Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 53(2): 417-428.e4, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32735844

RESUMO

Psychological stress has adverse effects on various human diseases, including those of the cardiovascular system. However, the mechanisms by which stress influences disease activity remain unclear. Here, using vaso-occlusive episodes (VOEs) of sickle cell disease as a vascular disease model, we show that stress promotes VOEs by eliciting a glucocorticoid hormonal response that augments gut permeability, leading to microbiota-dependent interleukin-17A (IL-17A) secretion from T helper 17 (Th17) cells of the lamina propria, followed by the expansion of the circulating pool of aged neutrophils that trigger VOEs. We identify segmented filamentous bacteria as the commensal essential for the stress-induced expansion of aged neutrophils that enhance VOEs in mice. Importantly, the inhibition of glucocorticoids synthesis, blockade of IL-17A, or depletion of the Th17 cell-inducing gut microbiota markedly reduces stress-induced VOEs. These results offer potential therapeutic targets to limit the impact of psychological stress on acute vascular occlusion.


Assuntos
Anemia Falciforme/patologia , Microbioma Gastrointestinal/imunologia , Interleucina-17/metabolismo , Estresse Psicológico/patologia , Células Th17/imunologia , Anemia Falciforme/psicologia , Animais , Bactérias/imunologia , Linhagem Celular , Vida Livre de Germes , Glucocorticoides/biossíntese , Fator Estimulador de Colônias de Granulócitos/metabolismo , Células HEK293 , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Inflamação/imunologia , Inflamação/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia
2.
Nature ; 607(7919): 578-584, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35636458

RESUMO

The nervous and immune systems are intricately linked1. Although psychological stress is known to modulate immune function, mechanistic pathways linking stress networks in the brain to peripheral leukocytes remain poorly understood2. Here we show that distinct brain regions shape leukocyte distribution and function throughout the body during acute stress in mice. Using optogenetics and chemogenetics, we demonstrate that motor circuits induce rapid neutrophil mobilization from the bone marrow to peripheral tissues through skeletal-muscle-derived neutrophil-attracting chemokines. Conversely, the paraventricular hypothalamus controls monocyte and lymphocyte egress from secondary lymphoid organs and blood to the bone marrow through direct, cell-intrinsic glucocorticoid signalling. These stress-induced, counter-directional, population-wide leukocyte shifts are associated with altered disease susceptibility. On the one hand, acute stress changes innate immunity by reprogramming neutrophils and directing their recruitment to sites of injury. On the other hand, corticotropin-releasing hormone neuron-mediated leukocyte shifts protect against the acquisition of autoimmunity, but impair immunity to SARS-CoV-2 and influenza infection. Collectively, these data show that distinct brain regions differentially and rapidly tailor the leukocyte landscape during psychological stress, therefore calibrating the ability of the immune system to respond to physical threats.


Assuntos
Encéfalo , Medo , Leucócitos , Neurônios Motores , Vias Neurais , Estresse Psicológico , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Encéfalo/citologia , Encéfalo/fisiologia , COVID-19/imunologia , Quimiocinas/imunologia , Suscetibilidade a Doenças , Medo/fisiologia , Glucocorticoides/metabolismo , Humanos , Leucócitos/citologia , Leucócitos/imunologia , Linfócitos/citologia , Linfócitos/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Camundongos , Monócitos/citologia , Monócitos/imunologia , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Neutrófilos/citologia , Neutrófilos/imunologia , Optogenética , Infecções por Orthomyxoviridae/imunologia , Núcleo Hipotalâmico Paraventricular/fisiologia , SARS-CoV-2/imunologia , Estresse Psicológico/imunologia , Estresse Psicológico/fisiopatologia
3.
Nature ; 589(7843): 591-596, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33361809

RESUMO

Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.


Assuntos
Vias Autônomas , Movimento Celular , Células-Tronco Hematopoéticas/citologia , Nociceptividade/fisiologia , Nociceptores/fisiologia , Sistema Nervoso Simpático/citologia , Adenilil Ciclases/metabolismo , Animais , Vias Autônomas/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Capsaicina/farmacologia , Movimento Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nociceptividade/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco , Sistema Nervoso Simpático/efeitos dos fármacos
4.
Development ; 145(2)2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29358215

RESUMO

Hematopoietic stem cells (HSCs) develop in discrete anatomical niches, migrating during embryogenesis from the aorta-gonad-mesonephros (AGM) region to the fetal liver, and finally to the bone marrow, where most HSCs reside throughout adult life. These niches provide supportive microenvironments that specify, expand and maintain HSCs. Understanding the constituents and molecular regulation of HSC niches is of considerable importance as it could shed new light on the mechanistic principles of HSC emergence and maintenance, and provide novel strategies for regenerative medicine. However, controversy exists concerning the cellular complexity of the bone marrow niche, and our understanding of the different HSC niches during development remains limited. In this Review, we summarize and discuss what is known about the heterogeneity of the HSC niches at distinct stages of their ontogeny, from the embryo to the adult bone marrow, drawing predominantly on data from mouse studies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Aorta/embriologia , Linhagem da Célula , Feminino , Gônadas/embriologia , Neoplasias Hematológicas/patologia , Sistema Hematopoético/embriologia , Humanos , Masculino , Mesonefro/embriologia , Camundongos , Placenta/citologia , Placenta/fisiologia , Gravidez , Células Estromais/citologia , Células Estromais/fisiologia , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia
5.
Nature ; 525(7570): 528-32, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26374999

RESUMO

Blood polymorphonuclear neutrophils provide immune protection against pathogens, but may also promote tissue injury in inflammatory diseases. Although neutrophils are generally considered to be a relatively homogeneous population, evidence for heterogeneity is emerging. Under steady-state conditions, neutrophil heterogeneity may arise from ageing and replenishment by newly released neutrophils from the bone marrow. Aged neutrophils upregulate CXCR4, a receptor allowing their clearance in the bone marrow, with feedback inhibition of neutrophil production via the IL-17/G-CSF axis, and rhythmic modulation of the haematopoietic stem-cell niche. The aged subset also expresses low levels of L-selectin. Previous studies have suggested that in vitro-aged neutrophils exhibit impaired migration and reduced pro-inflammatory properties. Here, using in vivo ageing analyses in mice, we show that neutrophil pro-inflammatory activity correlates positively with their ageing whilst in circulation. Aged neutrophils represent an overly active subset exhibiting enhanced αMß2 integrin activation and neutrophil extracellular trap formation under inflammatory conditions. Neutrophil ageing is driven by the microbiota via Toll-like receptor and myeloid differentiation factor 88-mediated signalling pathways. Depletion of the microbiota significantly reduces the number of circulating aged neutrophils and dramatically improves the pathogenesis and inflammation-related organ damage in models of sickle-cell disease or endotoxin-induced septic shock. These results identify a role for the microbiota in regulating a disease-promoting neutrophil subset.


Assuntos
Senescência Celular/imunologia , Microbiota/imunologia , Neutrófilos/citologia , Neutrófilos/imunologia , Anemia Falciforme/sangue , Anemia Falciforme/microbiologia , Anemia Falciforme/patologia , Animais , Modelos Animais de Doenças , Eritrócitos Anormais/patologia , Inflamação/imunologia , Inflamação/patologia , Antígeno de Macrófago 1/metabolismo , Masculino , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Choque Séptico/imunologia , Choque Séptico/microbiologia , Choque Séptico/patologia , Transdução de Sinais , Receptores Toll-Like/imunologia
6.
Blood ; 127(7): 801-9, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26758915

RESUMO

Sickle cell disease (SCD) is a severe genetic blood disorder characterized by hemolytic anemia, episodic vaso-occlusion, and progressive organ damage. Current management of the disease remains symptomatic or preventative. Specific treatment targeting major complications such as vaso-occlusion is still lacking. Recent studies have identified various cellular and molecular factors that contribute to the pathophysiology of SCD. Here, we review the role of these elements and discuss the opportunities for therapeutic intervention.


Assuntos
Anemia Falciforme/sangue , Anemia Falciforme/terapia , Plaquetas/metabolismo , Neutrófilos/metabolismo , Anemia Falciforme/patologia , Plaquetas/patologia , Humanos , Inflamação/sangue , Inflamação/patologia , Inflamação/terapia , Neutrófilos/patologia
7.
Proc Natl Acad Sci U S A ; 112(52): E7239-48, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26669445

RESUMO

Obesity-associated inflammation is accompanied by the accumulation of adipose tissue macrophages (ATMs), which is believed to predispose obese individuals to insulin resistance. CD11b (integrin αM) is highly expressed on monocytes and macrophages and is critical for their migration and function. We found here that high-fat diet-induced insulin resistance was significantly reduced in CD11b-deficient mice. Interestingly, the recruitment of monocytes to adipose tissue is impaired when CD11b is deficient, although the cellularity of ATMs in CD11b-deficient mice is higher than that in wild-type mice. We further found that the increase in ATMs is caused mainly by their vigorous proliferation in the absence of CD11b. Moreover, the proliferation and alternative activation of ATMs are regulated by the IL-4/STAT6 axis, which is inhibited by CD11b through the activity of phosphatase SHP-1. Thus, CD11b plays a critical role in obesity-induced insulin resistance by limiting the proliferation and alternative activation of ATMs.


Assuntos
Antígeno CD11b/genética , Proliferação de Células/genética , Resistência à Insulina/genética , Macrófagos/metabolismo , Obesidade/genética , Tecido Adiposo/metabolismo , Animais , Antígeno CD11b/metabolismo , Citometria de Fluxo , Expressão Gênica , Immunoblotting , Interleucina-4/metabolismo , Ativação de Macrófagos/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Obesidade/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT6/metabolismo
8.
J Immunol ; 192(1): 103-9, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24293629

RESUMO

Mesenchymal stem cells (MSCs) possess potent immunosuppression capacity and could exert strong therapeutic effects in many diseases, especially inflammatory disorders, in animal models and clinical settings. Although inflammatory cytokines are critical in inducing the immune modulatory properties of MSCs, detailed molecular mechanisms are yet to be fully understood. TGF-ß is a well-known anti-inflammatory cytokine and exists in various inflammatory processes; therefore, we investigated whether it could synergize with MSCs in suppressing immune responses. To our surprise, we found that TGF-ß actually reversed the immunosuppressive effect of MSCs on anti-CD3 activated splenocytes. Using TGF-ß unresponsive MSCs, we demonstrated that the TGF-ß directly acted on MSCs. Furthermore, we showed that the effect of TGF-ß is exerted through inhibiting inflammatory cytokines induced inducible NO synthase (iNOS) expression in a SMAD3-dependent manner. Interestingly, we found that TGF-ß produced by MSCs could act in an autocrine manner to reduce inflammatory cytokine-induced inducible NO synthase expression by MSCs themselves. Therefore, our study revealed a previously unrecognized property of TGF-ß in promoting immune responses in the presence of MSCs.


Assuntos
Tolerância Imunológica/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Comunicação Autócrina/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Tolerância Imunológica/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Proteína Smad3/metabolismo , Transcrição Gênica
9.
Stem Cells ; 32(2): 327-37, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24123709

RESUMO

An imbalance between normal adipogenesis and osteogenesis by mesenchymal stem cells (MSCs) has been shown to be related to various human metabolic diseases, such as obesity and osteoporosis; however, the underlying mechanisms remain elusive. We found that the interaction between osteopontin (OPN), an arginine-glycine-aspartate-containing glycoprotein, and integrin αv/ß1 plays a critical role in the lineage determination of MSCs. Although OPN is a well-established marker during osteogenesis, its role in MSC differentiation is still unknown. Our study reveals that blockade of OPN function promoted robust adipogenic differentiation, while inhibiting osteogenic differentiation. Re-expression of OPN restored a normal balance between adipogenesis and osteogenesis in OPN(-/-) MSCs. Retarded bone formation by OPN(-/-) MSCs was also verified by in vivo implantation with hydroxyapatite-tricalcium phosphate, a bone-forming matrix. The role of extracellular OPN in MSC differentiation was further demonstrated by supplementation and neutralization of OPN. Blocking well-known OPN receptors integrin αv/ß1 but not CD44 also affected MSC differentiation. Further studies revealed that OPN inhibits the C/EBPs signaling pathway through integrin αv/ß1. Consistent with these in vitro results, OPN(-/-) mice had a higher fat to total body weight ratio than did wild-type mice. Therefore, our study demonstrates a novel role for OPN-integrin αv/ß1 in regulating MSC differentiation.


Assuntos
Adipogenia/genética , Osteogênese/genética , Osteopontina/metabolismo , Receptores de Vitronectina/metabolismo , Adipócitos/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Humanos , Células-Tronco Mesenquimais , Camundongos , Osteoblastos/metabolismo , Osteopontina/genética , Mapas de Interação de Proteínas/genética , Receptores de Vitronectina/genética
10.
J Biol Chem ; 288(16): 11074-9, 2013 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-23449975

RESUMO

MSCs possess potent immunosuppressive capacity. We have reported that mouse MSCs inhibit T cell proliferation and function via nitric oxide. This immune regulatory capacity of MSCs is induced by the inflammatory cytokines IFNγ together with either TNFα or IL-1ß. This effect of inflammatory cytokines on MSCs is extraordinary; logarithmic increases in the expression of iNOS and chemokines are often observed. To investigate the molecular mechanisms underlying this robust effect of cytokines, we examined the expression of microRNAs in MSCs before and after cytokine treatment. We found that miR-155 is most significantly up-regulated. Furthermore, our results showed that miR-155 inhibits the immunosuppressive capacity of MSCs by reducing iNOS expression. We further demonstrated that miR-155 targets TAK1-binding protein 2 (TAB2) to regulate iNOS expression. Additionally, knockdown of TAB2 reduced iNOS expression. In summary, our study demonstrated that miR-155 inhibits the immunosuppressive capacity of MSCs by reducing iNOS expression by targeting TAB2. Our data revealed a novel role of miR-155 in regulating the immune modulatory activities of MSCs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Tolerância Imunológica/fisiologia , Células-Tronco Mesenquimais/imunologia , MicroRNAs/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Regulação Enzimológica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Brain Behav Immun ; 38: 125-32, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24480719

RESUMO

Chronic stress has dramatic impacts on the immune system and consequently contributes to the onset and progression of a variety of diseases, including cancer, immune disorders, and infections. Recent studies in animals and humans have demonstrated that mesenchymal stem cells (MSCs) significantly modulate the immune system. Here we show that administration of MSCs in vivo prevents lymphocyte depletion induced by physical restraint stress (12:12-h stress-rest, 2 repetitions) in mice. This effect was found to be exerted not through modulation of glucocorticoid levels in the circulation, but rather through direct effects on lymphocyte apoptosis. By testing various possible protective mechanisms, we found that IL-4 provides a strong anti-apoptosis signal to lymphocytes in the presence of dexamethasone. When neutralizing antibody against IL-4 was co-administered with MSCs to restraint-stressed mice, the protective effect of MSCs was diminished. Furthermore, in mice deficient in STAT6, a key molecule in IL-4 receptor-mediated signaling, MSCs had no effect on restraint stress-induced lymphocyte depletion. Additionally, MSCs administered to stressed mice promoted IL-4 production by splenocytes. This study reveals that MSCs can effectively prevent stress-induced lymphocyte apoptosis in an IL-4-dependent manner and provides novel information for the development of countermeasures against the deleterious effects of stress on the immune system.


Assuntos
Interleucina-4/metabolismo , Linfócitos/metabolismo , Transplante de Células-Tronco Mesenquimais , Estresse Psicológico/imunologia , Animais , Apoptose , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Restrição Física , Baço/metabolismo
12.
bioRxiv ; 2023 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-37961493

RESUMO

Haematopoietic stem cells (HSCs) reside in specialized microenvironments, also referred to as niches, and it has been widely believed that HSC numbers are determined by the niche size alone 1-5 . However, the vast excess of the number of niche cells over that of HSCs raises questions about this model. We initially established a mathematical model of niche availability and occupancy, which predicted that HSC numbers are restricted at both systemic and local levels. To address this question experimentally, we developed a femoral bone transplantation system, enabling us to increase the number of available HSC niches. We found that the addition of niches does not alter total HSC numbers in the body, regardless of whether the endogenous (host) niche is intact or defective, suggesting that HSC numbers are limited at the systemic level. Additionally, HSC numbers in transplanted wild-type femurs did not increase beyond physiological levels when HSCs were mobilized from defective endogenous niches to the periphery, indicating that HSC numbers are also constrained at the local level. Our study demonstrates that HSC numbers are not solely determined by niche availability, thereby rewriting the long-standing model for the regulation of HSC numbers.

13.
Cell Stem Cell ; 29(2): 232-247.e7, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35065706

RESUMO

Host microbiota crosstalk is essential for the production and functional modulation of blood-cell lineages. Whether, and if so how, the microbiota influences hematopoietic stem cells (HSCs) is unclear. Here, we show that the microbiota regulates HSC self-renewal and differentiation under stress conditions by modulating local iron availability in the bone marrow (BM). In microbiota-depleted mice, HSC self-renewal was enhanced during regeneration, while the commitment toward differentiation was dramatically compromised. Mechanistically, microbiota depletion selectively impaired the recycling of red blood cells (RBCs) by BM macrophages, resulting in reduced local iron levels without affecting systemic iron homeostasis. Limiting iron availability in food (in vivo) or in culture (ex vivo), or by CD169+ macrophage depletion, enhanced HSC self-renewal and expansion. These results reveal an intricate interplay between the microbiota, macrophages, and iron, and their essential roles in regulating critical HSC fate decisions under stress.


Assuntos
Medula Óssea , Microbiota , Animais , Medula Óssea/fisiologia , Diferenciação Celular , Células-Tronco Hematopoéticas , Ferro , Camundongos
14.
Oncogene ; 41(13): 1866-1881, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35145233

RESUMO

Cancer treatments have been revolutionized by the emergence of immune checkpoint blockade therapies. However, only a minority of patients with various tumor types have benefited from such treatments. New strategies focusing on the immune contexture of the tumor tissue microenvironment hold great promises. Here, we created IFNα-overexpressing mesenchymal stromal cells (IFNα-MSCs). Upon direct injection into tumors, we found that these cells are powerful in eliminating several types of tumors. Interestingly, the intra-tumoral injection of IFNα-MSCs could also induce specific anti-tumor effects on distant tumors. These IFNα-MSCs promoted tumor cells to produce CXCL10, which in turn potentiates the infiltration of CD8+ T cells in the tumor site. Furthermore, IFNα-MSCs enhanced the expression of granzyme B (GZMB) in CD8+ T cells and invigorated their cytotoxicity in a Stat3-dependent manner. Genetic ablation of Stat3 in CD8+ T cells impaired the effect of IFNα-MSCs on GZMB expression. Importantly, the combination of IFNα-MSCs and PD-L1 blockade induced an even stronger anti-tumor immunity. Therefore, IFNα-MSCs represent a novel tumor immunotherapy strategy, especially when combined with PD-L1 blockade.


Assuntos
Células-Tronco Mesenquimais , Neoplasias , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Humanos , Imunoterapia , Interferon-alfa , Células-Tronco Mesenquimais/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Microambiente Tumoral
15.
J Exp Med ; 218(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33045060

RESUMO

Sickle cell disease (SCD) is a common hereditary hematologic disorder. SCD patients suffer from acute vaso-occlusive episodes (VOEs), chronic organ damage, and premature death, with few therapeutic options. Although severe pain is a major clinical manifestation of SCD, it remains unknown whether nociception plays a role in SCD pathogenesis. To address this question, we generated nociceptor-deficient SCD mice and found, unexpectedly, that the absence of nociception led to more severe and more lethal VOE, indicating that somatosensory nerves protect SCD mice from VOE. Mechanistically, the beneficial effects of sensory nerves were induced by the neuropeptide calcitonin gene-related peptide (CGRP), which acted on hematopoietic cells. Additionally, oral capsaicin consumption, which can activate somatosensory nerves by binding to TRPV1, dramatically alleviated acute VOE and significantly prevented chronic liver and kidney damage in SCD mice. Thus, the manipulation of nociception may provide a promising approach to treat SCD.


Assuntos
Anemia Falciforme , Capsaicina/farmacologia , Nociceptores/metabolismo , Células Receptoras Sensoriais/metabolismo , Doenças Vasculares , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Humanos , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/prevenção & controle , Camundongos , Camundongos Knockout , Dor/tratamento farmacológico , Dor/genética , Dor/metabolismo , Dor/patologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/prevenção & controle , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/genética , Doenças Vasculares/metabolismo
16.
Nat Commun ; 12(1): 2522, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33947846

RESUMO

Haematopoietic stem cells (HSCs) tightly regulate their quiescence, proliferation, and differentiation to generate blood cells during the entire lifetime. The mechanisms by which these critical activities are balanced are still unclear. Here, we report that Macrophage-Erythroblast Attacher (MAEA, also known as EMP), a receptor thus far only identified in erythroblastic island, is a membrane-associated E3 ubiquitin ligase subunit essential for HSC maintenance and lymphoid potential. Maea is highly expressed in HSCs and its deletion in mice severely impairs HSC quiescence and leads to a lethal myeloproliferative syndrome. Mechanistically, we have found that the surface expression of several haematopoietic cytokine receptors (e.g. MPL, FLT3) is stabilised in the absence of Maea, thereby prolonging their intracellular signalling. This is associated with impaired autophagy flux in HSCs but not in mature haematopoietic cells. Administration of receptor kinase inhibitor or autophagy-inducing compounds rescues the functional defects of Maea-deficient HSCs. Our results suggest that MAEA provides E3 ubiquitin ligase activity, guarding HSC function by restricting cytokine receptor signalling via autophagy.


Assuntos
Autofagossomos/genética , Autofagia/genética , Moléculas de Adesão Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Proteínas do Citoesqueleto/genética , Perfilação da Expressão Gênica , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Estabilidade Proteica , Receptores de Trombopoetina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Tirosina Quinase 3 Semelhante a fms/metabolismo
17.
J Vis Exp ; (176)2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34747411

RESUMO

Type 1 diabetes mellitus (T1DM) is caused by autoimmune destruction of pancreatic ß cells, which results in little or no insulin production. Islet transplantation plays an important role in the treatment of T1DM, with the improved glycometabolic control, the reduced progression of complications, the reduction of hypoglycemic episodes when compared with traditional insulin therapy. The results of phase III clinical trial also demonstrated the safety and efficacy of islet allotransplantation in T1DM. However, the shortage of pancreas donors limits its widespread use. Animals as a source of islets such as the pig offer an alternative choice. Because the architecture of the pig pancreas is different from the islets of mice or humans, the pig islet isolation procedure is still challenging. Since the translation of alternative porcine islet sources (xenogeneic) to the clinical setting for treating T1DM through cellular transplantation is of great importance, a cost-effective, standardized, and reproducible protocol for isolating porcine islets is urgently needed. This manuscript describes a simplified and cost-effective method to isolate and purify adult porcine islets based on the previous protocols that have successfully transplanted porcine islets to non-human primates. This will be a beginners guide without the use of specialized equipment such as a COBE 2991 Cell Processor.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Diabetes Mellitus Tipo 1/cirurgia , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Pâncreas , Suínos , Transplante Heterólogo/métodos
18.
Nat Prod Res ; 34(2): 197-203, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30856343

RESUMO

Two new norneolignans, (7S,8R)-3-methoxy-3',4,9-trihydroxy-4',7-epoxy-8,3'-neolignane-1'-carboxylic acid (1) and (7R,8R)-3-methoxyl-4,9-dihydroxy-3':7,4':8-diepoxyneolignan-1'-carboxylic acid methyl ester (2) were isolated from Callicarpa kwangtungensis, together with ten known compounds, genistin (3), daidzin (4), silybin A (5), isosilybin A (6), isosilybin B (7), p-hydroxybenzaldehyde (8), syringic acid (9), lanceolatin A (10), icariside C5 (11), and (3S,6E,10R)-10-ß-D-glucopyranosyloxy-3,11-dihydroxy-3,7,11-trimethyldodeca-1,6-diene (12). Compounds 1 and 2 were evaluated for their effects on the inhibition of nitric oxide (NO) production in lipopolysaccharide induced RAW264.7 cells. Compounds 1 and 2 exhibited inhibitory activity with IC50 values of 31.45 ± 0.38 and 40.72 ± 0.54 µM, respectively.


Assuntos
Callicarpa/química , Lignanas/isolamento & purificação , Óxido Nítrico/antagonistas & inibidores , Animais , Concentração Inibidora 50 , Lignanas/análise , Lignanas/química , Lignanas/farmacologia , Lipopolissacarídeos , Macrófagos/metabolismo , Camundongos , Estrutura Molecular , Óxido Nítrico/biossíntese , Células RAW 264.7
19.
Complement Ther Med ; 52: 102481, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32951731

RESUMO

Sickle Cell Disease (SCD) is a chronic hemolytic disorder associated with frequent pain episodes, end organ damage and a shortened lifespan. Currently there exist no disease specific targeted therapies for the treatment of acute vaso-occlusive crisis (VOC) and management with analgesics and hydration is purely supportive. Improvement in understanding of disease pathophysiology has resulted in a great interest in disease modifying novel therapies and many are being evaluated in clinical trials. Here we report the results from the pre-specified mid-point analysis of the Phase 2 study of Intravenous Gamma Globulin (IVIG) for the treatment of acute VOC in patients with SCD and lessons learned.


Assuntos
Anemia Falciforme/tratamento farmacológico , Imunoglobulinas Intravenosas/uso terapêutico , Manejo da Dor/métodos , gama-Globulinas/uso terapêutico , Adolescente , Adulto , Anemia Falciforme/complicações , Criança , Método Duplo-Cego , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Masculino , Adulto Jovem
20.
Dev Cell ; 53(5): 503-513.e5, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32413329

RESUMO

Bone marrow (BM) mesenchymal stem and progenitor cells (MSPCs) are a critical constituent of the hematopoietic stem cell (HSC) niche. Previous studies have suggested that the zinc-finger epithelial-mesenchymal transition transcription factor Snai2 (also known as Slug) regulated HSCs autonomously. Here, we show that Snai2 expression in the BM is restricted to the BM stromal compartment where it regulates the HSC niche. Germline or MSPC-selective Snai2 deletion reduces the functional MSPC pool and their mesenchymal lineage output and impairs HSC niche function during homeostasis and after stress. RNA sequencing analysis revealed that Spp1 (osteopontin) expression is markedly upregulated in Snai2-deficient MSPCs. Genetic deletion of Spp1 in Snai2-deficient mice rescues MSPCs' functions. Thus, SNAI2 is a critical regulator of the transcriptional network maintaining MSPCs by the suppression of osteopontin expression.


Assuntos
Células da Medula Óssea/metabolismo , Osteopontina/genética , Fatores de Transcrição da Família Snail/metabolismo , Nicho de Células-Tronco , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Osteopontina/metabolismo , Fatores de Transcrição da Família Snail/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA