from the neonatal to adult stage. We found that loss of Pla2g2a in the BALB/c background decreased IL-5 from Th2-type ILC2s and NKT2s but increased bacterial-reactive NKT17 cells and MAIT cells, and decreased the number of early-generated B1a cells and MZ B cells and the CD4/CD8 T cell ratio. Low IL-5 by decreased Th2-type cells in Pla2g2a loss led to low early-generated B1a cell growth from the neonatal to adult stage. In anti-thymocyte/Thy-1 autoreactive μκ transgenic (ATAμκ Tg) Pla2g2a+ BALB/c background C.B17 mice generated NKT2 cells that continuously control CD1d+ B1 B cells through old aging and lost CD1d in B1 B cells generating strong B1 ATA B cell leukemia/lymphoma. Pla2g2a-deficient ATAμκTg C57BL/6 mice suppressed the initial B1a cell increase, with low/negative spontaneous leukemia/lymphoma generation. These data confirmed that the presence of Pla2g2a to control bacteria is important to allow the neonatal to adult stage. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase early generated B1a cells.
Collapse
Affiliation(s)
- Susan A Shinton
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | | | - Kyoko Hayakawa
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA.
| |
Collapse
|
7
|
Hortal AM, Oeste CL, Cifuentes C, Alcoceba M, Fernández-Pisonero I, Clavaín L, Tercero R, Mendoza P, Domínguez V, García-Flores M, Pintado B, Abia D, García-Macías C, Navarro-Bailón A, Bustelo XR, González M, Alarcón B. Overexpression of wild type RRAS2, without oncogenic mutations, drives chronic lymphocytic leukemia. Mol Cancer 2022; 21:35. [PMID: 35120522 PMCID: PMC8815240 DOI: 10.1186/s12943-022-01496-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background Chronic lymphocytic leukemia (CLL) is the most frequent, and still incurable, form of leukemia in the Western World. It is widely accepted that cancer results from an evolutionary process shaped by the acquisition of driver mutations which confer selective growth advantage to cells that harbor them. Clear examples are missense mutations in classic RAS genes (KRAS, HRAS and NRAS) that underlie the development of approximately 13% of human cancers. Although autonomous B cell antigen receptor (BCR) signaling is involved and mutations in many tumor suppressor genes and oncogenes have been identified, an oncogenic driver gene has not still been identified for CLL. Methods Conditional knock-in mice were generated to overexpress wild type RRAS2 and prove its driver role. RT-qPCR analysis of a human CLL sample cohort was carried out to measure RRAS2 transcriptional expression. Sanger DNA sequencing was used to identify a SNP in the 3’UTR region of RRAS2 in human CLL samples. RNAseq of murine CLL was carried out to identify activated pathways, molecular mechanisms and to pinpoint somatic mutations accompanying RRAS2 overexpression. Flow cytometry was used for phenotypic characterization and shRNA techniques to knockdown RRAS2 expression in human CLL. Results RRAS2 mRNA is found overexpressed in its wild type form in 82% of the human CLL samples analyzed (n = 178, mean and median = 5-fold) as well as in the explored metadata. A single nucleotide polymorphism (rs8570) in the 3’UTR of the RRAS2 mRNA has been identified in CLL patients, linking higher expression of RRAS2 with more aggressive disease. Deliberate overexpression of wild type RRAS2 in mice, but not an oncogenic Q72L mutation in the coding sequence, provokes the development of CLL. Overexpression of wild type RRAS2 in mice is accompanied by a strong convergent selection of somatic mutations in genes that have been identified in human CLL. R-RAS2 protein is physically bound to the BCR and mediates BCR signals in CLL. Conclusions The results indicate that overexpression of wild type RRAS2 is behind the development of CLL. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01496-x.
Collapse
|
8
|
Mazzarello AN, Gentner-Göbel E, Dühren-von Minden M, Tarasenko TN, Nicolò A, Ferrer G, Vergani S, Liu Y, Bagnara D, Rai KR, Burger JA, McGuire PJ, Maity PC, Jumaa H, Chiorazzi N. B-cell receptor isotypes differentially associate with cell signaling, kinetics, and outcome in chronic lymphocytic leukemia. J Clin Invest 2021; 132:149308. [PMID: 34813501 PMCID: PMC8759784 DOI: 10.1172/jci149308] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), the B cell receptor (BCR) plays a critical role in disease development and progression, as indicated by the therapeutic efficacy of drugs blocking BCR signaling. However, the mechanism(s) underlying BCR responsiveness are not completely defined. Selective engagement of membrane IgM or IgD on CLL cells, each coexpressed by more than 90% of cases, leads to distinct signaling events. Since both IgM and IgD carry the same antigen-binding domains, the divergent actions of the receptors are attributed to differences in immunoglobulin (Ig) structure or the outcome of signal transduction. We showed that IgM, not IgD, level and organization associated with CLL-cell birth rate and the type and consequences of BCR signaling in humans and mice. The latter IgM-driven effects were abrogated when BCR signaling was inhibited. Collectively, these studies demonstrated a critical, selective role for IgM in BCR signaling and B cell fate decisions, possibly opening new avenues for CLL therapy.
Collapse
Affiliation(s)
- Andrea N Mazzarello
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | | | | | - Tatyana N Tarasenko
- Metabolism, Infection and Immunity Section, National Institutes of Health, Bethesda, United States of America
| | | | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Stefano Vergani
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Yun Liu
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Davide Bagnara
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Jan A Burger
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Peter J McGuire
- National Institutes of Health, Bethesda, United States of America
| | - Palash C Maity
- Institute for Immunology, University Hospital Ulm, Ulm, Germany
| | - Hassan Jumaa
- Institute for Immunology, University Hospital Ulm, Ulm, Germany
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| |
Collapse
|
9
|
Rubio AJ, Porter T, Zhong X. Duality of B Cell-CXCL13 Axis in Tumor Immunology. Front Immunol 2020; 11:521110. [PMID: 33193299 PMCID: PMC7609404 DOI: 10.3389/fimmu.2020.521110] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor immunity is a rapidly evolving area of research consisting of many possible permutations of immune cell tumor interactions that are dependent upon cell type, tumor type, and stage in tumor progression. At the same time, the majority of cancer immunotherapies have been focused on modulating the T cell-mediated antitumor immune response and have largely ignored the potential utility that B cells possess with respect to tumor immunity. Therefore, this motivated an exploration into the role that B cells and their accompanying chemokine, CXCL13, play in tumor immunity across multiple tumor types. Both B cells and CXCL13 possess dualistic impacts on tumor progression and tumor immunity which is furthered detail in this review. Specifically, various B cells subtypes are able to suppress or enhance several important immunological functions. Paradoxically, CXCL13 has been shown to drive several pro-growth and invasive signaling pathways across multiple tumor types, while also, correlating with improved survival and immune cell tumor localization in other tumor types. Potential tools for better elucidating the mechanisms by which B cells and CXCL13 impact the antitumor immune response are also discussed. In addition, multiples strategies are proposed for modulating the B cell-CXCL13 axis for cancer immunotherapies.
Collapse
Affiliation(s)
- Angel J. Rubio
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| | - Tyrone Porter
- Department of Biomedical Engineering, University of Texas Austin, Austin, TX, United States
| | - Xuemei Zhong
- Hematology and Medical Oncology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
10
|
CD3+ B-1a Cells as a Mediator of Disease Progression in Autoimmune-Prone Mice. Mediators Inflamm 2018; 2018:9289417. [PMID: 30670930 PMCID: PMC6323491 DOI: 10.1155/2018/9289417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022] Open
Abstract
B-1a cells are distinguishable from conventional B cells, which are designated B-2 cells, on the basis of their developmental origin, surface marker expression, and functions. In addition to the unique expression of the CD5 antigen, B-1a cells are characterized by the expression level of CD23. Although B-1a cells are considered to be independent of T cells and produce natural autoantibodies that induce the clinical manifestations of autoimmune diseases, there is much debate on the role of B-1a cells in the development of autoimmune diseases. We examined the involvement of B-1a cells in autoimmune-prone mice with the lpr gene. MRL/lpr and B6/lpr mice exhibited lupus and lymphoproliferative syndromes because of the massive accumulation of CD3+CD4-CD8-B220+ T cells. Interestingly, the B220+CD23-CD5+ (B-1a) cell population in the peripheral blood and peritoneal cavity increased with age and disease progression. Ninety percent of B-1a cells were CD3 positive (CD3+ B-1a cells) and did not produce tumor necrosis factor alpha, interferon gamma, or interleukin-10. To test the possible involvement of CD3+ B-1a cells in autoimmune disease, we tried to eliminate the peripheral cells by hypotonic shock through repeated intraperitoneal injections of distilled water. The fraction of peritoneal CD3+ B-1a cells decreased, and symptoms of the autoimmune disease were much milder in the distilled water-treated MRL/lpr mice. These results suggest that CD3+ B-1a cells could be mediators of disease progression in autoimmune-prone mice.
Collapse
|
11
|
Hofmann K, Clauder AK, Manz RA. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front Immunol 2018; 9:835. [PMID: 29740441 PMCID: PMC5924791 DOI: 10.3389/fimmu.2018.00835] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/05/2018] [Indexed: 12/29/2022] Open
Abstract
Success with B cell depletion using rituximab has proven the concept that B lineage cells represent a valid target for the treatment of autoimmune diseases, and has promoted the development of other B cell targeting agents. Present data confirm that B cell depletion is beneficial in various autoimmune disorders and also show that it can worsen the disease course in some patients. These findings suggest that B lineage cells not only produce pathogenic autoantibodies, but also significantly contribute to the regulation of inflammation. In this review, we will discuss the multiple pro- and anti-inflammatory roles of B lineage cells play in autoimmune diseases, in the context of recent findings using B lineage targeting therapies.
Collapse
Affiliation(s)
- Katharina Hofmann
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| |
Collapse
|
12
|
Choi SC, Xu Z, Li W, Yang H, Roopenian DC, Morse HC, Morel L. Relative Contributions of B Cells and Dendritic Cells from Lupus-Prone Mice to CD4 + T Cell Polarization. THE JOURNAL OF IMMUNOLOGY 2018; 200:3087-3099. [PMID: 29563177 DOI: 10.4049/jimmunol.1701179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/28/2018] [Indexed: 11/19/2022]
Abstract
Mouse models of lupus have shown that multiple immune cell types contribute to autoimmune disease. This study sought to investigate the involvement of B cells and dendritic cells in supporting the expansion of inflammatory and regulatory CD4+ T cells that are critical for lupus pathogenesis. We used lupus-prone B6.NZM2410.Sle1.Sle2.Sle3 (TC) and congenic C57BL/6J (B6) control mice to investigate how the genetic predisposition of these two cell types controls the activity of normal B6 T cells. Using an allogeneic in vitro assay, we showed that TC B1-a and conventional B cells expanded Th17 cells significantly more than their B6 counterparts. This expansion was dependent on CD86 and IL-6 expression and mapped to the Sle1 lupus-susceptibility locus. In vivo, TC B cells promoted greater differentiation of CD4+ T cells into Th1 and follicular helper T cells than did B6 B cells, but they limited the expansion of Foxp3 regulatory CD4+ T cells to a greater extent than did B6 B cells. Finally, when normal B6 CD4+ T cells were introduced into Rag1-/- mice, TC myeloid/stromal cells caused their heightened activation, decreased Foxp3 regulatory CD4+ T cell differentiation, and increased renal infiltration of Th1 and Th17 cells in comparison with B6 myeloid/stromal cells. The results show that B cells from lupus mice amplify inflammatory CD4+ T cells in a nonredundant manner with myeloid/stromal cells.
Collapse
Affiliation(s)
- Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Zhiwei Xu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Hong Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | | | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610;
| |
Collapse
|
13
|
Choi SC, Morel L. B cell contribution of the CD4 + T cell inflammatory phenotypes in systemic lupus erythematosus. Autoimmunity 2017; 50:37-41. [PMID: 28166683 DOI: 10.1080/08916934.2017.1280028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease in which the effector molecules responsible for tissue damage are antibodies directed against a large number of self-antigens, among which nucleic acids complexed with proteins play a prominent role. These pathogenic autoantibodies are produced by plasma cells differentiated from activated autoreactive B cells, a process that requires complex interactions between multiple components of the immune systems. A key step in the activation of autoreactive B cells is provided by CD4+ T cells through cytokines and cell-to-cell contact. Lupus CD4+ T cells are autoreactive and they present an activated inflammatory phenotype that has been shown to contribute to disease. In addition to their role in antibody production, B cells have other effector functions, the most important ones being antigen presentation to and co-stimulation of CD4+ T cells, as well as the secretion of cytokines. Here, we review what is known, largely based on mouse models, how these B cell effector functions contribute to the CD4+ T cell inflammatory phenotypes in lupus. When possible, we compare CD4+ T cell activation by B cells and by dendritic cells, and speculate how these interactions may contribute to the disease process.
Collapse
Affiliation(s)
- Seung-Chul Choi
- a Department of Pathology, Immunology, and Laboratory Medicine , University of Florida , Gainesville , FL , USA
| | - Laurence Morel
- a Department of Pathology, Immunology, and Laboratory Medicine , University of Florida , Gainesville , FL , USA
| |
Collapse
|
14
|
Dufaud C, Rivera J, Rohatgi S, Pirofski LA. Naïve B cells reduce fungal dissemination in Cryptococcus neoformans infected Rag1 -/- mice. Virulence 2017; 9:173-184. [PMID: 28837391 PMCID: PMC5955176 DOI: 10.1080/21505594.2017.1370529] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IgM and B-1 cell deficient mice exhibit early C. neoformans dissemination from lungs to brain, but a definitive role for B cells in conferring resistance to C. neoformans dissemination has not been established. To address this question, we developed an intranasal (i.n.) C. neoformans infection model in B and T cell deficient Rag1-/- mice and found they also exhibit earlier fungal dissemination and higher brain CFU than wild-type C57Bl/6 (wild-type) mice. To probe the effect of B cells on fungal dissemination, Rag1-/- mice were given splenic (intravenously) or peritoneal (intraperitoneally) B cells from wild-type mice and infected i.n. with C. neoformans 7 d later. Mice that received B cells had lung histopathology resembling wild type mice 14 d post-infection, and B-1, not B-2 or T cells in their lungs, and serum and lung IgM and IgG 21 d post-infection. Lung CFU were comparable in wild-type, Rag1-/-, and Rag1-/- mice that received B cells 21 d post-infection, but brain CFU were significantly lower in mice that received B cells than Rag1-/- mice that did not. To determine if natural antibody can promote immunity in our model, we measured alveolar macrophage phagocytosis of C. neoformans in Rag1-/- mice treated with naive wild-type IgM-sufficient or sIgM-/- IgM-deficient sera before infection. Compared to IgM-deficient sera, IgM-sufficient sera significantly increased phagocytosis. Our data establish B cells are able to reduce early C. neoformans dissemination in mice and suggest natural IgM may be a key mediator of early antifungal immunity in the lungs.
Collapse
Affiliation(s)
- Chad Dufaud
- a Department of Immunology and Microbial Sciences , Scripps Research Institute , La Jolla , CA , USA
| | - Johanna Rivera
- b Division of Infectious Diseases , Albert Einstein College of Medicine and Montefiore Medical Center , Bronx , NY , USA
| | - Soma Rohatgi
- c Department of Biotechnology IIT-Roorkee , Uttarakhand , India
| | - Liise-Anne Pirofski
- a Department of Immunology and Microbial Sciences , Scripps Research Institute , La Jolla , CA , USA.,d Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
15
|
Dysregulated Lymphoid Cell Populations in Mouse Models of Systemic Lupus Erythematosus. Clin Rev Allergy Immunol 2017; 53:181-197. [DOI: 10.1007/s12016-017-8605-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Oleksyn D, Zhao J, Vosoughi A, Zhao JC, Misra R, Pentland AP, Ryan D, Anolik J, Ritchlin C, Looney J, Anandarajah AP, Schwartz G, Calvi LM, Georger M, Mohan C, Sanz I, Chen L. PKK deficiency in B cells prevents lupus development in Sle lupus mice. Immunol Lett 2017; 185:1-11. [PMID: 28274793 DOI: 10.1016/j.imlet.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies that can result in damage to multiple organs. It is well documented that B cells play a critical role in the development of the disease. We previously showed that protein kinase C associated kinase (PKK) is required for B1 cell development as well as for the survival of recirculating mature B cells and B-lymphoma cells. Here, we investigated the role of PKK in lupus development in a lupus mouse model. We demonstrate that the conditional deletion of PKK in B cells prevents lupus development in Sle1Sle3 mice. The loss of PKK in Sle mice resulted in the amelioration of multiple classical lupus-associated phenotypes and histologic features of lupus nephritis, including marked reduction in the levels of serum autoantibodies, proteinuria, spleen size, peritoneal B-1 cell population and the number of activated CD4 T cells. In addition, the abundance of autoreactive plasma cells normally seen in Sle lupus mice was also significantly decreased in the PKK-deficient Sle mice. Sle B cells deficient in PKK display defective proliferation responses to BCR and LPS stimulation. Consistently, B cell receptor-mediated NF-κB activation, which is required for the survival of activated B cells, was impaired in the PKK-deficient B cells. Taken together, our work uncovers a critical role of PKK in lupus development and suggests that targeting the PKK-mediated pathway may represent a promising therapeutic strategy for lupus treatment.
Collapse
Affiliation(s)
- D Oleksyn
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Zhao
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A Vosoughi
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J C Zhao
- Department of Biology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - R Misra
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Pentland
- Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - D Ryan
- Department of Pathology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Anolik
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Ritchlin
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Looney
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Anandarajah
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - G Schwartz
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L M Calvi
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - M Georger
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, United States
| | - I Sanz
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L Chen
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States; Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States.
| |
Collapse
|