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

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
Immunity ; 34(6): 879-92, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21658975

RESUMO

Cytotoxic lymphocyte-mediated apoptosis is dependent on the delivery of perforin to secretory granules and its ability to form calcium-dependent pores in the target cell after granule exocytosis. It is unclear how cytotoxic lymphocytes synthesize and store perforin without incurring damage or death. We discovered that the extreme C terminus of perforin was essential for rapid trafficking from the endoplasmic reticulum to the Golgi compartment. Substitution of the C-terminal tryptophan residue resulted in retention of perforin in the ER followed by calcium-dependent toxic activity that eliminated host cells. We also found that N-linked glycosylation of perforin was critical for transport from the Golgi to secretory granules. Overall, an intact C terminus and N-linked glycosylation provide accurate and efficient export of perforin from the endoplasmic reticulum to the secretory granules and are critical for cytotoxic lymphocyte survival.


Assuntos
Movimento Celular , Exocitose , Perforina/imunologia , Polissacarídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Autólise/imunologia , Linhagem Celular , Retículo Endoplasmático/imunologia , Glicosilação , Humanos , Camundongos , Camundongos Knockout , Mutação , Perforina/deficiência , Ratos
4.
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
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.
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
7.
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
8.
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
9.
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
10.
J Immunol ; 184(3): 1218-26, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20048124

RESUMO

Commensal bacteria play an important role in formation of the immune system, but the mechanisms involved are incompletely understood. In this study, we analyze CD1d-restricted invariant NKT (iNKT) cells in germfree mice and in two colonies of C57BL/6 mice termed conventional flora and restricted flora (RF), stably bearing commensal microbial communities of diverse but distinct composition. In germfree mice, iNKT cells were moderately reduced, suggesting that commensal microbiota were partially required for the antigenic drive in maintaining systemic iNKT cells. Surprisingly, even greater depletion of iNKT cell population occurred in RF mice. This was in part attributable to reduced RF levels of intestinal microbial taxa (Sphingomonas spp.) known to express antigenic glycosphingolipid products. However, memory and activated CD8(+) T cells were also expanded in RF mice, prompting us to test whether CD8(+) T cell activity might be further depleting iNKT cells. Indeed, iNKT cell numbers were restored in RF mice bearing the CD8alpha(-/-) genotype or in adult wild-type RF mice acutely depleted with anti-CD8 Ab. Moreover, iNKT cells were restored in RF mice bearing the Prf1(-/-) phenotype, a key component of cytolytic function. These findings indicate that commensal microbiota, through positive (antigenic drive) and negative (cytolytic depletion by CD8(+) T cells) mechanisms, profoundly shape the iNKT cell compartment. Because individuals greatly vary in the composition of their microbial communities, enteric microbiota may play an important epigenetic role in the striking differences in iNKT cell abundance in humans and therefore in their potential contribution to host immune status.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/microbiologia , Animais , Antígenos CD8/biossíntese , Linfócitos T CD8-Positivos/transplante , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Citotoxicidade Imunológica/genética , Vida Livre de Germes/imunologia , Memória Imunológica/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/citologia , Perforina/biossíntese , Perforina/deficiência , Perforina/genética
11.
J Immunol ; 184(5): 2528-38, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20097864

RESUMO

Malaria results in >1,000,000 deaths per year worldwide. Although no licensed vaccine exists, much effort is currently focused on subunit vaccines that elicit CD8 T cell responses directed against Plasmodium parasite liver stage Ags. Multiple immune-effector molecules play a role in antimicrobial immunity mediated by memory CD8 T cells, including IFN-gamma, perforin, TRAIL, Fas ligand, and TNF-alpha. However, it is not known which pathways are required for memory CD8 T cell-mediated immunity against liver stage Plasmodium infection. In this study, we used a novel immunization strategy to generate memory CD8 T cells in the BALB/c mouse model of P. berghei or P. yoelii sporozoite infection to examine the role of immune-effector molecules in resistance to the liver stage infection. Our studies reveal that endogenous memory CD8 T cell-mediated protection against both parasite species is, in part, dependent on IFN-gamma, whereas perforin was only critical in protection against P. yoelii. We further show that neutralization of TNF-alpha in immunized mice markedly reduces memory CD8 T cell-mediated protection against both parasite species. Thus, our studies identify IFN-gamma and TNF-alpha as important components of the noncytolytic pathways that underlie memory CD8 T cell-mediated immunity against liver stage Plasmodium infection. Our studies also show that the effector pathways that memory CD8 T cells use to eliminate liver stage infection are, in part, Plasmodium species specific.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Esporozoítos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Culicidae/parasitologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Feminino , Memória Imunológica/imunologia , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/imunologia , Listeria monocytogenes/imunologia , Hepatopatias Parasitárias/imunologia , Hepatopatias Parasitárias/parasitologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Perforina/deficiência , Perforina/genética , Perforina/imunologia , Transdução de Sinais/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
12.
J Virol ; 84(1): 312-20, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19828618

RESUMO

Although much is known about lymphocytic choriomeningitis virus (LCMV) infection and the subsequent immune response in its natural murine host, some crucial aspects of LCMV-mediated pathogenesis remain undefined, including the underlying basis of the characteristic central nervous system disease that occurs following intracerebral (i.c.) challenge. We show that the classic seizures and paresis that occur following i.c. infection of adult, immunocompetent mice with LCMV are accompanied by anatomical and histological changes that are consistent with brain herniation, likely of the uncal subtype, as a causative basis for disease and precipitous death. Both by water weight determinations and by magnetic resonance imaging of infected brain tissues, edema was detected only at the terminal stages of disease, likely caused by the leakage of cerebrospinal fluid from the ventricles into the parenchyma. Furthermore, death was accompanied by unilateral pupillary dilation, which is indicative of uncal herniation. While immunohistochemical analysis revealed periventricular inflammation and a loss of integrity of the blood-brain barrier (BBB), these events preceded seizures by 2 to 3 days. Moreover, surviving perforin knockout mice showed barrier permeability equivalent to that of moribund, immunocompetent mice; thus, BBB damage does not appear to be the basis of LCMV-induced neuropathogenesis. Importantly, brain herniation can occur in humans as a consequence of injuries that would be predicted to increase intracranial pressure, including inflammation, head trauma, and brain tumors. Thus, a mechanistic dissection of the basis of LCMV neuropathogenesis may be informative for the development of interventive therapies to prevent this typically fatal human condition.


Assuntos
Edema/etiologia , Encefalocele/etiologia , Coriomeningite Linfocítica/mortalidade , Vírus da Coriomeningite Linfocítica , Animais , Barreira Hematoencefálica/patologia , Edema/patologia , Encefalocele/patologia , Inflamação , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Mortalidade , Paresia , Perforina/deficiência , Convulsões
13.
Epilepsia ; 52(9): 1627-34, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21627645

RESUMO

PURPOSE: A link between seizure susceptibility, blood-brain barrier (BBB) failure, and the activation of peripheral white blood cells has been recently proposed. However, the molecular players involved in this cascade of events are unknown. We tested the hypothesis that immunosupression by splenectomy or lack of perforin, a downstream factor of natural killer (NK) and cytotoxic T cells, could reduce seizure onset. METHODS: Pilocarpine was used to induce seizures in adult rats wild-type and perforin-deficient mice. Splenectomy was performed prior to pilocarpine injection. Seizure onset was evaluated by electroencephalography (EEG) and joint time-frequency analysis. Spleens from control and pilocarpine-treated groups were analyzed for anatomical changes and CD3+ cell content. BBB damage was assessed by measuring albumin parenchymal extravasation. Fluorescence-activated cell sorting (FACS) analysis was performed on spleen and brain tissue of wild-type and perforin-deficient mice treated, or not, with pilocarpine. KEY FINDINGS: Splenectomy significantly reduced seizure-associated mortality. Histologic analysis of the spleens exposed to pilocarpine revealed altered white and red pulp anatomy and an increase in CD3+ T cells. Onset of status epilepticus (SE) and mortality were significantly decreased in perforin-deficient mice. Pilocarpine significantly increased spleen NK 1.1 and CD8+ cell percentage; in contrast, the brain inflammatory cell profile remained unchanged at the time of pilocarpine SE. BBB damage was reduced in the perforin-deficient pilocarpine-treated mice. SIGNIFICANCE: Immunosuppressant maneuvers such as splenectomy or lack of perforin decrease the onset or the severity of pilocarpine SE. Our results suggest that cytotoxic lymphocytes, and specifically the cytolytic factor perforin, may be key molecular players involved in the axis between peripheral intravascular inflammation and seizures.


Assuntos
Convulsões/etiologia , Convulsões/patologia , Linfócitos T Citotóxicos/fisiologia , Animais , Antígenos CD/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Eletroencefalografia , Citometria de Fluxo/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/deficiência , Pilocarpina , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/terapia , Esplenectomia/métodos , Linfócitos T Citotóxicos/efeitos dos fármacos
14.
Pediatr Blood Cancer ; 57(7): 1239-43, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21681935

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is an immunodysregulatory disorder for which more effective treatments are needed. The macrolide rapamycin has immunosuppressive properties, making it an attractive candidate for controlling the aberrant T cell activation that occurs in HLH. To investigate its therapeutic potential, we used rapamycin to treat Lymphocytic Choriomeningitis Virus (LCMV)-infected perforin-deficient (Prf1(-/-)) mice according to a well-established model of HLH. At the regimens tested, rapamycin did not improve weight loss, splenomegaly, hemophagocytosis, cytopenias, or proinflammatory cytokine production in LCMV-infected Prf1(-/-) animals. Thus, single agent rapamycin appears ineffective in treating the clinical and laboratory manifestations of LCMV-induced HLH.


Assuntos
Imunossupressores/uso terapêutico , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Sirolimo/uso terapêutico , Animais , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/imunologia , Separação Celular , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Vírus da Coriomeningite Linfocítica , Linfo-Histiocitose Hemofagocítica/imunologia , Linfo-Histiocitose Hemofagocítica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/deficiência
15.
J Immunol ; 183(9): 5870-8, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19812194

RESUMO

The production of IFN-gamma by CD8(+) T cells is an important hallmark of protective immunity induced by irradiation-attenuated sporozoites against malaria. Here, we demonstrate that protracted sterile protection conferred by a Plasmodium yoelii genetically attenuated parasite (PyGAP) vaccine was completely dependent on CD8(+) T lymphocytes but only partially dependent on IFN-gamma. We used live cell imaging to document that CD8(+) CTL from PyGAP-immunized mice directly killed hepatocyte infected with a liver stage parasite. Immunization studies with perforin and IFN-gamma knockout mice also indicated that the protection was largely dependent on perforin-mediated effector mechanisms rather than on IFN-gamma. This was further supported by our observation that both liver and spleen CD8(+) T cells from PyGAP-immunized mice induced massive apoptosis of liver stage-infected hepatocytes in vitro without the release of detectable IFN-gamma and TNF-alpha. Conversely, CD8(+) T cells isolated from naive mice that had survived wild-type P. yoelii sporozoite infection targeted mainly sporozoite-traversed and uninfected hepatocytes, revealing an immune evasion strategy that might be used by wild-type parasites to subvert host immune responses during natural infection. However, CTLs from wild-type sporozoite-challenged mice could recognize and kill infected hepatocytes that were pulsed with circumsporozoite protein. Additionally, protection in PyGAP-immunized mice directly correlated with the magnitude of effector memory CD8(+) T cells. Our findings implicate CTLs as key immune effectors in a highly protective PyGAP vaccine for malaria and emphasize the critical need to define surrogate markers for correlates of protection, apart from IFN-gamma.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/imunologia , Citotoxicidade Imunológica/genética , Hepatócitos/imunologia , Hepatócitos/parasitologia , Hepatopatias Parasitárias/prevenção & controle , Vacinas Antimaláricas/genética , Plasmodium yoelii/genética , Animais , Linfócitos T CD8-Positivos/parasitologia , Linfócitos T CD8-Positivos/patologia , Comunicação Celular/genética , Feminino , Hepatócitos/patologia , Memória Imunológica/genética , Interferon gama/deficiência , Interferon gama/genética , Hepatopatias Parasitárias/imunologia , Hepatopatias Parasitárias/parasitologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Perforina/deficiência , Perforina/genética , Plasmodium yoelii/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
16.
Immunol Cell Biol ; 88(7): 761-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20309009

RESUMO

Cytotoxic T lymphocytes (CTLs) represent one of the front lines of defense for the immune system, killing virus-infected and tumor-transformed cells. CTL use at least two mechanisms to induce apoptosis in their targets, one mediated by perforin and granzymes, and the other triggered by the death ligand, CD95 ligand (CD95L). Here, we used an in vivo cytotoxicity assay to measure specific clearance of antigen-bearing target cells in mice that had previously been immunized with noninfectious cell-associated antigens. We found that perforin was dispensable for efficient clearance of antigen-bearing cells from immunized mice, but only if CD95/CD95L was functional; however, there was a delay in target cell clearance in the absence of perforin. In addition, we observed ∼35% target cell clearance in the absence of both perforin and CD95L, which was only slightly abrogated in the presence of a neutralizing anti-tumor necrosis factor (TNF) antibody. The presence of a dominant negative Fas-associated death domain (FADD) did not block target cell clearance and therefore cannot be attributed to known death receptors. Taken together, these data suggest that perforin- and CD95L-dependent killing are complementary at early time points, each can compensate for the absence of the other at later time points, and that there is an additional component of antigen-restricted CTL killing independent of perforin, CD95L, and TNFα.


Assuntos
Citotoxicidade Imunológica/fisiologia , Perforina/fisiologia , Linfócitos T Citotóxicos/imunologia , Receptor fas/fisiologia , Transferência Adotiva , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Perforina/deficiência , Fator de Necrose Tumoral alfa/fisiologia , Receptor fas/deficiência
17.
J Immunol ; 181(12): 8595-603, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19050279

RESUMO

Optimal immunity to Mycobacterium tuberculosis (Mtb) infection requires CD8(+) T cells, and several current Mtb vaccine candidates are being engineered to elicit enhanced CD8(+) T cell responses. However, the function of these T cells and the mechanism by which they provide protection is still unknown. We have previously shown that CD8(+) T cells specific for the mycobacterial Ags CFP10 and TB10.4 accumulate in the lungs of mice following Mtb infection and have cytolytic activity in vivo. In this study, we determine which cytolytic pathways are used by these CD8(+) T cells during Mtb infection. We find that Mtb-specific CD8(+) T cells lacking perforin have reduced cytolytic capacity in vivo. In the absence of perforin, the residual cytolytic activity is CD95 and TNFR dependent. This is particularly true in Mtb-infected lung tissue where disruption of both perforin and CD95 eliminates target cell lysis. Moreover, adoptive transfer of immune CD8(+) T cells isolated from wild-type, but not perforin-deficient mice, protect recipient mice from Mtb infection. We conclude that CD8(+) T cells elicited following Mtb infection use several cytolytic pathways in a hierarchical and compensatory manner dominated by perforin-mediated cytolysis. Finally, although several cytolytic pathways are available, adoptively transferred Mtb-specific CD8(+) T cells require perforin-mediated cytolysis to protect animals from infection. These data show that CD8(+) T cell-mediated protection during Mtb infection requires more than the secretion of IFN-gamma and specifically defines the CD8(+) cytolytic mechanisms utilized and required in vivo.


Assuntos
Testes Imunológicos de Citotoxicidade , Epitopos de Linfócito T/imunologia , Mycobacterium tuberculosis/imunologia , Perforina/administração & dosagem , Perforina/fisiologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/microbiologia , Tuberculose Pulmonar/prevenção & controle , Transferência Adotiva , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Epitopos de Linfócito T/administração & dosagem , Feminino , Antígenos de Histocompatibilidade Classe I/administração & dosagem , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Perforina/deficiência , Linfócitos T Citotóxicos/transplante , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/metabolismo
18.
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
19.
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
20.
Immunology ; 128(1): 69-82, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19689737

RESUMO

Although CD8(+) cytotoxic T lymphocytes (CTL) exhibit both Fas ligand (FasL) -based and perforin-based lytic activities, the accepted hallmark of a fully active CTL remains its perforin killing machinery. Yet the origin, rationale for possessing both a slow-acting (FasL) and a fast-acting (perforin) killing mechanism has remained enigmatic. Here we have investigated perforin expression in CTL directly involved in acute tumour (i.e. leukaemias EL4 and L1210) allograft rejection occurring within the peritoneal cavity. We show that at the height of the immune response, the majority of conjugate-forming CD8(+) CTL express high levels of perforin messenger RNA and protein, and kill essentially via perforin. Later however, coinciding with complete rejection, fully cytocidal CTL emerge which exhibit a stark decrease in perforin and now kill preferentially via constitutively expressed FasL. Although late in emergence, and persistent, these powerful CTL are neither effector-memory nor memory CTL. This finding has implications for the monitoring of anti-transplant responses in clinical settings, based on assessing perforin expression in graft infiltrating CD8(+) T cells. The results show that as the immune response progresses in vivo, targeted cellular suicide mainly prunes high perforin-expressing CD8(+) cells, resulting in the gradual switch in effector CTL, from mostly perforin-based to largely Fas/FasL-based killers. Hence, two kinds of CD8(+) CTL have two killing strategies.


Assuntos
Leucemia Experimental/imunologia , Perforina/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Proteína Ligante Fas/imunologia , Rejeição de Enxerto/imunologia , Memória Imunológica/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Perforina/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA