Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Lett ; 525: 84-96, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-34740608

RESUMO

Wnt/ß-catenin signaling is a highly conserved pathway that regulates cell proliferation, differentiation, apoptosis, stem cell self-renewal, tissue homeostasis, and wound healing. Dysregulation of the Wnt pathway is intricately involved in almost all stages of tumorigenesis in various cancers. Through direct and/or indirect effects on effector T cells, T-regulatory cells, T-helper cells, dendritic cells, and other cytokine-expressing immune cells, abnormal activation of Wnt/ß-catenin signaling benefits immune exclusion and hinders T-cell-mediated antitumor immune responses. Activation of Wnt signaling results in increased resistance to immunotherapies. In this review, we summarize the process by which Wnt signaling affects cancer and immune surveillance, and the potential for targeting the Wnt-signaling pathway via cancer immunotherapy.


Assuntos
Carcinogênese/genética , Imunoterapia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Carcinogênese/imunologia , Proliferação de Células/genética , Autorrenovação Celular/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/terapia , Células Th1/imunologia , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/imunologia
2.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-33951416

RESUMO

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Assuntos
COVID-19/imunologia , COVID-19/virologia , Autorrenovação Celular/imunologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , SARS-CoV-2/imunologia , Biomarcadores , COVID-19/metabolismo , Citocinas/metabolismo , Suscetibilidade a Doenças/imunologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Transdução de Sinais
3.
Nat Immunol ; 22(6): 723-734, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33958784

RESUMO

Continuous supply of immune cells throughout life relies on the delicate balance in the hematopoietic stem cell (HSC) pool between long-term maintenance and meeting the demands of both normal blood production and unexpected stress conditions. Here we identified distinct subsets of human long-term (LT)-HSCs that responded differently to regeneration-mediated stress: an immune checkpoint ligand CD112lo subset that exhibited a transient engraftment restraint (termed latency) before contributing to hematopoietic reconstitution and a primed CD112hi subset that responded rapidly. This functional heterogeneity and CD112 expression are regulated by INKA1 through direct interaction with PAK4 and SIRT1, inducing epigenetic changes and defining an alternative state of LT-HSC quiescence that serves to preserve self-renewal and regenerative capacity upon regeneration-mediated stress. Collectively, our data uncovered the molecular intricacies underlying HSC heterogeneity and self-renewal regulation and point to latency as an orchestrated physiological response that balances blood cell demands with preserving a stem cell reservoir.


Assuntos
Autorrenovação Celular/imunologia , Células-Tronco Hematopoéticas/fisiologia , Reconstituição Imune , Células-Tronco Multipotentes/fisiologia , Estresse Fisiológico/imunologia , Adulto , Animais , Autorrenovação Celular/genética , Células Cultivadas , Epigênese Genética/imunologia , Feminino , Sangue Fetal/citologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Hematopoese , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Separação Imunomagnética , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Nectinas/metabolismo , Cultura Primária de Células , RNA-Seq , Análise de Célula Única , Sirtuína 1/metabolismo , Estresse Fisiológico/genética , Transplante Heterólogo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
5.
Theranostics ; 10(26): 11950-11962, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204322

RESUMO

Rationale: Mesenchymal stem cells (MSCs) have been the focus of many studies because of their abilities to modulate immune responses, angiogenesis, and promote tumor growth and metastasis. Our previous work showed that gastric cancer MSCs (GCMSCs) promoted immune escape by secreting of IL-8, which induced programmed cell death ligand 1 (PD-L1) expression in GC cells. Mounting evidence has revealed that PD-L1 expression is related to intrinsic tumor cell properties. Here, we investigated whether GCMSCs maintained a pool of cancer stem cells (CSCs) through PD-L1 signaling and the specific underlying molecular mechanism. Methods: Stem cell surface markers, aldehyde dehydrogenase (ALDH) activity, migration and sphere formation abilities were tested to evaluate the stemness of GC cells. PD-L1-expressing lentivirus and PD-L1 specific siRNA were used to analyze the effects of PD-L1 on GC cells stemness. Annexin V/PI double staining was used to assess apoptosis of GC cells induced by chemotherapy. Co-Immunoprecipitation (Co-IP) and Mass spectrometry were employed to determine the PD-L1 binding partner in GC cells. PD-L1Negative and PD-L1Positive cells were sorted by flow cytometry and used for limiting dilution assays to verify the effect of PD-L1 on tumorigenic ability in GC cells. Results: The results showed that GCMSCs enhanced the CSC-like properties of GC cells through PD-L1, which led to the resistance of GC cells to chemotherapy. PD-L1 associated with CTCF to contribute to the stemness and self-renewal of GC cells. In vivo, PD-L1Positive GC cells had greater stemness potential and tumorigenicity than PD-L1Negative GC cells. The results also indicated that GC cells were heterogeneous, and that PD-L1 in GC cells had different reactivity to GCMSCs. Conclusions: Overall, our data indicated that GCMSCs enriched CSC-like cells in GC cells, which gives a new insight into the mechanism of GCMSCs prompting GC progression and provides a potential combined therapeutic target.


Assuntos
Antígeno B7-H1/metabolismo , Fator de Ligação a CCCTC/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Gástricas/imunologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/patologia , Linhagem Celular Tumoral , Autorrenovação Celular/imunologia , Meios de Cultivo Condicionados/metabolismo , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/imunologia , Mucosa Gástrica/citologia , Mucosa Gástrica/imunologia , Mucosa Gástrica/patologia , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Masculino , Camundongos , Células-Tronco Neoplásicas/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Invest Dermatol ; 140(12): 2433-2441.e5, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32311397

RESUMO

Mast cells (MCs) are tissue-resident hematopoietic cells intensely studied for their role as effectors in allergic immune responses. Yolk sac-derived embryonic MCs first populate tissues and are later replaced by definitive MCs. We show that definitive MC progenitors expand locally in skin and form clonal colonies that cover stable territories. In MC-deficient skin, colonies grow by proliferation of MCs at the border of the clonal territory. Clonal growth ceases at common borders of neighboring colonies. In steady state, colony self-renewal is independent of bone marrow contribution, and the clonal architecture remains fixed if not disturbed by skin inflammation. Inflammatory cues increase MC density setpoint, stimulating the influx of new progenitors from the bone marrow as well as proliferation of skin-resident cells. The expanding new arrivals disrespect territories of preexisting MC clones. We conclude that during a limited window early in development, definitive MC precursors efficiently enter the skin, expand, and self-maintain, occupying stable territories. In adulthood, circulating progenitors, excluded from steady-state skin, are recruited only into inflamed skin where they clonally expand alongside proliferating skin-resident MCs, disorganizing the original architecture of clonal territories.


Assuntos
Células-Tronco Adultas/fisiologia , Autorrenovação Celular/imunologia , Dermatite/imunologia , Mastócitos/imunologia , Pele/patologia , Animais , Medula Óssea , Células Cultivadas , Dermatite/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Células-Tronco Embrionárias/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mórula/citologia , Pele/citologia , Pele/imunologia , Acetato de Tetradecanoilforbol/imunologia
7.
Cancer Discov ; 10(3): 371-381, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31919052

RESUMO

Glioblastoma (GBM) is a lethal brain tumor containing a subpopulation of glioma stem cells (GSC). Pan-cancer analyses have revealed that stemness of cancer cells correlates positively with immunosuppressive pathways in many solid tumors, including GBM, prompting us to conduct a gain-of-function screen of epigenetic regulators that may influence GSC self-renewal and tumor immunity. The circadian regulator CLOCK emerged as a top hit in enhancing stem-cell self-renewal, which was amplified in about 5% of human GBM cases. CLOCK and its heterodimeric partner BMAL1 enhanced GSC self-renewal and triggered protumor immunity via transcriptional upregulation of OLFML3, a novel chemokine recruiting immune-suppressive microglia into the tumor microenvironment. In GBM models, CLOCK or OLFML3 depletion reduced intratumoral microglia density and extended overall survival. We conclude that the CLOCK-BMAL1 complex contributes to key GBM hallmarks of GSC maintenance and immunosuppression and, together with its downstream target OLFML3, represents new therapeutic targets for this disease. SIGNIFICANCE: Circadian regulator CLOCK drives GSC self-renewal and metabolism and promotes microglia infiltration through direct regulation of a novel microglia-attracting chemokine, OLFML3. CLOCK and/or OLFML3 may represent novel therapeutic targets for GBM.This article is highlighted in the In This Issue feature, p. 327.


Assuntos
Fatores de Transcrição ARNTL/genética , Proteínas CLOCK/genética , Glioblastoma/genética , Glicoproteínas/genética , Animais , Linhagem Celular Tumoral , Autorrenovação Celular/genética , Autorrenovação Celular/imunologia , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/terapia , Xenoenxertos , Humanos , Imunidade Celular/imunologia , Camundongos , Microglia/imunologia , Microglia/metabolismo , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
8.
Int J Cancer ; 146(2): 424-438, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31241171

RESUMO

Stem cell chemoresistance remains challenging the efficacy of the front-line temozolomide against glioblastoma. Novel therapies are urgently needed to fight those cells in order to control tumor relapse. Here, we report that anti-O-acetyl-GD2 adjuvant immunotherapy controls glioma stem-like cell-driven chemoresistance. Using patient-derived glioblastoma cells, we found that glioma stem-like cells overexpressed O-acetyl-GD2. As a result, monoclonal antibody 8B6 immunotherapy significantly increased temozolomide genotoxicity and tumor cell death in vitro by enhancing temozolomide tumor uptake. Furthermore, the combination therapy decreased the expression of the glioma stem-like cell markers CD133 and Nestin and compromised glioma stem-like cell self-renewal capabilities. When tested in vivo, adjuvant 8B6 immunotherapy prevented the extension of the temozolomide-resistant glioma stem-like cell pool within the tumor bulk in vivo and was more effective than the single agent therapies. This is the first report demonstrating that anti-O-acetyl-GD2 monoclonal antibody 8B6 targets glioblastoma in a manner that control temozolomide-resistance driven by glioma stem-like cells. Together our results offer a proof of concept for using anti-O-acetyl GD2 reagents in glioblastoma to develop more efficient combination therapies for malignant gliomas.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Gangliosídeos/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Autorrenovação Celular/efeitos dos fármacos , Autorrenovação Celular/imunologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/imunologia , Sinergismo Farmacológico , Gangliosídeos/imunologia , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Camundongos , Células-Tronco Neoplásicas/imunologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Stem Cells Dev ; 29(6): 327-335, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31856674

RESUMO

Head and neck squamous cell carcinomas (HNSCCs) are malignancies that originate in the mucosal lining of the upper aerodigestive tract. Despite advances in therapeutic interventions, survival rates among HNSCC patients have remained static for years. Cancer stem cells (CSCs) are tumor-initiating cells that are highly resistant to treatment, and are hypothesized to contribute to a significant fraction of tumor recurrences. Consequently, further investigations of how CSCs mediate recurrence may provide insights into novel druggable targets. A key element of recurrence involves the tumor's ability to evade immunosurveillance. Recent published reports suggest that CSCs possess immunosuppressive properties, however, the underlying mechanism have yet to be fully elucidated. To date, most groups have focused on the role of CSC-derived secretory proteins, such as cytokines and growth factors. Here, we review the established immunoregulatory role of exosomes derived from mixed tumor cell populations, and propose further study of CSC-derived exosomes may be warranted. Such studies may yield novel insights into new druggable targets, or lay the foundation for future exosome-based diagnostics.


Assuntos
Carcinoma de Células Escamosas/imunologia , Exossomos/imunologia , Neoplasias de Cabeça e Pescoço/imunologia , Células-Tronco Neoplásicas/imunologia , Carcinoma de Células Escamosas/patologia , Diferenciação Celular/imunologia , Autorrenovação Celular/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Recidiva Local de Neoplasia , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia
10.
J Exp Med ; 216(8): 1791-1808, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31164392

RESUMO

Chronic infection and cancer are associated with suppressed T cell responses in the presence of cognate antigen. Recent work identified memory-like CXCR5+ TCF1+ CD8+ T cells that sustain T cell responses during persistent infection and proliferate upon anti-PD1 treatment. Approaches to expand these cells are sought. We show that blockade of interferon type 1 (IFN-I) receptor leads to CXCR5+ CD8+ T cell expansion in an IL-27- and STAT1-dependent manner. IFNAR1 blockade promoted accelerated cell division and retention of TCF1 in virus-specific CD8+ T cells. We found that CD8+ T cell-intrinsic IL-27 signaling safeguards the ability of TCF1hi cells to maintain proliferation and avoid terminal differentiation or programmed cell death. Mechanistically, IL-27 endowed rapidly dividing cells with IRF1, a transcription factor that was required for sustained division in a cell-intrinsic manner. These findings reveal that IL-27 opposes IFN-I to uncouple effector differentiation from cell division and suggest that IL-27 signaling could be exploited to augment self-renewing T cells in chronic infections and cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Autorrenovação Celular/imunologia , Interleucinas/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Memória Imunológica , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interleucinas/genética , Coriomeningite Linfocítica/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptores CXCR5/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transcriptoma
11.
Arthritis Rheumatol ; 71(1): 133-142, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29984480

RESUMO

OBJECTIVE: A major characteristic of the autoimmune disease primary Sjögren's syndrome (SS) is salivary gland (SG) hypofunction. The inability of resident SG stem cells (SGSCs) to maintain homeostasis and saliva production has never been explained and limits our comprehension of mechanisms underlying primary SS. The present study was undertaken to investigate the role of salivary gland stem cells in hyposalivation in primary SS. METHODS: SGSCs were isolated from parotid biopsy samples from controls and patients classified as having primary SS or incomplete primary SS, according to the American College of Rheumatology/European League Against Rheumatism criteria. Self-renewal and differentiation assays were used to determine SGSC regenerative potential, RNA was extracted for sequencing analysis, single telomere length analysis was conducted to determine telomere length, and frozen tissue samples were used for immunohistochemical analysis. RESULTS: SGSCs isolated from primary SS parotid gland biopsy samples were regeneratively inferior to healthy control specimens. We demonstrated that SGSCs from samples from patients with primary SS are not only lower in number and less able to differentiate, but are likely to be senescent, as revealed by telomere length analysis, RNA sequencing, and immunostaining. We further found that SGSCs exposed to primary SS-associated proinflammatory cytokines we induced to proliferate, express senescence-associated genes, and subsequently differentiate into intercalated duct cells. We also localized p16+ senescent cells to the intercalated ducts in primary SS SG tissue, suggesting a block in SGSC differentiation into acinar cells. CONCLUSION: This study represents the first characterization of SGSCs in primary SS, and also the first demonstration of a linkage between an autoimmune disease and a parenchymal premature-aging phenotype. The knowledge garnered in this study indicates that disease-modifying antirheumatic drugs used to treat primary SS are not likely to restore saliva production, and should be supplemented with fresh SGSCs to recover saliva production.


Assuntos
Autorrenovação Celular/imunologia , Senescência Celular/imunologia , Glândula Parótida/imunologia , Síndrome de Sjogren/imunologia , Células-Tronco/imunologia , Estudos de Casos e Controles , Autorrenovação Celular/genética , Senescência Celular/genética , Citocinas/imunologia , Humanos , Imuno-Histoquímica , Glândula Parótida/citologia , Glândula Parótida/metabolismo , Glândulas Salivares , Análise de Sequência de RNA , Células-Tronco/metabolismo , Telômero/metabolismo
12.
Biomed Pharmacother ; 107: 1318-1325, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30257347

RESUMO

Colorectal carcinoma (CRC) is the third most common malignant tumor in the world. In recent years, the morbidity and mortality of CRC have increased in the world due to increasingly ageing population, modern dietary habits, environmental change, genetic disorders and chronic intestinal inflammation. Despite recent advances in earlier detection and improvements in chemotherapy, the 5-year survival rate of patients with metastatic CRC remains low. Therefore, novel effective treatment strategies for primary or metastatic CRC have emerged to enhance cure rate as well as elongation of patient's survival. Immunotherapy has been proposed for a potentially effective therapeutic approach to the treatment of CRC. Tumor vaccination in preclinical and clinical studies has supported the antitumor activity induced by immunization with CRC cell vaccines. Epithelial cell molecule Mucin 1 (MUC1), a transmembrane glycoprotein aberrantly overexpressed in various cancers including CRC, has been used as a candidate target antigen in the peptide, dendritic cell, and whole tumor vaccines. Several clinical trials in progress reveal the immunogenicity and suitability of MUC1 that acted as immunotherapeutic vaccines for CRC/colorectal cancer stem cells (CCSC). The present review summarizes the potential roles of MUC1 on CRC/CCSC vaccines according to the latest data. Moreover, this review also discusses the novel strategies for targeting CCSC via inducing an immune response against MUC1 to achieve the best prevention and treatment effects in animal models and clinical trails.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias Colorretais/metabolismo , Imunoterapia/métodos , Mucina-1/metabolismo , Proliferação de Células , Autorrenovação Celular/imunologia , Ensaios Clínicos como Assunto , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Humanos , Mucina-1/imunologia , Taxa de Sobrevida
13.
Mamm Genome ; 29(11-12): 771-776, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30182300

RESUMO

Emerging evidence indicates that thymocyte self-renewal induced by progenitor deprivation carries an oncogenic risk that is modulated by intra-thymic competition from differentiation-committed cells. Here we discuss formative studies demonstrating that, in mice, early thymocytes acquire self-renewing potential when thymic progenitor supply is sub-physiological and the importance of cellular competition with this at-risk cell population to prevent lymphoid malignancy. We also consider the possibility that increased thymic residency time, established under conditions of limited cellular competition, may have contributed to oncogenesis observed in early SCID-X1 trials when combined with insertional activation of proto-oncogenes such as LMO2.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinogênese/genética , Proteínas com Domínio LIM/genética , Neoplasias/imunologia , Timócitos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Carcinogênese/imunologia , Autorrenovação Celular/imunologia , Transformação Celular Neoplásica/imunologia , Modelos Animais de Doenças , Terapia Genética , Células-Tronco Hematopoéticas/imunologia , Humanos , Proteínas com Domínio LIM/imunologia , Camundongos , Neoplasias/genética , Células-Tronco Neoplásicas/imunologia
14.
Front Immunol ; 9: 1064, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29868023

RESUMO

Studies performed in animal models and in humans indicate that the innate arm of the immune system provides an essential role in the initial protection against potential insults and in maintaining tolerance to self-antigens. In the B cell compartment, several subsets engage in both adaptive and innate functions. Whereas B cell subsets are recognized to play important roles in autoimmune diseases, understanding the intricacies of their effector functions remains challenging. In addition to B-1a cells and marginal zone B cells, the B cell compartment comprises other B cells with innate-like functions, including innate response activator B cells, T-bet positive B cells, natural killer-like B cells, IL-17-producing B cells, and human self-reactive VH4-34-expressing B cells. Herein, we summarize the functions of recently described B cell populations that can exert innate-like roles in both animal models and humans. We also highlight the importance of the cross talk between innate-like B cells and other adaptive and innate branches of the immune system in various autoimmune and inflammatory diseases. In as much as innate immunity seems to be important in resolving inflammation, it is possible that targeting certain innate-like B cell subsets could represent a novel therapeutic approach for inducing resolution of inflammation of autoimmune and inflammatory responses.


Assuntos
Autoimunidade , Subpopulações de Linfócitos B/imunologia , Comunicação Celular/imunologia , Imunidade Inata , Animais , Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Autorrenovação Celular/imunologia , Quimiotaxia de Leucócito/imunologia , Humanos , Tolerância Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Transdução de Sinais , Proteínas com Domínio T/metabolismo
15.
Immunity ; 48(4): 730-744.e5, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29669251

RESUMO

Although characterization of T cell exhaustion has unlocked powerful immunotherapies, the mechanisms sustaining adaptations of short-lived innate cells to chronic inflammatory settings remain unknown. During murine chronic viral infection, we found that concerted events in bone marrow and spleen mediated by type I interferon (IFN-I) and Toll-like receptor 7 (TLR7) maintained a pool of functionally exhausted plasmacytoid dendritic cells (pDCs). In the bone marrow, IFN-I compromised the number and the developmental capacity of pDC progenitors, which generated dysfunctional pDCs. Concurrently, exhausted pDCs in the periphery were maintained by self-renewal via IFN-I- and TLR7-induced proliferation of CD4- subsets. On the other hand, pDC functional loss was mediated by TLR7, leading to compromised IFN-I production and resistance to secondary infection. These findings unveil the mechanisms sustaining a self-perpetuating pool of functionally exhausted pDCs and provide a framework for deciphering long-term exhaustion of other short-lived innate cells during chronic inflammation.


Assuntos
Autorrenovação Celular/imunologia , Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Células 3T3 , Animais , Proteínas de Transporte/biossíntese , Linhagem Celular , Proliferação de Células , Proteínas de Ligação a DNA/biossíntese , Células Dendríticas/citologia , Humanos , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/biossíntese , Proteínas Repressoras , Transdução de Sinais/imunologia , Fator de Transcrição 4/biossíntese , Fatores de Transcrição/biossíntese
16.
Cell Stem Cell ; 20(4): 450-461.e4, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28089910

RESUMO

Tumors contain hostile inflammatory signals generated by aberrant proliferation, necrosis, and hypoxia. These signals are sensed and acted upon acutely by the Toll-like receptors (TLRs) to halt proliferation and activate an immune response. Despite the presence of TLR ligands within the microenvironment, tumors progress, and the mechanisms that permit this growth remain largely unknown. We report that self-renewing cancer stem cells (CSCs) in glioblastoma have low TLR4 expression that allows them to survive by disregarding inflammatory signals. Non-CSCs express high levels of TLR4 and respond to ligands. TLR4 signaling suppresses CSC properties by reducing retinoblastoma binding protein 5 (RBBP5), which is elevated in CSCs. RBBP5 activates core stem cell transcription factors, is necessary and sufficient for self-renewal, and is suppressed by TLR4 overexpression in CSCs. Our findings provide a mechanism through which CSCs persist in hostile environments because of an inability to respond to inflammatory signals.


Assuntos
Autorrenovação Celular/imunologia , Glioblastoma/imunologia , Glioblastoma/patologia , Evasão da Resposta Imune , Imunidade Inata , Células-Tronco Neoplásicas/patologia , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação a DNA , Feminino , Humanos , Camundongos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
17.
Nat Med ; 23(1): 18-27, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-28060797

RESUMO

T memory stem (TSCM) cells are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector T cell subsets. Cumulative evidence in mice, nonhuman primates and humans indicates that TSCM cells are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM cells, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM cell stemness could be leveraged therapeutically to enhance the efficacy of vaccines and adoptive T cell therapies for cancer and infectious diseases or, conversely, how it could be disrupted to treat TSCM cell driven and sustained diseases, such as autoimmunity, adult T cell leukemia and HIV-1.


Assuntos
Autorrenovação Celular/imunologia , Memória Imunológica/imunologia , Células-Tronco/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Diferenciação Celular , Infecções por HIV/imunologia , Infecções por HIV/terapia , Humanos , Leucemia de Células T/imunologia , Leucemia de Células T/terapia , Linfócitos T/transplante
18.
Clin Cancer Res ; 23(10): 2516-2527, 2017 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-27780858

RESUMO

Purpose: Locoregional recurrence is a frequent treatment outcome for patients with advanced head and neck squamous cell carcinoma (HNSCC). Emerging evidence suggests that tumor recurrence is mediated by a small subpopulation of uniquely tumorigenic cells, that is, cancer stem cells (CSC), that are resistant to conventional chemotherapy, endowed with self-renewal and multipotency.Experimental Design: Here, we evaluated the efficacy of MEDI0641, a novel antibody-drug conjugate targeted to 5T4 and carrying a DNA-damaging "payload" (pyrrolobenzodiazepine) in preclinical models of HNSCC.Results: Analysis of a tissue microarray containing 77 HNSCC with follow-up of up to 12 years revealed that patients with 5T4high tumors displayed lower overall survival than those with 5T4low tumors (P = 0.038). 5T4 is more highly expressed in head and neck CSC (ALDHhighCD44high) than in control cells (non-CSC). Treatment with MEDI0641 caused a significant reduction in the CSC fraction in HNSCC cells (UM-SCC-11B, UM-SCC-22B) in vitro Notably, a single intravenous dose of 1 mg/kg MEDI0641 caused long-lasting tumor regression in three patient-derived xenograft (PDX) models of HNSCC. MEDI0641 ablated CSC in the PDX-SCC-M0 model, reduced it by five-fold in the PDX-SCC-M1, and two-fold in the PDX-SCC-M11 model. Importantly, mice (n = 12) treated with neoadjuvant, single administration of MEDI0641 prior to surgical tumor removal showed no recurrence for more than 200 days, whereas the control group had 7 recurrences (in 12 mice; P = 0.0047).Conclusions: Collectively, these findings demonstrate that an anti-5T4 antibody-drug conjugate reduces the fraction of CSCs and prevents local recurrence and suggest a novel therapeutic approach for patients with HNSCC. Clin Cancer Res; 23(10); 2516-27. ©2016 AACR.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Imunoconjugados/administração & dosagem , Glicoproteínas de Membrana/imunologia , Animais , Benzodiazepinas/administração & dosagem , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Autorrenovação Celular/genética , Autorrenovação Celular/imunologia , Dano ao DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imunoconjugados/imunologia , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Pirróis/administração & dosagem , Carcinoma de Células Escamosas de Cabeça e Pescoço , Análise Serial de Tecidos , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Nat Med ; 22(11): 1285-1293, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27694932

RESUMO

Successful recovery from lung injury requires the repair and regeneration of alveolar epithelial cells to restore the integrity of gas-exchanging regions within the lung and preserve organ function. Improper regeneration of the alveolar epithelium is often associated with severe pulmonary fibrosis, the latter of which involves the recruitment and activation of fibroblasts, as well as matrix accumulation. Type 2 alveolar epithelial cells (AEC2s) are stem cells in the adult lung that contribute to the lung repair process. The mechanisms that regulate AEC2 renewal are incompletely understood. We provide evidence that expression of the innate immune receptor Toll-like receptor 4 (TLR4) and the extracellular matrix glycosaminoglycan hyaluronan (HA) on AEC2s are important for AEC2 renewal, repair of lung injury and limiting the extent of fibrosis. Either deletion of TLR4 or HA synthase 2 in surfactant-protein-C-positive AEC2s leads to impaired renewal capacity, severe fibrosis and mortality. Furthermore, AEC2s from patients with severe pulmonary fibrosis have reduced cell surface HA and impaired renewal capacity, suggesting that HA and TLR4 are key contributors to lung stem cell renewal and that severe pulmonary fibrosis is the result of distal epithelial stem cell failure.


Assuntos
Células Epiteliais Alveolares/imunologia , Autorrenovação Celular/imunologia , Ácido Hialurônico/metabolismo , Lesão Pulmonar/imunologia , Fibrose Pulmonar/imunologia , Proteína C Associada a Surfactante Pulmonar/metabolismo , Células-Tronco/imunologia , Receptor 4 Toll-Like/imunologia , Células Epiteliais Alveolares/metabolismo , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Linhagem Celular , Proliferação de Células , Autorrenovação Celular/genética , Citometria de Fluxo , Glucuronosiltransferase/genética , Humanos , Hialuronan Sintases , Hidroxiprolina/metabolismo , Fibrose Pulmonar Idiopática/imunologia , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Imunidade Inata , Interleucina-6/imunologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Camundongos , Camundongos Knockout , Organoides , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Células-Tronco/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
20.
J Immunol ; 196(10): 4052-63, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27076678

RESUMO

Tuberculosis-associated immune reconstitution inflammatory syndrome (TB-IRIS) complicates combination antiretroviral therapy (cART) in up to 25% of patients with HIV/TB coinfection. Monocytes and IL-18, a signature cytokine of inflammasome activation, are implicated in TB-IRIS pathogenesis. In this study, we investigated inflammasome activation both pre- and post-cART in TB-IRIS patients. HIV/TB patients exhibited higher proportions of monocytes expressing activated caspase-1 (casp1) pre-cART, compared with HIV patients without TB, and patients who developed TB-IRIS exhibited the greatest increase in casp1 expression. CD64(+) monocytes were a marker of increased casp1 expression. Furthermore, IL-1ß, another marker of inflammasome activation, was also elevated during TB-IRIS. TB-IRIS patients also exhibited greater upregulation of NLRP3 and AIM2 inflammasome mRNA, compared with controls. Analysis of plasma mitochondrial DNA levels showed that TB-IRIS patients experienced greater cell death, especially pre-cART. Plasma NO levels were lower both pre- and post-cART in TB-IRIS patients, providing evidence of inadequate inflammasome regulation. Plasma IL-18 levels pre-cART correlated inversely with NO levels but positively with monocyte casp1 expression and mitochondrial DNA levels, and expression of IL-18Rα on CD4(+) T cells and NK cells was higher in TB-IRIS patients, providing evidence that IL-18 is a marker of inflammasome activation. We propose that inflammasome activation in monocytes/macrophages of HIV/TB patients increases with ineffective T cell-dependent activation of monocytes/macrophages, priming them for an excessive inflammatory response after cART is commenced, which is greatest in patients with TB-IRIS.


Assuntos
Autorrenovação Celular/imunologia , Infecções por HIV/imunologia , Inflamassomos/metabolismo , Inflamação/imunologia , Macrófagos/fisiologia , Tuberculose/imunologia , Antirretrovirais/uso terapêutico , Caspase 1/metabolismo , Morte Celular , Células Cultivadas , DNA Mitocondrial/sangue , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Inflamação/complicações , Inflamação/tratamento farmacológico , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Óxido Nítrico/sangue , Síndrome , Tuberculose/complicações , Tuberculose/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA