Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.254
Filtrar
1.
Front Immunol ; 13: 929000, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35837386

RESUMO

Dendritic cells (DC) are uniquely capable of initiating and directing immune responses. The range of their activities grounds in the heterogeneity of DC subsets and their functional plasticity. Numerical and functional DC changes influence the development and progression of disease, and correction of such dysregulations has the potential to treat disease causally. In this review, we discuss the major advances in our understanding of the regulation of DC lineage formation, differentiation, and function in the skin. We describe the alteration of DC in disease as well as possibilities for therapeutic reprogramming with a focus on tolerogenic DC. Because regulatory T cells (Treg) are indispensable partners of DC in the induction and control of tolerance, we pay special attention to the interactions with these cells. Above all, we would like to arouse fascination for this cell type and its therapeutic potential in skin diseases.


Assuntos
Células Dendríticas , Tolerância Imunológica , Diferenciação Celular , Tolerância Imunológica/fisiologia , Pele , Linfócitos T Reguladores
2.
Front Immunol ; 13: 829406, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35619720

RESUMO

Background: Vascularized composite allografts (VCAs) allow reconstruction of devastating injuries and amputations, yet require lifelong immunosuppression that is associated with significant morbidity. Induction of immune tolerance of VCAs would permit widespread use of these procedures. VCAs are acquired from deceased donors most likely to be fully-MHC-mismatched (in contrast to living-related renal transplant donor-recipient pairs matched at one MHC haplotype). After achieving VCA tolerance in a swine model equivalent to clinical living-related renal transplants (single-haplotype MHC mismatches: e.g., "mother-daughter"/haploidentical), we tested our protocol in MHC class I, class II, and fully-MHC-mismatched pairs. Although class II mismatched swine demonstrated similar results as the haploidentical scenario (stable mixed chimerism and tolerance), our protocol failed to prevent rejection of class I and full mismatch VCAs. Here, we describe a new adapted conditioning protocol that successfully achieved tolerance across MHC class-I-mismatch barriers in swine. Methods: Swine were treated with non-myeloablative total body and thymic irradiation two days prior to infusion of bone marrow cells from an MHC class I-mismatched donor. They also received a short-term treatment with CTLA4-Ig (Belatacept®) and anti-IL6R mAb (Tociluzimab®) and were transplanted with an osteomyocutaneous VCA from the same donor. Results: Stable mixed chimerism and tolerance of MHC class-I-mismatched VCAs was achieved in 3 recipients. Allograft tolerance was associated with a sustained lack of anti-donor T cell response and a concomitant expansion of double negative CD4-CD8- T cells producing IL-10. Conclusions: This study demonstrates the first successful mixed chimerism-induced VCA tolerance in a large animal model across a MHC class-I-mismatch. Future studies aimed at fully-mismatched donor-recipient pairs are under investigation with this protocol.


Assuntos
Aloenxertos Compostos , Transplante de Rim , Animais , Quimerismo , Aloenxertos Compostos/transplante , Tolerância Imunológica/fisiologia , Suínos , Tolerância ao Transplante
3.
Adv Drug Deliv Rev ; 179: 114021, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34710529

RESUMO

The gastrointestinal tract (GIT) affects not only local diseases in the GIT but also various systemic diseases. Factors that can affect the health and disease of both GIT and the human body include 1) the mucosal immune system composed of the gut-associated lymphoid tissues and the lamina propria, 2) the intestinal barrier composed of mucus and intestinal epithelium, and 3) the gut microbiota. Selective delivery of drugs, including antigens, immune-modulators, intestinal barrier enhancers, and gut-microbiome manipulators, has shown promising results for oral vaccines, immune tolerance, treatment of inflammatory bowel diseases, and other systemic diseases, including cancer. However, physicochemical and biological barriers of the GIT present significant challenges for successful translation. With the advances of novel nanomaterials, oral nanomedicine has emerged as an attractive option to not only overcome these barriers but also to selectively deliver drugs to the target sites in GIT. In this review, we discuss the GIT factors and physicochemical and biological barriers in the GIT. Furthermore, we present the recent progress of oral nanomedicine for oral vaccines, immune tolerance, and anti-inflammation therapies. We also discuss recent advances in oral nanomedicine designed to fortify the intestinal barrier functions and modulate the gut microbiota and microbial metabolites. Finally, we opine about the future directions of oral nano-immunotherapy.


Assuntos
Microbioma Gastrointestinal/fisiologia , Imunidade nas Mucosas/fisiologia , Mucosa Intestinal/fisiologia , Sistemas de Liberação de Fármacos por Nanopartículas/farmacocinética , Administração Oral , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacocinética , Humanos , Tolerância Imunológica/fisiologia , Mucosa/metabolismo , Vacinas/administração & dosagem
4.
Exp Cell Res ; 409(1): 112886, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34673000

RESUMO

Chimeric antigen receptor (CAR) T cells have been successfully used for the treatment of hematological malignancies including acute and chronic lymphoblastic leukemia. However, results of CAR T cell projects in solid tumors have been less impressive to date, partly because of immunosuppressive tumor microenvironment (TME). It is widely known that high adenosine production is an important factor causing tumor-induced immunosuppression in TME, and adenosine mediates the suppression of anti-tumor T cell responses via binding and signaling through adenosine 2a receptor (A2aR). Previous studies have shown that adenosine generated by cancer cells significantly inhibits T cell anti-tumor activity through binding and then activating adenosine 2A receptors (A2aRs) of T cells. Based on the previous work, in our study, we evaluated whether A2aR disruption by shRNA could enhance the anti-tumor function of anti-mesothelin (MSLN) CAR T cells both in vitro and in vivo. For this goal above, we used MSLN-positive human ovarian serous carcinoma cells (SKOV3) and human colon cancer cells (HCT116) as target cancer cells while MSLN-negative human ovarian cancer cells (ES2) as non-target cancer cells. We observed that targeting cell-intrinsic A2aR through shRNA overexpression caused significant A2aR disruption in CAR T cells and profoundly increased CAR T cell efficacy in both CAR T cell cytokine production and cytotoxicity towards MSLN-positive cancer cells in vitro. More importantly, in SKOV3 xenograft mouse models, anti-MSLN CAR-T cells significantly reduced the tumor burden compared with non-transduced T cells, and the anti-tumor activity of A2aR-disrupted anti-MSLN CAR-T cells was stronger than that of wild-type anti-MSLN CAR-T cells. Altogether, our study showed enhanced anti-tumor efficacy caused by shRNA-mediated A2aR disruption in anti-MSLN CAR T cells both in vitro and in vivo, which proved that shRNA-mediated modification of gene expression might be an excellent strategy for improving CAR T cell function in immunosuppressive tumor microenvironment (TME) and could potentially improve the outcome of treatment in clinical trials.


Assuntos
Carcinoma Epitelial do Ovário/metabolismo , Mesotelina/metabolismo , Neoplasias Ovarianas/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Células HCT116 , Células HEK293 , Humanos , Tolerância Imunológica/fisiologia , Imunoterapia Adotiva/métodos , Camundongos , Microambiente Tumoral/fisiologia
5.
Theranostics ; 11(17): 8587-8604, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373760

RESUMO

Background: Sonodynamic therapy (SDT) is a promising strategy to inhibit tumor growth and activate antitumor immune responses for immunotherapy. However, the hypoxic and immunosuppressive tumor microenvironment limits its therapeutic efficacy and suppresses immune response. Methods: In this study, mitochondria-targeted and ultrasound-responsive nanoparticles were developed to co-deliver oxygen (O2) and nitric oxide (NO) to enhance SDT and immune response. This system (PIH-NO) was constructed with a human serum albumin-based NO donor (HSA-NO) to encapsulate perfluorodecalin (FDC) and the sonosensitizer (IR780). In vitro, the burst release of O2 and NO with US treatment to generate reactive oxygen species (ROS), the mitochondria targeting properties and mitochondrial dysfunction were evaluated in tumor cells. Moreover, in vivo, tumor accumulation, therapeutic efficacy, the immunosuppressive tumor microenvironment, immunogenic cell death, and immune activation after PIH-NO treatment were also studied in 4T1 tumor bearing mice. Results: PIH-NO could accumulate in the mitochondria and relive hypoxia. After US irradiation, O2 and NO displayed burst release to enhance SDT, generated strongly oxidizing peroxynitrite anions, and led to mitochondrial dysfunction. The release of NO increased blood perfusion and enhanced the accumulation of the formed nanoparticles. Owing to O2 and NO release with US, PIH-NO enhanced SDT to inhibit tumor growth and amplify immunogenic cell death in vitro and in vivo. Additionally, PIH-NO promoted the maturation of dendritic cells and increased the number of infiltrating immune cells. More importantly, PIH-NO polarized M2 macrophages into M1 phenotype and depleted myeloid-derived suppressor cells to reverse immunosuppression and enhance immune response. Conclusion: Our findings provide a simple strategy to co-deliver O2 and NO to enhance SDT and reverse immunosuppression, leading to an increase in the immune response for cancer immunotherapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanomedicina Teranóstica/métodos , Terapia por Ultrassom/métodos , Animais , Linhagem Celular Tumoral , China , Humanos , Hipóxia/metabolismo , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/fisiologia , Imunidade , Imunoterapia/métodos , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Nanopartículas/uso terapêutico , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacologia , Oxigênio/metabolismo , Oxigênio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral/fisiologia , Ultrassonografia
6.
Life Sci ; 287: 119726, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34144058

RESUMO

Autoimmune diseases (AD), which are classified as chronic injuries, are caused by a specific auto-reactive reaction. The etiology of most ADs is not well understood. Meanwhile, Autophagy is a protective response defining as a catabolic method by lysosomes tending to maintain homeostasis acts by recycling and discrediting cell compartments. Autophagy plays a crucial role in controlling immune homeostasis by eliminating intracellular pathogens and presenting antigens to immune cognition. MicroRNAs are commonly known as endogenous non-coding small RNAs, which span 18-25 nt and take part in the gene expression at the post-transcriptional level regulation. miRNAs play important roles in different processes like, cell differentiation, duplicating, and apoptosis. Moreover, miRNAs are the critical molecules for the regular function of the immune system by modulating immune tolerance mechanisms and autoimmunity. Recent findings support the role of dysregulated miRNAs in the pathogenesis of ADs and in the regulation of autophagy. In this review, we will focus on the role of the miRNAs in the regulation of autophagy and then will explain the role of dysregulated miRNAs in the initiation of the ADs by modulating autophagy.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/fisiologia , Autofagia/fisiologia , MicroRNAs/fisiologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/metabolismo , Humanos , Tolerância Imunológica/fisiologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo
7.
Reprod Biomed Online ; 43(2): 319-328, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34103260

RESUMO

RESEARCH QUESTION: What is the potential role of immune cells and their inflammatory cytokines in the pathogenesis, development and establishment of endometriosis? DESIGN: Peritoneal fluid from 59 women (43 with endometriosis and 16 controls) who had undergone laparoscopic surgery was analysed. Changes in the population of innate and adaptive immune cells, cytokines, chemokines and growth factor expression were measured by flow cytometry, Luminex Technology and enzyme-linked immunosorbent assay. RESULTS: No differences were found in the frequencies of the innate and adaptive immune cells between women with and without endometriosis. In the peritoneal fluid of women with endometriosis, IL-1ß, IL-1RN, IL-2, IL-4, IL-8, IL-10, IL-12 (p70), IL-17α, FGF2, G-CSF, MCP-1, MIP-1α and TNF-α were significantly increased compared with controls. A correlation between IL-2, MCP-1, MIP-1α, TNF-α and the severity of endometriosis was observed. The concentration of neopterin, a possible biomarker for this disease, was increased in women with endometriosis compared with controls. CONCLUSIONS: The functional activity of immune cells seemed to be reduced despite their numbers remaining unchanged. The data indicate that a shift of TH cytokine profile occurs, which increases the TH1-TH2 ratio. This is driven by the increased levels of the cytokines (TNF-α and IL-2) in women with severe endometriosis.


Assuntos
Endometriose/imunologia , Tolerância Imunológica/fisiologia , Doenças Peritoneais/imunologia , Adolescente , Adulto , Líquido Ascítico/imunologia , Líquido Ascítico/metabolismo , Líquido Ascítico/patologia , Estudos de Casos e Controles , Quimiocinas/metabolismo , Citocinas/metabolismo , Endometriose/metabolismo , Endometriose/patologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Matadoras Naturais/patologia , Células Matadoras Naturais/fisiologia , Leucócitos/patologia , Leucócitos/fisiologia , Doenças Peritoneais/metabolismo , Doenças Peritoneais/patologia , Transdução de Sinais/imunologia , Adulto Jovem
8.
Diabetes ; 70(6): 1211-1219, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34016597

RESUMO

Critical insights into the etiology of type 1 diabetes (T1D) came from genome-wide association studies that unequivocally connected genetic susceptibility to immune cell function. At the top of the susceptibility are genes involved in regulatory T-cell (Treg) function and development. The advances in epigenetic and transcriptional analyses have provided increasing evidence for Treg dysfunction in T1D. These are well supported by functional studies in mouse models and analysis of peripheral blood during T1D. For these reasons, Treg-based therapies are at the forefront of research and development and have a tangible probability to deliver a long-sought-after successful immune-targeted treatment for T1D. The current challenge in the field is whether we can directly assess Treg function at the tissue site or make informative interpretations based on peripheral data. Future studies focused on Treg function in pancreatic lymph nodes and pancreas could provide key insight into the ultimate mechanisms underlying Treg failure in T1D. In this Perspective we will provide an overview of current literature regarding Treg development and function in T1D and how this knowledge has been applied to Treg therapies.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Linfócitos T Reguladores/fisiologia , Animais , Autoimunidade/fisiologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Endocrinologia/métodos , Endocrinologia/tendências , Humanos , Tolerância Imunológica/fisiologia , Imunoterapia Adotiva/métodos , Imunoterapia Adotiva/tendências , Camundongos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Pâncreas/imunologia , Pâncreas/metabolismo , Pâncreas/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante
9.
Med Sci (Paris) ; 37(1): 35-40, 2021 Jan.
Artigo em Francês | MEDLINE | ID: mdl-33492216

RESUMO

Mucosal immunity has to deal with a patchy mix of commensal but also eventually pathogenic bugs. Immunoglobulins of the A class (IgA) are opposing to this duality a functional balance going from tolerance to protective response or even to hyper-inflammation. Recent reports have shown the binding of polyreactive natural IgA, but also of affinity maturated protective IgA to the commensal microbiota, to superantigens and also to vaccinal antigens. Diverse types of humoral responses thus altogether contribute to the homeostasis of mucosal immunity. Their knowledge has to be taken into consideration for defining strategies of immuno-intervention, for mucosal vaccination as much as for immunotherapy of chronic inflammatory bowel disease.


TITLE: Homéostasie de la réponse IgA et microbiote. ABSTRACT: L'immunité muqueuse s'établit en réponse à un ensemble de microorganismes qui sont surtout commensaux mais aussi, parfois, pathogènes. À cette dualité, les immunoglobulines de classe A (IgA) opposent une balance fonctionnelle allant de la tolérance à la protection, voire à une hyper-inflammation. Des travaux récents ont révélé la liaison d'IgA polyréactives naturelles ou, à l'inverse, d'IgA spécifiquement affines et protectrices, au microbiote commensal, mais aussi à des super-antigènes ou encore à des vaccins muqueux. Différents types de réponse humorale s'associent ainsi pour composer, ensemble, l'homéostasie de l'immunité muqueuse. Leur connaissance devrait ainsi influencer les stratégies de vaccination muqueuse et également les immunothérapies ciblant les maladies inflammatoires chroniques de l'intestin.


Assuntos
Imunidade/fisiologia , Imunoglobulina A/metabolismo , Microbiota/fisiologia , Animais , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Homeostase/fisiologia , Humanos , Tolerância Imunológica/fisiologia , Imunidade nas Mucosas/fisiologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Microbiota/imunologia
10.
Mol Nutr Food Res ; 65(5): e2000036, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32996681

RESUMO

The gastrointestinal tract harbors a complex resident microbial ecosystem, comprising over 500 species, spanning commensals, mutualist, opportunistic, and professional pathogens thriving on undigested food components originating from the diet and endogenous secretions. Despite this high concentration of food and bacterial antigens, a healthy gut has a near absent level of inflammation, a status called intestinal immune homeostasis. This immune homeostasis is built and maintained in the presence, and interestingly, with cooperation of the microbiota. The microbiota ferments undigested food components into a wide variety of metabolites, some of which interact with the intestinal immune system. In particular short-chain fatty acids, aryl hydrocarbon receptor ligands, and bile acid metabolites have been involved in the induction of intestinal immune homeostasis. The production of these metabolites is influenced by the microbial load and community structure, as well as the availability of substrates and the gut environment which are directly or indirectly modulated by food intake. In this manuscript, the factors that influence the production of these metabolites and their interaction with the immune cells that play key roles in maintaining intestinal immune homeostasis in the healthy gut are reviewed.


Assuntos
Alimentos , Microbioma Gastrointestinal/fisiologia , Tolerância Imunológica/fisiologia , Intestinos/imunologia , Ácidos e Sais Biliares/metabolismo , Fermentação , Microbiologia de Alimentos , Humanos , Fatores Imunológicos/metabolismo , Intestinos/citologia , Filogenia , Triptofano/metabolismo
11.
Neurochem Int ; 142: 104919, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33242538

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease which causes a non-reversible cognitive impairment and dementia. The primary cause of late-onset AD remains unknown although its pathology was discovered over a century ago. Recently, the vascular hypothesis of AD has received backing from evidence emerging from neuroimaging studies which have revealed the presence of a significant hypoperfusion in the brain regions vulnerable to AD pathology. In fact, hypoxia can explain many of the pathological changes evident in AD pathology, e.g. the deposition of ß-amyloid plaques and chronic low-grade inflammation. Hypoxia-inducible factor-1α (HIF-1α) stimulates inflammatory responses and modulates both innate and adaptive immunity. It is known that hypoxia-induced inflammation evokes compensatory anti-inflammatory response involving tissue-resident microglia/macrophages and infiltrated immune cells. Hypoxia/HIF-1α induce immunosuppression by (i) increasing the expression of immunosuppressive genes, (ii) stimulating adenosinergic signaling, (iii) enhancing aerobic glycolysis, i.e. lactate production, and (iv) augmenting the secretion of immunosuppressive exosomes. Interestingly, it seems that these common mechanisms are also involved in the pathogenesis of AD. In AD pathology, an enhanced immunosuppression appears, e.g. as a shift in microglia/macrophage phenotypes towards the anti-inflammatory M2 phenotype and an increase in the numbers of regulatory T cells (Treg). The augmented anti-inflammatory capacity promotes the resolution of acute inflammation but persistent inflammation has crucial effects not only on immune cells but also harmful responses to the homeostasis of AD brain. I will examine in detail the mechanisms of the hypoperfusion/hypoxia-induced immunosuppressive state in general and especially, in its association with AD pathogenesis. These immunological observations support the vascular hypothesis of AD pathology.


Assuntos
Doença de Alzheimer/imunologia , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Circulação Cerebrovascular/fisiologia , Tolerância Imunológica/fisiologia , Acoplamento Neurovascular/fisiologia , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/metabolismo , Humanos , Rede Nervosa/irrigação sanguínea , Rede Nervosa/imunologia , Rede Nervosa/metabolismo
12.
Acta Pharmacol Sin ; 42(1): 27-35, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32123300

RESUMO

Herbal and dietary supplements (HDS)-induced liver injury has been a great concern all over the world. Polygonum multiflorum Thunb., a well-known Chinese herbal medicine, is recently drawn increasing attention because of its hepatotoxicity. According to the clinical and experimental studies, P. multiflorum-induced liver injury (PM-DILI) is considered to be immune-mediated idiosyncratic liver injury, but the role of immune response and the underlying mechanisms are not completely elucidated. Previous studies focused on the direct toxicity of PM-DILI by using animal models with intrinsic drug-induced liver injury (DILI). However, most epidemiological and clinical evidence demonstrate that PM-DILI is immune-mediated idiosyncratic liver injury. The aim of this review is to assess current epidemiological, clinical and experimental evidence about the possible role of innate and adaptive immunity in the idiosyncratic hepatotoxicity of P. multiflorum. The potential effects of factors associated with immune tolerance, including immune checkpoint molecules and regulatory immune cells on the individual's susceptibility to PM-DILI are also discussed. We conclude by giving our hypothesis of possible immune mechanisms of PM-DILI and providing suggestions for future studies on valuable biomarkers identification and proper immune models establishment.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Medicamentos de Ervas Chinesas/efeitos adversos , Fallopia multiflora/efeitos adversos , Imunidade Inata/efeitos dos fármacos , Fígado/efeitos dos fármacos , Imunidade Adaptativa/genética , Animais , Povo Asiático , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/toxicidade , Fallopia multiflora/toxicidade , Antígeno HLA-B35/genética , Humanos , Tolerância Imunológica/fisiologia , Lipopolissacarídeos/toxicidade
13.
Annu Rev Med ; 72: 215-228, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33106103

RESUMO

The introduction of targeted biologic therapies has changed the treatment landscape for autoimmune diseases (ADs) substantially, but although these therapies provide more specificity, they require continuous administration, rarely restore organ function or reverse disability, and are not curative. Over the last 25 years, hematopoietic stem cell transplantation (HSCT) has been increasingly used to treat patients in whom the risk:benefit ratio of HSCT is acceptable. In contrast to chronic suppression of immune function, this intensive one-off procedure aims to provide treatment-free remissions by the reinduction of self-tolerance. The European Society for Blood and Marrow Transplantation (EBMT) Autoimmune Diseases Working Party (ADWP) has been central to development of this approach, with over 3,300 HSCT registrations for ADs. Recent data have improved the evidence base to support autologous HSCT in multiple sclerosis, systemic sclerosis, and Crohn's disease, along with a wide range of rarer disease indications, and autologous HSCT has become an integral part of treatment algorithms in various ADs.


Assuntos
Doenças Autoimunes/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Tolerância Imunológica/fisiologia , Doenças Autoimunes/imunologia , Humanos , Transplante Autólogo
14.
Inflammation ; 44(1): 1-12, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32954452

RESUMO

Autophagy exerts its dual role in eukaryotic cells and exerts its cytoprotective action through degradation mechanism and by regulating catabolic processes which results in elimination of pathogens. Under suitable conditions, autophagy is associated with recycling of cytoplasmic components which causes regeneration of energy whereas deregulated autophagy exerts its implicated role in development and pathogenesis of auto-immune diseases such as rheumatoid arthritis. The immune, innate, and adaptive responses are regulated through the development, proliferation, and growth of lymphocytes. Such innate and adaptive responses can act as mediator of arthritis; along with this, stimulation of osteoclast-mediated bone resorption takes place via transferring citrullinated peptides towards MHC (major histocompatibility complex) compartments, thereby resulting in degradation of bone. Processes such as apoptosis resistance are also regulated through autophagy. In this review, the current knowledge based on role of autophagy in pathogenesis of rheumatoid arthritis is summarized along with proteins associated.


Assuntos
Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Autofagia/fisiologia , Tolerância Imunológica/fisiologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Humanos
15.
Placenta ; 104: 81-88, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33296735

RESUMO

Chronic placental inflammatory lesions lead to poor obstetric outcomes. These lesions often proceed undetected until examination of placental tissues after delivery and are mediated by CXCR3, a seven-transmembrane G protein-coupled receptor, and its chemokine ligands - CXCL9, CXCL10 and CXCL11. CXCR3-chemokine ligand interaction disrupts feto-maternal immune tolerance and activate obnoxious immunological responses similar to transplant rejection and graft-versus-host disease. The resultant chronic inflammatory responses manifest in different parts of the placenta characterised by the presence of incompatible immunocompetent cells from the feto-maternal unit i.e. maternal CD8+ T cells in the chorionic membrane or plate (chronic chorioamnionitis); foetal Hofbauer cells and maternal CD8+ T cells in the chorionic villous tree (villitis of unknown aetiology); maternal CD8+ T and plasma cells in the basal plate (chronic deciduitis); and maternal CD8+ T cells, histiocytes and T regulatory cells in the intervillous space (chronic intervillositis). This review critically examines how the CXCR3-chemokine ligand interaction disrupts feto-maternal immune tolerance, initiates a series of chronic placental inflammatory lesions, and consequently activates the pathways to intrauterine growth restriction, stillbirth, spontaneous abortion, preterm prelabour rupture of membranes, preterm labour and birth. The possibility of interrupting these signalling pathways through the use of CXCR3 chemokine inhibitors to prevent adverse reproductive sequelae as well as the potential clinical utility of CXCR3 chemokines as non-invasive predictive clinical biomarkers are also highlighted.


Assuntos
Quimiocinas CXC/metabolismo , Tolerância Imunológica/fisiologia , Inflamação/metabolismo , Placenta/metabolismo , Receptores CXCR3/metabolismo , Feminino , Humanos , Inflamação/imunologia , Placenta/imunologia , Gravidez
16.
Cancer Res ; 81(4): 801-805, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33177063

RESUMO

The tumor microenvironment (TME) promotes tumor development via complex intercellular signaling, aiding tumor growth and suppressing immunity. Eph receptors (Eph) and their ephrin ligands control cell interactions during normal development, and reemerge in tumors and the TME, where they are implicated in invasion, metastasis, and angiogenesis. Recent studies also indicate roles for Ephs in suppressing immune responses by controlling tumor interactions with innate and adaptive immune cells within the TME. Accordingly, inhibiting these functions can promote immune response and efficacy of immune checkpoint inhibition. This research highlights Ephs as potential targets to enhance efficacy of immune-based therapies in patients with cancer.


Assuntos
Tolerância Imunológica/fisiologia , Neoplasias/imunologia , Neoplasias/patologia , Receptores da Família Eph/fisiologia , Microambiente Tumoral/imunologia , Animais , Efrinas/metabolismo , Humanos , Imunidade/genética , Neoplasias/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Evasão Tumoral/genética , Evasão Tumoral/imunologia , Microambiente Tumoral/genética
17.
PLoS One ; 15(12): e0240773, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378402

RESUMO

Infectious diseases are a threat to elderly individuals, whose immune systems weaken with age. Among the various infectious diseases, Clostridium difficile infection is associated with a high rate of mortality in elderly individuals and is a serious health problem worldwide, owing to the increasing infection rates. Probiotic use has been proposed as an effective countermeasure for C. difficile infection. The aim of this study was to evaluate the effects of heat-killed Enterococcus faecalis T-110 on intestinal immunity, intestinal flora, and intestinal infections, especially C. difficile infections, in naturally ageing animals, for extrapolating the results to elderly human subjects. Twenty female hamsters were randomly distributed into two groups. Group 1 was fed a basal diet and group 2 was fed a basal diet supplemented with heat-killed E. faecalis for 7 days. Heat-killed E. faecalis T-110 improved the gut immunity and microflora, especially Clostridium perfringens and C. difficile, in naturally aged hamsters. Therefore, heat-killed E. faecalis T-110 use may be a countermeasure against age-related immune dysfunction and intestinal infections, especially C. difficile infection, in elderly humans. However, further investigation in this regard is needed in humans.


Assuntos
Envelhecimento/imunologia , Infecções por Clostridium/prevenção & controle , Diarreia/prevenção & controle , Enterococcus faecalis/imunologia , Probióticos/administração & dosagem , Fatores Etários , Idoso , Animais , Clostridioides difficile/imunologia , Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Clostridium perfringens/imunologia , Clostridium perfringens/isolamento & purificação , Cricetinae , Diarreia/imunologia , Diarreia/microbiologia , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/imunologia , Temperatura Alta , Humanos , Tolerância Imunológica/fisiologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia
18.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105907

RESUMO

Aryl hydrocarbon receptor (AhR) was identified in the early 1970s as a receptor for the ubiquitous environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin), which is a member of halogenated aromatic hydrocarbons (HAHs). TCDD was found to be highly toxic to the immune system, causing thymic involution and suppression of a variety of T and B cell responses. The fact that environmental chemicals cause immunosuppression led to the emergence of a new field, immunotoxicology. While studies carried out in early 1980s demonstrated that TCDD induces suppressor T cells that attenuate the immune response to antigens, further studies on these cells were abandoned due to a lack of specific markers to identify such cells. Thus, it was not until 2001 when FoxP3 was identified as a master regulator of Regulatory T cells (Tregs) that the effect of AhR activation on immunoregulation was rekindled. The more recent research on AhR has led to the emergence of AhR as not only an environmental sensor but also as a key regulator of immune response, especially the differentiation of Tregs vs. Th17 cells, by a variety of endogenous, microbial, dietary, and environmental ligands. This review not only discusses how the role of AhR emerged from it being an environmental sensor to become a key immunoregulator, but also confers the identification of new AhR ligands, which are providing novel insights into the mechanisms of Treg vs. Th17 differentiation. Lastly, we discuss how AhR ligands can trigger epigenetic pathways, which may provide new opportunities to regulate inflammation and treat autoimmune diseases.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Tolerância Imunológica/fisiologia , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/imunologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Epigênese Genética , Humanos , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/imunologia , Inflamação/imunologia , Linfócitos T Reguladores/imunologia
19.
J Neuroinflammation ; 17(1): 287, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33008419

RESUMO

BACKGROUND: Current methods to repair ablation-type peripheral nerve injuries (PNIs) using peripheral nerve allografts (PNAs) often result in poor functional recovery due to immunological rejection as well as to slow and inaccurate outgrowth of regenerating axonal sprouts. In contrast, ablation-type PNIs repaired by PNAs, using a multistep protocol in which one step employs the membrane fusogen polyethylene glycol (PEG), permanently restore sciatic-mediated behaviors within weeks. Axons and cells within PEG-fused PNAs remain viable, even though outbred host and donor tissues are neither immunosuppressed nor tissue matched. PEG-fused PNAs exhibit significantly reduced T cell and macrophage infiltration, expression of major histocompatibility complex I/II and consistently low apoptosis. In this study, we analyzed the coding transcriptome of PEG-fused PNAs to examine possible mechanisms underlying immunosuppression. METHODS: Ablation-type sciatic PNIs in adult Sprague-Dawley rats were repaired using PNAs and a PEG-fusion protocol combined with neurorrhaphy. Electrophysiological and behavioral tests confirmed successful PEG-fusion of PNAs. RNA sequencing analyzed differential expression profiles of protein-coding genes between PEG-fused PNAs and negative control PNAs (not treated with PEG) at 14 days PO, along with unoperated control nerves. Sequencing results were validated by quantitative reverse transcription PCR (RT-qPCR), and in some cases, immunohistochemistry. RESULTS: PEG-fused PNAs display significant downregulation of many gene transcripts associated with innate and adaptive allorejection responses. Schwann cell-associated transcripts are often upregulated, and cellular processes such as extracellular matrix remodeling and cell/tissue development are particularly enriched. Transcripts encoding several potentially immunosuppressive proteins (e.g., thrombospondins 1 and 2) also are upregulated in PEG-fused PNAs. CONCLUSIONS: This study is the first to characterize the coding transcriptome of PEG-fused PNAs and to identify possible links between alterations of the extracellular matrix and suppression of the allorejection response. The results establish an initial molecular basis to understand mechanisms underlying PEG-mediated immunosuppression.


Assuntos
Aloenxertos/fisiologia , Perfilação da Expressão Gênica/métodos , Tolerância Imunológica/fisiologia , Polietilenoglicóis/administração & dosagem , Nervo Isquiático/fisiologia , Nervo Isquiático/transplante , Animais , Feminino , Mapas de Interação de Proteínas/fisiologia , Ratos , Ratos Sprague-Dawley
20.
Sci Rep ; 10(1): 17023, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046725

RESUMO

In this study, we investigated the effects of Formaldehyde (FA) exposure on splenic immune responses wherein helper T cells become activated and differentiate into effector T and regulatory T cells. BALB/c mice were exposed to two FA concentrations (1.38 mg/m3 and 5.36 mg/m3) for 4 h/day and 5 days/week for 2 weeks. FA-induced immune responses were examined by the production of cytokines, expression of mRNAs, and distributions of helper T cells and regulatory T cells. Moreover, expression of calcineurin and NFATs, regulatory T cell-related signalling proteins, were evaluated. FA exposure suppressed Th2-, Th1-, and Th17-related splenic cytokines in a dose-dependent manner. mRNA expression of splenic cytokines was also decreased by FA exposure, which correlated with decreased cytokine expression. In parallel, FA exposure promoted T cell differentiation into regulatory T cells in a dose-dependent manner supported by the expression of calcineurin and NFAT1. Taken together, our results indicated that FA exposure increases the number of regulatory T cells via calcineurin-NFAT signalling, thereby leading to effector T cell activity suppression with decreased T cell-related cytokine secretion and mRNA expression. These findings provide insight into the mechanisms underlying the adverse effects of FA and accordingly have general implications for human health, particularly in occupational settings.


Assuntos
Calcineurina/metabolismo , Formaldeído/farmacologia , Formaldeído/toxicidade , Tolerância Imunológica/efeitos dos fármacos , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Formaldeído/efeitos adversos , Formaldeído/imunologia , Formaldeído/metabolismo , Tolerância Imunológica/fisiologia , Imunidade/efeitos dos fármacos , Imunidade/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...