Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Immunol ; 25(2): 256-267, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38172258

RESUMO

The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Humanos , Animais , Camundongos , Interleucina-33/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Alarminas , Subpopulações de Linfócitos T/metabolismo , Antivirais
2.
EMBO J ; 40(13): e106272, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33942347

RESUMO

Cellular stress has been associated with inflammation, yet precise underlying mechanisms remain elusive. In this study, various unrelated stress inducers were employed to screen for sensors linking altered cellular homeostasis and inflammation. We identified the intracellular pattern recognition receptors NOD1/2, which sense bacterial peptidoglycans, as general stress sensors detecting perturbations of cellular homeostasis. NOD1/2 activation upon such perturbations required generation of the endogenous metabolite sphingosine-1-phosphate (S1P). Unlike peptidoglycan sensing via the leucine-rich repeats domain, cytosolic S1P directly bound to the nucleotide binding domains of NOD1/2, triggering NF-κB activation and inflammatory responses. In sum, we unveiled a hitherto unknown role of NOD1/2 in surveillance of cellular homeostasis through sensing of the cytosolic metabolite S1P. We propose S1P, an endogenous metabolite, as a novel NOD1/2 activator and NOD1/2 as molecular hubs integrating bacterial and metabolic cues.


Assuntos
Inflamação/metabolismo , Lisofosfolipídeos/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Esfingosina/análogos & derivados , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Células HEK293 , Células HeLa , Humanos , Camundongos , NF-kappa B/metabolismo , Peptidoglicano/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/metabolismo , Células THP-1
3.
Science ; 366(6472)2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31857448

RESUMO

Pseudomonas aeruginosa rapidly adapts to altered conditions by quorum sensing (QS), a communication system that it uses to collectively modify its behavior through the production, release, and detection of signaling molecules. QS molecules can also be sensed by hosts, although the respective receptors and signaling pathways are poorly understood. We describe a pattern of regulation in the host by the aryl hydrocarbon receptor (AhR) that is critically dependent on qualitative and quantitative sensing of P. aeruginosa quorum. QS molecules bind to AhR and distinctly modulate its activity. This is mirrored upon infection with P. aeruginosa collected from diverse growth stages and with QS mutants. We propose that by spying on bacterial quorum, AhR acts as a major sensor of infection dynamics, capable of orchestrating host defense according to the status quo of infection.


Assuntos
Interações Hospedeiro-Patógeno , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Percepção de Quorum/fisiologia , Receptores de Hidrocarboneto Arílico/fisiologia , Células A549 , Animais , Humanos , Larva , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Pseudomonas aeruginosa/genética , Percepção de Quorum/genética , Receptores de Hidrocarboneto Arílico/genética , Peixe-Zebra
4.
J Neuroinflammation ; 15(1): 114, 2018 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-29673365

RESUMO

BACKGROUND: The success of clinical trials of selective B cell depletion in patients with relapsing multiple sclerosis (MS) indicates B cells are important contributors to peripheral immune responses involved in the development of new relapses. Such B cell contribution to peripheral inflammation likely involves antibody-independent mechanisms. Of growing interest is the potential that B cells, within the MS central nervous system (CNS), may also contribute to the propagation of CNS-compartmentalized inflammation in progressive (non-relapsing) disease. B cells are known to persist in the inflamed MS CNS and are more recently described as concentrated in meningeal immune-cell aggregates, adjacent to the subpial cortical injury which has been associated with progressive disease. How B cells are fostered within the MS CNS and how they may contribute locally to the propagation of CNS-compartmentalized inflammation remain to be elucidated. METHODS: We considered whether activated human astrocytes might contribute to B cell survival and function through soluble factors. B cells from healthy controls (HC) and untreated MS patients were exposed to primary human astrocytes that were either maintained under basal culture conditions (non-activated) or pre-activated with standard inflammatory signals. B cell exposure to astrocytes included direct co-culture, co-culture in transwells, or exposure to astrocyte-conditioned medium. Following the different exposures, B cell survival and expression of T cell co-stimulatory molecules were assessed by flow cytometry, as was the ability of differentially exposed B cells to induce activation of allogeneic T cells. RESULTS: Secreted factors from both non-activated and activated human astrocytes robustly supported human B cell survival. Soluble products of pre-activated astrocytes also induced B cell upregulation of antigen-presenting cell machinery, and these B cells, in turn, were more efficient activators of T cells. Astrocyte-soluble factors could support survival and activation of B cell subsets implicated in MS, including memory B cells from patients with both relapsing and progressive forms of disease. CONCLUSIONS: Our findings point to a potential mechanism whereby activated astrocytes in the inflamed MS CNS not only promote a B cell fostering environment, but also actively support the ability of B cells to contribute to the propagation of CNS-compartmentalized inflammation, now thought to play key roles in progressive disease.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/fisiologia , Sistema Nervoso Central/citologia , Citocinas/farmacologia , Esclerose Múltipla/patologia , Linfócitos B/classificação , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Feminino , Feto/citologia , Citometria de Fluxo , Humanos , Ativação Linfocitária/efeitos dos fármacos , Masculino , Esclerose Múltipla/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/fisiologia
5.
J Immunol ; 198(8): 3109-3117, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28264972

RESUMO

The development of rheumatoid arthritis (RA) is linked to functional changes in synovial fibroblasts (SF) and local infiltration of T lymphocytes. Fibroblasts possess the capacity to suppress T cell responses, although the molecular mechanisms of this suppression remain incompletely understood. In this study, we aimed to define the mechanisms by which noninflammatory SF modulate Th cell responses and to determine the immunosuppressive efficacy of RASF. Hence, the influence of SF from osteoarthritis or RA patients on total Th cells or different Th cell subsets of healthy donors was analyzed in vitro. We show that SF strongly suppressed the proliferation of Th cells and the secretion of IFN-γ in a cell contact-independent manner. In cocultures of SF and Th cells, tryptophan was completely depleted within a few days, resulting in eukaryotic initiation factor 2α phosphorylation, TCRζ-chain downregulation, and proliferation arrest. Blocking IDO1 activity completely restored Th cell proliferation, but not IFN-γ production. Interestingly, only the proliferation of Th1 cells, but not of Th2 or Th17 cells, was affected. Finally, RASF had a significantly lower IDO1 expression and a weaker Th cell suppressive capacity compared with osteoarthritis SF. We postulate that the suppression of Th cell growth by SF through tryptophan catabolism may play an important role in preventing inappropriate Th cell responses under normal conditions. However, expansion of Th17 cells that do not induce IDO1-mediated suppression and the reduced capacity of RASF to restrict Th cell proliferation through tryptophan metabolism may support the initiation and propagation of synovitis in RA patients.


Assuntos
Artrite Reumatoide/imunologia , Fibroblastos/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células Th1/imunologia , Triptofano/metabolismo , Diferenciação Celular/imunologia , Cromatografia Líquida de Alta Pressão , Técnicas de Cocultura , Fibroblastos/metabolismo , Humanos , Immunoblotting , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Ativação Linfocitária/imunologia , Osteoartrite/imunologia , Reação em Cadeia da Polimerase , Membrana Sinovial/imunologia , Células Th17/imunologia , Células Th2/imunologia , Triptofano/imunologia
6.
mBio ; 7(6)2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27879332

RESUMO

Mycobacterium bovis Bacille Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB), yet its moderate efficacy against pulmonary TB calls for improved vaccination strategies. Mucosal BCG vaccination generates superior protection against TB in animal models; however, the mechanisms of protection remain elusive. Tissue-resident memory T (TRM) cells have been implicated in protective immune responses against viral infections, but the role of TRM cells following mycobacterial infection is unknown. Using a mouse model of TB, we compared protection and lung cellular infiltrates of parenteral and mucosal BCG vaccination. Adoptive transfer and gene expression analyses of lung airway cells were performed to determine the protective capacities and phenotypes of different memory T cell subsets. In comparison to subcutaneous vaccination, intratracheal and intranasal BCG vaccination generated T effector memory and TRM cells in the lung, as defined by surface marker phenotype. Adoptive mucosal transfer of these airway-resident memory T cells into naive mice mediated protection against TB. Whereas airway-resident memory CD4+ T cells displayed a mixture of effector and regulatory phenotype, airway-resident memory CD8+ T cells displayed prototypical TRM features. Our data demonstrate a key role for mucosal vaccination-induced airway-resident T cells in the host defense against pulmonary TB. These results have direct implications for the design of refined vaccination strategies. IMPORTANCE: BCG remains the only licensed vaccine against TB. Parenterally administered BCG has variable efficacy against pulmonary TB, and thus, improved prevention strategies and a more refined understanding of correlates of vaccine protection are required. Induction of memory T cells has been shown to be essential for protective TB vaccines. Mimicking the natural infection route by mucosal vaccination has been known to generate superior protection against TB in animal models; however, the mechanisms of protection have remained elusive. Here we performed an in-depth analysis to dissect the immunological mechanisms associated with superior mucosal protection in the mouse model of TB. We found that mucosal, and not subcutaneous, BCG vaccination generates lung-resident memory T cell populations that confer protection against pulmonary TB. We establish a comprehensive phenotypic characterization of these populations, providing a framework for future vaccine development.


Assuntos
Administração Intranasal , Vacina BCG/administração & dosagem , Memória Imunológica , Pulmão/imunologia , Linfócitos T/imunologia , Tuberculose/prevenção & controle , Adjuvantes Imunológicos , Transferência Adotiva , Animais , Modelos Animais de Doenças , Injeções Subcutâneas , Camundongos , Resultado do Tratamento , Tuberculose/imunologia
8.
mBio ; 6(5): e01187-15, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26374119

RESUMO

UNLABELLED: An estimated one-third of the world's population is currently latently infected with Mycobacterium tuberculosis. Latent M. tuberculosis infection (LTBI) progresses into active tuberculosis (TB) disease in ~5 to 10% of infected individuals. Diagnostic and prognostic biomarkers to monitor disease progression are urgently needed to ensure better care for TB patients and to decrease the spread of TB. Biomarker development is primarily based on transcriptomics. Our understanding of biology combined with evolving technical advances in high-throughput techniques led us to investigate the possibility of additional platforms (epigenetics and proteomics) in the quest to (i) understand the biology of the TB host response and (ii) search for multiplatform biosignatures in TB. We engaged in a pilot study to interrogate the DNA methylome, transcriptome, and proteome in selected monocytes and granulocytes from TB patients and healthy LTBI participants. Our study provides first insights into the levels and sources of diversity in the epigenome and proteome among TB patients and LTBI controls, despite limitations due to small sample size. Functionally the differences between the infection phenotypes (LTBI versus active TB) observed in the different platforms were congruent, thereby suggesting regulation of function not only at the transcriptional level but also by DNA methylation and microRNA. Thus, our data argue for the development of a large-scale study of the DNA methylome, with particular attention to study design in accounting for variation based on gender, age, and cell type. IMPORTANCE: DNA methylation modifies the transcriptional program of cells. We have focused on two major populations of leukocytes involved in immune response to infectious diseases, granulocytes and monocytes, both of which are professional phagocytes that engulf and kill bacteria. We have interrogated how DNA methylation, gene expression, and protein translation differ in these two cell populations between healthy individuals and patients suffering from TB. To better understand the underlying biologic mechanisms, we harnessed a statistical enrichment analysis, taking advantage of predefined and well-characterized gene sets. Not only were there clear differences on various levels between the two populations, but there were also differences between TB patients and healthy controls in the transcriptome, proteome, and, for the first time, DNA methylome in these cells. Our pilot study emphasizes the value of a large-scale study of the DNA methylome taking into account our findings.


Assuntos
Biomarcadores/análise , Epigenômica/métodos , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno , Mycobacterium tuberculosis/crescimento & desenvolvimento , Proteômica/métodos , Tuberculose/patologia , Humanos , Dados de Sequência Molecular , Projetos Piloto , Análise de Sequência de DNA
9.
J Immunol ; 190(6): 2510-8, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23396942

RESUMO

The mechanisms whereby immune cells infiltrating the CNS in multiple sclerosis patients contribute to tissue injury remain to be defined. CD4 T cells are key players of this inflammatory response. Myelin-specific CD4 T cells expressing CD56, a surrogate marker of NK cells, were shown to be cytotoxic to human oligodendrocytes. Our aim was to identify NK-associated molecules expressed by human CD4 T cells that confer this oligodendrocyte-directed cytotoxicity. We observed that myelin-reactive CD4 T cell lines, as well as short-term PHA-activated CD4 T cells, can express NKG2C, the activating receptor interacting with HLA-E, a nonclassical MHC class I molecule. These cells coexpress CD56 and NKG2D, have elevated levels of cytotoxic molecules FasL, granzyme B, and perforin compared with their NKG2C-negative counterparts, and mediate significant in vitro cytotoxicity toward human oligodendrocytes, which upregulated HLA-E upon inflammatory cytokine treatment. A significantly elevated proportion of ex vivo peripheral blood CD4 T cells, but not CD8 T cells or NK cells, from multiple sclerosis patients express NKG2C compared with controls. In addition, immunohistochemical analyses showed that multiple sclerosis brain tissues display HLA-E(+) oligodendrocytes and NKG2C(+) CD4 T cells. Our results implicate a novel mechanism through which infiltrating CD4 T cells contribute to tissue injury in multiple sclerosis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Esclerose Múltipla/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/fisiologia , Oligodendroglia/imunologia , Regulação para Cima/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Antígeno CD56/metabolismo , Antígeno CD56/fisiologia , Linhagem Celular , Movimento Celular/genética , Citotoxicidade Imunológica/genética , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/biossíntese , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Regulação para Cima/genética , Antígenos HLA-E
10.
Biochim Biophys Acta ; 1812(2): 151-61, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20637863

RESUMO

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) characterized by multi-focal demyelination, axonal loss, and immune cell infiltration. Numerous immune mediators are detected within MS lesions, including CD4(+) and CD8(+) T lymphocytes suggesting that they participate in the related pathogenesis. Although CD4(+) T lymphocytes are traditionally considered the main actors in MS immunopathology, multiple lines of evidence suggest that CD8(+) T lymphocytes are also implicated in the pathogenesis. In this review, we outline the recent literature pertaining to the potential roles of CD8(+) T lymphocytes both in MS and its animal models. The CD8(+) T lymphocytes detected in MS lesions demonstrate characteristics of activated and clonally expanded cells supporting the notion that these cells actively contribute to the observed injury. Moreover, several experimental in vivo models mediated by CD8(+) T lymphocytes recapitulate important features of the human disease. Whether the CD8(+) T cells can induce or aggravate tissue destruction in the CNS needs to be fully explored. Strengthening our understanding of the pathogenic potential of CD8(+) T cells in MS should provide promising new avenues for the treatment of this disabling inflammatory disease.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Animais , Apresentação de Antígeno , Autoantígenos/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Genes MHC Classe I , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/etiologia , Esclerose Múltipla/patologia , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Oligodendroglia/imunologia , Oligodendroglia/patologia
11.
J Immunol ; 185(10): 5693-703, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20926794

RESUMO

The contribution of local factors to the activation of immune cells infiltrating the CNS of patients with multiple sclerosis (MS) remains to be defined. The cytokine IL-15 is pivotal in the maintenance and activation of CD8 T lymphocytes, a prominent lymphocyte population found in MS lesions. We investigated whether astrocytes are a functional source of IL-15 sufficient to enhance CD8 T lymphocyte responses and whether they provide IL-15 in the inflamed CNS of patients with MS. We observed that human astrocytes in primary cultures increased surface IL-15 levels upon activation with combinations of proinflammatory cytokines. Expanded human myelin autoreactive CD8 T lymphocytes cultured with such activated astrocytes displayed elevated lytic enzyme content, NKG2D expression, and Ag-specific cytotoxicity. These functional enhancements were abrogated by anti-IL-15-blocking Abs. Immunohistochemical analysis of brain tissue sections obtained from patients with MS demonstrated colocalization for IL-15 and the astrocyte marker glial fibrillary acidic protein within white matter lesions. The majority of astrocytes (80-90%) present in demyelinating MS lesions expressed IL-15, whereas few astrocytes in normal control brain sections had detectable IL-15. IL-15 could be detected in the majority of Iba-1-expressing microglia in the control sections, albeit in lower numbers when compared with microglia/macrophages in MS lesions. Furthermore, infiltrating CD8 T lymphocytes in MS lesions were in close proximity to IL-15-expressing cells. Astrocyte production of IL-15 resulting in the activation of CD8 T lymphocytes ascribes a role for these cells as contributors to the exacerbation of tissue damage during MS pathogenesis.


Assuntos
Astrócitos/imunologia , Linfócitos T CD8-Positivos/imunologia , Interleucina-15/imunologia , Ativação Linfocitária/imunologia , Esclerose Múltipla/imunologia , Astrócitos/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Separação Celular , Técnicas de Cocultura , Citocinas/imunologia , Citocinas/metabolismo , Citocinas/farmacologia , Citotoxicidade Imunológica/imunologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interleucina-15/biossíntese , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Ann Neurol ; 66(5): 617-29, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19938104

RESUMO

OBJECTIVE: The serum of most neuromyelitis optica (NMO) patients contains autoantibodies (NMO-IgGs) directed against the aquaporin-4 (AQP4) water channel located on astrocyte foot processes in the perivessel and subpial areas of the brain. Our objectives were to determine the source of central nervous system (CNS) NMO-IgGs and their role in disease pathogenesis. METHODS: Fluorescence-activated cell sorting and single-cell reverse transcriptase polymerase chain reaction were used to identify overrepresented plasma cell immunoglobulin (Ig) sequences in the cerebrospinal fluid (CSF) of an NMO patient after a first clinical attack. Monoclonal recombinant antibodies (rAbs) were generated from the paired heavy and light chain sequences and tested for target specificity and Fc effector function. The effect of CSF rAbs on CNS immunopathology was investigated by delivering single rAbs to rats with experimental autoimmune encephalomyelitis (EAE). RESULTS: Repertoire analysis revealed a dynamic, clonally expanded plasma cell population with features of an antigen-targeted response. Using multiple independent assays, 6 of 11 rAbs generated from CSF plasma cell clones specifically bound to AQP4. AQP4-specific rAbs recognized conformational epitopes and mediated both AQP4-directed antibody-dependent cellular cytotoxicity and complement-mediated lysis. When administered to rats with EAE, an AQP4-specific NMO CSF rAb induced NMO immunopathology: perivascular astrocyte depletion, myelinolysis, and complement and Ig deposition. INTERPRETATION: Molecular characterization of the CSF plasma cell repertoire in an early NMO patient demonstrates that AQP4-specific Ig is synthesized intrathecally at disease onset and directly contributes to CNS pathology. AQP4 is now the first confirmed antigenic target in human demyelinating disease.


Assuntos
Aquaporina 4/líquido cefalorraquidiano , Aquaporina 4/imunologia , Autoanticorpos/líquido cefalorraquidiano , Neuromielite Óptica/líquido cefalorraquidiano , Neuromielite Óptica/diagnóstico , Sequência de Aminoácidos , Animais , Aquaporina 4/genética , Biomarcadores/líquido cefalorraquidiano , Linhagem Celular , Células Cultivadas , Feminino , Feto , Humanos , Imunoglobulina G/líquido cefalorraquidiano , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Ratos , Ratos Endogâmicos Lew , Punção Espinal , Fatores de Tempo
13.
Ann N Y Acad Sci ; 1173: 478-86, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19758189

RESUMO

NMO-IgG autoantibody is now considered a useful serum biomarker of neuromyelitis optica (NMO). A series of clinical and pathological observations suggest that NMO-IgG may play a central role in NMO physiopathology. Although the exact role and function of NMO-IgG in vivo remain to be determined, recent reports suggest that this antibody that targets the aquaporin-4 (AQP-4) water channel expressed by astrocytes is able to destabilize the blood-brain barrier and contribute to astrocyte and oligodendrocyte damage. This review summarizes the latest molecular effector functions attributed to the NMO-IgG/anti-AQP4 antibody and discusses their potential role in NMO pathology.


Assuntos
Aquaporina 4/imunologia , Autoanticorpos/imunologia , Imunoglobulina G/imunologia , Neuromielite Óptica/imunologia , Animais , Aquaporina 4/metabolismo , Astrócitos/imunologia , Astrócitos/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Endocitose/imunologia , Humanos , Modelos Imunológicos , Neuromielite Óptica/metabolismo
14.
Autoimmun Rev ; 9(2): 132-5, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19389490

RESUMO

NMO-IgG, the auto-antibody specific to the aquaporin-4 (AQP4) water channel associated with the autoimmune inflammatory disease neuromyelitis optica (NMO), is considered to be an accurate serum biomarker and is thought to be an important contributor to NMO pathology. In this review, we summarize recent evidences from our group and others indicating that NMO-IgG can be implicated at several levels in the immuno-pathology of NMO. NMO-IgG/anti-AQP4 antibodies may compromise the integrity of the blood-brain barrier and consequently facilitate and enhance the perivascular inflammation characteristic of NMO. Lastly, NMO-IgG can induce astrocyte injury which may lead to the accumulation of excitatory/toxic molecules and accordingly damage oligodendrocytes and compromise myelin integrity.


Assuntos
Aquaporina 4/imunologia , Astrócitos/metabolismo , Neuromielite Óptica/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Astrócitos/imunologia , Astrócitos/patologia , Autoanticorpos/sangue , Biomarcadores/sangue , Barreira Hematoencefálica , Doenças Desmielinizantes , Humanos , Modelos Imunológicos , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia , Neuromielite Óptica/fisiopatologia
15.
J Immunol ; 181(8): 5730-7, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832732

RESUMO

Autoantibody neuromyelitis optica-IgG (NMO-IgG) recognizing aquaporin-4 (AQP4) is implicated as playing a central role in the physiopathology of NMO. The aim of this in vitro-based study was to characterize functional consequences of interaction between NMO-IgG and cells of the neurovascular unit (astrocytes and brain endothelium) that would provide insight into recognized features of NMO, namely altered blood-brain barrier (BBB) permeability and granulocyte recruitment. We used sera from NMO and longitudinally extensive transverse myelitis cases shown to bind in a characteristic perivascular pattern to primate cerebellar slices. Using flow cytometry, we found that sera from NMO-IgG-positive patients reacted with CNS-derived human fetal astrocytes, whereas sera from multiple sclerosis patients did not. We demonstrated that NMO-IgG binding to astrocytes alters aquaporin-4 polarized expression and increases permeability of a human BBB endothelium/astrocyte barrier. We further demonstrated that NMO-IgG binding to human fetal astrocytes can result in NK cell degranulation, astrocyte killing by Ab-dependent cellular cytotoxicity and complement-dependent granulocyte attraction through the BBB model. Our study highlights important functional roles for NMO-IgG that could account for pathological lesions and BBB dysfunction observed in NMO.


Assuntos
Aquaporina 4/imunologia , Astrócitos/imunologia , Autoanticorpos/imunologia , Barreira Hematoencefálica/imunologia , Granulócitos/imunologia , Imunoglobulina G/imunologia , Neuromielite Óptica/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Astrócitos/patologia , Autoanticorpos/sangue , Barreira Hematoencefálica/patologia , Degranulação Celular/imunologia , Cerebelo/irrigação sanguínea , Cerebelo/imunologia , Cerebelo/patologia , Proteínas do Sistema Complemento/imunologia , Feminino , Feto/imunologia , Feto/patologia , Granulócitos/patologia , Humanos , Imunidade Celular/imunologia , Imunoglobulina G/sangue , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/sangue , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia , Permeabilidade , Primatas
16.
J Neuropathol Exp Neurol ; 67(6): 590-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18520777

RESUMO

Neuronal injury and loss are recognized features of neuroinflammatory disorders, including acute and chronic encephalitides and multiple sclerosis; destruction of astrocytes has been demonstrated in cases of Rasmussen encephalitis. Here, we show that innate immune cells (i.e. natural killer [NK] and gammadelta T cells) cause loss of neurons from primary human neuron-enriched cultures by destroying the supporting astrocytes. Interleukin 2-activated NK cells caused loss of astrocytes within 1 hour, whereas neurons were lost at 4 hours. Time-lapse imaging indicated that delayed neuron loss was due to early destruction of supporting astrocytes. Selective blocking of astrocyte death with anti-NKG2D antibodies reduced neuron loss, as did blocking of CD54 on astrocytes. gammadelta T cells also induced astrocyte cytotoxicity, leading to subsequent neuronal displacement. In astrocytes, NK cells caused caspase-dependent fragmentation of the intermediate filament proteins glial fibrillary acidic protein and vimentin, whereas anti-CD3-activated T cells produced fragmentation to a lesser extent and without measurable cytotoxicity. Glial fibrillary acidic protein fragmentation was also demonstrated in lysates from chronic multiple sclerosis plaques but not from normal control white matter. These data suggest that non-major histocompatibility complex-restricted immune effector cells may contribute to neuron loss in neuroinflammatory disorders indirectly through injury of glia.


Assuntos
Astrócitos/patologia , Citotoxicidade Imunológica , Imunidade Inata , Células Matadoras Naturais/imunologia , Neurônios/patologia , Linfócitos T/imunologia , Western Blotting , Citometria de Fluxo , Proteína Glial Fibrilar Ácida/metabolismo , Antígenos de Histocompatibilidade Classe I , Humanos , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Vimentina
17.
J Neurosci ; 27(5): 1220-8, 2007 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-17267578

RESUMO

NKG2D is an activating or coactivating receptor expressed on human natural killer (NK) cells, CD8+ T cells, and gamma/delta T cells. NKG2D ligands have been detected on many tumor cell types and can be induced on nontransformed cells by environmental signals including DNA damage and inflammation. We investigated the contribution of NKG2D-NKG2D ligand interaction on CNS-directed immune responses. We observed that primary cultures of human adult oligodendrocytes and fetal astrocytes expressed ligands for NKG2D in vitro whereas neurons, microglia, and adult astrocytes did not. Disruption of the NKG2D-NKG2D ligand interaction using blocking antibodies significantly inhibited killing of primary human oligodendrocytes mediated by activated human NK cells, gamma/delta T cells, and allo-reactive CD8+ T cells. NKG2D ligands [major histocompatibility complex class I chain-related molecules A and B (MICA/B)] were detected in groups of cells and colocalized with an oligodendrocyte marker (adenomatous polyposis coli) in white matter sections obtained from multiple sclerosis lesions but not in normal control samples. CD8+ T cells could be detected in close proximity to MICA/B+ cells within multiple sclerosis lesions, supporting an in vivo interaction between these immune effectors and stressed MICA/B-expressing oligodendrocytes. These results imply that NKG2D-NKG2D ligand interaction can potentially contribute to cytotoxic responses mediated by activated immune effector cells in the inflamed CNS, as observed in multiple sclerosis.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/metabolismo , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Oligodendroglia/metabolismo , Receptores Imunológicos/fisiologia , Adulto , Idoso , Células Cultivadas , Citotoxicidade Imunológica/genética , Embrião de Mamíferos , Feminino , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Células Matadoras Naturais/imunologia , Ligantes , Ativação Linfocitária/genética , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Oligodendroglia/imunologia , Oligodendroglia/patologia , Ligação Proteica/genética , Receptores Imunológicos/genética , Receptores de Células Matadoras Naturais
18.
J Immunol Methods ; 310(1-2): 53-61, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16464465

RESUMO

Human self-reactive T cells are potentially involved in many autoimmune diseases. Although ex vivo detection of self-reactive T cells is possible, exhaustive functional characterization of these cells is impeded by their low frequency. In vitro expansion of antigen (Ag) specific T cells is typically achieved by using autologous (fresh or frozen) irradiated peripheral blood mononuclear cells (PBMCs), EBV-immortalized B cells or dendritic cells in the presence of Ag. These approaches require a large blood volume. We explored a method successfully applied for tumor specific T cells using in vitro expanded autologous B cells. PBMCs were stimulated with irradiated CD40L-expressing fibroblasts and IL-4, resulting in an enriched population of B cell that expressed high levels of MHC and co-stimulatory molecules, essential hallmarks of antigen presenting cells (APCs). Expanded B cells were loaded with Ag, irradiated and then used as APCs to stimulate T cells. The specificity of T cell lines was assessed by comparing their proliferation and IFN-gamma secretion when cultured with antigen-loaded B cells vs. unloaded B cells. T cell lines exhibiting antigen-specific proliferation and/or IFN-gamma secretion were expanded. Using this method, MBP and MOG specific CD4(+) and CD8(+) T cell lines were obtained from multiple donors in comparable numbers to those obtained using the traditional approach (i.e. fresh PBMCs as APCs) and were kept in culture for many weeks. We have shown that myelin specific CD4(+) and CD8(+) T cells can be expanded from a relatively small volume of blood (50-100 ml) from multiple donors using expanded B cells as APCs.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Esclerose Múltipla/imunologia , Bainha de Mielina/imunologia , Sequência de Aminoácidos , Animais , Linfócitos B/citologia , Linfócitos T CD4-Positivos/citologia , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/citologia , Técnicas de Cultura de Células/métodos , Proliferação de Células , Citometria de Fluxo , Humanos , Interferon gama/imunologia , Interleucina-4/imunologia , Camundongos , Dados de Sequência Molecular , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Células NIH 3T3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA