Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Immunol ; 41(5): 987-991, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33570715

RESUMO

Familial hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome resulting from defective cytotoxicity. A previously healthy 3-month-old female presented with fever, irritability, abdominal distention, and tachypnea. She ultimately met all eight HLH-2004 diagnostic criteria, accompanied by elevated CXCL9. Initial empiric anti-inflammatory treatment included anakinra and IVIg, which stabilized ferritin and cytopenias. She had molecular and genetic confirmation of perforin deficiency and was started on dexamethasone and etoposide per HLH-94. She clinically improved, though CXCL9 and sIL-2Ra remained elevated. She was readmitted at week 8 for relapsed HLH without clear trigger and HLH-94 induction therapy was reinitiated. Her systemic HLH symptoms failed to respond and she soon developed symptomatic CNS HLH. She was incidentally found to have multifocal lung and kidney nodules, which were sterile and consisted largely of histiocytes and activated, oligoclonal CD8 T cells. The patient had a laboratory response to salvage therapy with alemtuzumab and emapalumab, but progressive neurologic decline led to withdrawal of care. This report highlights HLH foci manifest as pulmonary/renal nodules, demonstrates the utility of monitoring an array of HLH biomarkers, and suggests possible benefit of earlier salvage therapy.


Assuntos
Linfo-Histiocitose Hemofagocítica/diagnóstico , Alemtuzumab/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Dexametasona/uso terapêutico , Etoposídeo/uso terapêutico , Evolução Fatal , Feminino , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Lactente , Inflamação/diagnóstico , Inflamação/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Perforina/deficiência , Terapia de Salvação
2.
J Clin Invest ; 130(10): 5425-5443, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32925169

RESUMO

Late-onset inflammatory toxicities resembling hemophagocytic lymphohistiocytosis (HLH) or macrophage activation syndrome (MAS) occur after chimeric antigen receptor T cell (CAR T cell) infusion and represent a therapeutic challenge. Given the established link between perforin deficiency and primary HLH, we investigated the role of perforin in anti-CD19 CAR T cell efficacy and HLH-like toxicities in a syngeneic murine model. Perforin contributed to both CD8+ and CD4+ CAR T cell cytotoxicity but was not required for in vitro or in vivo leukemia clearance. Upon CAR-mediated in vitro activation, perforin-deficient CAR T cells produced higher amounts of proinflammatory cytokines compared with WT CAR T cells. Following in vivo clearance of leukemia, perforin-deficient CAR T cells reexpanded, resulting in splenomegaly with disruption of normal splenic architecture and the presence of hemophagocytes, which are findings reminiscent of HLH. Notably, a substantial fraction of patients who received anti-CD22 CAR T cells also experienced biphasic inflammation, with the second phase occurring after the resolution of cytokine release syndrome, resembling clinical manifestations of HLH. Elevated inflammatory cytokines such as IL-1ß and IL-18 and concurrent late CAR T cell expansion characterized the HLH-like syndromes occurring in the murine model and in humans. Thus, a murine model of perforin-deficient CAR T cells recapitulated late-onset inflammatory toxicities occurring in human CAR T cell recipients, providing therapeutically relevant mechanistic insights.


Assuntos
Imunoterapia Adotiva/efeitos adversos , Perforina/deficiência , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Mediadores da Inflamação/metabolismo , Linfo-Histiocitose Hemofagocítica/etiologia , Linfo-Histiocitose Hemofagocítica/imunologia , Linfo-Histiocitose Hemofagocítica/patologia , Síndrome de Ativação Macrofágica/etiologia , Síndrome de Ativação Macrofágica/imunologia , Síndrome de Ativação Macrofágica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Perforina/genética , Linfócitos T/patologia
3.
Blood ; 136(6): 657-668, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32530039

RESUMO

Cytokine storm syndromes (CSS) are severe hyperinflammatory conditions characterized by excessive immune system activation leading to organ damage and death. Hemophagocytic lymphohistiocytosis (HLH), a disease often associated with inherited defects in cell-mediated cytotoxicity, serves as a prototypical CSS for which the 5-year survival is only 60%. Frontline therapy for HLH consists of the glucocorticoid dexamethasone (DEX) and the chemotherapeutic agent etoposide. Many patients, however, are refractory to this treatment or relapse after an initial response. Notably, many cytokines that are elevated in HLH activate the JAK/STAT pathway, and the JAK1/2 inhibitor ruxolitinib (RUX) has shown efficacy in murine HLH models and humans with refractory disease. We recently reported that cytokine-induced JAK/STAT signaling mediates DEX resistance in T cell acute lymphoblastic leukemia (T-ALL) cells, and that this could be effectively reversed by RUX. On the basis of these findings, we hypothesized that cytokine-mediated JAK/STAT signaling might similarly contribute to DEX resistance in HLH, and that RUX treatment would overcome this phenomenon. Using ex vivo assays, a murine model of HLH, and primary patient samples, we demonstrate that the hypercytokinemia of HLH reduces the apoptotic potential of CD8 T cells leading to relative DEX resistance. Upon exposure to RUX, this apoptotic potential is restored, thereby sensitizing CD8 T cells to DEX-induced apoptosis in vitro and significantly reducing tissue immunopathology and HLH disease manifestations in vivo. Our findings provide rationale for combining DEX and RUX to enhance the lymphotoxic effects of DEX and thus improve the outcomes for patients with HLH and related CSS.


Assuntos
Apoptose/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Síndrome da Liberação de Citocina/tratamento farmacológico , Dexametasona/uso terapêutico , Inibidores de Janus Quinases/uso terapêutico , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Pirazóis/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Animais , Linfócitos T CD8-Positivos/imunologia , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/fisiopatologia , Citocinas/fisiologia , Dexametasona/administração & dosagem , Dexametasona/farmacologia , Modelos Animais de Doenças , Resistência a Medicamentos/efeitos dos fármacos , Quimioterapia Combinada , Humanos , Interleucina-2/farmacologia , Inibidores de Janus Quinases/administração & dosagem , Inibidores de Janus Quinases/farmacologia , Janus Quinases , Coriomeningite Linfocítica/complicações , Coriomeningite Linfocítica/fisiopatologia , Linfo-Histiocitose Hemofagocítica/complicações , Linfo-Histiocitose Hemofagocítica/enzimologia , Linfo-Histiocitose Hemofagocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrilas , Perforina/deficiência , Pirazóis/administração & dosagem , Pirazóis/farmacologia , Pirimidinas , Fator de Transcrição STAT5/fisiologia , Organismos Livres de Patógenos Específicos
4.
Front Immunol ; 11: 846, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32528465

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is one of the main causes of cirrhosis and major risk factors for hepatocellular carcinoma and liver-related death. Despite substantial clinical and basic research, the pathogenesis of obesity-related NAFLD remains poorly understood. In this study, we show that perforin can act as an immune regulator to prevent the progression of NAFLD. Aged perforin-deficient (Prf-/-) mice have increased lipid accumulation in the liver compared to WT mice. With high-fat diet (HFD) challenge, Prf-/- mice have increased liver weight, more severe liver damage, and increased liver inflammation when compared with WT controls. Mechanistic studies revealed that perforin specifically regulates intrinsic IFN-γ production in CD4 T cells, not CD8 T cells. We found that CD4 T cell depletion reduces liver injury and ameliorates the inflammation and metabolic morbidities in Prf-/- mice. Furthermore, improved liver characteristics in HFD Prf-/- and IFN-γR-/- double knockout mice confirmed that IFN-γ is a key factor for mediating perforin regulation of NAFLD progression. Overall, our findings reveal the important regulatory role perforin plays in the progression of obesity-related NAFLD and highlight novel strategies for treating NAFLD.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Progressão da Doença , Interferon gama/metabolismo , Hepatopatia Gordurosa não Alcoólica/imunologia , Perforina/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Hepatite/etiologia , Interferon gama/genética , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/sangue , Obesidade/metabolismo , Perforina/deficiência , Perforina/genética
5.
J Pediatr Hematol Oncol ; 41(6): 482-488, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31219909

RESUMO

Primary, or familial, hemophagocytic lymphohistiocytosis (P-HLH) is a rare inherited autosomal-recessive immune deficiency which generally manifests during infancy or early childhood. Recent literature suggests an increased number of reports of late-onset P-HLH, especially in association with infection and underlying malignancy. The authors describe a case of subcutaneous T-cell lymphoma in a 8-year-old child that was complicated by primary, perforin-deficient HLH. In contrast, we examined retrospective data for 19 cases of late-onset P-HLH with available treatment data and compared the results of conservative medical therapy with hematopoietic stem cell transplant (HSCT) postremission therapy. Our patient displayed compound heterozygous mutations in PRF1 that have not been described in the literature previously: allele 1 [c.786_801del(p.Gln263fs)] and allele 2 [c.886T>C(p.Tyr296His)]. Of the 19 cases analyzed, 14 achieved remission. Postremission, 7 of 14 (50%) received HSCT and were reported alive at a median time of 24 months, 5 of 14 (36%) received medical therapy and were reported alive at a median time of 24 months, and 2 of 14 (14%) received medical therapy and died at a median of 73 months postremission. Our retrospective literature review suggests that some patients can survive late-onset, perforin-deficient, P-HLH without the potential lifelong risks of HSCT when in the first remission.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/estatística & dados numéricos , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Perforina/deficiência , Criança , Humanos , Linfo-Histiocitose Hemofagocítica/metabolismo , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Prognóstico , Indução de Remissão
6.
Biochem Biophys Res Commun ; 512(2): 367-372, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-30894275

