Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Rep Med ; 2(1): 100185, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33521699

RESUMO

BCG vaccination can strengthen protection against pathogens through the induction of epigenetic and metabolic reprogramming of innate immune cells, a process called trained immunity. We and others recently demonstrated that mucosal or intravenous BCG better protects rhesus macaques from Mycobacterium tuberculosis infection and TB disease than standard intradermal vaccination, correlating with local adaptive immune signatures. In line with prior mouse data, here, we show in rhesus macaques that intravenous BCG enhances innate cytokine production associated with changes in H3K27 acetylation typical of trained immunity. Alternative delivery of BCG does not alter the cytokine production of unfractionated bronchial lavage cells. However, mucosal but not intradermal vaccination, either with BCG or the M. tuberculosis-derived candidate MTBVAC, enhances innate cytokine production by blood- and bone marrow-derived monocytes associated with metabolic rewiring, typical of trained immunity. These results provide support to strategies for improving TB vaccination and, more broadly, modulating innate immunity via mucosal surfaces.


Assuntos
Vacina BCG/administração & dosagem , Imunidade nas Mucosas , Mycobacterium tuberculosis/imunologia , Mucosa Respiratória/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Tuberculose Pulmonar/prevenção & controle , Acetilação , Administração Intranasal , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/microbiologia , Reprogramação Celular/genética , Reprogramação Celular/imunologia , Feminino , Regulação da Expressão Gênica , Histonas/genética , Histonas/imunologia , Injeções Intravenosas , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/microbiologia , Macaca mulatta , Masculino , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , Mycobacterium tuberculosis/patogenicidade , Mucosa Respiratória/microbiologia , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
2.
Cell Rep Med ; 2(1): 100187, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33521701

RESUMO

To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.


Assuntos
Vacina BCG/administração & dosagem , Imunidade nas Mucosas , Mycobacterium tuberculosis/imunologia , Mucosa Respiratória/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Tuberculose Pulmonar/prevenção & controle , Administração Intranasal , Animais , Reprogramação Celular/genética , Reprogramação Celular/imunologia , Feminino , Regulação da Expressão Gênica , Injeções Intradérmicas , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/microbiologia , Macaca mulatta , Masculino , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , Mycobacterium tuberculosis/patogenicidade , Mucosa Respiratória/microbiologia , Células Th1/imunologia , Células Th1/microbiologia , Células Th17/imunologia , Células Th17/microbiologia , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
3.
Clin Transl Immunology ; 9(7): e1150, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32642064

RESUMO

OBJECTIVES: Loss of vaccine-induced antibodies (Abs) after chemotherapy against paediatric acute lymphoblastic leukaemia (ALL) is common and often necessitates re-immunisation after cessation of treatment. Even so, some ALL survivors fail to mount or to maintain protective Abs. Germinal centres (GCs) are clusters of proliferating B cells in follicles of secondary lymphoid tissues (SLTs) formed during adaptive immune responses and the origins of long-lived memory B and plasma cells that are the source of Abs. Furthermore, productive GC reactions depend on T follicular helper (TFH) cells. To understand why chemotherapy induces deficits in Ab responses, we examined how SLTs were affected by chemotherapy. METHODS: Rhesus macaques were infused with either three cycles of the anthracycline doxorubicin or saline, followed by immunisation with a de novo and booster antigen. Spleen and lymph nodes were removed, and memory B, bulk T and TFH cells were examined. RESULTS: Despite adequate GC morphology, a diminished memory and IgG+ B-cell population along with diminished total and booster vaccine-specific IgG-producing memory B cells were noted in the spleens of macaques with past doxorubicin exposure compared to the saline-treated controls (P < 0.05). Intact bulk T and TFH cells were found in the SLTs of treated macaques, which displayed higher CD40L upregulation capacity by their splenic CXCR5+ helper T cells (P < 0.01). In contrast to the spleen, the immune cell populations studied were comparable between the lymph nodes of both saline- and doxorubicin-treated macaques. CONCLUSION: Our findings suggest that the splenic memory B-cell subset, compared to its lymph node counterpart, is more severely altered by anthracycline treatment.

4.
NPJ Vaccines ; 5(1): 39, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32435513

RESUMO

Tuberculosis (TB) still is the principal cause of death from infectious disease and improved vaccination strategies are required to reduce the disease burden and break TB transmission. Here, we investigated different routes of administration of vectored subunit vaccines based on chimpanzee-derived adenovirus serotype-3 (ChAd3) for homologous prime-boosting and modified vaccinia virus Ankara (MVA) for heterologous boosting with both vaccine vectors expressing the same antigens from Mycobacterium tuberculosis (Ag85B, ESAT6, Rv2626, Rv1733, RpfD). Prime-boost strategies were evaluated for immunogenicity and protective efficacy in highly susceptible rhesus macaques. A fully parenteral administration regimen was compared to exclusive respiratory mucosal administration, while parenteral ChAd3-5Ag prime-boosting and mucosal MVA-5Ag boosting were applied as a push-and-pull strategy from the periphery to the lung. Immune analyses corroborated compartmentalized responses induced by parenteral versus mucosal vaccination. Despite eliciting TB-specific immune responses, none of the investigational regimes conferred a protective effect by standard readouts of TB compared to non-vaccinated controls, while lack of protection by BCG underpinned the stringency of this non-human primate test modality. Yet, TB manifestation after full parenteral vaccination was significantly less compared to exclusive mucosal vaccination.

5.
Front Immunol ; 10: 2479, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31736945

RESUMO

While tuberculosis continues to afflict mankind, the immunological mechanisms underlying TB disease development are still incompletely understood. Advanced preclinical models for TB research include both rhesus and cynomolgus macaques (Macaca mulatta and Macaca fascicularis, respectively), with rhesus typically being more susceptible to acute progressive TB disease than cynomolgus macaques. To determine which immune mechanisms are responsible for this dissimilar disease development, we profiled a broad range of innate and adaptive responses, both local and peripheral, following experimental pulmonary Mycobacterium tuberculosis (Mtb) infection of both species. While T-cell and antibody responses appeared indistinguishable, we identified anti-inflammatory skewing of peripheral monocytes in rhesus and a more prominent local pro-inflammatory cytokine release profile in cynomolgus macaques associated with divergent TB disease outcome. Importantly, these differences were detectable both before and early after infection. This work shows that inflammatory and innate immune status prior to and at early stages after infection, critically affects outcome of TB infection.


Assuntos
Macaca fascicularis/imunologia , Macaca mulatta/imunologia , Mycobacterium tuberculosis , Tuberculose Pulmonar/imunologia , Animais , Citocinas/imunologia , Imunidade Inata , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
6.
Nat Med ; 25(2): 255-262, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30664782

RESUMO

Tuberculosis (TB) remains the deadliest infectious disease1, and the widely used Bacillus Calmette-Guérin (BCG) vaccine fails to curb the epidemic. An improved vaccination strategy could provide a cost-effective intervention to break the transmission cycle and prevent antimicrobial resistance2,3. Limited knowledge of the host responses critically involved in protective immunity hampers the development of improved TB vaccination regimens. Therefore, assessment of new strategies in preclinical models to select the best candidate vaccines before clinical vaccine testing remains indispensable. We have previously established in rhesus macaques (Macaca mulatta) that pulmonary mucosal BCG delivery reduces TB disease where standard intradermal injection fails4,5. Here, we show that pulmonary BCG prevents infection by using a repeated limiting-dose Mycobacterium tuberculosis challenge model and identify polyfunctional T-helper type 17 (TH17) cells, interleukin-10 and immunoglobulin A as correlates of local protective immunity. These findings warrant further research into mucosal immunization strategies and their translation to clinical application to more effectively prevent the spread of TB.


Assuntos
Vacina BCG/imunologia , Tuberculose/imunologia , Tuberculose/prevenção & controle , Animais , Carga Bacteriana , Relação Dose-Resposta Imunológica , Imunidade Humoral , Interferon gama/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macaca mulatta , Masculino , Mucosa/imunologia , Vacinação
7.
Sci Rep ; 7(1): 992, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28428630

RESUMO

Autoantibodies against ion channels are the cause of numerous neurologic autoimmune disorders. Frequently, such pathogenic autoantibodies have a restricted epitope-specificity. In such cases, competing antibody formats devoid of pathogenic effector functions (blocker antibodies) have the potential to treat disease by displacing autoantibodies from their target. Here, we have used a model of the neuromuscular autoimmune disease myasthenia gravis in rhesus monkeys (Macaca mulatta) to test the therapeutic potential of a new blocker antibody: MG was induced by passive transfer of pathogenic acetylcholine receptor-specific monoclonal antibody IgG1-637. The effect of the blocker antibody (IgG4Δhinge-637, the hinge-deleted IgG4 version of IgG1-637) was assessed using decrement measurements and single-fiber electromyography. Three daily doses of 1.7 mg/kg IgG1-637 (cumulative dose 5 mg/kg) induced impairment of neuromuscular transmission, as demonstrated by significantly increased jitter, synaptic transmission failures (blockings) and a decrease in the amplitude of the compound muscle action potentials during repeated stimulations (decrement), without showing overt symptoms of muscle weakness. Treatment with three daily doses of 10 mg/kg IgG4Δhinge-637 significantly reduced the IgG1-637-induced increase in jitter, blockings and decrement. Together, these results represent proof-of principle data for therapy of acetylcholine receptor-myasthenia gravis with a monovalent antibody format that blocks binding of pathogenic autoantibodies.


Assuntos
Autoanticorpos/metabolismo , Imunoglobulina G/administração & dosagem , Miastenia Gravis/tratamento farmacológico , Receptores Colinérgicos/metabolismo , Animais , Células CHO , Antagonistas Colinérgicos , Cricetulus , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Éxons Codificadores da Região de Dobradiça , Humanos , Imunoglobulina G/farmacologia , Macaca mulatta , Miastenia Gravis/imunologia , Miastenia Gravis/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Resultado do Tratamento
8.
Front Immunol ; 7: 462, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27872622

RESUMO

Aging western societies are facing an increasing prevalence of chronic inflammatory and degenerative diseases for which often no effective treatments exist, resulting in increasing health-care expenditure. Despite high investments in drug development, the number of promising new drug candidates decreases. We propose that preclinical research in non-human primates can help to bridge the gap between drug discovery and drug prescription. Translational research covers various stages of drug development of which preclinical efficacy tests in valid animal models is usually the last stage. Preclinical research in non-human primates may be essential in the evaluation of new drugs or therapies when a relevant rodent model is not available. Non-human primate models for life-threatening or severely debilitating diseases in humans are available at the Biomedical Primate Research Centre (BPRC). These have been instrumental in translational research for several decades. In order to stimulate European health research and innovation from bench to bedside, the European Commission has invested heavily in access to non-human primate research for more than 20 years. BPRC has hosted European users in a series of transnational access programs covering a wide range of research areas with the common theme being immune-mediated inflammatory disorders. We present an overview of the results and give an account of the studies performed as part of European Union Framework Programme (EU FP)-funded translational non-human primate research performed at the BPRC. These data illustrate the value of translational non-human primate research for the development of new therapies and emphasize the importance of EU FP funding in drug development.

10.
Inflammation ; 38(6): 2191-202, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26140903

RESUMO

Kidney allograft biopsies were analyzed for the presence of B cell clusters/aggregates using CD20 staining. Few B cells were found in the diffuse interstitial infiltrates, but clusters of B cells were found in nodular infiltrates. These nodular infiltrates were smaller shortly after transplantation, and their size increased over time. At the time of clinical rejection, the nodules often presented as tertiary lymphoid structures (TLS) with lymphoid-like follicles. The presence of small B cell clusters during the first 2 months after transplantation was not associated with early rejection. Even in animals that did not reject their allograft, TLS-like structures were present and could disappear over time. Although TLS were more often found in samples with interstitial fibrosis and tubular atrophy (IFTA), TLS were also present in samples without IFTA. The presence and density of clusters resembling tertiary lymphoid structures most likely reflect an ongoing immune response inside the graft and do not necessarily signify a poor graft outcome or IFTA.


Assuntos
Linfócitos B/imunologia , Rejeição de Enxerto/imunologia , Transplante de Rim/efeitos adversos , Rim/imunologia , Rim/cirurgia , Tecido Linfoide/imunologia , Aloenxertos , Animais , Antígenos CD20/metabolismo , Atrofia , Linfócitos B/metabolismo , Linfócitos B/patologia , Biomarcadores/metabolismo , Biópsia , Fibrose , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Macaca mulatta , Modelos Animais , Fatores de Tempo
11.
Eur J Pharmacol ; 759: 69-83, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25814254

RESUMO

The immune system plays a central role in the defense against environmental threats - such as infection with viruses, parasites or bacteria - but can also be a cause of disease, such as in the case of allergic or autoimmune disorders. In the past decades the impressive development of biotechnology has provided scientists with biological tools for the development of highly selective treatments for the different types of disorders. However, despite some clear successes the translation of scientific discoveries into effective treatments has remained challenging. The often-disappointing predictive validity of the preclinical animal models that are used in the selection of the most promising vaccine or drug candidates is the Achilles heel in the therapy development process. This publication summarizes the relevance and usage of non-human primates as pre-clinical model in infectious and autoimmune diseases, in particular for biologicals, which due to their high species-specificity are inactive in lower species.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Doenças Transmissíveis/tratamento farmacológico , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Doenças Autoimunes/imunologia , Doenças Transmissíveis/imunologia , Humanos , Primatas , Especificidade da Espécie
12.
J Immunol ; 194(4): 1454-66, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589073

RESUMO

Costimulatory and coinhibitory receptor-ligand pairs on T cells and APC control the immune response. We have investigated whether selective blockade of CD28-CD80/86 costimulatory interactions, which preserves the coinhibitory CTLA4-CD80/86 interactions and the function of regulatory T (Treg) cells, abrogates the induction of experimental autoimmune encephalomyelitis (EAE) in rhesus monkeys. EAE was induced by intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein (rhMOG) in CFA on day 0. FR104 is a monovalent, PEGylated-humanized Fab' Ab fragment against human CD28, cross-reactive with rhesus monkey CD28. FR104 or placebo was administered on days 0, 7, 14, and 21. FR104 levels remained high until the end of the study (day 42). Placebo-treated animals all developed clinical EAE between days 12 and 27. FR104-treated animals did not develop clinical EAE and were sacrificed at the end of the study resulting in a significantly prolonged survival. FR104 treatment diminished T and B cell responses against rhMOG, significantly reduced CNS inflammation and prevented demyelination. The inflammatory profile in the cerebrospinal fluid and brain material was also strongly reduced. Recrudescence of latent virus was investigated in blood, spleen, and brain. No differences between groups were observed for the ß-herpesvirus CMV and the polyomaviruses SV40 and SA12. Cross-sectional measurement of lymphocryptovirus, the rhesus monkey EBV, demonstrated elevated levels in the blood of FR104-treated animals. Blocking rhesus monkey CD28 with FR104 mitigated autoreactive T and B cell activation and prevented CNS pathology in the rhMOG/CFA EAE model in rhesus monkeys.


Assuntos
Antígenos CD28/antagonistas & inibidores , Encefalomielite Autoimune Experimental/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Linfócitos B , Encefalomielite Autoimune Experimental/virologia , Humanos , Macaca mulatta , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/imunologia , Viroses/complicações , Latência Viral
13.
Drug Discov Today ; 19(9): 1394-401, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24704460

RESUMO

The poor translational validity of autoimmune-mediated inflammatory disease (AIMID) models in inbred and specific pathogen-free (SPF) rodents underlies the high attrition of new treatments for the corresponding human disease. Experimental autoimmune encephalomyelitis (EAE) is a frequently used preclinical AIMID model. We discuss here how crucial information needed for the innovation of current preclinical models can be obtained from postclinical analysis of the nonhuman primate EAE model, highlighting the mechanistic reasons why some therapies fail and others succeed. These new insights can also help identify new targets for treatment.


Assuntos
Doenças Autoimunes/fisiopatologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Animais , Desenho de Fármacos , Humanos , Primatas , Roedores , Especificidade da Espécie , Pesquisa Translacional Biomédica/métodos
14.
PLoS One ; 8(8): e71549, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977076

RESUMO

The overlapping epidemiology of multiple sclerosis (MS) and Epstein-Barr virus (EBV), the increased risk to develop MS after infectious mononucleosis (IM) and the localization of EBV-infected B-cells within the MS brain suggest a causal link between EBV and MS. However, the underlying mechanism is unknown. We hypothesize that EBV-infected B-cells are capable of eliciting a central nervous system (CNS) targeting autoimmune reaction. To test this hypothesis we have developed a novel experimental model in rhesus monkeys of IM-like disease induced by infusing autologous B-lymphoblastoid cells (B-LCL). Herpesvirus papio (HVP) is a lymphocryptovirus related to EBV and was used to generate rhesus monkey B-LCL. Three groups of five animals were included; each group received three intravenous infusions of B-LCL that were either pulsed with the encephalitogenic self peptide MOG(34-56) (group A), a mimicry peptide (981-1003) of the major capsid protein of cytomegalovirus (CMVmcp(981-1003); group B) or the citrullinated MOG(34-56) (cMOG(34-56); group C). Groups A and B received on day 98 a single immunization with MOG(34-56) in incomplete Freund's adjuvant (IFA). Group C monkeys were euthanized just prior to day 98 without booster immunization. We observed self-peptide-specific proliferation of T-cells, superimposed on similar strong proliferation of CD3(+)CD8(+) T-cells against the B-LCL as observed in IM. The brains of several monkeys contained perivascular inflammatory lesions of variable size, comprising CD3(+) and CD68(+) cells. Moreover, clusters of CD3(+) and CD20(+) cells were detected in the meninges. The only evident clinical sign was substantial loss of bodyweight (>15%), a symptom observed both in early autoimmune encephalitis and IM. In conclusion, this model suggests that EBV-induced B-LCL can elicit a CNS targeting inflammatory (auto)immune reaction.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos B/virologia , Encefalite/imunologia , Lymphocryptovirus/imunologia , Macaca mulatta/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Peptídeos/imunologia , Animais , Autoanticorpos/imunologia , Linfócitos B/imunologia , Proliferação de Células , Sistema Nervoso Central/patologia , Citrulina/imunologia , Estudos de Coortes , Encefalite/virologia , Humanos , Imunidade Celular/imunologia , Imunidade Inata/imunologia , Imunoglobulina G/imunologia , Macaca mulatta/virologia , Linfócitos T/imunologia
15.
J Neuroimmune Pharmacol ; 8(5): 1251-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23821341

RESUMO

The experimental autoimmune encephalitis (EAE) model is used for preclinical research into the pathogenesis of multiple sclerosis (MS), mostly in inbred, specific pathogen free (SPF)-raised laboratory mice. However, the naive state of the laboratory mouse immune system is considered a major hurdle in the translation of principles from the EAE model to the MS patient. Non-human primates (NHP) have an immune system harboring T- and B-cell memory against environmental antigens, similar as in humans. We sought to further refine existing NHP EAE models, which may help to bridge the gab between mouse EAE models and MS. We report here on new EAE models in three NHP species: rhesus monkeys (Macaca mulatta), cynomolgus monkeys (Macaca fascicularis) and common marmosets (Callithrix jacchus). EAE was induced with recombinant human myelin oligodendrocyte glycoprotein extracellular domain (1-125) (rhMOG) formulated in incomplete Freund's adjuvant (IFA). IFA lacks the bacterial antigens that are present in complete Freund's adjuvant (CFA), which are notorious for the induction of discomforting side effects. Clinically evident EAE could be induced in two out of five rhesus monkeys, six out of six cynomolgus monkeys and six out of six common marmosets. In each of these species, the presence of an early, high anti-rhMOG IgM response is correlated with EAE with an earlier onset and more severe disease course. Animals without an early high IgM response either did not develop disease (rhesus monkeys) or developed only mild signs of neurological deficit (marmoset and cynomolgus monkeys).


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Adjuvante de Freund/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Encéfalo/patologia , Callithrix , Encefalomielite Autoimune Experimental/patologia , Adjuvante de Freund/farmacologia , Humanos , Imunoglobulina M/imunologia , Imuno-Histoquímica , Macaca fascicularis , Macaca mulatta , Proteínas Recombinantes/imunologia , Medula Espinal/patologia
16.
J Immunol ; 190(5): 1961-73, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23365083

RESUMO

The immune system is characterized by the preferential migration of lymphocytes through specific tissues (i.e., tissue tropism). Tissue tropism is mediated, in part, by the α(4) integrins expressed by T lymphocytes. The α(4)ß(1) integrin mediates migration of memory T lymphocytes into the CNS, whereas the α(4)ß(7) integrin mediates migration preferentially into gastrointestinal tissue. This paradigm was established primarily from investigations in rodents; thus, the objective of this investigation was to determine if blocking the α(4)ß(7) integrin exclusively would affect migration of T lymphocytes into the CNS of primates. The effects of the dual α(4)ß(1) and α(4)ß(7) antagonist natalizumab were compared with those of the α(4)ß(7) antagonist vedolizumab on experimental autoimmune encephalomyelitis in the rhesus monkey. Animals received an initial i.v. bolus of placebo, natalizumab (30 mg/kg), or vedolizumab (30 mg/kg) before intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein and then Ab once weekly thereafter. Natalizumab prevented CNS inflammation and demyelination significantly (p < 0.05), compared with time-matched placebo control animals, whereas vedolizumab did not inhibit these effects, despite saturating the α(4)ß(7) integrin in each animal for the duration of the investigation. These results demonstrate that blocking α(4)ß(7) exclusively does not inhibit immune surveillance of the CNS in primates.


Assuntos
Autoimunidade/efeitos dos fármacos , Inibição de Migração Celular/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Integrina alfa4beta1/antagonistas & inibidores , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Humanos , Vigilância Imunológica/efeitos dos fármacos , Injeções Intravenosas , Integrina alfa4beta1/imunologia , Macaca mulatta , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , Glicoproteína Mielina-Oligodendrócito/imunologia , Natalizumab , Especificidade de Órgãos , Placebos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
17.
Transpl Immunol ; 25(2-3): 133-40, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21708251

RESUMO

MT204 is a humanized IgG1 antibody specific for interleukin-2 (IL-2) of human and rhesus monkey origin. It potently antagonizes IL-2 signaling in both CD25(+) and CD25(-) cells by a unique mode of action. MT204 can not only prevent soluble IL-2 from binding to the intermediate affinity IL-2 receptors but can also antagonize IL-2 that is already bound to the CD25 subunit of high affinity IL-2 receptors on the cell surface. As initial steps toward development of a therapeutic antibody, pharmacokinetics (PK) and tolerability of MT204, as well as efficacy were investigated in rhesus monkeys. MT204 was infused by single escalating (2, 10 and 50mg/kg) or repeat dose administrations (50mg/kg, 1 ×/week for 3 weeks). Over the course of the experiment, the animals were regularly observed for clinical adverse reaction and bled for laboratory investigations (PK, hematology, chemical chemistry and leukocyte subset analysis). For the efficacy study, six rhesus monkeys were grafted with MHC-mismatched rhesus skin and infused with MT204 (50mg/kg), on the day of transplantation and again on days 5 and 12 post grafting. Efficacy was determined by assessment of graft necrosis at different time-points. No obvious adverse effects were observed clinically after single infusion, or after three repeated infusions of 50mg/kg and no MT204-related toxic effects were revealed by hematology or clinical chemistry. In the efficacy study, MT204-treated animals showed a significant delay in graft rejection versus control animals (p=0.025 by Log-rank test). The characteristics of MT204, displaying linear pharmacokinetics, half-life in the range expected for a human IgG, benign safety profile and signs of efficacy, warrant further development of this antibody for therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica , Interleucina-2/antagonistas & inibidores , Transplante de Pele , Animais , Interleucina-2/imunologia , Macaca mulatta , Transplante Homólogo
18.
Transplantation ; 85(8): 1185-92, 2008 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-18431240

RESUMO

BACKGROUND: Translational research in a relevant preclinical model is recommended before Treg-inducing protocols can be implemented in humans. We have characterized rhesus monkey CD25 cells phenotypically and functionally. METHODS: The phenotype of CD4(+)CD25(high) cells was determined by FACS, focusing on established markers of mouse and human Treg cells. Percentages of cells positive for CD45RA, CD62L, and intracellular CTLA-4 and FOXP3 were compared between CD4(+)CD25(high) and CD4(+)CD25(-) cells. CD25 cells stimulated with anti-CD3, ConA, and/or allogeneic peripheral blood mononuclear cells were mixed with freshly isolated CD25 cells. The suppressive activity of the CD25 cells in vitro was assessed using several experimental conditions. RESULTS: Rhesus monkey CD4(+)CD25(high) cells expressed high intracellular levels of CTLA-4 and FOXP3, whereas expression was negligible in CD4(+)CD25(-) cells. The CD25(high) population was mostly CD45RA(-), indicative of a memory phenotype. The CD25(+) cells were anergic, because they showed low proliferative responses, no interleukin-2 production, and some interferon-gamma and interleukin-10 production. Proliferation of CD4(+)CD25(-) cells stimulated by anti-CD3 or allogeneic cells was decreased when CD4(+)CD25(-) cells were added at a 1:1 ratio. In addition, we found that CD25 cells inhibited the interleukin-2 and interferon-gamma production by anti-CD3-stimulated CD25 cells in a dose-dependent fashion, through a cell-cell contact-dependent mechanism. CONCLUSIONS: Rhesus monkey CD4(+)CD25(+) cells have similar phenotypic and functional characteristics as natural Tregs in humans. These findings allow testing of Treg expansion and induction protocols in a relevant preclinical model, the rhesus monkey.


Assuntos
Macaca mulatta/imunologia , Linfócitos T Reguladores/fisiologia , Animais , Antígenos CD/análise , Antígenos de Diferenciação/análise , Antígeno CTLA-4 , Comunicação Celular , Citocinas/biossíntese , Memória Imunológica , Antígenos Comuns de Leucócito/análise , Linfócitos T Reguladores/imunologia
19.
Transplantation ; 82(9): 1194-201, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17102771

RESUMO

BACKGROUND: Costimulation blockade with antibodies directed against human CD40 and CD86 leads to prolonged kidney allograft survival in rhesus monkeys, but fails to induce permanent graft acceptance. We have tested whether costimulation blockade is more effective after peripheral T-cell ablation with antithymocyte globulin (ATG), with the aim to remove already primed autoreactive cells present in the normal repertoire. METHODS: Rhesus monkeys were transplanted with a mismatched kidney allograft. ATG was given around the time of transplantation (day -1 and 0). Costimulation blockade with anti-CD40+anti-CD86 was given at tapering dosages from day -1 to 56. Cyclosporin A (CsA) was given from day 42 onwards and first rejections occurring after day 42 were treated with prednisone. RESULTS: We observed accelerated rejection in ATG-treated monkeys, compared to animals receiving only costimulation blockade. The accelerated rejection of the kidney allograft occurred despite the application of rejection therapy with steroids and CsA. Three of the five ATG-treated animals were found seropositive for donor-specific alloantibodies. Early biopsies (day 21) from animals treated with ATG and anti-CD40+anti-CD86 show substantially reduced expression of cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and forkhead box P3 (FOXP3) in focal infiltrates as compared to animals treated with only costimulation blockade. Furthermore, we observed the rapid reappearance of CD8 T-cells with a memory phenotype (disappearance of naive CD95/CD11a T-cells) in peripheral blood. CONCLUSION: We conclude that (subtotal) T-cell depletion using ATG does not add to costimulation blockade induced kidney allograft survival.


Assuntos
Anticorpos Bloqueadores/administração & dosagem , Soro Antilinfocitário/administração & dosagem , Sobrevivência de Enxerto/efeitos dos fármacos , Terapia de Imunossupressão , Transplante de Rim , Animais , Antígenos CD/análise , Antígenos de Diferenciação/análise , Antígeno B7-2/efeitos dos fármacos , Antígenos CD40/antagonistas & inibidores , Antígeno CTLA-4 , Sinergismo Farmacológico , Fatores de Transcrição Forkhead/análise , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/prevenção & controle , Memória Imunológica , Rim/química , Rim/patologia , Depleção Linfocítica , Macaca mulatta , Linfócitos T/química , Linfócitos T/efeitos dos fármacos
20.
Transplantation ; 79(11): 1623-6, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15940054

RESUMO

Costimulation blockade as a single immunosuppressive treatment modality is not sufficient to prevent graft rejection. Here, we report an induction therapy using antagonistic antibodies against CD40 and CD86, given twice weekly from day -1 until day 56, followed by a delayed 12-week course of low-dose cyclosporine A (CsA) treatment in the rhesus monkey kidney-allograft model. Low-dose CsA treatment was initiated on day 42 and tapered until total cessation of all treatment on day 126. Treatment with anti-CD40/86 alone resulted in graft survival of 61, 71, 75, 78, and 116 days. Costimulation blockade followed by CsA resulted in more than 3-year drug-free survival in two of four animals. None of the animals developed donor-specific alloantibodies. Transforming growth factor-beta producing cells are present in early as well as in late kidney-graft biopsies and could play a role in the observed long-term drug-free graft survival.


Assuntos
Sobrevivência de Enxerto/imunologia , Transplante de Rim/imunologia , Animais , Anticorpos/uso terapêutico , Antígenos CD/imunologia , Antígeno B7-2 , Antígenos CD40/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Rim/patologia , Macaca mulatta , Glicoproteínas de Membrana/imunologia , Fatores de Tempo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...