Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7(1): 12168, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28939858

RESUMO

Increased availability of homeostatic cytokines is considered a major mechanism by which lymphodepletion enhances the efficacy of adoptive T cell therapy (ACT). IL-7 is one such cytokine capable of augmenting the function of tumor-reactive CD8+ T cells. However, whether host-derived IL-7 plays a role in driving the proper function of CD4+ T cells in an ACT setting remains unclear. Here we report that lymphodepleting chemotherapy by cyclophosphamide (CTX) does not lead to increased availability of the endogenous IL-7 in mice. Despite of a paucity of IL-7 in the immune milieu, CTX preconditioning allowed adoptively transferred naïve tumor-specific CD4+ T cells to undergo effector differentiation and regain IL-7Rα expression, giving rise to IL-7-responsive polyfunctional CD4+ effector cells. Correspondingly, supplementation of exogenous recombinant IL-7 markedly amplified and sustained polyfunctional CD4+ effector cells, resulting in improved therapeutic outcome in a mouse lymphoma model. We further demonstrated that the immune-enhancing effects of IL-7 were also applicable to donor CD4+ T cells pre-activated under Th1 polarizing condition. These findings suggest caution in relying on the endogenous IL-7 to enhance donor T cell expansion and persistence after lymphodepleting chemotherapy, and highlight the usefulness of recombinant IL-7 as an adjuvant for adoptive immunotherapy.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Imunoterapia Adotiva/métodos , Interleucina-7/uso terapêutico , Linfoma/terapia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/transplante , Linfoma/imunologia , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/uso terapêutico
2.
Oncotarget ; 8(67): 111931-111942, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29340102

RESUMO

In recent years the combined use of chemotherapy and immunotherapy, collectively termed chemoimmunotherapy, has emerged as a promising treatment option for patients with cancer. Antibiotics are commonly used to reduce infection-related complications in patients undergoing chemotherapy. Intriguingly, accumulating evidence has implicated gut microbiota as a critical determinant of host antitumor immune responses, raising the question as to whether the use of broad-spectrum antibiotics would invariably diminish tumor response to chemoimmunotherapies. We investigated the impact of antibiotics on the therapeutic outcomes of cyclophosphamide (CTX) chemotherapy and adoptive T-cell therapy (ACT) where CTX was used as the host-conditioning regimen in mice. We show that antibiotic prophylaxis dampened the endogenous T cell responses elicited by CTX, and reduced the efficacy of CTX against B-cell lymphoma. In the ACT setting, antibiotics administration impaired the therapeutic effects of adoptively transferred tumor-specific CD4+ T cells in mice with implanted colorectal tumors. In contrast, long-term antibiotic exposure did not affect the efficacy of ACT using CD19-targeting chimeric antigen receptor (CAR) T cells in mice with systemic B-cell lymphoma, although it correlated with prolonged CAR expression and sustained B-cell aplasia. Our study demonstrates that chemoimmunotherapies may have variable reliance on intestinal microbiota for T cell activation and function, and thus have different sensitivities to antibiotic prophylaxis. These findings may have implications for the judicial use of antibiotics in cancer patients receiving chemoimmunotherapies.

3.
Crit Rev Immunol ; 36(2): 179-191, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27910767

RESUMO

The alkylating agent melphalan is used in the treatment of hematological malignancies, especially multiple myeloma. In the past, the usefulness of melphalan has been solely attributed to its cytotoxicity on fastgrowing cancerous cells. Although the immunomodulatory effects of melphalan were suggested many years ago, only recently has this aspect of melphalan's activity begun to be elucidated at the molecular level. Emerging evidence indicates that melphalan can foster an immunogenic microenvironment by inducing immunogenic cell death (ICD) as characterized by membrane translocation of endoplasmic reticulum protein calreticulin (CRT) and by release of chromatin-binding protein high-mobility group box 1 (HMGB1). In addition, the lympho-depletive effect of melphalan can induce the release of pro-inflammatory cytokines and growth factors, deplete regulatory T cells, and create space to facilitate the expansion of infused tumor-reactive T cells. These features suggest that melphalan can be used as a preparative chemotherapy for adoptive T-cell therapy. This notion is supported by our recent work demonstrating that the combination of melphalan and adoptive transfer of tumor-reactive CD4+ T cells can mediate potent antitumor effects in animal models. This review summarizes the recent advances in understanding and utilizing the immunomodulatory effects of melphalan.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Imunoterapia Adotiva , Melfalan/uso terapêutico , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos
4.
Oncoimmunology ; 5(6): e1171445, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27471650

RESUMO

The functional status of CD4(+) T cells is a critical determinant of antitumor immunity. Polyfunctional CD4(+) T cells possess the ability to concomitantly produce multiple Th1-type cytokines, exhibiting a functional attribute desirable for cancer immunotherapy. However, the mechanisms by which these cells are induced are neither defined nor it is clear if these cells can be used therapeutically to treat cancer. Here, we report that CD4(+) T cells exposed to exogenous IL-7 during antigenic stimulation can acquire a polyfunctional phenotype, characterized by their ability to simultaneously express IFNγ, IL-2, TNFα and granzyme B. This IL-7-driven polyfunctional phenotype was associated with increased histone acetylation in the promoters of the effector genes, indicative of increased chromatin accessibility. Moreover, forced expression of a constitutively active (CA) form of STAT5 recapitulated IL-7 in inducing CD4(+) T-cell polyfunctionality. Conversely, the expression of a dominant negative (DN) form of STAT5 abolished the ability of IL-7 to induce polyfunctional CD4(+) T cells. These in-vitro-generated polyfunctional CD4(+) T cells can traffic to tumor and expand intratumorally in response to immunization. Importantly, adoptive transfer of polyfunctional CD4(+) T cells following lymphodepletive chemotherapy was able to eradicate large established tumors. This beneficial outcome was associated with the occurrence of antigen epitope spreading, activation of the endogenous CD8(+) T cells and persistence of donor CD4(+) T cells exhibiting memory stem cell attributes. These findings indicate that IL-7 signaling can impart polyfunctionality and stemness potential to CD4(+) T cells, revealing a previously unknown property of IL-7 that can be exploited in adoptive T-cell immunotherapy.

5.
Vitam Horm ; 99: 171-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26279376

RESUMO

Bone morphogenetic proteins (BMPs) are growth factors belonging to the TGF-ß (transforming growth factor ß) superfamily. BMPs were found to regulate multiple cell processes such as proliferation, survival, differentiation, and apoptosis. They were originally described to play a pivotal role in inducing bone, cartilage, ligament, and tendon formation at both heterotopic and orthotopic sites but were found to play a significant role in embryogenesis and development of multiple tissues and organs. Activities of BMPs are regulated by a number of secreted proteins, which modulate their availability to bind cellular receptors. The functions of individual BMPs are highly redundant due to binding the same receptors and inducing overlapping signal transduction pathways. Recently, BMPs were found to regulate cells of the innate and adaptive immune system. BMPs are involved in thymic development of T cells at the early, double negative, as well as later, double positive, stages of thymopoesis. They specifically modulate thymic development of regulatory T cells (T(reg)). In the periphery, BMPs affect T cell activation, promoting generation of T(reg) cells. We found that mice deficient for one of the receptors activated by BMPs demonstrated slower growth of transplantable melanoma tumors.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/fisiologia , Transdução de Sinais/fisiologia , Timo/crescimento & desenvolvimento , Animais , Proteínas Morfogenéticas Ósseas/imunologia , Linfócitos T CD4-Positivos/citologia , Regulação da Expressão Gênica/fisiologia , Humanos , Timo/citologia , Timo/imunologia
6.
Dis Model Mech ; 8(7): 691-700, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26035381

RESUMO

Acute schistosomiasis is characterized by pro-inflammatory responses against tissue- or organ-trapped parasite eggs along with granuloma formation. Here, we describe studies in Cx3cr1(-/-) mice and demonstrate the role of Cx3cr1 in the pathoetiology of granuloma formation during acute schistosomiasis. Mice deficient in Cx3cr1 were protected from granuloma formation and hepatic injury induced by Schistosoma japonicum eggs, as manifested by reduced body weight loss and attenuated hepatomegaly along with preserved liver function. Notably, S. japonicum infection induced high levels of hepatic Cx3cr1 expression, which was predominantly expressed by infiltrating macrophages. Loss of Cx3cr1 rendered macrophages preferentially towards M2 polarization, which then led to a characteristic switch of the host immune defense from a conventional Th1 to a typical Th2 response during acute schistosomiasis. This immune switch caused by Cx3cr1 deficiency was probably associated with enhanced STAT6/PPAR-γ signaling and increased expression of indoleamine 2,3-dioxygenase (IDO), an enzyme that promotes M2 polarization of macrophages. Taken together, our data provide evidence suggesting that CX3CR1 could be a viable therapeutic target for treatment of acute schistosomiasis.


Assuntos
Hepatopatias Parasitárias/etiologia , Macrófagos/imunologia , Receptores de Interleucina-8A/deficiência , Esquistossomose Japônica/imunologia , Esquistossomose Japônica/patologia , Doença Aguda , Animais , Modelos Animais de Doenças , Feminino , Granuloma/etiologia , Granuloma/imunologia , Granuloma/patologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cirrose Hepática Experimental/etiologia , Cirrose Hepática Experimental/imunologia , Cirrose Hepática Experimental/patologia , Hepatopatias Parasitárias/imunologia , Hepatopatias Parasitárias/patologia , Macrófagos/classificação , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óvulo/imunologia , PPAR gama/metabolismo , Receptores de Interleucina-8A/genética , Fator de Transcrição STAT6/metabolismo , Schistosoma japonicum/imunologia , Esquistossomose Japônica/complicações , Transdução de Sinais , Células Th2/imunologia , Células Th2/patologia
7.
J Immunotoxicol ; 11(4): 319-27, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24350726

RESUMO

Immunotherapy is becoming an increasingly attractive therapeutic alternative for conventional cancer therapy. In recent years Foxp3(+) regulatory T-cells (T(R)) were identified as the major obstacle to effective cancer immunotherapy. The abundance of these cells in peripheral blood is increased in patients with multiple types of cancer and their prevalence among tumor-infiltrating lymphocytes correlated with poor clinical prognosis. In contrast, removal or inactivation of T(R) cells led to enhanced anti-tumor immune response and better efficacy of cancer vaccines. This study reports that Bone Morphogenic Protein Receptor 1α (BMPR1α, Alk-3) is expressed by activated effector CD4(+) and T(R) cells and modulates functions of both cell types. Bone Morphogenic Proteins (BMPs) belong to the transforming growth factor (TGF)-ß family of cytokines that also include TGFß and activins. BMPs play crucial roles in embryonic development, tissue differentiation and homeostasis, and development of cancer. It was demonstrated that BMPs and activins synergize with TGFß to regulate thymic T-cell development, maintain T(R) cells, and control peripheral tolerance. Inactivation of BMPR1α in T-cells results in impaired thymic and peripheral generation of T(R) cells. BMPR1α-deficient activated T-cells produced a higher level of interferon (IFN)-γ than BMPR1α-sufficient T-cells. Moreover, transplanted B16 melanoma tumors grew smaller in mice lacking expression of BMPR1α in T-cells and tumors had few infiltrating TR cells and a higher proportion of CD8(+) T-cells than wild-type mice.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/imunologia , Proteínas Morfogenéticas Ósseas/imunologia , Imunoterapia/métodos , Neoplasias Experimentais/terapia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Diferenciação Celular/genética , Humanos , Ativação Linfocitária/genética , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Tolerância Periférica/genética , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Carga Tumoral
8.
Am J Transl Res ; 5(4): 427-40, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23724166

RESUMO

As a versatile regulatory mechanism, sumoylation has been found to be essential for ordered diverse cellular processes. However, the exact impact of sumoylation on endothelial function largely remained elusive. Here we investigated the role of small ubiquitin-like modifier 1 (SUMO1) mediated sumoylation in the regulation of endothelial function by examining its effect on angiogenesis and homeostatic responses. Adenoviral-mediated SUMO1 expression in porcine aortic endothelial cells (PAECs) dose-dependently promoted proliferation, migration and tube formation. In line with these results in PAECs, Matrigel plug assays in SUMO1 transgenic mice demonstrated a significant higher capacity for vascular neogenesis as compared with that of control littermates. Moreover, SUMO1 expression protected PAECs from serum starvation or H2O2-induced apoptosis. Mechanistic studies demonstrated that SUMO1 sumoylation modulates ERK1/2 activation and MMP13 expression as well as Jak2/STAT5 signaling to promote angiogenesis. SUMO1 sumoylation also suppressed NFκB and c-JUN transcriptional activity to provide protection for PAECs against oxidative stress-induced apoptosis. Given that sumoylation is a reversible process, dynamic regulation of the sumoylation function could be a novel strategy to modulate endothelial function in disease states.

9.
J Immunol ; 187(1): 248-57, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642545

RESUMO

Despite their importance for the functioning of the immune system, thymic development and peripheral maintenance of Foxp3(+) regulatory T (T(R)) cells are poorly understood. We have found that connexin 43 (Cx43), expressed by thymic T(R) cells progenitors, supports T(R) development. Mice with deletion of the Cx43 gene induced in T cells produce only few T(R) cells and had increased proportion of activated T cells in the lymph nodes, suggesting impaired peripheral tolerance. Reduction of the T(R) cell numbers was accompanied by increased presence of CD4(+)CD25(+)GITR(+)Foxp3(-) T cells, which did not produce inflammatory cytokines and lost suppressor function. These results strongly argue that we have discovered a novel signaling pathway, controlled by Cx43, that enhances the generation of T(R) cells. We propose that a possible mechanism of Cx43 activity is by regulating Foxp3 expression in T(R) lineage cells.


Assuntos
Diferenciação Celular/imunologia , Conexina 43/fisiologia , Fatores de Transcrição Forkhead/biossíntese , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Regulação para Cima/imunologia , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Células Cultivadas , Conexina 43/biossíntese , Conexina 43/genética , Regulação da Expressão Gênica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação/imunologia , Transdução de Sinais/genética , Linfócitos T Reguladores/citologia , Regulação para Cima/genética
10.
PLoS One ; 5(10): e13623, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-21049016

RESUMO

The presence of Foxp3(+) regulatory CD4(+) T cells in tumor lesions is considered one of the major causes of ineffective immune response in cancer. It is not clear whether intratumoral T(reg) cells represent T(reg) cells pre-existing in healthy mice, or arise from tumor-specific effector CD4(+) T cells and thus representing adaptive T(reg) cells. The generation of T(reg) population in tumors could be further complicated by recent evidence showing that both in humans and mice the peripheral population of T(reg) cells is heterogenous and consists of subsets which may differentially respond to tumor-derived antigens. We have studied T(reg) cells in cancer in experimental mice that express naturally selected, polyclonal repertoire of CD4(+) T cells and which preserve the heterogeneity of the T(reg) population. The majority of T(reg) cells present in healthy mice maintained a stable suppressor phenotype, expressed high level of Foxp3 and an exclusive set of TCRs not used by naive CD4(+) T cells. A small T(reg) subset, utilized TCRs shared with effector T cells and expressed a lower level of Foxp3. We show that response to tumor-derived antigens induced efficient clonal recruitment and expansion of antigen-specific effector and T(reg) cells. However, the population of T(reg) cells in tumors was dominated by cells expressing TCRs shared with effector CD4(+) T cells. In contrast, T(reg) cells expressing an exclusive set of TCRs, that dominate in healthy mice, accounted for only a small fraction of all T(reg) cells in tumor lesions. Our results suggest that the T(reg) repertoire in tumors is generated by conversion of effector CD4(+) T cells or expansion of a minor subset of T(reg) cells. In conclusion, successful cancer immunotherapy may depend on the ability to block upregulation of Foxp3 in effector CD4(+) T cells and/or selectively inhibiting the expansion of a minor T(reg) subset.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Melanoma Experimental/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Separação Celular , Células Clonais , Primers do DNA , Citometria de Fluxo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Polimorfismo Conformacional de Fita Simples , Receptores de Antígenos de Linfócitos T/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA