275 results on '"Hodes RJ"'
Search Results
2. The role of B7-CD28 co-stimulation in tumor rejection.
- Author
-
Yu, X, Abe, R, and Hodes, RJ
- Abstract
The role of B7 co-stimulatory signaling in in vivo tumor rejection remains incompletely characterized. In particular, the relative competence of B7-1 (CD80) and B7-2 (CD86) to provide effective co-stimulus is not well defined, and the identification of the T cell co-stimulatory receptor that mediates B7 co-stimulation in tumor rejection has not been addressed. These issues were studied by assessing rejection of B7-negative or B7-transfected tumor cells in CD28-expressing or CD28-deficient hosts. B7-negative EL4 tumor cells grew progressively in normal syngeneic C57BL/6 (B6) mice. In contrast EL4 cells transfected with either full length B7-1 or full length B7-2 were rejected, indicating that both B7-1 and B7-2 are competent to mediate rejection of EL4 tumor cells. Expression of truncated B7-1 or B7-2 products, with complete deletion of cytoplasmic domains, was as effective as expression of full length B7-1 or B7-2 in mediating rejection. In contrast to the rejection of B7-transfected EL4 cells observed in CD28-expressing syngeneic hosts, B7-1- and B7-2-positive EL4 cells as well as control EL4 cells grew progressively in CD28-deficient mice, demonstrating the requirement for host expression of CD28 in B7-mediated tumor rejection. These results indicate that interaction of host CD28 with co-stimulatory extracellular B7-1 or B7-2 ligands expressed on tumor cells can play a necessary role in mediating tumor rejection. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
3. Self recognition in allogeneic radiation bone marrow chimeras. A radiation- resistant host element dictates the self specificity and immune response gene phenotype of T-helper cells
- Author
-
Singer, A, Hathcock, KS, and Hodes, RJ
- Abstract
The specificity of the self-recognition repertoire in fully allogeneic (A {arrow} B), semiallogeneic (A {arrow} A x B and A x B {arrow} A), and double donor (A + B {arrow} A) radiation bone marrow chimeras was assessed by the ability of their spleen cells to generate in vitro primary plaque-forming cell (PFC) responses to trinitrophenyl- keyhole limpet hemocyanin. In contrast to spleen cells from semiallogeneic and double donor chimeras, intact spleen cells from fully allogeneic BI0 {arrow} B10.A and B10.A {arrow} B10 chimeras were not capable of generating responses to trinitrophenyl (TNP)-keyhole limpet hemocyanin. However, cultures containing a mixture of both B10 {arrow} B10.A and B10.A {arrow} B10 spleen cells did respond, demonstrating that all the cell populations required for the in vitro generation of T-dependent PFC responses were able to differentiate into functional competence in a fully allogeneic major histocompatibility complex (MHC) environment. The self recognition repertoire of T-helper cells from fully allogeneic A {arrow} B chimeras was determined to be specific for the recognition of host, not donor, MHC determinants in that they were able to collaborate with cells expressing only host MHC determinants but not with cells expressing only donor MHC determinants, even though the functional lymphocytes in these chimeras were shown to be of donor origin. Experiments utilizing double donor A + B {arrow} A chimeras further demonstrated that the ability of chimeric T cells to recognize allogeneic MHC determinants as self structures was a function of a radiation-resistant host element and not simply a consequence of the tolerization of T cell precursors to allogeneic MHC determinants, because strain A lymphocytes isolated from A + B {arrow} A chimeras were tolerant to both A and B MHC determinants but were restricted to the self recognition of syngeneic host type A MHC determinants. Finally, the Ir gene phenotype expressed by B10 {arrow} B10.A and B10.A {arrow} B10 chimeric lymphocytes was determined by their ability to function in the Ir gene controlled response to TNP-poly-L-(Tyr,Glu)-poly-D,L-Ala-poly- L-Lys [(T,G)-A--L]. The ability of lymphocytes to function in TNP-(T,G)-A--L responses was not determined by their genotype but rather paralleled the specificity of their self recognition repertoire for high responder (H-2 (b)) determinants. The possible degeneracy of the MHC-specific self recognition repertoire is discussed, and a model is proposed for Ir gene regulation in which expression of Ir gene function by lymphocytes is an antigen-nonspecific consequence of the specificity and cross-reactivity of their self recognition repertoire.
- Published
- 1981
- Full Text
- View/download PDF
4. Role of the major histocompatibility complex in T cell activation of B cell subpopulations. Major histocompatibility complex-restricted and -unrestricted B cell responses are mediated by distinct B cell subpopulations
- Author
-
Asano, Y, Singer, A, and Hodes, RJ
- Abstract
The present study has evaluated the identity of the B cell subpopulations participating in T dependent antibody responses that differ in their requirements for major histocompatibility complex-restricted T cell recognition. In vitro responses of keyhole limpet hemocyanin (KLH)-primed T cells and trinitrophenyl (TNP)-primed B cells were studied to both low and high concentrations of the antigen TNP-KLH. It was first demonstrated that for responses to low concentrations of TNP-KLH, (A × B)F(1) {arrow} parent(A) chimeric helper T cells were restricted in their ability to recognize parent(A) but not parent(B) H-2 determinants expressed by both B cells and antigen-presenting cells (APC). In contrast, at higher antigen concentrations, helper T cells were not restricted in their interaction with B cells. It was then determined whether these observed differences in T cell recognition resulted from the activation of distinct B cell subpopulations with different activation requirements. At low concentrations of TNP-KLH it was demonstrated that Lyb-5(-) B cells were activated, and that it was thus the activation of the Lyb-5(-) subpopulation that required T cell recognition of B cell H-2 under these conditions. In contrast, responses to high concentration of antigen required the participation of Lyb-5(+) B cells, and these Lyb-5(+) B cells were activated by a pathway that required H-2- restricted T cell interaction with APC, but not with B cells. The findings presented here have demonstrated that Lyb-5(-) and Lyb-5(+) B cells constitute B cell subpopulations that differ significantly in their activation requirements for T cell-dependent antibody responses to TNP-KLH. In so doing, these findings have established that the function of genetic restrictions in immune response regulation is critically dependent upon the activation pathways employed by functionally distinct subpopulations of B, as well as T, lymphocytes.
- Published
- 1981
- Full Text
- View/download PDF
5. Role of the major histocompatibility complex in T cell activation of B cell subpopulations. lyb-5(+) and lyb-5(-) B cell subpopulations differ in their requirement for major histocompatibility complex-restricted T cell recognition
- Author
-
Singer, A, Morrissey, PJ, Hathcock, KS, Ahmed, A, Scher, I, and Hodes, RJ
- Abstract
This report has examined the requirements for T helper (T(H)) cell recognition of major histocompatibility complex (MHC) determinants expressed by B cells for the activation of unprimed Lyb-5(+) and Lyb-5(-) B cell subpopulations . The generation of primary T(H) cell-dependent plaque-forming cell responses in vitro microculture required the presence of Lyb-5(+) B cells because B cell populations that were deprived, either genetically or serologically, of the Lyb-5(+) subpopulation were not activated in these responses. Cell-mixing experiments in which A X B {arrow} A chimeric T(H) cells were mixed with purified populations of parental accessory cells and parental B cells demonstrated that the in vitro activation of Lyb-5(+) B cells did not require T(H) cell recognition of B cell MHC determinants, although it did require T(H) cell recognition of accessory cell MHC determinants . In contrast to the failure of Lyb-5(-) B cells to be activated in primary T(H) cell-dependent responses in vitro microculture, isolated populations of Lyb-5(-) B cells were triggered by T(H) cells in vivo in short-term adoptive transfer experiments . By the use of A X B {arrow} A chimeric T(H) cells and parental strain B adoptive hosts, it was possible in vivo to distinguish genetically restricted T(H) cell recognition of B cells from genetically restricted T(H) cell recognition of accessory cells. Similar to the results obtained in vitro, the activation in vivo of unfractionated (Lyb-5(+) plus Lyb-5(-)) B cell populations did not require T(H) cell recognition of B cell MHC determinants . In contrast, in the same in vivo responses activation of isolated populations of Lyb-5(-) B cells did require T(H) cell recognition of B cell MHC determinants. The most straightforward interpretation of these experiments is that T(H) cell recognition of B cell MHC determinants is required for the activation of Lyb-5(-) B cells but is not required for the activation of Lyb-5(+) B cells . To better understand why T(H) cell activation of one B cell subpopulation is genetically restricted, whereas activation of another subpopulation is not, the response of Lyb-5(+) and Lyb-5(-) B cells to the soluble activating factors present in concanavalin A-induced spleen cell supernates (Con A SN) was examined. It was observed that Lyb-5(-) B cells, as opposed to Lyb-5(+) B cells, were unable to respond in microculture to the nonspecific T(H) cell- activating factors present in Con A SN, even though they were able to nonspecifically respond under the same conditions to trinitrophenyllipopolysaccharide. It was observed that the ability of B cell subpopulations to respond to nonspecific soluble T cell factors paralleled their ability to be activated by T(H) cells in a genetically unrestricted manner. Thus, the present experiments demonstrate that activation by T(H) cells of Lyb-5(-) B cells is MHC restricted, whereas activation of Lyb-5(+) B cells is not. These experiments suggest that one possible explanation for such differences is that activation of Lyb-5(+) B cells does not require direct interaction with T(H) cells because they can be activated by soluble activation signals that T(H) cells secrete.
- Published
- 1981
- Full Text
- View/download PDF
6. Cellular and genetic control of antibody responses in vitro. III. Immune response gene regulation of accessory cell function
- Author
-
Singer, A, Cowing, C, Hathcock, KS, Dickler, HB, and Hodes, RJ
- Abstract
The possibility was investigated that Ir genes regulate the function of cells other than T or B cells in the primary IgM responses to the synthetic antigens trinitrophenylated poly-L-(Tyr,Glu)-poly-D,L-Ala--poly-L-Lys [TNP-(T,G)-A--L]and trinitrophenylated poly-,-(His,Glu)-poly-D, L-Ala--poly-L-Lys [TNP-(H,G)-A--L]. The primary responses of (B10 × B10.A)F(1) spleen cells to both antigens were abrogated by Sephadex G-10 passage, and restored by the addition of spleen adherent cells. The cell type in the spleen adherent cell population active in reconstituting the responses to TNP-(T,G)-A--L and TNP-(H,G)-A--L was a non-T, non-B, radiation-resistant, glass-adherent spleen cell. The responses of Sephadex G-10-passed (responder x nonresponder)F(1) spleen cells to TNP-(T,G)-A--L or TNP-(H,G)-A--L were reconstituted by spleen adherent cells from only responder strains. Spleen adherent cells from F(1) mice reconstituted the responses to both antigens. Spleen adherent cells from each of the strains tested reconstituted the non- Ir gene-controlled response to a third antigen, TNP-keyhole limpet hemocyanin. The inability of spleen adherent cells from nonresponder strains to reconstitute the responses to either TNP-(T,G)-A--L or TNP-(H,G)-A--L was not a result of active suppression induced by the presence of nonresponder adherent cells, since a mixture of responder and nonresponder spleen adherent cells reconstituted the responses to both antigens. The use of spleen adherent cells from recombinant strains demonstrated that the autosomal dominant genes controlling the ability of spleen adherent cells to function as accessory cells in the responses to TNP-(T,G)-A--L and TNP-(H,G)-A--L are located in the K or I-A regions of the responder H-2 complex, the same region(s) of H-2 as the Ir genes controlling overall in vitro and in vivo responsiveness to these antigens.
- Published
- 1978
- Full Text
- View/download PDF
7. ANALYSIS OF 2 DISTINCT B-CELL ACTIVATION PATHWAYS MEDIATED BY A MONOCLONAL T-HELPER CELL .2. T-HELPER CELL SECRETION OF INTERLEUKIN-4 SELECTIVELY INHIBITS ANTIGEN-SPECIFIC B-CELL ACTIVATION BY COGNATE, BUT NOT NONCOGNATE, INTERACTIONS WITH T-CELLS
- Author
-
Asano, Y., Nakayama, T., Kubo, M., Fujisawa, I., Hajime Karasuyama, Singer, A., Hodes, Rj, and Tada, T.
8. The T-cell antigen receptor: a complex signal-transducing molecule
- Author
-
Bonifacino, Juan S., Merćep, Mladen, Sussman, Jeffrey J., Klausner, Richar D., Ashwell, Jonathan D., Hamaoka T, Hodes RJ, Klein G, Sugimura T, Takayama S, and Yamamura Y
- Subjects
T cell receptor ,IL-2 ,mutants ,hybridoma ,activation ,hydrolysis - Abstract
The T cell antigen-specific receptor (TCR) on most mature T cells is a multisubunit complex composed of 7 chains: alpha, beta, gamma, delta, epsilon, and either a zeta-zeta homodimer (zeta 2) or a zeta-eta heterodimer (zeta eta). We have derived a series of TCR variants and mutants from the antigen-specific murine T cell hybridoma, 2B4.11, permitting detailed analyses of the assembly and transport of the TCR. Loss of the zeta chain resulted in markedly reduced cell surface TCR expression. This was due to enhanced degradation of the other TCR chains in a post-Golgi, probably lysosomal, compartment. Loss of the beta chain, delta chain, or the combination of delta and zeta chains, also resulted in loss of cell surface TCR expression. Unlike the zeta loss variants, in these cases the other TCR chains were retained in the ER. Epsilon, gamma, and zeta could survive for prolonged periods in the ER, while the alpha, beta, and delta chains were rapidly and efficiently degraded. In another series of studies, variants that were deficient in the eta chain but that expressed normal levels of zeta 2-containing TCRs were analyzed for their functional properties. A positive relationship was found between the presence of zeta eta and the ability to respond to mitogenic stimuli with increases in phosphoinositide hydrolysis and, in one well characterized variant, increases in intracellular Ca2+. Despite this, late biological responses such as interleukin 2 (IL-2) production and inhibition of transformed growth were relatively normal. These results call into question the putative cause-and-effect relationship between some early biochemical events and these late biological responses. Further, they suggest a model in which zeta eta-containing TCR complexes are largely responsible for activation-induced phosphoinositide hydrolysis.
- Published
- 1989
9. Tumor suppressor p53 controls thymic NKT17 development.
- Author
-
Celli S, Watanabe M, and Hodes RJ
- Abstract
The tumor suppressor p53 antagonizes tumorigenesis, notably including the suppression of T cell lymphomas while its role on physiological T cell biology including thymic T cell development has not been fully understood. Invariant natural killer T (iNKT) cells develop in the thymus as innate-like αβ-T cells which consist of NKT1, NKT2 and NKT17 subsets. We found that the tumor suppressor p53 regulates specifically thymic NKT17 development. p53 is highly expressed in NKT17 relative to other T cell populations. Loss of p53 in the T cell lineage resulted in increased thymic NKT17 cell number with retention of lineage specific cytokine production, while development of NKT1, NKT2 and conventional T cells was not affected. Of interest, γH2AX expression was higher in NKT17 than NKT1 and NKT2 at steady state, and it was further increased in p53-deficient NKT17, suggesting that NKT17 development involves selectively greater DNA damage or genomic instability and that p53 expression might be in response to these damage signals. Taken together, our results indicated that the tumor suppressor p53 is active in selectively controlling thymic NKT17 development, with absence of p53 leading to an increase in thymic NKT17 cells expressing high levels of DNA damage response.
- Published
- 2024
- Full Text
- View/download PDF
10. Lessons From COVID-19 Testing Research: The Power of Rapid Response.
- Author
-
Compton WM, Hooper MW, Hodes RJ, and Pérez-Stable EJ
- Subjects
- Humans, COVID-19 epidemiology, COVID-19 Testing methods, SARS-CoV-2
- Published
- 2024
- Full Text
- View/download PDF
11. The All of Us Research Program is an opportunity to enhance the diversity of US biomedical research.
- Author
-
Bianchi DW, Brennan PF, Chiang MF, Criswell LA, D'Souza RN, Gibbons GH, Gilman JK, Gordon JA, Green ED, Gregurick S, Hodes RJ, Kilmarx PH, Koob GF, Koroshetz WJ, Langevin HM, Lorsch JR, Marrazzo JM, Pérez-Stable EJ, Rathmell WK, Rodgers GP, Rutter JL, Simoni JM, Tromberg BJ, Tucci DL, Volkow ND, Woychik R, Zenk SN, Kozlowski E, Peterson RS, Ginsburg GS, and Denny JC
- Subjects
- Humans, Mentors, Population Health, Biomedical Research
- Published
- 2024
- Full Text
- View/download PDF
12. TRAF6 and TRAF2/3 Binding Motifs in CD40 Differentially Regulate B Cell Function in T-Dependent Antibody Responses and Dendritic Cell Function in Experimental Autoimmune Encephalomyelitis.
- Author
-
Lu Y, Chiang J, Zhang R, Roche PA, and Hodes RJ
- Subjects
- Mice, Animals, TNF Receptor-Associated Factor 2 genetics, Signal Transduction, Antibody Formation, CD40 Antigens metabolism, Dendritic Cells metabolism, TNF Receptor-Associated Factor 6, Encephalomyelitis, Autoimmune, Experimental
- Abstract
Expression of the costimulatory molecule CD40 on both B cells and dendritic cells (DCs) is required for induction of experimental autoimmune encephalomyelitis (EAE), and cell-autonomous CD40 expression on B cells is required for primary T-dependent (TD) Ab responses. We now ask whether the function of CD40 expressed by different cell types in these responses is mediated by the same or different cytoplasmic domains. CD40 has been reported to possess multiple cytoplasmic domains, including distinct TRAF6 and TRAF2/3 binding motifs. To elucidate the in vivo function of these motifs in B cells and DCs involved in EAE and TD germinal center responses, we have generated knock-in mice containing distinct CD40 cytoplasmic domain TRAF-binding site mutations and have used these animals, together with bone marrow chimeric mice, to assess the roles that these motifs play in CD40 function. We found that both TRAF2/3 and TRAF6 motifs of CD40 are critically involved in EAE induction and demonstrated that this is mediated by a role of both motifs for priming of pathogenic T cells by DCs. In contrast, the TRAF2/3 binding motif, but not the TRAF6 binding motif, is required for B cell CD40 function in TD high-affinity Ab responses. These data demonstrate that the requirements for expression of specific TRAF-binding CD40 motifs differ for B cells or DCs that function in specific immune responses and thus identify targets for intervention to modulate these responses., (Copyright © 2023 by The American Association of Immunologists, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
13. CD40 Drives Central Nervous System Autoimmune Disease by Inducing Complementary Effector Programs via B Cells and Dendritic Cells.
- Author
-
Lu Y, Xu M, Dorrier CE, Zhang R, Mayer CT, Wagner D, McGavern DB, and Hodes RJ
- Subjects
- Humans, Animals, Mice, CD40 Antigens, Lymphocyte Count, Dendritic Cells, Autoimmune Diseases of the Nervous System, Central Nervous System Diseases, Encephalomyelitis, Autoimmune, Experimental
- Abstract
Costimulatory CD40 plays an essential role in autoimmune diseases, including experimental autoimmune encephalomyelitis (EAE), a murine model of human multiple sclerosis (MS). However, how CD40 drives autoimmune disease pathogenesis is not well defined. Here, we used a conditional knockout approach to determine how CD40 orchestrates a CNS autoimmune disease induced by recombinant human myelin oligodendrocyte glycoprotein (rhMOG). We found that deletion of CD40 in either dendritic cells (DCs) or B cells profoundly reduced EAE disease pathogenesis. Mechanistically, CD40 expression on DCs was required for priming pathogenic Th cells in peripheral draining lymph nodes and promoting their appearance in the CNS. By contrast, B cell CD40 was essential for class-switched MOG-specific Ab production, which played a crucial role in disease pathogenesis. In fact, passive transfer of MOG-immune serum or IgG into mice lacking CD40 on B cells but not DCs reconstituted autoimmune disease, which was associated with inundation of the spinal cord parenchyma by Ig and complement. These data demonstrate that CD40 supports distinct effector programs in B cells and DCs that converge to drive a CNS autoimmune disease and identify targets for intervention., (Copyright © 2022 by The American Association of Immunologists, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
14. Antigen-presenting T cells provide critical B7 co-stimulation for thymic iNKT cell development via CD28-dependent trogocytosis.
- Author
-
Watanabe M, Celli S, Alkhaleel FA, and Hodes RJ
- Subjects
- CD28 Antigens, Trogocytosis, Antigen-Presenting Cells, Thymus Gland, Natural Killer T-Cells
- Abstract
Invariant natural killer T (iNKT) cell development in the thymus depends on T cell receptor recognition of CD1d ligand on CD4/CD8 double-positive thymocytes. We previously reported that B7-CD28 co-stimulation is required for thymic iNKT cell development, but the cellular and molecular mechanisms underlying this co-stimulatory requirement are not understood. Here we report that CD28 expression on CD1d-expressing antigen-presenting T cells is required for thymic iNKT cell development. Mechanistically, antigen-presenting T cells provide co-stimulation through an unconventional mechanism, acquiring B7 molecules via CD28-dependent trogocytosis from B7-expressing thymic epithelial cells, dendritic cells, and B cells and providing critical B7 co-stimulation to developing iNKT cells. Thus, the present study demonstrates a mechanism of B7 co-stimulation in thymic T cell development by antigen-presenting T cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
15. Harnessing the Power of Community-Engaged Science to Facilitate Access and Uptake of COVID-19 Testing: RADx-UP.
- Author
-
Webb Hooper M, Compton WM, Walsh ER, Hodes RJ, and Pérez-Stable EJ
- Subjects
- Humans, COVID-19 diagnosis
- Published
- 2022
- Full Text
- View/download PDF
16. An NIH Response to COVID-19 That Engages Communities and Scientists.
- Author
-
Pérez-Stable EJ, Hodes RJ, and Schwetz TA
- Subjects
- Humans, COVID-19, Physicians
- Published
- 2022
- Full Text
- View/download PDF
17. B7-CD28 co-stimulation modulates central tolerance via thymic clonal deletion and Treg generation through distinct mechanisms.
- Author
-
Watanabe M, Lu Y, Breen M, and Hodes RJ
- Subjects
- Animals, Antigen-Presenting Cells metabolism, Autoimmunity physiology, CD28 Antigens genetics, Cell Differentiation immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Flow Cytometry, Gene Knock-In Techniques, Mice, Mice, Knockout, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta metabolism, Signal Transduction immunology, T-Lymphocytes, Regulatory metabolism, Thymocytes physiology, Thymus Gland cytology, Thymus Gland metabolism, B7-1 Antigen metabolism, CD28 Antigens metabolism, Central Tolerance, Clonal Deletion, T-Lymphocytes, Regulatory immunology, Thymus Gland immunology
- Abstract
The molecular and cellular mechanisms mediating thymic central tolerance and prevention of autoimmunity are not fully understood. Here we show that B7-CD28 co-stimulation and B7 expression by specific antigen-presenting cell (APC) types are required for clonal deletion and for regulatory T (Treg) cell generation from endogenous tissue-restricted antigen (TRA)-specific thymocytes. While B7-CD28 interaction is required for both clonal deletion and Treg induction, these two processes differ in their CD28 signaling requirements and in their dependence on B7-expressing dendritic cells, B cells, and thymic epithelial cells. Meanwhile, defective thymic clonal deletion due to altered B7-CD28 signaling results in the accumulation of mature, peripheral TRA-specific T cells capable of mediating destructive autoimmunity. Our findings thus reveal a function of B7-CD28 co-stimulation in shaping the T cell repertoire and limiting autoimmunity through both thymic clonal deletion and Treg cell generation.
- Published
- 2020
- Full Text
- View/download PDF
18. A Blueprint for Characterizing Senescence.
- Author
-
Roy AL, Sierra F, Howcroft K, Singer DS, Sharpless N, Hodes RJ, Wilder EL, and Anderson JM
- Subjects
- Biomarkers metabolism, Clinical Trials as Topic, Humans, Models, Biological, Cellular Senescence
- Abstract
Given the heterogeneity of senescent cells, our knowledge of both the drivers and consequences of cellular senescence in tissues and organs remains limited, as is our understanding of how this process could be harnessed for human health. Here we identified five broad areas that would help propel the field forward., (Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
19. National Institutes of Health social and behavioral research in response to the SARS-CoV2 Pandemic.
- Author
-
Riley WT, Borja SE, Hooper MW, Lei M, Spotts EL, Phillips JRW, Gordon JA, Hodes RJ, Lauer MS, Schwetz TA, and Perez-Stable E
- Subjects
- Behavior Control methods, Betacoronavirus, COVID-19, Health Status Disparities, Humans, National Institutes of Health (U.S.), SARS-CoV-2, United States epidemiology, Behavioral Research methods, Behavioral Research trends, Communicable Disease Control economics, Communicable Disease Control organization & administration, Coronavirus Infections economics, Coronavirus Infections epidemiology, Coronavirus Infections prevention & control, Coronavirus Infections psychology, Pandemics economics, Pandemics prevention & control, Pneumonia, Viral economics, Pneumonia, Viral epidemiology, Pneumonia, Viral prevention & control, Pneumonia, Viral psychology, Public Health trends, Social Sciences methods, Social Sciences trends, Telemedicine methods, Telemedicine trends
- Abstract
The COVID-19 pandemic has been mitigated primarily using social and behavioral intervention strategies, and these strategies have social and economic impacts, as well as potential downstream health impacts that require further study. Digital and community-based interventions are being increasingly relied upon to address these health impacts and bridge the gap in health care access despite insufficient research of these interventions as a replacement for, not an adjunct to, in-person clinical care. As SARS-CoV-2 testing expands, research on encouraging uptake and appropriate interpretation of these test results is needed. All of these issues are disproportionately impacting underserved, vulnerable, and health disparities populations. This commentary describes the various initiatives of the National Institutes of Health to address these social, behavioral, economic, and health disparities impacts of the pandemic, the findings from which can improve our response to the current pandemic and prepare us better for future infectious disease outbreaks., (Published by Oxford University Press on behalf of the Society of Behavioral Medicine 2020.)
- Published
- 2020
- Full Text
- View/download PDF
20. Rapid Scaling Up of Covid-19 Diagnostic Testing in the United States - The NIH RADx Initiative.
- Author
-
Tromberg BJ, Schwetz TA, Pérez-Stable EJ, Hodes RJ, Woychik RP, Bright RA, Fleurence RL, and Collins FS
- Subjects
- COVID-19, COVID-19 Testing, Humans, Pandemics, Reverse Transcriptase Polymerase Chain Reaction, Serologic Tests, United States, Clinical Laboratory Techniques statistics & numerical data, Clinical Laboratory Techniques trends, Coronavirus Infections diagnosis, Pneumonia, Viral diagnosis, Point-of-Care Testing trends
- Published
- 2020
- Full Text
- View/download PDF
21. TCR Repertoires of Thymic Conventional and Regulatory T Cells: Identification and Characterization of Both Unique and Shared TCR Sequences.
- Author
-
Ko A, Watanabe M, Nguyen T, Shi A, Achour A, Zhang B, Sun X, Wang Q, Zhuang Y, Weng NP, and Hodes RJ
- Subjects
- Animals, CD4-Positive T-Lymphocytes metabolism, High-Throughput Nucleotide Sequencing, Machine Learning, Mice, Receptors, Antigen, T-Cell, alpha-beta metabolism, T-Lymphocytes, Regulatory metabolism, CD4-Positive T-Lymphocytes immunology, Receptors, Antigen, T-Cell, alpha-beta genetics, T-Lymphocytes, Regulatory immunology
- Abstract
Thymic regulatory T cells (tTreg) are critical in the maintenance of normal T cell immunity and tolerance. The role of TCR in tTreg selection remains incompletely understood. In this study, we assessed TCRα and TCRβ sequences of mouse tTreg and thymic conventional CD4
+ T cells (Tconv) by high-throughput sequencing. We identified αβ TCR sequences that were unique to either tTreg or Tconv and found that these were distinct as recognized by machine learning algorithm and by preferentially used amino acid trimers in αβ CDR3 of tTreg. In addition, a proportion of αβ TCR sequences expressed by tTreg were also found in Tconv, and machine learning classified the great majority of these shared αβ TCR sequences as characteristic of Tconv and not tTreg. These findings identify two populations of tTreg, one in which the regulatory T cell fate is associated with unique properties of the TCR and another with TCR properties characteristic of Tconv for which tTreg fate is determined by factors beyond TCR sequence., (Copyright © 2020 by The American Association of Immunologists, Inc.)- Published
- 2020
- Full Text
- View/download PDF
22. Transient induction of telomerase expression mediates senescence and reduces tumorigenesis in primary fibroblasts.
- Author
-
Sun L, Chiang JY, Choi JY, Xiong ZM, Mao X, Collins FS, Hodes RJ, and Cao K
- Subjects
- Animals, Cell Cycle genetics, Cells, Cultured, Fibroblasts pathology, Gene Expression, Gene Expression Regulation, Gene Knockdown Techniques, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Telomere pathology, Cell Transformation, Neoplastic genetics, Cellular Senescence genetics, Telomerase genetics, Telomerase metabolism
- Abstract
Telomerase is an enzymatic ribonucleoprotein complex that acts as a reverse transcriptase in the elongation of telomeres. Telomerase activity is well documented in embryonic stem cells and the vast majority of tumor cells, but its role in somatic cells remains to be understood. Here, we report an unexpected function of telomerase during cellular senescence and tumorigenesis. We crossed Tert heterozygous knockout mice ( mTert
+/- ) for 26 generations, during which time there was progressive shortening of telomeres, and obtained primary skin fibroblasts from mTert+/+ and mTert-/- progeny of the 26th cross. As a consequence of insufficient telomerase activities in prior generations, both mTert+/+ and mTert-/- fibroblasts showed comparable and extremely short telomere length. However, mTert-/- cells approached cellular senescence faster and exhibited a significantly higher rate of malignant transformation than mTert+/+ cells. Furthermore, an evident up-regulation of telomerase reverse-transcriptase (TERT) expression was detected in mTert+/+ cells at the presenescence stage. Moreover, removal or down-regulation of TERT expression in mTert+/+ and human primary fibroblast cells via CRISPR/Cas9 or shRNA recapitulated mTert-/- phenotypes of accelerated senescence and transformation, and overexpression of TERT in mTert-/- cells rescued these phenotypes. Taking these data together, this study suggests that TERT has a previously underappreciated, protective role in buffering senescence stresses due to short, dysfunctional telomeres, and preventing malignant transformation., Competing Interests: The authors declare no conflict of interest., (Copyright © 2019 the Author(s). Published by PNAS.)- Published
- 2019
- Full Text
- View/download PDF
23. The NIH Blueprint for Neuroscience Research Seeks Community Input on Future Neuroscience Investments.
- Author
-
Mott MC, Austin CP, Bianchi DW, Cashion AK, Gordon JA, Heemskerk JE, Hodes RJ, Koob GF, Riley WT, Sieving PA, Shurtleff D, Somerman MJ, Volkow ND, Anderson KC, Owens DF, and Koroshetz WJ
- Subjects
- Humans, United States, Community-Institutional Relations trends, National Institutes of Health (U.S.), Neurosciences trends, Research Support as Topic
- Published
- 2019
- Full Text
- View/download PDF
24. Aging Research: Collaborations Forge a Promising Future.
- Author
-
Kelley MS, Bernard MA, and Hodes RJ
- Subjects
- Aged, Aged, 80 and over, Humans, Interdisciplinary Research, National Institutes of Health (U.S.), United States, Aging, Biomedical Research trends, Health Services Research trends, Health Services for the Aged trends, Translational Research, Biomedical trends
- Abstract
The National Institute on Aging (NIA), one of 27 institutes and centers at the National Institutes of Health (NIH), was founded in 1974 to conduct and support research on aging and the health and well-being of older people. The Institute's interests span the fundamental processes that contribute to aging and their impact on systems; diseases and conditions for which aging is a risk factor; and interventions that may prevent, delay, or treat these conditions or otherwise contribute to an extension of healthy, active years of life. Multiple fruitful research collaborations within and outside the federal government, spanning the breadth of the Institute's research activities, have marked NIA's growth over the past 40 years, as well as its current areas of ongoing research. This article discusses several highlights of these collaborations, including the Health and Retirement Study, geroscience research, falls injury prevention in elderly adults, and implementation of the National Plan to Address Alzheimer's Disease, from the perspective of past accomplishments and trends for the future., (Published 2017. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2017
- Full Text
- View/download PDF
25. Co-stimulatory function in primary germinal center responses: CD40 and B7 are required on distinct antigen-presenting cells.
- Author
-
Watanabe M, Fujihara C, Radtke AJ, Chiang YJ, Bhatia S, Germain RN, and Hodes RJ
- Subjects
- Animals, Antibody Formation physiology, B-Lymphocytes physiology, Dendritic Cells physiology, Immunoglobulin G metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, T-Cell Antigen Receptor Specificity physiology, T-Lymphocytes physiology, Antigen-Presenting Cells physiology, B7 Antigens physiology, CD40 Antigens physiology, Germinal Center physiology
- Abstract
T cell-dependent germinal center (GC) responses require coordinated interactions of T cells with two antigen-presenting cell (APC) populations, B cells and dendritic cells (DCs), in the presence of B7- and CD40-dependent co-stimulatory pathways. Contrary to the prevailing paradigm, we found unique cellular requirements for B7 and CD40 expression in primary GC responses to vaccine immunization with protein antigen and adjuvant: B7 was required on DCs but was not required on B cells, whereas CD40 was required on B cells but not on DCs in the generation of antigen-specific follicular helper T cells, antigen-specific GC B cells, and high-affinity class-switched antibody production. There was, in fact, no requirement for coexpression of B7 and CD40 on the same cell in these responses. Our findings support a substantially revised model for co-stimulatory function in the primary GC response, with crucial and distinct contributions of B7- and CD40-dependent pathways expressed by different APC populations and with important implications for understanding how to optimize vaccine responses or limit autoimmunity., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2017
- Full Text
- View/download PDF
26. Telomere Shortening, Inflammatory Cytokines, and Anti-Cytomegalovirus Antibody Follow Distinct Age-Associated Trajectories in Humans.
- Author
-
Lustig A, Liu HB, Metter EJ, An Y, Swaby MA, Elango P, Ferrucci L, Hodes RJ, and Weng NP
- Abstract
A number of biological parameters have been cited as hallmarks of immune aging. However, it is not clear whether these multiple biological changes are the result of common underlying aging processes and follow correlated trajectories, or whether the patterns of change for multiple parameters vary across individuals and reflect heterogeneity in the aging process. Here, we have studied parameters of immune system aging through longitudinal analysis of telomere length, inflammatory cytokines, and antibody titer to cytomegalovirus (CMV) in 465 subjects ranging in age from 21 to 88 years at the first visit, with an average of 13 years (7-19 years) follow-up. We observed a highly variable rate of change in telomere length of PBMCs with a relatively slow average rate of telomere shortening (-16 bp/year). Similarly, there were significant increases with age in vivo in three inflammation-related cytokines (interferon gamma, IL-6, and IL-10) and in anti-CMV IgG titer, which varied widely across individuals as well. We further observed positive correlative changes among different inflammatory cytokines. However, we did not find significant correlations among the rate of changes in telomere length, inflammatory cytokines, and anti-CMV IgG titers. Our findings thus reveal that age-related trajectories of telomere attrition, elevated circulating inflammatory cytokines, and anti-CMV IgG are independent and that aging individuals do not show a uniform pattern of change in these variables. Immune aging processes are complex and vary across individuals, and the use of multiple biomarkers is essential to evaluation of biological aging of the immune system.
- Published
- 2017
- Full Text
- View/download PDF
27. Disease drivers of aging.
- Author
-
Hodes RJ, Sierra F, Austad SN, Epel E, Neigh GN, Erlandson KM, Schafer MJ, LeBrasseur NK, Wiley C, Campisi J, Sehl ME, Scalia R, Eguchi S, Kasinath BS, Halter JB, Cohen HJ, Demark-Wahnefried W, Ahles TA, Barzilai N, Hurria A, and Hunt PW
- Subjects
- Chronic Disease, Humans, Acquired Immunodeficiency Syndrome genetics, Acquired Immunodeficiency Syndrome metabolism, Acquired Immunodeficiency Syndrome pathology, Aging genetics, Aging metabolism, Aging pathology, Diabetes Mellitus genetics, Diabetes Mellitus metabolism, Diabetes Mellitus pathology, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology
- Abstract
It has long been known that aging, at both the cellular and organismal levels, contributes to the development and progression of the pathology of many chronic diseases. However, much less research has examined the inverse relationship-the contribution of chronic diseases and their treatments to the progression of aging-related phenotypes. Here, we discuss the impact of three chronic diseases (cancer, HIV/AIDS, and diabetes) and their treatments on aging, putative mechanisms by which these effects are mediated, and the open questions and future research directions required to understand the relationships between these diseases and aging., (© 2016 New York Academy of Sciences.)
- Published
- 2016
- Full Text
- View/download PDF
28. T-cell development is regulated by the coordinated function of proximal and distal Lck promoters active at different developmental stages.
- Author
-
Chiang YJ and Hodes RJ
- Subjects
- Animals, CD4 Antigens metabolism, CD8 Antigens metabolism, Cell Differentiation, Cell Lineage, Cells, Cultured, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Promoter Regions, Genetic genetics, Protein Binding, Proto-Oncogene Proteins c-fyn genetics, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Proto-Oncogene Proteins c-fyn metabolism, T-Lymphocytes physiology, Thymocytes physiology, Thymus Gland immunology
- Abstract
Expression of Lck, a T-cell lineage-specific tyrosine kinase critical for T-cell development and activation, can be mediated by either proximal or distal lck promoter. We generated BAC transgenic mice in which BAC lck promoter was deleted and bred these transgenes to an Lck knockout background. Lck-PROX mice, in which only the proximal promoter is functional, have maximal Lck protein and normal thymic development through CD4
- CD8- double negative (DN) and CD4+ CD8+ double positive (DP) stages, but undetectable Lck later in development and reduced mature single positive thymocytes. In contrast, Lck-DIST mice, in which only distal promoter was functional, are deficient in Lck protein in DN and DP thymocytes and severely defective in early T-cell development, with a block at the DN3-DN4 beta checkpoint equivalent to complete Lck knockouts. The ability of the proximal lck promoter to support thymic development is independent of Fyn; while, in contrast, the distal lck promoter alone is completely unable to support development in the absence of Fyn. Notably, normal thymocyte development is restored by presence of both proximal and distal promoters, even when independently expressed on different lck genes. These results define distinct and complementary requirements for proximal and distal lck promoters during T-cell development., (Published 2016. This article is a U.S. Government work and is in the public domain in the USA.)- Published
- 2016
- Full Text
- View/download PDF
29. Accelerating Medicines Partnership: Alzheimer's Disease (AMP-AD) Knowledge Portal Aids Alzheimer's Drug Discovery through Open Data Sharing.
- Author
-
Hodes RJ and Buckholtz N
- Subjects
- Animals, Drug Design, Humans, Public-Private Sector Partnerships, Alzheimer Disease drug therapy, Drug Discovery methods, Information Dissemination
- Published
- 2016
- Full Text
- View/download PDF
30. Social protection: potential for improving HIV outcomes among adolescents.
- Author
-
Cluver LD, Hodes RJ, Sherr L, Orkin FM, Meinck F, Lim Ah Ken P, Winder-Rossi NE, Wolfe J, and Vicari M
- Subjects
- Adolescent, Africa, Eastern, Female, HIV Infections prevention & control, Humans, Male, Mass Screening, Risk Reduction Behavior, HIV Infections therapy, Social Support
- Abstract
Introduction: Advances in biomedical technologies provide potential for adolescent HIV prevention and HIV-positive survival. The UNAIDS 90-90-90 treatment targets provide a new roadmap for ending the HIV epidemic, principally through antiretroviral treatment, HIV testing and viral suppression among people with HIV. However, while imperative, HIV treatment and testing will not be sufficient to address the epidemic among adolescents in Southern and Eastern Africa. In particular, use of condoms and adherence to antiretroviral therapy (ART) remain haphazard, with evidence that social and structural deprivation is negatively impacting adolescents' capacity to protect themselves and others. This paper examines the evidence for and potential of interventions addressing these structural deprivations., Discussion: New evidence is emerging around social protection interventions, including cash transfers, parenting support and educational support ("cash, care and classroom"). These interventions have the potential to reduce the social and economic drivers of HIV risk, improve utilization of prevention technologies and improve adherence to ART for adolescent populations in the hyper-endemic settings of Southern and Eastern Africa. Studies show that the integration of social and economic interventions has high acceptability and reach and that it holds powerful potential for improved HIV, health and development outcomes., Conclusions: Social protection is a largely untapped means of reducing HIV-risk behaviours and increasing uptake of and adherence to biomedical prevention and treatment technologies. There is now sufficient evidence to include social protection programming as a key strategy not only to mitigate the negative impacts of the HIV epidemic among families, but also to contribute to HIV prevention among adolescents and potentially to remove social and economic barriers to accessing treatment. We urge a further research and programming agenda: to actively combine programmes that increase availability of biomedical solutions with social protection policies that can boost their utilization.
- Published
- 2015
- Full Text
- View/download PDF
31. ATM deficiency promotes development of murine B-cell lymphomas that resemble diffuse large B-cell lymphoma in humans.
- Author
-
Hathcock KS, Padilla-Nash HM, Camps J, Shin DM, Triner D, Shaffer AL 3rd, Maul RW, Steinberg SM, Gearhart PJ, Staudt LM, Morse HC 3rd, Ried T, and Hodes RJ
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins immunology, Caspases genetics, Caspases immunology, Cell Line, Tumor, Chromosomal Instability immunology, Genetic Loci immunology, Humans, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Mice, Mice, Knockout, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, NF-kappa B genetics, NF-kappa B immunology, Neoplasm Proteins genetics, Neoplasm Proteins immunology, Receptors, Antigen, B-Cell genetics, Receptors, Antigen, B-Cell immunology, T-Lymphocytes immunology, T-Lymphocytes pathology, Tumor Suppressor Proteins immunology, Ataxia Telangiectasia Mutated Proteins deficiency, Immunologic Surveillance, Lymphoma, Large B-Cell, Diffuse immunology, Tumor Suppressor Proteins deficiency
- Abstract
The serine-threonine kinase ataxia-telangiectasia mutated (ATM) plays a central role in maintaining genomic integrity. In mice, ATM deficiency is exclusively associated with T-cell lymphoma development, whereas B-cell tumors predominate in human ataxia-telangiectasia patients. We demonstrate in this study that when T cells are removed as targets for lymphomagenesis and as mediators of immune surveillance, ATM-deficient mice exclusively develop early-onset immunoglobulin M(+) B-cell lymphomas that do not transplant to immunocompetent mice and that histologically and genetically resemble the activated B cell-like (ABC) subset of human diffuse large B-cell lymphoma (DLBCL). These B-cell lymphomas show considerable chromosomal instability and a recurrent genomic amplification of a 4.48-Mb region on chromosome 18 that contains Malt1 and is orthologous to a region similarly amplified in human ABC DLBCL. Of importance, amplification of Malt1 in these lymphomas correlates with their dependence on nuclear factor (NF)-κB, MALT1, and B-cell receptor (BCR) signaling for survival, paralleling human ABC DLBCL. Further, like some human ABC DLBCLs, these mouse B-cell lymphomas also exhibit constitutive BCR-dependent NF-κB activation. This study reveals that ATM protects against development of B-cell lymphomas that model human ABC DLBCL and identifies a potential role for T cells in preventing the emergence of these tumors.
- Published
- 2015
- Full Text
- View/download PDF
32. 'HIV is like a tsotsi. ARVs are your guns': associations between HIV-disclosure and adherence to antiretroviral treatment among adolescents in South Africa.
- Author
-
Cluver LD, Hodes RJ, Toska E, Kidia KK, Orkin FM, Sherr L, and Meinck F
- Subjects
- Adolescent, Anti-Retroviral Agents therapeutic use, Child, Cross-Sectional Studies, Female, HIV Infections drug therapy, Humans, Male, Qualitative Research, South Africa epidemiology, Surveys and Questionnaires, Truth Disclosure, Young Adult, HIV Infections psychology, Health Knowledge, Attitudes, Practice, Medication Adherence statistics & numerical data
- Abstract
Objectives: WHO guidelines recommend disclosure to HIV-positive children by school age in order to improve antiretroviral therapy (ART) adherence. However, quantitative evidence remains limited for adolescents. This study examines associations between adolescent knowledge of HIV-positive status and ART-adherence in South Africa., Design: A cross-sectional study of the largest known community-traced sample of HIV-positive adolescents. Six hundred and eighty-four ART-initiated adolescents aged 10-19 years (52% female, 79% perinatally infected) were interviewed., Methods: In a low-resource health district, all adolescents who had ever initiated ART in a stratified sample of 39 health facilities were identified and traced to 150 communities [n = 1102, 351 excluded, 27 deceased, 40 (5.5%) refusals]. Quantitative interviews used standardized questionnaires and clinic records. Quantitative analyses used multivariate logistic regressions, and qualitative analyses used grounded theory for 18 months of interviews, focus groups and participant observations with 64 adolescents, caregivers and healthcare workers., Results: About 36% of adolescents reported past-week ART nonadherence, and 70% of adolescents knew their status. Adherence was associated with fewer opportunistic infection symptoms [odds ratio (OR) 0.55; 95% CI 0.40-0.76]. Adolescent knowledge of HIV-positive status was associated with higher adherence, independently of all cofactors (OR 2.18; 95% CI 1.47-3.24). Among perinatally infected adolescents who knew their status (n = 362/540), disclosure prior to age 12 was associated with higher adherence (OR 2.65; 95% CI 1.34-5.22). Qualitative findings suggested that disclosure was undertaken sensitively in clinical and family settings, but that adults lacked awareness about adolescent understandings of HIV status., Conclusion: Early and full disclosure is strongly associated with improved adherence amongst ART-initiated adolescents. Disclosure may be an essential tool in improving adolescent adherence and reducing mortality and onwards transmission.
- Published
- 2015
- Full Text
- View/download PDF
33. Regulation of T cell development by c-Cbl: essential role of Lck.
- Author
-
Chiang YJ and Hodes RJ
- Subjects
- Animals, Cell Differentiation genetics, Cells, Cultured, Histocompatibility Antigens metabolism, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) genetics, Mice, Mice, Knockout, Phosphorylation genetics, Proto-Oncogene Proteins c-cbl genetics, Receptors, Antigen, T-Cell metabolism, Signal Transduction genetics, ZAP-70 Protein-Tyrosine Kinase genetics, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Proto-Oncogene Proteins c-cbl metabolism, T-Lymphocytes immunology, Thymocytes immunology, ZAP-70 Protein-Tyrosine Kinase metabolism
- Abstract
A canonical pre-TCR/TCR signaling pathway critical for thymic T cell development involves sequential phosphorylation and signaling through Lck, Zap70, Lat and Slp76. However, we and others have previously reported that genomic deletion of c-Cbl (Cbl) partially or completely reverses the defects in thymic development in mice deficient in Zap70, Slp76, Lat or Vav1, indicating the presence of alternative pathways normally suppressed by Cbl. To further elucidate pre-TCR/TCR signaling pathways involved in thymic development, we characterized the effect of Cbl inactivation on developmental and signaling defects in mice deficient in proximal signaling molecules Lck and Zap70. Inactivation of Cbl partially reversed defective T cell development in Zap70 (-/-) mice and reversed defects in phosphorylation of Erk, Plc-γ1, Vav1 and Akt, in TCR-stimulated Cbl (-/-) Zap70 (-/-) thymocytes. Recent reports identified an essential role of Lck in associating with CD4 and CD8 coreceptors and mediating the requirement for MHC restriction in TCR recognition. Since TCR recognition has been shown to be MHC-restricted in Cbl (-/-) mice, it was of interest to determine whether the requirement for Lck remained unmodified by Cbl deletion. Indeed, in contrast to the effect of Cbl inactivation in partially or fully bypassing requirements for other TCR signaling components, inactivation of Cbl did not reverse either defective T cell development or defective phosphorylation of TCR signaling molecules in Lck (-/-) mice. Thus, Lck, which plays a unique role in enforcing MHC restriction, is essential for thymic development in presence or absence of Cbl, ensuring MHC restriction of T cells derived from either pathway., (Published by Oxford University Press on behalf of The Japanese Society for Immunology 2014.)
- Published
- 2015
- Full Text
- View/download PDF
34. Age-associated telomere attrition of lymphocytes in vivo is co-ordinated with changes in telomerase activity, composition of lymphocyte subsets and health conditions.
- Author
-
Lin Y, Damjanovic A, Metter EJ, Nguyen H, Truong T, Najarro K, Morris C, Longo DL, Zhan M, Ferrucci L, Hodes RJ, and Weng NP
- Subjects
- Adult, Aged, Aged, 80 and over, Aging immunology, Aging metabolism, B-Lymphocytes enzymology, B-Lymphocytes physiology, Follow-Up Studies, Humans, Leukocytes, Mononuclear metabolism, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Lymphocyte Subsets immunology, Middle Aged, Monocytes physiology, T-Lymphocytes enzymology, T-Lymphocytes physiology, Telomere Homeostasis physiology, Young Adult, Aging genetics, Lymphocyte Subsets enzymology, Telomerase blood, Telomere Homeostasis immunology
- Abstract
Telomeres are essential in maintaining chromosome integrity and in controlling cellular replication. Attrition of telomere length in peripheral blood mononuclear cells (PBMCs) with age is well documented from cross-sectional studies. But the actual in vivo changes in telomere lengths and its relationship with the contributing factors within the individuals with age have not been fully addressed. In the present paper, we report a longitudinal analysis of telomere length in the PBMCs, lymphocytes and monocytes of 216 human subjects aged from 20-90 years assessed at 0-, 5- and 12-year follow-up. For the 5- and 12-year follow-up, telomere length in the PBMCs decreased in 34% and 46%, exhibited no detectable change in 56% and 47% and increased in 10% and 7% of the subjects respectively. The rate of telomere change was distinct for T-cells, B-cells and monocytes for any given subject. Telomerase activity declined with age in the resting T-cells and B-cells and the activated T-cells. Finally, a significant portion of telomere attrition in T-cells with age was explained by a decline in the telomerase activity, decreased naïve cells and the change in physiological conditions such as elevated blood glucose and interleukin (IL)-6 levels. These findings show that changes in the telomere length of the PBMCs with age in vivo occur at different rates in different individuals and cell types and reveal that changes in the telomere length in the T-cells with age is influenced by the telomerase activity, naïve T-cell percentage and changes in health conditions.
- Published
- 2015
- Full Text
- View/download PDF
35. CD28-CD80/86 and CD40-CD40L Interactions Promote Thymic Tolerance by Regulating Medullary Epithelial Cell and Thymocyte Development.
- Author
-
Williams JA, Tai X, and Hodes RJ
- Subjects
- Animals, Cell Differentiation, Humans, Immune Tolerance, Receptor Cross-Talk, Receptors, Antigen, T-Cell metabolism, Signal Transduction, B7-1 Antigen metabolism, B7-2 Antigen metabolism, CD28 Antigens metabolism, CD40 Antigens metabolism, CD40 Ligand metabolism, Epithelial Cells immunology, T-Lymphocytes immunology, Thymocytes immunology
- Abstract
Development and central tolerance of T lymphocytes in the thymus requires both TCR signals and collaboration with signals generated through costimulatory molecule interactions. In this review, we discuss the importance of CD28-CD80/86 and CD40-CD40L costimulatory interactions in promoting normal thymic development. This discussion includes roles in the generation of a normal thymic medulla, in the development of specific T-cells subsets, including iNKT and T regulatory cells, and in the generation of a tolerant mature T-cell repertoire. We discuss recent contributions to the understanding of CD28-CD80/86 and CD40-CD40L costimulatory interactions in thymic development, and we highlight the ways in which the many important roles mediated by these interactions collaborate to promote normal thymic development.
- Published
- 2015
- Full Text
- View/download PDF
36. T cell-B cell thymic cross-talk: maintenance and function of thymic B cells requires cognate CD40-CD40 ligand interaction.
- Author
-
Fujihara C, Williams JA, Watanabe M, Jeon H, Sharrow SO, and Hodes RJ
- Subjects
- Animals, Autoantigens immunology, CD40 Antigens genetics, CD40 Ligand genetics, Cell Communication, Cell Proliferation, Cell Survival, Cells, Cultured, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Protein Binding genetics, B-Lymphocytes immunology, CD40 Antigens metabolism, CD40 Ligand metabolism, T-Lymphocytes immunology, Thymus Gland immunology
- Abstract
Thymic development requires bidirectional interaction or cross-talk between developing T cells and thymic stromal cells, a relationship that has been best characterized for the interaction between thymocytes and thymic epithelial cells. We have characterized in this article the requirement for similar cross-talk in the maintenance and function of thymic B cells, another population that plays a role in selection of developing thymic T cells. We found that maintenance of thymic B cells is strongly dependent on the presence of mature single-positive thymocytes and on the interactions of these T cells with specific Ag ligand. Maintenance of thymic B cell number is strongly dependent on B cell-autonomous expression of CD40, but not MHC class II, indicating that direct engagement of CD40 on thymic B cells is necessary to support their maintenance and proliferation. Thymic B cells can mediate negative selection of superantigen-specific, self-reactive, single-positive thymocytes, and we show that CD40 expression on B cells is critical for this negative selection. Cross-talk with thymic T cells is thus required to support the thymic B cell population through a pathway that requires cell-autonomous expression of CD40, and that reciprocally functions in negative selection of autoreactive T cells.
- Published
- 2014
- Full Text
- View/download PDF
37. Downmodulation of tumor suppressor p53 by T cell receptor signaling is critical for antigen-specific CD4(+) T cell responses.
- Author
-
Watanabe M, Moon KD, Vacchio MS, Hathcock KS, and Hodes RJ
- Subjects
- Animals, Antibody Specificity immunology, Cell Cycle genetics, Cell Cycle immunology, Cell Proliferation, Cells, Cultured, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, RNA, Messenger biosynthesis, Receptors, Interleukin-2 immunology, Signal Transduction immunology, Transcription, Genetic, Tumor Suppressor Protein p53 biosynthesis, CD4-Positive T-Lymphocytes immunology, Interleukin-2 immunology, Proto-Oncogene Proteins c-mdm2 genetics, Receptors, Antigen, T-Cell immunology, Tumor Suppressor Protein p53 genetics
- Abstract
Antigen specificity is critical in immune response and requires integration of antigen-specific signals with antigen-nonspecific signals such as those provided by cytokines. The mechanism integrating these pathways is incompletely understood. We report here that antigen-specific proliferative responses of CD4(+) T cells required downmodulation of tumor suppressor p53. In the absence of T cell receptor (TCR) signal, IL-2 induced sustained increase in p53 protein, which prevented proliferative responses despite strong signaling through the IL-2 receptor. In contrast, TCR signaling resulted in early termination of p53 protein expression by decreasing p53 mRNA as well as strong transcriptional induction of the p53-regulating protein Mdm2. Downmodulation of p53 in response to antigen stimulation was in fact critical for antigen-specific T cell proliferation, and preventing p53 degradation by inhibiting Mdm2 resulted in sustained p53 protein and prevented antigen-specific T cell proliferation. It is thus termination of p53 by TCR signaling that allows proliferative responses, enforcing antigen specificity., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
38. Thymic medullary epithelium and thymocyte self-tolerance require cooperation between CD28-CD80/86 and CD40-CD40L costimulatory pathways.
- Author
-
Williams JA, Zhang J, Jeon H, Nitta T, Ohigashi I, Klug D, Kruhlak MJ, Choudhury B, Sharrow SO, Granger L, Adams A, Eckhaus MA, Jenkinson SR, Richie ER, Gress RE, Takahama Y, and Hodes RJ
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Epithelial Cells immunology, Killer Cells, Natural immunology, Male, Mice, Mice, Inbred BALB C, Mice, Knockout, NF-kappa B immunology, Receptors, Antigen, T-Cell immunology, Signal Transduction immunology, T-Lymphocytes, Regulatory immunology, Up-Regulation immunology, B7-1 Antigen immunology, B7-2 Antigen immunology, CD28 Antigens immunology, CD40 Antigens immunology, CD40 Ligand immunology, Epithelium immunology, Self Tolerance immunology, Thymocytes immunology
- Abstract
A critical process during thymic development of the T cell repertoire is the induction of self-tolerance. Tolerance in developing T cells is highly dependent on medullary thymic epithelial cells (mTEC), and mTEC development in turn requires signals from mature single-positive thymocytes, a bidirectional relationship termed thymus crosstalk. We show that CD28-CD80/86 and CD40-CD40L costimulatory interactions, which mediate negative selection and self-tolerance, upregulate expression of LTα, LTβ, and receptor activator for NF-κB in the thymus and are necessary for medullary development. Combined absence of CD28-CD80/86 and CD40-CD40L results in profound deficiency in mTEC development comparable to that observed in the absence of single-positive thymocytes. This requirement for costimulatory signaling is maintained even in a TCR transgenic model of high-affinity TCR-ligand interactions. CD4 thymocytes maturing in the altered thymic epithelial environment of CD40/CD80/86 knockout mice are highly autoreactive in vitro and are lethal in congenic adoptive transfer in vivo, demonstrating a critical role for these costimulatory pathways in self-tolerance as well as thymic epithelial development. These findings demonstrate that cooperativity between CD28-CD80/86 and CD40-CD40L pathways is required for normal medullary epithelium and for maintenance of self-tolerance in thymocyte development.
- Published
- 2014
- Full Text
- View/download PDF
39. A novel T cell subset with trans-rearranged Vγ-Cβ TCRs shows Vβ expression is dispensable for lineage choice and MHC restriction.
- Author
-
Bowen S, Sun P, Livak F, Sharrow S, and Hodes RJ
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins deficiency, Ataxia Telangiectasia Mutated Proteins genetics, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Gene Expression, Genes, MHC Class I, Genes, MHC Class II, Mice, Mice, Knockout, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta metabolism, Transcriptional Activation, Cell Lineage genetics, Gene Rearrangement, beta-Chain T-Cell Antigen Receptor, Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor, Major Histocompatibility Complex immunology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism
- Abstract
αβ T cells, which express the α-β TCR heterodimer, express CD4 or CD8 coreceptors on cells that are MHC class I or MHC class II dependent. In contrast, γδ T cells do not express CD4 or CD8 and develop independently of MHC interaction. The factors that determine αβ and γδ lineage choice are not fully understood, and the determinants of MHC restriction of TCR specificity have been controversial. In this study we have identified a naturally occurring population of T cells expressing Vγ-Cβ receptor chains on the cell surface, the products of genomic trans-rearrangement between the Vγ2 gene and a variety of Dβ or Jβ genes, in place of an intact TCRβ-chain and in association with TCRα. Identification of this population allowed an analysis of the role of TCR variable regions in determining T cell lineage choice and MHC restriction. We found that Vγ2(+)Cβ(+) cells are positive for either CD4 or CD8 and are selected in an MHC class II- or MHC class I-dependent manner, respectively, thus following the differentiation pathway of αβ and not γδ cells and demonstrating that Vβ V region sequences are not required for selection of an MHC-restricted repertoire.
- Published
- 2014
- Full Text
- View/download PDF
40. TRAF3 enforces the requirement for T cell cross-talk in thymic medullary epithelial development.
- Author
-
Jenkinson SR, Williams JA, Jeon H, Zhang J, Nitta T, Ohigashi I, Kruhlak M, Zuklys S, Sharrow S, Adams A, Granger L, Choi Y, Siebenlist U, Bishop GA, Hollander GA, Takahama Y, and Hodes RJ
- Subjects
- Animals, CD40 Antigens genetics, Flow Cytometry, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Knockout, TNF Receptor-Associated Factor 3 deficiency, Thymocytes immunology, Cell Differentiation immunology, Receptor Cross-Talk immunology, Self Tolerance immunology, T-Lymphocytes immunology, TNF Receptor-Associated Factor 3 immunology, Thymocytes metabolism
- Abstract
Induction of self-tolerance in developing T cells depends on medullary thymic epithelial cells (mTECs), whose development, in turn, requires signals from single-positive (SP) thymocytes. Thus, the absence of SP thymocytes in Tcra(-/-) mice results in a profound deficiency in mTECs. Here, we have probed the mechanism that underlies this requirement for cross-talk with thymocytes in medullary development. Previous studies have implicated nonclassical NF-κB as a pathway important in the development of mTECs, because mice lacking RelB, NIK, or IKKα, critical components of this pathway, have an almost complete absence of mTECs, with resulting autoimmune pathology. We therefore assessed the effect of selective deletion in TEC of TNF receptor-associated factor 3 (TRAF3), an inhibitor of nonclassical NF-κB signaling. Deletion of TRAF3 in thymic epithelial cells allowed RelB-dependent development of normal numbers of AIRE-expressing mTECs in the complete absence of SP thymocytes. Thus, mTEC development can occur in the absence of cross-talk with SP thymocytes, and signals provided by SP T cells are needed to overcome TRAF3-imposed arrest in mTEC development mediated by inhibition of nonclassical NF-κB. We further observed that TRAF3 deletion is also capable of overcoming all requirements for LTβR and CD40, which are otherwise necessary for mTEC development, but is not sufficient to overcome the requirement for RANKL, indicating a role for RANKL that is distinct from the signals provided by SP thymocytes. We conclude that TRAF3 plays a central role in regulation of mTEC development by imposing requirements for SP T cells and costimulation-mediated cross-talk in generation of the medullary compartment.
- Published
- 2013
- Full Text
- View/download PDF
41. ATM influences the efficiency of TCRβ rearrangement, subsequent TCRβ-dependent T cell development, and generation of the pre-selection TCRβ CDR3 repertoire.
- Author
-
Hathcock KS, Bowen S, Livak F, and Hodes RJ
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins deficiency, Ataxia Telangiectasia Mutated Proteins genetics, Cell Division genetics, Cell Line, Cell Survival genetics, DNA Breaks, Double-Stranded, Gene Order, Mice, Mice, Knockout, Thymocytes metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Complementarity Determining Regions genetics, Gene Rearrangement, beta-Chain T-Cell Antigen Receptor, Receptors, Antigen, T-Cell, alpha-beta genetics, T-Lymphocytes metabolism
- Abstract
Generation and resolution of DNA double-strand breaks is required to assemble antigen-specific receptors from the genes encoding V, D, and J gene segments during recombination. The present report investigates the requirement for ataxia telangiectasia-mutated (ATM) kinase, a component of DNA double-strand break repair, during TCRβ recombination and in subsequent TCRβ-dependent repertoire generation and thymocyte development. CD4(-)CD8(-) double negative stage 2/3 thymocytes from ATM-deficient mice have both an increased frequency of cells with DNA break foci at TCRβ loci and reduced Vβ-DJβ rearrangement. Sequencing of TCRβ complementarity-determining region 3 demonstrates that ATM-deficient CD4(+)CD8(+) double positive thymocytes and peripheral T cells have altered processing of coding ends for both in-frame and out-of-frame TCRβ rearrangements, providing the unique demonstration that ATM deficiency alters the expressed TCRβ repertoire by a selection-independent mechanism. ATMKO thymi exhibit a partial developmental block in DN cells as they negotiate the β-selection checkpoint to become double negative stage 4 and CD4(+)CD8(+) thymocytes, resulting in reduced numbers of CD4(+)CD8(+) cells. Importantly, expression of a rearranged TCRβ transgene substantially reverses this defect in CD4(+)CD8(+) cells, directly linking a requirement for ATM during endogenous TCRβ rearrangement to subsequent TCRβ-dependent stages of development. These results demonstrate that ATM plays an important role in TCRβ rearrangement, generation of the TCRβ CDR3 repertoire, and efficient TCRβ-dependent T cell development.
- Published
- 2013
- Full Text
- View/download PDF
42. Concurrent V(D)J recombination and DNA end instability increase interchromosomal trans-rearrangements in ATM-deficient thymocytes.
- Author
-
Bowen S, Wangsa D, Ried T, Livak F, and Hodes RJ
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins deficiency, Cell Cycle Proteins genetics, DNA metabolism, DNA Breaks, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Mice, Mice, Knockout, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics, T-Lymphocytes immunology, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Cell Cycle Proteins physiology, DNA-Binding Proteins physiology, Gene Rearrangement, T-Lymphocyte, Protein Serine-Threonine Kinases physiology, Thymocytes immunology, Tumor Suppressor Proteins physiology, V(D)J Recombination
- Abstract
During the CD4(-)CD8(-) (DN) stage of T-cell development, RAG-dependent DNA breaks and V(D)J recombination occur at three T-cell receptor (TCR) loci: TCRβ, TCRγ and TCRδ. During this stage, abnormal trans-rearrangements also take place between TCR loci, occurring at increased frequency in absence of the DNA damage response mediator ataxia telangiectasia mutated (ATM). Here, we use this model of physiologic trans-rearrangement to study factors that predispose to rearrangement and the role of ATM in preventing chromosomal translocations. The frequency of DN thymocytes with DNA damage foci at multiple TCR loci simultaneously is increased 2- to 3-fold in the absence of ATM. However, trans-rearrangement is increased 10 000- to 100 000-fold, indicating that ATM function extends beyond timely resolution of DNA breaks. RAG-mediated synaptic complex formation occurs between recombination signal sequences with unequal 12 and 23 base spacer sequences (12/23 rule). TCR trans-rearrangements violate this rule, as we observed similar frequencies of 12/23 and aberrant 12/12 or 23/23 recombination products. This suggests that trans-rearrangements are not the result of trans-synaptic complex formation, but they are instead because of unstable cis synaptic complexes that form simultaneously at distinct TCR loci. Thus, ATM suppresses trans-rearrangement primarily through stabilization of DNA breaks at TCR loci.
- Published
- 2013
- Full Text
- View/download PDF
43. The NIH toolbox: setting a standard for biomedical research.
- Author
-
Hodes RJ, Insel TR, and Landis SC
- Subjects
- Biomedical Research methods, Humans, Internet, United States, Biomedical Research standards, National Institutes of Health (U.S.), Neurosciences standards
- Abstract
This special issue of Neurology(®) marks the unveiling of a multi-year effort to develop the NIH Toolbox for Assessment of Neurological and Behavioral Function (NIH Toolbox). Constructed based on state-of-the-art psychometric research and novel testing methods, this approach to functional neurologic measurement is as innovative in concept as it is in design. This initiative and the resulting set of instruments, supported through the NIH Blueprint for Neuroscience Research (NIH Blueprint) and built by a development team of more than 250 scientists from almost 100 academic institutions, promises to provide long overdue economies of scale and efficiency to the clinical research enterprise. The NIH Toolbox achieves that end by providing psychometrically sound, cutting-edge, adaptable measures that enable uniformity of measurement, data sharing, and integration of findings in the research setting.
- Published
- 2013
- Full Text
- View/download PDF
44. Prohibitins and the cytoplasmic domain of CD86 cooperate to mediate CD86 signaling in B lymphocytes.
- Author
-
Lucas CR, Cordero-Nieves HM, Erbe RS, McAlees JW, Bhatia S, Hodes RJ, Campbell KS, and Sanders VM
- Subjects
- Active Transport, Cell Nucleus, Animals, B7-2 Antigen chemistry, CD40 Antigens metabolism, Cell Line, Cell Nucleus metabolism, Female, Gene Expression Regulation, Mice, NF-kappa B metabolism, Phospholipase C gamma metabolism, Prohibitins, Protein Binding, Protein Kinase C metabolism, Repressor Proteins genetics, B-Lymphocytes metabolism, B7-2 Antigen metabolism, Protein Interaction Domains and Motifs, Repressor Proteins metabolism, Signal Transduction
- Abstract
CD86 engagement on a CD40L/IL-4-primed murine B cell activates signaling intermediates that promote NF-κB activation to increase Oct-2 and mature IgG1 mRNA and protein expression, as well as the rate of IgG1 transcription, without affecting class switch recombination. One of the most proximal signaling intermediates identified is phospholipase Cγ2, a protein reported to bind tyrosine residues, which are absent in the cytoplasmic domain of CD86. Using a proteomics-based identification approach, we show that the tyrosine-containing transmembrane adaptor proteins prohibitin (Phb)1 and Phb2 bind to CD86. The basal expression of Phb1/2 and association with CD86 was low in resting B cells, whereas the level of expression and association increased primarily after priming with CD40. The CD86-induced increase in Oct-2 and IgG1 was less when either Phb1/2 expression was reduced by short hairpin RNA or the cytoplasmic domain of CD86 was truncated or mutated at serine/threonine protein kinase C phosphorylation sites, which did not affect Phb1/2 binding to CD86. Using this approach, we also show that Phb1/2 and the CD86 cytoplasmic domain are required for the CD86-induced phosphorylation of IκBα, which we previously reported leads to NF-κB p50/p65 activation, whereas only Phb1/2 was required for the CD86-induced phosphorylation of phospholipase Cγ2 and protein kinase Cα/β(II), which we have previously reported leads to NF-κB (p65) phosphorylation and subsequent nuclear translocation. Taken together, these findings suggest that Phb1/2 and the CD86 cytoplasmic domain cooperate to mediate CD86 signaling in a B cell through differential phosphorylation of distal signaling intermediates required to increase IgG1.
- Published
- 2013
- Full Text
- View/download PDF
45. Editorial overview.
- Author
-
Akbar AN and Hodes RJ
- Subjects
- Humans, Aging physiology, Immune System physiology
- Published
- 2012
- Full Text
- View/download PDF
46. National Institute on Aging at middle age--its past, present, and future.
- Author
-
Nagy CL, Bernard MA, and Hodes RJ
- Subjects
- Alzheimer Disease prevention & control, Forecasting, History, 20th Century, History, 21st Century, Humans, Organizational Objectives, Translational Research, Biomedical history, United States, Vulnerable Populations, Aging, Biomedical Research, National Institutes of Health (U.S.) history
- Abstract
The National Institute on Aging (NIA) at the National Institutes of Health (NIH) leads the federal effort conducting and supporting research on aging. It is also designated as the lead within NIH for research on Alzheimer's disease. Since NIA's establishment in 1974, it has grown to a billion dollar enterprise featuring a balanced program of basic, clinical, and behavioral and social science. Investigator-initiated research and strategic investments have been critical to the NIA's success in bringing new insights and understandings to aging processes and diseases and conditions associated with advancing age. In recent years, constraints in the growth of resources have posed new challenges as the NIA and NIH leadership seek to maintain a robust and productive program. This article will review the history of the NIA, discuss current programs and priorities, and point to new directions in research, looking ahead., (© 2012, Copyright the Authors Journal compilation © 2012, The American Geriatrics Society.)
- Published
- 2012
- Full Text
- View/download PDF
47. Exon 1 disruption alters tissue-specific expression of mouse p53 and results in selective development of B cell lymphomas.
- Author
-
Chiang YJ, Difilippantonio MJ, Tessarollo L, Morse HC, and Hodes RJ
- Subjects
- Animals, B-Lymphocytes metabolism, Blotting, Western, DNA Primers genetics, Exons genetics, Flow Cytometry, Genotype, Humans, Karyotyping, Lymphoma, B-Cell genetics, Mice, Mice, Knockout, Reverse Transcriptase Polymerase Chain Reaction, Thymus Gland metabolism, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Disease Models, Animal, Gene Expression Regulation genetics, Lymphoma, B-Cell metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 is a tumor suppressor gene mutated in >50% of human cancers, while p53 deficiency in mice results in cancers and accelerated mortality. Thymic T cell lymphoma is the most common malignancy in p53-deficient mice, making it difficult to study the role of p53 in other malignancies. To overcome this limitation, we attempted to generate mice with a reversible p53 knockout (p53(rev/rev)) by inserting a floxed transcriptional stop into the first exon of p53, anticipating that this would allow tissue-specific Cre-mediated expression of p53. Contrary to expectations, functional p53 protein was expressed in the thymus and multiple other tissues of p53(rev/rev) mice in the absence of Cre, whereas B cells expressed p53 protein only in the presence of B cell-specific CD19-Cre. In the absence of Cre, 76% of p53(rev/rev) mice developed splenic marginal zone B cell lymphomas, indicating sensitivity of this B cell subset to transformation caused by p53 deficiency. 5'-RACE identified p53 mRNA transcribed from a novel start site utilized in thymocytes but not normal B cells or B cell lymphomas from p53(rev/rev) mice. The p53(rev/rev) mouse thus demonstrates an effect of p53 deficiency in development of splenic marginal zone lymphomas and provides a model for study of p53-deficient human B cell lymphomas.
- Published
- 2012
- Full Text
- View/download PDF
48. Cbl enforces Vav1 dependence and a restricted pathway of T cell development.
- Author
-
Chiang J and Hodes RJ
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Blotting, Western, Membrane Proteins metabolism, Mice, Mice, Knockout, Phosphoproteins metabolism, Phosphorylation, Proto-Oncogene Proteins c-cbl immunology, Proto-Oncogene Proteins c-vav immunology, Signal Transduction genetics, Signal Transduction physiology, T-Lymphocytes immunology, ZAP-70 Protein-Tyrosine Kinase metabolism, Proto-Oncogene Proteins c-cbl metabolism, Proto-Oncogene Proteins c-vav metabolism, T-Lymphocytes metabolism
- Abstract
Extensive studies of pre-TCR- and TCR-dependent signaling have led to characterization of a pathway deemed essential for efficient T cell development, and comprised of a cascade of sequential events involving phosphorylation of Lck and ZAP-70, followed by phosphorylation of LAT and SLP-76, and subsequent additional downstream events. Of interest, however, reports from our lab as well as others have indicated that the requirements for ZAP-70, LAT, and SLP-76 are partially reversed by inactivation of c-Cbl (Cbl), an E3 ubiquitin ligase that targets multiple molecules for ubiquitination and degradation. Analysis of signaling events in these Cbl knockout models, including the recently reported analysis of SLP-76 transgenes defective in interaction with Vav1, suggested that activation of Vav1 might be a critical event in alternative pathways of T cell development. To extend the analysis of signaling requirements for thymic development, we have therefore assessed the effect of Cbl inactivation on the T cell developmental defects that occur in Vav1-deficient mice. The defects in Vav1-deficient thymic development, including a marked defect in DN3-DN4 transition, were completely reversed by Cbl inactivation, accompanied by enhanced phosphorylation of PLC-γ1 and ERKs in response to pre-TCR/TCR cross-linking of Vav1⁻/⁻Cbl⁻/⁻ DP thymocytes. Taken together, these results suggest a substantially modified paradigm for pre-TCR/TCR signaling and T cell development. The observed consensus pathways of T cell development, including requirements for ZAP-70, LAT, SLP-76, and Vav1, appear to reflect the restriction by Cbl of an otherwise much broader set of molecular pathways capable of mediating T cell development.
- Published
- 2011
- Full Text
- View/download PDF
49. The requirement for pre-TCR during thymic differentiation enforces a developmental pause that is essential for V-DJβ rearrangement.
- Author
-
Hathcock KS, Farrington L, Ivanova I, Livak F, Selimyan R, Sen R, Williams J, Tai X, and Hodes RJ
- Subjects
- Animals, B7-2 Antigen genetics, B7-2 Antigen metabolism, CD28 Antigens genetics, CD28 Antigens metabolism, DNA-Binding Proteins metabolism, Gene Expression Regulation immunology, Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor, Histones chemistry, Histones metabolism, Lysine, Methylation, Mice, Mice, Transgenic, T-Lymphocytes cytology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Thymus Gland metabolism, Transcription, Genetic immunology, Cell Differentiation, Gene Rearrangement, beta-Chain T-Cell Antigen Receptor, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta metabolism, Thymus Gland cytology
- Abstract
T cell development occurs in the thymus and is critically dependent on productive TCRβ rearrangement and pre-TCR expression in DN3 cells. The requirement for pre-TCR expression results in the arrest of thymocytes at the DN3 stage (β checkpoint), which is uniquely permissive for V-DJβ recombination; only cells expressing pre-TCR survive and develop beyond the DN3 stage. In addition, the requirement for TCRβ rearrangement and pre-TCR expression enforces suppression of TCRβ rearrangement on a second allele, allelic exclusion, thus ensuring that each T cell expresses only a single TCRβ product. However, it is not known whether pre-TCR expression is essential for allelic exclusion or alternatively if allelic exclusion is enforced by developmental changes that can occur in the absence of pre-TCR. We asked if thymocytes that were differentiated without pre-TCR expression, and therefore without pause at the β checkpoint, would suppress all V-DJβ rearrangement. We previously reported that premature CD28 signaling in murine CD4(-)CD8(-) (DN) thymocytes supports differentiation of CD4(+)CD8(+) (DP) cells in the absence of pre-TCR expression. The present study uses this model to define requirements for TCRβ rearrangement and allelic exclusion. We demonstrate that if cells exit the DN3 developmental stage before TCRβ rearrangement occurs, V-DJβ rearrangement never occurs, even in DP cells that are permissive for D-Jβ and TCRα rearrangement. These results demonstrate that pre-TCR expression is not essential for thymic differentiation to DP cells or for V-DJβ suppression. However, the requirement for pre-TCR signals and the exclusion of alternative stimuli such as CD28 enforce a developmental "pause" in early DN3 cells that is essential for productive TCRβ rearrangement to occur.
- Published
- 2011
- Full Text
- View/download PDF
50. Decreased glucocerebrosidase activity in Gaucher disease parallels quantitative enzyme loss due to abnormal interaction with TCP1 and c-Cbl.
- Author
-
Lu J, Chiang J, Iyer RR, Thompson E, Kaneski CR, Xu DS, Yang C, Chen M, Hodes RJ, Lonser RR, Brady RO, and Zhuang Z
- Subjects
- Acetylcysteine analogs & derivatives, Acetylcysteine pharmacology, Cell Line, Chaperonin Containing TCP-1 genetics, Cysteine Proteinase Inhibitors pharmacology, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts physiology, Gaucher Disease genetics, Humans, Molecular Chaperones metabolism, Proteasome Endopeptidase Complex metabolism, Proto-Oncogene Proteins c-cbl genetics, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Chaperonin Containing TCP-1 metabolism, Gaucher Disease enzymology, Glucosylceramidase metabolism, Proto-Oncogene Proteins c-cbl metabolism
- Abstract
Gaucher disease (GD), the most common lysosomal storage disorder of humans, is caused by mutations in the gene coding for the enzyme glucocerebrosidase (GCase). Clinical manifestations vary among patients with the three types of GD, and phenotypic heterogeneity occurs even among patients with identical mutations. To gain insight into why phenotypic heterogeneity occurs in GD, we investigated mechanisms underlying the net loss of GCase catalytic activity in cultured skin fibroblasts derived from patients with the three types of GD. The findings indicate that the loss of catalytic activity of GCase correlates with its quantitative reduction, rather than a decrease in functional capacity of mutant enzyme. Use of a proteasome inhibitor, lactacystin, resulted in increased expression of GCase, suggesting a mechanism of protein degradation in GD. Furthermore, reduced binding of GCase to TCP1 ring complex (TRiC), a regulator of correct protein folding, may result in defective maturation of nascent GCase in GD cells. Additionally, increased interaction between GCase and c-Cbl, an E3 ubiquitin ligase, may be involved in the degradation and loss of GCase in GD. The findings suggest that specific molecular mediators involved in GCase maturation and degradation could be responsible for phenotypic variation among patients with the same genotypes and that these mediators could be therapeutically targeted to increase GCase activity in patients with GD.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.