RESUMO

Perforin plays an important role in autoimmune and infectious diseases, but its function in immune inflammatory responses after spinal cord injury (SCI) has received insufficient attention. The goal of this study is to determine the influence of perforin after spinal cord injury (SCI) on secondary inflammation. Compared recovery from SCI in perforin knockout (Prf1-/-) and wild-type(WT)mice, WT mice had significantly lower the Basso mouse score (BMS), CatWalk XT, and motor-evoked potentials (MEPs) than Prf1-/- mice. Spinal cord lesions were also more obvious through glial fibrillary acidic protein (GFAP), Nissl, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining. Furthermore, the blood-spinal cord barrier (BSCB) disruption was more severe and inflammatory cytokine levels were higher. Flow cytometry indicated that perforin mainly originated from CD8 T cells. With flow cytometry and enzyme-linked immunosorbent assay (ELISA), human cerebrospinal fluid (CSF) yielded similar results. Together, this study firstly demonstrated that CD8 T cell-derived perforin is detrimental to SCI recovery in the mouse model. Mechanistically, this effect occurs because perforin increases BSCB permeability, causing inflammatory cells and related cytokines to infiltrate and disrupt the nervous system.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Perforina/imunologia , Traumatismos da Medula Espinal/imunologia , Animais , Apoptose , Barreira Hematoneural/imunologia , Barreira Hematoneural/lesões , Barreira Hematoneural/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/líquido cefalorraquidiano , Perforina/deficiência , Perforina/genética , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia
7.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1182-1191, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30658162

RESUMO

Pyrimidine nucleotides are essential for a vast number of cellular processes and dysregulation of pyrimidine metabolism has been associated with a variety of clinical abnormalities. Inborn errors of pyrimidine metabolism affecting enzymes in the pyrimidine de novo and degradation pathway have been identified but no patients have been described with a deficiency in proteins affecting the cellular import of ribonucleosides. In this manuscript, we report the elucidation of the genetic basis of the observed uridine-cytidineuria in a patient presenting with fever, hepatosplenomegaly, persistent lactate acidosis, severely disturbed liver enzymes and ultimately multi-organ failure. Sequence analysis of genes encoding proteins directly involved in the metabolism of uridine and cytidine showed two variants c.1528C > T (p.R510C) and c.1682G > A (p.R561Q) in SLC28A1, encoding concentrative nucleotide transporter 1 (hCNT1). Functional analysis showed that these variants affected the three-dimensional structure of hCNT1, altered glycosylation and decreased the half-life of the mutant proteins which resulted in impaired transport activity. Co-transfection of both variants, mimicking the trans disposition of c.1528C > T (p.R510C) and c.1682G > A (p.R561Q) in the patient, significantly impaired hCNT1 biological function. Whole genome sequencing identified two pathogenic variants c.50delT; p.(Leu17Argfs*34) and c.853_855del; p.(Lys285del) in the PRF1 gene, indicating that our patient was also suffering from Familial Hemophagocytic Lymphohistiocytosis type 2. The identification of two co-existing monogenic defects might have resulted in a blended phenotype. Thus, the clinical presentation of isolated hCNT1 deficiency remains to be established.


Assuntos
Proteínas de Membrana Transportadoras/deficiência , Insuficiência de Múltiplos Órgãos/metabolismo , Perforina/deficiência , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Pirimidinas/metabolismo , Evolução Fatal , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas de Membrana Transportadoras/genética , Insuficiência de Múltiplos Órgãos/genética , Perforina/genética , Fenótipo , Erros Inatos do Metabolismo da Purina-Pirimidina/genética
8.
Cell Death Differ ; 25(9): 1536-1548, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743559

RESUMO

Cytotoxic CD8+ T (Tc) cells are the main executors of transformed and cancer cells during cancer immunotherapy. The latest clinical results evidence a high efficacy of novel immunotherapy agents that modulate Tc cell activity against bad prognosis cancers. However, it has not been determined yet whether the efficacy of these treatments can be affected by selection of tumoural cells with mutations in the cell death machinery, known to promote drug resistance and cancer recurrence. Here, using a model of prophylactic tumour vaccination based on the LCMV-gp33 antigen and the mouse EL4 T lymphoma, we analysed the molecular mechanism employed by Tc cells to eliminate cancer cells in vivo and the impact of mutations in the apoptotic machinery on tumour development. First of all, we found that Tc cells, and perf and gzmB are required to efficiently eliminate EL4.gp33 cells after LCMV immunisation during short-term assays (1-4 h), and to prevent tumour development in the long term. Furthermore, we show that antigen-pulsed chemoresistant EL4 cells overexpressing Bcl-XL or a dominant negative form of caspase-3 are specifically eliminated from the peritoneum of infected animals, as fast as parental EL4 cells. Notably, antigen-specific Tc cells control the tumour growth of the mutated cells, as efficiently as in the case of parental cells. Altogether, expression of the anti-apoptotic mutations does not confer any advantage for tumour cells neither in the short-term survival nor in long-term tumour formation. Although the mechanism involved in the elimination of the apoptosis-resistant tumour cells is not completely elucidated, neither necroptosis nor pyroptosis seem to be involved. Our results provide the first experimental proof that chemoresistant cancer cells with mutations in the main cell death pathways are efficiently eliminated by Ag-specific Tc cells in vivo during immunotherapy and, thus, provide the molecular basis to treat chemoresistant cancer cells with CD8 Tc-based immunotherapy.


Assuntos
Antígenos Virais/imunologia , Resistencia a Medicamentos Antineoplásicos/genética , Glicoproteínas/imunologia , Neoplasias/terapia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/transplante , Proteínas Virais/imunologia , Animais , Apoptose , Caspase 3/metabolismo , Linhagem Celular Tumoral , Granzimas/deficiência , Granzimas/genética , Granzimas/metabolismo , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/imunologia , Neoplasias/mortalidade , Perforina/deficiência , Perforina/genética , Perforina/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia , Transplante Heterólogo , Proteína bcl-X/metabolismo
9.
J Pediatr Hematol Oncol ; 39(2): 143-146, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27271812

RESUMO

Neurological manifestations in familial hemophagocytic lymphohistiocytosis (FHL) are common, seen in up to 73% of patients in their course of disease. However, in majority of the cases central nervous system manifestations are associated with other clinical and laboratory parameters of hemophagocytic lymphohistiocytosis. We report here a case with FHL2 in whom hemophagocytic lymphohistiocytosis was a presenting manifestation which responded to specific therapy, however, there was isolated central nervous system relapse while patient was in remission and off therapy. FHL2 was confirmed on the basis of reduced perforin expression and homozygous mutation in PRF1at codon 637 in exon 3 (c.673C>T p.Arg225Trp).


Assuntos
Doenças Desmielinizantes/etiologia , Linfo-Histiocitose Hemofagocítica/genética , Transtornos dos Movimentos/etiologia , Debilidade Muscular/etiologia , Mutação de Sentido Incorreto , Perforina/genética , Adulto , Tronco Encefálico/diagnóstico por imagem , Pré-Escolar , Códon/genética , Ciclosporina/uso terapêutico , Doenças Desmielinizantes/diagnóstico por imagem , Dexametasona/uso terapêutico , Éxons/genética , Feminino , Ferritinas/sangue , Lobo Frontal/diagnóstico por imagem , Humanos , Imunossupressores/uso terapêutico , Linfo-Histiocitose Hemofagocítica/sangue , Linfo-Histiocitose Hemofagocítica/diagnóstico por imagem , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Imageamento por Ressonância Magnética , Masculino , Metotrexato/uso terapêutico , Perforina/deficiência , Recidiva , Infecções Respiratórias/etiologia
10.
Proc Natl Acad Sci U S A ; 113(48): 13821-13826, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27856754

RESUMO

Epstein-Barr Virus (EBV) infects human B cells and drives them into continuous proliferation. Two key viral factors in this process are the latent membrane proteins LMP1 and LMP2A, which mimic constitutively activated CD40 receptor and B-cell receptor signaling, respectively. EBV-infected B cells elicit a powerful T-cell response that clears the infected B cells and leads to life-long immunity. Insufficient immune surveillance of EBV-infected B cells causes life-threatening lymphoproliferative disorders, including mostly germinal center (GC)-derived B-cell lymphomas. We have modeled acute EBV infection of naive and GC B cells in mice through timed expression of LMP1 and LMP2A. Although lethal when induced in all B cells, induction of LMP1 and LMP2A in just a small fraction of naive B cells initiated a phase of rapid B-cell expansion followed by a proliferative T-cell response, clearing the LMP-expressing B cells. Interfering with T-cell activity prevented clearance of LMP-expressing B cells. This was also true for perforin deficiency, which in the human causes a life-threatening EBV-related immunoproliferative syndrome. LMP expression in GC B cells impeded the GC reaction but, upon loss of T-cell surveillance, led to fatal B-cell expansion. Thus, timed expression of LMP1 together with LMP2A in subsets of mouse B cells allows one to study major clinically relevant features of human EBV infection in vivo, opening the way to new therapeutic approaches.


Assuntos
Linfócitos B/virologia , Infecções por Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Proteínas da Matriz Viral/genética , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Antígenos CD40/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Regulação Viral da Expressão Gênica , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Herpesvirus Humano 4/patogenicidade , Humanos , Camundongos , Perforina/deficiência , Perforina/genética , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/virologia , Proteínas da Matriz Viral/biossíntese
11.
Cell Death Dis ; 7: e2193, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27077809

RESUMO

CD4(+) T lymphocytes are key players in the adaptive immune system and can differentiate into a variety of effector and regulatory T cells. Here, we provide evidence that a novel differentiation pathway of CD4(+) T cells shifts the balance from a destructive T-cell response to one that favors regulation in an immune-mediated liver injury model. Peripheral CD4(-)CD8(-)NK1.1(-) double-negative T cells (DNT) was increased following Concanavalin A administration in mice. Adoptive transfer of DNT led to significant protection from hepatocyte necrosis by direct inhibition on the activation of lymphocytes, a process that occurred primarily through the perforin-granzyme B route. These DNT converted from CD4(+) rather than CD8(+) T cells, a process primarily regulated by OX40. DNT migrated to the liver through the CXCR3-CXCL9/CXCL10 interaction. In conclusion, we elucidated a novel differentiation pathway from activated CD4(+) T cells to regulatory DNT cells for maintaining homeostasis of the immune system in vivo, and provided key evidence that utilizing this novel differentiation pathway has potential application in the prevention and treatment of autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T/citologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Concanavalina A/farmacologia , Citocinas/sangue , Granzimas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligante OX40/deficiência , Ligante OX40/genética , Perforina/deficiência , Perforina/genética , Perforina/farmacologia , RNA Mensageiro/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
12.
J Exp Med ; 213(2): 225-33, 2016 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-26755706

RESUMO

Type I interferon (IFN) is crucial in host antiviral defense. Previous studies have described the pleiotropic role of type I IFNs on innate and adaptive immune cells during viral infection. Here, we demonstrate that natural killer (NK) cells from mice lacking the type I IFN-α receptor (Ifnar(-/-)) or STAT1 (which signals downstream of IFNAR) are defective in expansion and memory cell formation after mouse cytomegalovirus (MCMV) infection. Despite comparable proliferation, Ifnar(-/-) NK cells showed diminished protection against MCMV infection and exhibited more apoptosis compared with wild-type NK cells. Furthermore, we show that Ifnar(-/-) NK cells express increased levels of NK group 2 member D (NKG2D) ligands during viral infection and are susceptible to NK cell-mediated fratricide in a perforin- and NKG2D-dependent manner. Adoptive transfer of Ifnar(-/-) NK cells into NK cell-deficient mice reverses the defect in survival and expansion. Our study reveals a novel type I IFN-dependent mechanism by which NK cells evade mechanisms of cell death after viral infection.


Assuntos
Infecções por Herpesviridae/imunologia , Interferon Tipo I/metabolismo , Células Matadoras Naturais/imunologia , Muromegalovirus , Animais , Apoptose , Proliferação de Células , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Perforina/deficiência , Perforina/genética , Perforina/metabolismo , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
13.
Med Microbiol Immunol ; 204(3): 335-44, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25809566

RESUMO

Natural killer (NK) and CD8(+) T cells play a crucial role in the control of mouse cytomegalovirus (MCMV) infection. These effector cells exert their functions by releasing antiviral cytokines and by cytolytic mechanisms including perforin activation. In addition to their role in virus control, NK cells play an immunoregulatory role since they shape the CD8(+) T cell response to MCMV. To investigate the role of perforin-dependent cytolytic mechanism in NK cell modulation of CD8(+) T cell response during acute MCMV infection, we have used perforin-deficient C57BL/6 mice (Prf1(-/-)) and have shown that virus control by CD8(+) T cells in Prf1(-/-) mice is more efficient if NK cells are activated by the engagement of the Ly49H receptor with the m157 MCMV protein. A lack of perforin results in severe liver inflammation after MCMV infection, which is characterized by immunopathological lesions that are more pronounced in Prf1(-/-) mice infected with virus unable to activate NK cells. This immunopathology is caused by an abundant infiltration of activated CD8(+) T cells. The depletion of CD8(+) T cells has markedly reduced pathohistological lesions in the liver and improved the survival of Prf1(-/-) mice in spite of an increased viral load. Altogether, the results of our study suggest that a lack of perforin and absence of the specific activation of NK cells during acute MCMV infection lead to an unleashed CD8(+) T cell response that is detrimental for the host.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Muromegalovirus/imunologia , Perforina/metabolismo , Animais , Citocinas/metabolismo , Deleção de Genes , Infecções por Herpesviridae/mortalidade , Infecções por Herpesviridae/patologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Ativação Linfocitária/imunologia , Depleção Linfocítica , Camundongos , Camundongos Knockout , Muromegalovirus/genética , Mutação , Perforina/deficiência , Perforina/genética , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Carga Viral
14.
Methods Mol Biol ; 1070: 191-201, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24092441

RESUMO

Patients with small-cell lung cancer (SCLC) and prostate cancer (PCa) as well as other solid tumors may have micro- or macro-metastatic spread at an early stage of the disease. SCLC and PCa xenograft transfer models in immunodeficient mice fail to model this metastatic spread in vivo. In both tumor types the depletion of NK cells found in immunodeficient mice results in an increased number of spontaneous metastases, mirroring the clinical situation where NK cell activity in patients is related to metastatic spread of the disease. As a result NK cell activity directly influences treatment options and mortality. Newly developed immunodeficient mouse strains lacking functional T- and B-cells (rag2 knockout) however presenting functional NK cells (perforin knockout) are superior in producing spontaneous metastasis of SCLC and PCa cells compared to the system using SCID mice.


Assuntos
Proteínas de Ligação a DNA/deficiência , Neoplasias Pulmonares/secundário , Perforina/deficiência , Neoplasias da Próstata/secundário , Carcinoma de Pequenas Células do Pulmão/secundário , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Humanos , Injeções , Pulmão/patologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Knockout , Transplante de Neoplasias/patologia , Inclusão em Parafina , Perforina/metabolismo , Neoplasias da Próstata/patologia , Carcinoma de Pequenas Células do Pulmão/patologia , Tela Subcutânea/patologia , Fixação de Tecidos
15.
PLoS One ; 8(5): e63340, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691028

RESUMO

CD4 T cells are critical for clearing experimental Chlamydia muridarum genital tract infections. Two independent in vitro CD4 T cell mechanisms have been identified for terminating Chlamydia replication in epithelial cells. One mechanism, requiring IFN-γ and T cell-epithelial cell contact, terminates infection by triggering epithelial production of nitric oxide to chlamydiacidal levels; the second is dependent on T cell degranulation. We recently demonstrated that there are two independent in vivo clearance mechanisms singly sufficient for clearing genital tract infections within six weeks; one dependent on iNOS, the other on Plac8. Redundant genital tract clearance mechanisms bring into question negative results in single-gene knockout mice. Two groups have shown that perforin-knockout mice were not compromised in their ability to clear C. muridarum genital tract infections. Because cell lysis would be detrimental to epithelial nitric oxide production we hypothesized that perforin was not critical for iNOS-dependent clearance, but posited that perforin could play a role in Plac8-dependent clearance. We tested whether the Plac8-dependent clearance was perforin-dependent by pharmacologically inhibiting iNOS in perforin-knockout mice. In vitro we found that perforin was detrimental to iNOS-dependent CD4 T cell termination of Chlamydia replication in epithelial cells. In vivo, unexpectedly, clearance in perforin knockout mice was delayed to the end of week 7 regardless of iNOS status. The discordant in vitro/in vivo results suggest that the perforin's contribution to bacterial clearance in vivo is not though enhancing CD4 T cell termination of Chlamydia replication in epithelial cells, but likely via a mechanism independent of T cell-epithelial cell interactions.


Assuntos
Chlamydia muridarum/fisiologia , Perforina/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Perforina/deficiência , Perforina/genética , Infecções do Sistema Genital/imunologia , Infecções do Sistema Genital/metabolismo , Infecções do Sistema Genital/microbiologia
16.
ASN Neuro ; 5(1): e00105, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23289514

RESUMO

MS (multiple sclerosis) is the most prevalent autoimmune disease of the CNS (central nervous system) historically characterized as an inflammatory and demyelinating disease. More recently, extensive neuronal pathology has lead to its classification as a neurodegenerative disease as well. While the immune system initiates the autoimmune response it remains unclear how it orchestrates neuronal damage. In our previous studies, using in vitro cultured embryonic neurons, we demonstrated that MBP (myelin basic protein)-specific encephalitogenic CD4 T-cells induce early neuronal damage. In an extension of those studies, here we show that polarized CD4 Th1 and Th17 cells as wells as CD8 T-cells and NK (natural killer) cells induce microtubule destabilization within neurites in a contact-independent manner. Owing to the cytotoxic potential of these immune cells, we isolated the luminal components of lytic granules and determined that they were sufficient to drive microtubule destabilization. Since lytic granules contain cytolytic proteins, we determined that the induction of microtubule destabilization occurred prior to signs of apoptosis. Furthermore, we determined that microtubule destabilization was largely restricted to axons, sparing dendrites. This study demonstrated that lymphocytes with cytolytic activity have the capacity to directly drive MAD (microtubule axonal destabilization) in a bystander manner that is independent of neuronal death.


Assuntos
Axônios/fisiologia , Encefalomielite Autoimune Experimental/patologia , Linfócitos/metabolismo , Microtúbulos/metabolismo , Neurônios/fisiologia , Animais , Proteínas de Bactérias/genética , Linfócitos T CD4-Positivos , Morte Celular/imunologia , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Citocinas/farmacologia , Embrião de Mamíferos , Encefalomielite Autoimune Experimental/imunologia , Granzimas/deficiência , Marcação In Situ das Extremidades Cortadas , Células Matadoras Naturais , Proteínas Luminescentes/genética , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Modelos Biológicos , Proteína Básica da Mielina/genética , Neurônios/citologia , Perforina/deficiência , Células Th1 , Células Th17/metabolismo
17.
Immunol Lett ; 148(2): 138-43, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23089550

RESUMO

Protective immunity against Salmonella infection is known to require CD4 Th1 cells and B cells, but the role of MHC class-I-restricted CD8 T cells is less clear. Previous studies have suggested that CD8 T cells participate in secondary, but not primary, bacterial clearance. However, these studies have used experimental models that are difficult to interpret and do not clearly isolate the role of MHC class-I-restricted CD8 T cells from other cell populations. Here, we examined the role of class-I-restricted T cells in protection against Salmonella infection using mice lacking all classical MHC class-Ia molecules, perforin, or granzyme B. Immunized K(b)D(b)-, perforin-, granzyme B-, or perforin/granzyme B-deficient mice were able to resolve secondary infection with virulent Salmonella, demonstrating that class-I-restricted CTLs are not required for acquired immunity. However, during primary infection with attenuated bacteria, bacterial clearance was delayed in each of these mouse strains when compared to wild-type mice. Taken together, these data demonstrate that CD8 T cells are not required for acquired immunity to Salmonella, but can play a protective role in resolving primary infection with attenuated bacteria.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Salmonelose Animal/imunologia , Salmonella typhi/imunologia , Animais , Carga Bacteriana , Citotoxicidade Imunológica/imunologia , Granzimas/deficiência , Granzimas/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/deficiência , Perforina/genética , Salmonella typhi/patogenicidade
18.
Cancer Res ; 72(5): 1103-15, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22237627

RESUMO

In metronomic chemotherapy, frequent drug administration at lower than maximally tolerated doses can improve activity while reducing the dose-limiting toxicity of conventional dosing schedules. Although the antitumor activity produced by metronomic chemotherapy is attributed widely to antiangiogenesis, the significance of this mechanism remains somewhat unclear. In this study, we show that a 6-day repeating metronomic schedule of cyclophosphamide administration activates a potent antitumor immune response associated with brain tumor recruitment of natural killer (NK) cells, macrophages, and dendritic cells that leads to marked tumor regression. Tumor regression was blocked in nonobese diabetic/severe combined immunodeficient (NOD/SCID-γ) mice, which are deficient or dysfunctional in all these immune cell types. Furthermore, regression was blunted by NK cell depletion in immunocompetent syngeneic mice or in perforin-deficient mice, which are compromised for NK, NKT, and T-cell cytolytic functions. Unexpectedly, we found that VEGF receptor inhibitors blocked both innate immune cell recruitment and the associated tumor regression response. Cyclophosphamide administered at a maximum tolerated dose activated a transient, weak innate immune response, arguing that persistent drug-induced cytotoxic damage or associated cytokine and chemokine responses are required for effective innate immunity-based tumor regression. Together, our results reveal an innate immunity-based mechanism of tumor regression that can be activated by a traditional cytotoxic chemotherapy administered on a metronomic schedule. These findings suggest the need to carefully evaluate the clinical effects of combination chemotherapies that incorporate antiangiogenesis drugs targeting VEGF receptor.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Ciclofosfamida/farmacologia , Glioblastoma/dietoterapia , Glioblastoma/imunologia , Imidazóis/farmacologia , Imunidade Inata/efeitos dos fármacos , Indazóis/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Administração Metronômica , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/administração & dosagem , Axitinibe , Linhagem Celular Tumoral , Ciclofosfamida/administração & dosagem , Humanos , Masculino , Dose Máxima Tolerável , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Perforina/deficiência , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Immunol ; 187(12): 6301-9, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22084442

RESUMO

CTLs and NK cells use the perforin/granzyme cytotoxic pathway to kill virally infected cells and tumors. Human regulatory T cells also express functional granzymes and perforin and can induce autologous target cell death in vitro. Perforin-deficient mice die of excessive immune responses after viral challenges, implicating a potential role for this pathway in immune regulation. To further investigate the role of granzyme B in immune regulation in response to viral infections, we characterized the immune response in wild-type, granzyme B-deficient, and perforin-deficient mice infected with Sendai virus. Interestingly, granzyme B-deficient mice, and to a lesser extent perforin-deficient mice, exhibited a significant increase in the number of Ag-specific CD8(+) T cells in the lungs and draining lymph nodes of virally infected animals. This increase was not the result of failure in viral clearance because viral titers in granzyme B-deficient mice were similar to wild-type mice and significantly less than perforin-deficient mice. Regulatory T cells from WT mice expressed high levels of granzyme B in response to infection, and depletion of regulatory T cells from these mice resulted in an increase in the number of Ag-specific CD8(+) T cells, similar to that observed in granzyme B-deficient mice. Furthermore, granzyme B-deficient regulatory T cells displayed defective suppression of CD8(+) T cell proliferation in vitro. Taken together, these results suggest a role for granzyme B in the regulatory T cell compartment in immune regulation to viral infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Granzimas/fisiologia , Vírus Sendai/imunologia , Carga Viral/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Granzimas/deficiência , Granzimas/genética , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Perforina/deficiência , Perforina/genética , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Infecções por Respirovirus/virologia , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/virologia , Carga Viral/genética , Redução de Peso/genética , Redução de Peso/imunologia
20.
PLoS One ; 6(9): e24772, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21966366

RESUMO

Immune responses have the important function of host defense and protection against pathogens. However, the immune response also causes inflammation and host tissue injury, termed immunopathology. For example, hepatitis B and C virus infection in humans cause immunopathological sequel with destruction of liver cells by the host's own immune response. Similarly, after infection with lymphocytic choriomeningitis virus (LCMV) in mice, the adaptive immune response causes liver cell damage, choriomeningitis and destruction of lymphoid organ architecture. The immunopathological sequel during LCMV infection has been attributed to cytotoxic CD8(+) T cells. However, we now show that during LCMV infection CD4(+) T cells selectively induced the destruction of splenic marginal zone and caused liver cell damage with elevated serum alanin-transferase (ALT) levels. The destruction of the splenic marginal zone by CD4(+) T cells included the reduction of marginal zone B cells, marginal zone macrophages and marginal zone metallophilic macrophages. Functionally, this resulted in an impaired production of neutralizing antibodies against LCMV. Furthermore, CD4(+) T cells reduced B cells with an IgM(high)IgD(low) phenotype (transitional stage 1 and 2, marginal zone B cells), whereas other B cell subtypes such as follicular type 1 and 2 and germinal center/memory B cells were not affected. Adoptive transfer of CD4(+) T cells lacking different important effector cytokines and cytolytic pathways such as IFNγ, TNFα, perforin and Fas-FasL interaction did reveal that these cytolytic pathways are redundant in the induction of immunopathological sequel in spleen. In conclusion, our results define an important role of CD4(+) T cells in the induction of immunopathology in liver and spleen. This includes the CD4(+) T cell mediated destruction of the splenic marginal zone with consecutively impaired protective neutralizing antibody responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Hepatite Viral Animal/imunologia , Tecido Linfoide/imunologia , Baço/imunologia , Transferência Adotiva , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Quimiocinas/genética , Quimiocinas/imunologia , Quimiocinas/metabolismo , Proteína Ligante Fas/deficiência , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Citometria de Fluxo , Hepatite Viral Animal/metabolismo , Hepatite Viral Animal/virologia , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Tecido Linfoide/metabolismo , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Perforina/deficiência , Perforina/genética , Perforina/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/metabolismo , Baço/virologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...