Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Transplant ; 31: 9636897221136149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36367048

RESUMO

CD8 T cells play a key role in cancer immunotherapy and allograft rejection. However, it is not clear how they kill cells and tissues that do not have the agonist peptide-major histocompatibility complex (MHC) on their surface, as in the settings of MHC class I deficient tumors and indirect rejection of MHC-mismatched transplants. CD8 T cells might respond to agonist antigen cross-presented on hematopoietic cells, leading to a "bystander" rejection. Alternatively, they may recognize agonist antigen cross-presented on recipient endothelial cells and kill the tissue's vital blood supply. The latter mechanism predicts that all non-vascularized grafts, grafts dependent on in-growth of recipient blood vessels, will be susceptible to CD8 T cell mediated indirect rejection. In contrast, we show here that non-vascularized transplants, bearing the same agonist antigen, are not universally susceptible to this rejection pathway. Non-vascularized skin, but not islet or heart tissue transplants were indirectly rejected by CD8 T cells. Furthermore, CD8 T cells were able to indirectly reject skin grafts when recipient MHC class I expression was restricted to bone marrow derived cells but not when it was restricted to radioresistant cells (e.g. endothelial cells). These findings argue against a major role for endothelial cell cross-presentation in killing of tissue that does not present the agonist peptide-MHC class I. Instead, the data suggests that cross-presentation by recipient hematopoietic cells underlies the CD8 T cell mediated killing of tissue that is unable to directly present the target peptide-MHC class I.


Assuntos
Medula Óssea , Células Endoteliais , Transplante Homólogo , Rejeição de Enxerto , Linfócitos T CD8-Positivos , Complexo Principal de Histocompatibilidade , Antígenos de Histocompatibilidade Classe I , Peptídeos , Linfócitos T CD4-Positivos
2.
Front Immunol ; 9: 837, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29740442

RESUMO

Stable mixed hematopoietic chimerism is a robust method for inducing donor-specific tolerance with the potential to prevent rejection of donor islets in recipients with autoimmune type-1 diabetes. However, with reduced intensity conditioning, fully allogeneic chimerism in a tolerance resistant autoimmune-prone non-obese diabetic (NOD) recipient has rarely been successful. In this setting, successful multilineage chimerism has required either partial major histocompatability complex matching, mega doses of bone marrow, or conditioning approaches that are not currently clinically feasible. Irradiation free protocols with moderate bone marrow doses have not generated full tolerance; donor skin grafts were rejected. We tested whether more efficient recipient T cell depletion would generate a more robust tolerance. We show that a combination of donor-specific transfusion-cyclophosphamide and multiple T cell depleting antibodies could induce stable high levels of fully allogeneic chimerism in NOD recipients. Less effective T cell depletion was associated with instability of chimerism. Stable chimeras appeared fully donor-specific tolerant, with clonal deletion of allospecific T cells and acceptance of donor skin grafts, while recovering substantial immunocompetence. The loss of chimerism months after transplant was significantly associated with a lower level of chimerism and donor T cells within the first 2 weeks after transplant. Thus, rapid and robust recipient T cell depletion allows for stable high levels of fully allogeneic chimerism and robust donor-specific tolerance in the stringent NOD model while using a clinically feasible protocol. In addition, these findings open the possibility of identifying recipients whose chimerism will later fail, stratifying patients for early intervention.


Assuntos
Diabetes Mellitus Experimental/imunologia , Depleção Linfocítica , Quimeras de Transplante/imunologia , Tolerância ao Transplante , Animais , Ciclofosfamida/administração & dosagem , Feminino , Imunossupressores/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Transplante de Pele , Linfócitos T/citologia , Condicionamento Pré-Transplante , Transplante Homólogo
3.
Eur J Immunol ; 44(8): 2263-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24777700

RESUMO

Cervical thymus mimics the thoracic thymus in supporting T-cell development and exists in a subset of mice and humans. Importantly, it remains unknown whether the cervical thymus can generate T cells that are self-tolerant in the complete absence of signals from the thoracic thymus. Using a fetal liver reconstitution model in thoracic thymectomized RAG(-/-) mice, we found that T cells could be generated without contribution from the thoracic thymus. However, these mice had decreased T cells, increased proportions of effector memory T cells and Treg phenotype cells, increased serum IgG1/2b, and increased frequency of T cells expressing IFN-γ, IL-17 or IL-10. Half of the mice that received a thoracic thymectomy and fetal liver cells, unlike sham surgery controls, developed substantial morbidity with age. Disease was associated with lymphopenia-driven activation rather than inherent defects in the cervical thymus, as both thoracic and cervical thymocytes could generate disease in lymphopenic recipients. Administration of the homeostatic cytokine IL-7 caused a rapid, transient increase in T-cell numbers and reduced the time to disease onset. Together the data suggests that the cervical thymus can function in the complete absence of the thoracic thymus; however, the T cells generated do not establish homeostasis.


Assuntos
Linfócitos T/imunologia , Tórax/imunologia , Timo/imunologia , Animais , Homeostase , Imunoglobulina G/imunologia , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-17/imunologia , Interleucina-7/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Timo/citologia
4.
Chimerism ; 4(1): 32-3, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23434631

RESUMO

Recently, our group reported the presence of microchimerism (Mc) in the human brain by performing quantitative PCR on female human brain tissues to amplify male DNA. We found brain Mc to be relatively frequent in humans and widely distributed in this organ. Our data also suggested a lower prevalence of brain Mc in women without Alzheimer disease than women without neurological disease. Altogether, these findings suggest that Mc could sometimes influence health and disease of the brain. As further research will be required to clarify this issue, here we discuss some of the questions that could be addressed to improve our understanding.


Assuntos
Doença de Alzheimer/imunologia , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Quimerismo , Doença de Alzheimer/epidemiologia , DNA/genética , Feminino , Humanos , Masculino , Gravidez
5.
PLoS One ; 7(9): e45592, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049819

RESUMO

In humans, naturally acquired microchimerism has been observed in many tissues and organs. Fetal microchimerism, however, has not been investigated in the human brain. Microchimerism of fetal as well as maternal origin has recently been reported in the mouse brain. In this study, we quantified male DNA in the human female brain as a marker for microchimerism of fetal origin (i.e. acquisition of male DNA by a woman while bearing a male fetus). Targeting the Y-chromosome-specific DYS14 gene, we performed real-time quantitative PCR in autopsied brain from women without clinical or pathologic evidence of neurologic disease (n=26), or women who had Alzheimer's disease (n=33). We report that 63% of the females (37 of 59) tested harbored male microchimerism in the brain. Male microchimerism was present in multiple brain regions. Results also suggested lower prevalence (p=0.03) and concentration (p=0.06) of male microchimerism in the brains of women with Alzheimer's disease than the brains of women without neurologic disease. In conclusion, male microchimerism is frequent and widely distributed in the human female brain.


Assuntos
Doença de Alzheimer/genética , Encéfalo/metabolismo , Quimerismo , Troca Materno-Fetal/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Autopsia , Encéfalo/patologia , Cromossomos Humanos Y , Feminino , Feto , Humanos , Masculino , Pessoa de Meia-Idade , Gravidez , Reação em Cadeia da Polimerase em Tempo Real
6.
Arthritis Rheum ; 64(2): 380-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21953057

RESUMO

OBJECTIVE: The rheumatoid nodule is a lesion commonly found on extraarticular areas prone to mechanic trauma. When present with inflammatory symmetric polyarthritis, it is pathognomonic of rheumatoid arthritis (RA), an autoimmune disease in which naturally acquired microchimerism has previously been described and can sometimes contribute to RA risk. Since RA patients harbor microchimerism in the blood, we hypothesized that microchimerism is also present in rheumatoid nodules and could play a role in rheumatoid nodule formation. This study was undertaken to investigate rheumatoid nodules for microchimerism. METHODS: Rheumatoid nodules were tested for microchimerism by real-time quantitative polymerase chain reaction (qPCR). The rheumatoid nodules of 29 female patients were tested for a Y chromosome-specific sequence. After HLA genotyping of patients and family members, rheumatoid nodules from 1 man and 14 women were tested by HLA-specific qPCR, targeting a nonshared HLA allele of the potential microchimerism source. Results were expressed as genome equivalents of microchimeric cells per 10(5) patient genome equivalents (GE/10(5)). RESULTS: Rheumatoid nodules from 21% of the female patients contained male DNA (range <0.5, 10.3 GE/10(5)). By HLA-specific qPCR, 60% of patients were microchimeric (range 0, 18.5 GE/10(5)). Combined microchimerism prevalence was 47%. A fetal or maternal source was identified in all patients who tested positive by HLA-specific qPCR. Unexpectedly, a few rheumatoid nodules also contained microchimerism without evidence of a fetal or maternal source, suggesting alternative sources. CONCLUSION: Our findings indicate that microchimerism is frequently present in the rheumatoid nodules of RA patients. Since microchimerism is genetically disparate, whether microchimerism in rheumatoid nodules serves as an allogeneic stimulus or allogeneic target warrants further investigation.


Assuntos
Artrite Reumatoide/genética , Quimerismo , Nódulo Reumatoide/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Artrite Reumatoide/imunologia , Feminino , Genótipo , Antígenos HLA/genética , Humanos , Masculino , Pessoa de Meia-Idade , Nódulo Reumatoide/imunologia
7.
Transplantation ; 92(9): 977-84, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21956197

RESUMO

BACKGROUND: Induction of allogeneic hematopoietic chimerism is a promising strategy to induce tolerance to donor islets for treating type 1 diabetes. Successful induction of chimerism requires overcoming host alloimmunity. In diabetes-prone nonobese diabetic (NOD) mice, this is challenging due to their general tolerance resistance. Although the adaptive alloimmunity of NOD mice is a known barrier to allogeneic chimerism, whether NOD natural killer (NK) cells are an additional barrier has not been examined. Because NOD NK cells exhibit functional defects, they may not inhibit chimerism generation. METHODS: Antibody depletion of NK cells in vivo, or transplantation of F1 hybrid donor cells to eliminate the "missing-self" trigger of NK cells, was preformed to test the NK-mediated rejection of donor bone marrow cells. We also studied the capacity of rapamycin to block the NK cell response against allogeneic cells in vivo. RESULTS: Depleting NK cells or rendering them unresponsive to the donor greatly improved the level of chimerism obtained in NOD mice. Rapamycin significantly reduced the resistance to allogeneic chimerism mounted by NOD NK cells; however, it was much less effective than NK cell depletion by antibodies. CONCLUSIONS: Contrary to the view that NOD NK cells are defective, we found these cells to be a substantial barrier to allogeneic chimerism in the presence or absence of adaptive immunity. Moreover, rapamycin will need to be combined with other approaches to fully overcome the NK cell barrier.


Assuntos
Quimerismo/efeitos dos fármacos , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/cirurgia , Imunossupressores/farmacologia , Transplante das Ilhotas Pancreáticas , Células Matadoras Naturais/patologia , Sirolimo/farmacologia , Imunidade Adaptativa/fisiologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Células-Tronco Hematopoéticas/patologia , Células Matadoras Naturais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Quimeras de Transplante/imunologia , Transplante Homólogo
8.
J Immunol ; 181(7): 4603-12, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802062

RESUMO

CD4 T cells frequently help to activate CD8 T and B cells that effect transplant rejection. However, CD4 T cells alone can reject transplants, either directly or indirectly. The relative effectiveness of indirect CD4 immunity in rejecting different types of allogeneic grafts is unknown. To address this, we used a TCR transgenic mouse model in which indirect CD4 alloimmunity alone can be studied. We challenged transgenic recipients with hematopoietic cells and shortly thereafter skin transplants that could only be rejected indirectly, and observed Ag-specific indirect donor B cell and skin rejection, but not T cell elimination, reflecting a state of split tolerance. Deficiency of indirect CD4 alloimmunity in donor T cell rejection was also apparent when acute indirect rejection of donor islets occurred despite generation and maintenance of mixed T cell chimerism, due to migration of the few passenger T cells into recipient circulation. Although passenger lymphocytes delayed indirect islet rejection, they enhanced rejection by a full repertoire capable of both direct and indirect reactivity. Interestingly, the persistence of chimerism was associated with the eventual development of tolerance, as demonstrated by acceptance of donor skin grafts given late to hematopoietic cell recipients, and hyporesponsiveness of transgenic T cells from islet recipients in vitro. Mechanistically, tolerance was recessive and associated with progressive down-regulation of CD4. Collectively, our data indicate that indirect CD4 immunity is not equally destructive toward different types of allogeneic grafts, the deficiency of which generates split tolerance. The futility of these responses can convert immunity into tolerance.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Isoantígenos/imunologia , Tolerância ao Transplante/imunologia , Animais , Quimera/imunologia , Feminino , Antígeno H-Y/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Transplante de Pele/imunologia , Transplante de Pele/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tolerância ao Transplante/genética
9.
J Immunol ; 180(8): 5177-86, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18390699

RESUMO

Hematopoietic chimerism is considered to generate robust allogeneic tolerance; however, tissue rejection by chimeras can occur. This "split tolerance" can result from immunity toward tissue-specific Ags not expressed by hematopoietic cells. Known to occur in chimeric recipients of skin grafts, it has not often been reported for other donor tissues. Because chimerism is viewed as a potential approach to induce islet transplantation tolerance, we generated mixed bone marrow chimerism in the tolerance-resistant NOD mouse and tested for split tolerance. An unusual multilevel split tolerance developed in NOD chimeras, but not chimeric B6 controls. NOD chimeras demonstrated persistent T cell chimerism but rejected other donor hematopoietic cells, including B cells. NOD chimeras also showed partial donor alloreactivity. Furthermore, NOD chimeras were split tolerant to donor skin transplants and even donor islet transplants, unlike control B6 chimeras. Surprisingly, islet rejection was not a result of autoimmunity, since NOD chimeras did not reject syngeneic islets. Split tolerance was linked to non-MHC genes of the NOD genetic background and was manifested recessively in F(1) studies. Also, NOD chimeras but not B6 chimeras could generate serum alloantibodies, although at greatly reduced levels compared with nonchimeric controls. Surprisingly, the alloantibody response was sufficiently cross-reactive that chimerism-induced humoral tolerance extended to third-party cells. These data identify split tolerance, generated by a tolerance-resistant genetic background, as an important new limitation to the chimerism approach. In contrast, the possibility of humoral tolerance to multiple donors is potentially beneficial.


Assuntos
Transplante de Medula Óssea/imunologia , Tolerância Imunológica , Transplante das Ilhotas Pancreáticas/imunologia , Isoantígenos/imunologia , Transplante de Pele/imunologia , Quimeras de Transplante/imunologia , Tolerância ao Transplante , Animais , Autoimunidade , Linfócitos B/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos NOD , Linfócitos T/imunologia , Quimeras de Transplante/metabolismo
10.
Biol Direct ; 2: 10, 2007 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-17437644

RESUMO

BACKGROUND: Transplant rejection has been considered to occur primarily because donor antigens are not present during the development of the recipient's immune system to induce tolerance. Thus, transplantation prior to recipient immune system development (pre-immunocompetence transplants) should induce natural tolerance to the donor. Surprisingly, tolerance was often not the outcome in such 'natural tolerance models'. We explored the ability of natural tolerance to prevent immune responses to alloantigens, and the reasons for the disparate outcomes of pre-immunocompetence transplants. RESULTS: We found that internal transplants mismatched for a single minor-H antigen and 'healed-in' before immune system development were not ignored but instead induced natural tolerance. In contrast, multiple minor-H or MHC mismatched transplants did not consistently induce natural tolerance unless they carried chimerism generating passenger lymphocytes. To determine whether the systemic nature of passenger lymphocytes was required for their tolerizing capacity, we generated a model of localized vs. systemic donor lymphocytes. We identified the peritoneal cavity as a site that protects allogeneic lymphocytes from killing by NK cells, and found that systemic chimerism, but not chimerism restricted to the peritoneum, was capable of generating natural tolerance. CONCLUSION: These data provide an explanation for the variable results with pre-immunocompetence transplants and suggest that natural tolerance to transplants is governed by the systemic vs. localized nature of donor antigen, the site of transplantation, and the antigenic disparity. Furthermore, in the absence of systemic lymphocyte chimerism the capacity to establish natural tolerance to allogeneic tissue appears strikingly limited. REVIEWERS: This article was reviewed by Matthias von Herrath, Irun Cohen, and Wei-Ping Min (nominated by David Scott).

11.
Blood ; 109(12): 5346-54, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17327412

RESUMO

Researchers designing antitumor treatments have long focused on eliciting tumor-specific CD8 cytotoxic T lymphocytes (CTL) because of their potent killing activity and their ability to reject transplanted organs. The resulting treatments, however, have generally been surprisingly poor at inducing complete tumor rejection, both in experimental models and in the clinic. Although a few scattered studies suggested that CD4 T "helper" cells might also serve as antitumor effectors, they have generally been studied mostly for their ability to enhance the activity of CTL. In this mouse study, we compared monoclonal populations of tumor-specific CD4 and CD8 T cells as effectors against several different tumors, and found that CD4 T cells eliminated tumors that were resistant to CD8-mediated rejection, even in cases where the tumors expressed major histocompatibility complex (MHC) class I molecules but not MHC class II. MHC class II expression on host tissues was critical, suggesting that the CD4 T cells act indirectly. Indeed, the CD4 T cells partnered with NK cells to obtain the maximal antitumor effect. These findings suggest that CD4 T cells can be powerful antitumor effector cells that can, in some cases, outperform CD8 T cells, which are the current "gold standard" effector cell in tumor immunotherapy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias/imunologia , Animais , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunoterapia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Transgênicos , Linfócitos T Citotóxicos/imunologia
12.
Eur J Immunol ; 37(5): 1233-42, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17390394

RESUMO

Stable mixed chimerism has been considered the most robust tolerance strategy. However, rejection of solid donor tissues by chimeras has been observed, a state termed split tolerance. Since new non-myeloablative mixed chimerism approaches are being actively pursued, we sought to determine whether they lead to full tolerance or split tolerance and to define the mechanisms involved. Fully mismatched mixed chimeras generated by induction with various lymphocyte-depleting antibodies along with either low-dose irradiation or busulfan and temporary sirolimus, maintained stable mixed chimerism but nevertheless rejected donor skin grafts. Generation of stable mixed chimerism using antibody targeting CD40L, but not depleting antibodies to CD4 and CD8, could prevent split tolerance when skin grafts were given together with donor bone marrow. Minor antigen matching abrogated the ability of effector T cells to reject donor skin grafts. A CFSE killing assay indicated that chimeras were both directly and indirectly tolerant of donor hematopoietic cell antigens, suggesting that minor mismatches triggered a tissue-specific response. Thus, split tolerance due to tissue-restricted polymorphic antigens prevents full tolerance in a number of non-myeloablative mixed chimerism protocols and a 'tolerizing' agent is required to overcome split tolerance. A model of the requirements for split tolerance is presented.


Assuntos
Quimera/imunologia , Quimerismo , Rejeição de Enxerto/prevenção & controle , Tolerância ao Transplante/imunologia , Animais , Antígenos CD/imunologia , Quimera/genética , Citometria de Fluxo , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Camundongos , Polimorfismo Genético , Transplante de Pele/imunologia , Tolerância ao Transplante/genética
13.
Front Biosci ; 9: 2947-63, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15353328

RESUMO

Each response made by our immune system is either to promote or to prevent immune reactivity. That the immune system can successfully achieve both goals under specific biological conditions depends on central and peripheral tolerance mechanisms. Over the years, various experimental systems have been generated to study peripheral CD4 T cell tolerance. The experimental approaches are clearly diverse, but can be broadly categorized into those involving, tolerogenic, antigen injections, the use of transgenic mice (antigen transgenics, TCR transgenics and the combination thereof) and transplantation models. Results of these studies suggested a number of potential mechanisms mediating peripheral CD4 T cell tolerance, including clonal deletion, anergy and immune regulation. While the available systems all presented with their own limitations, they nevertheless provided a foundation from which our understanding of peripheral CD4 T cell tolerance will be built. In addition, previous studies done to examine CD8 T cell tolerance left with us some potentially helpful clues and ideas about how CD4 T cell tolerance should be studied. However, our current understanding on the immunity/tolerance decision made by CD4 T cells in the periphery remains incomplete. Evidence accumulated thus far favors the view that peripheral tolerance, unlike central tolerance, may not primarily determine the selection of specificities within the baseline T cell repertoire but may instead regulate cellular responses within the repertoire, in terms of both expansion/contraction and differentiation. Nevertheless, more robust and refined models need to be developed before we can make definitive conclusions about the potential role of peripheral tolerance in repertoire selection mediated by deletion of self-reactive cells.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Animais , Antígenos/química , Doenças Autoimunes/imunologia , Linfócitos T CD8-Positivos/metabolismo , Transplante de Células , Humanos , Sistema Imunitário , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Transgenes , Tolerância ao Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...