Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
2.
J Immunol ; 211(10): 1561-1577, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37756544

RESUMO

Lipid accumulation in macrophages (Mφs) is a hallmark of atherosclerosis, yet how lipid accumulation affects inflammatory responses through rewiring of Mφ metabolism is poorly understood. We modeled lipid accumulation in cultured wild-type mouse thioglycolate-elicited peritoneal Mφs and bone marrow-derived Mφs with conditional (Lyz2-Cre) or complete genetic deficiency of Vhl, Hif1a, Nos2, and Nfe2l2. Transfection studies employed RAW264.7 cells. Mφs were cultured for 24 h with oxidized low-density lipoprotein (oxLDL) or cholesterol and then were stimulated with LPS. Transcriptomics revealed that oxLDL accumulation in Mφs downregulated inflammatory, hypoxia, and cholesterol metabolism pathways, whereas the antioxidant pathway, fatty acid oxidation, and ABC family proteins were upregulated. Metabolomics and extracellular metabolic flux assays showed that oxLDL accumulation suppressed LPS-induced glycolysis. Intracellular lipid accumulation in Mφs impaired LPS-induced inflammation by reducing both hypoxia-inducible factor 1-α (HIF-1α) stability and transactivation capacity; thus, the phenotype was not rescued in Vhl-/- Mφs. Intracellular lipid accumulation in Mφs also enhanced LPS-induced NF erythroid 2-related factor 2 (Nrf2)-mediated antioxidative defense that destabilizes HIF-1α, and Nrf2-deficient Mφs resisted the inhibitory effects of lipid accumulation on glycolysis and inflammatory gene expression. Furthermore, oxLDL shifted NADPH consumption from HIF-1α- to Nrf2-regulated apoenzymes. Thus, we postulate that repurposing NADPH consumption from HIF-1α to Nrf2 transcriptional pathways is critical in modulating inflammatory responses in Mφs with accumulated intracellular lipid. The relevance of our in vitro models was established by comparative transcriptomic analyses, which revealed that Mφs cultured with oxLDL and stimulated with LPS shared similar inflammatory and metabolic profiles with foamy Mφs derived from the atherosclerotic mouse and human aorta.


Assuntos
Aterosclerose , Hipercolesterolemia , Humanos , Camundongos , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Lipopolissacarídeos/metabolismo , NADP/metabolismo , Macrófagos/metabolismo , Lipoproteínas LDL/metabolismo , Glicólise , Aterosclerose/metabolismo , Colesterol/metabolismo , Antioxidantes/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
3.
J Immunol ; 211(3): 497-507, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37294291

RESUMO

Cachexia is a major cause of death in cancer and leads to wasting of cardiac and skeletal muscle, as well as adipose tissue. Various cellular and soluble mediators have been postulated in driving cachexia; however, the specific mechanisms behind this muscle wasting remain poorly understood. In this study, we found polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) to be critical for the development of cancer-associated cachexia. Significant expansion of PMN-MDSCs was observed in the cardiac and skeletal muscles of cachectic murine models. Importantly, the depletion of this cell subset, using depleting anti-Ly6G Abs, attenuated this cachectic phenotype. To elucidate the mechanistic involvement of PMN-MDSCs in cachexia, we examined major mediators, that is, IL-6, TNF-α, and arginase 1. By employing a PMN-MDSC-specific Cre-recombinase mouse model, we showed that PMN-MDSCs were not maintained by IL-6 signaling. In addition, PMN-MDSC-mediated cardiac and skeletal muscle loss was not abrogated by deficiency in TNF-α or arginase 1. Alternatively, we found PMN-MDSCs to be critical producers of activin A in cachexia, which was noticeably elevated in cachectic murine serum. Moreover, inhibition of the activin A signaling pathway completely protected against cardiac and skeletal muscle loss. Collectively, we demonstrate that PMN-MDSCs are active producers of activin A, which in turn induces cachectic muscle loss. Targeting this immune/hormonal axis will allow the development of novel therapeutic interventions for patients afflicted with this debilitating syndrome.


Assuntos
Células Supressoras Mieloides , Neoplasias , Animais , Camundongos , Células Supressoras Mieloides/metabolismo , Arginase/metabolismo , Caquexia , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Neoplasias/complicações , Neoplasias/metabolismo , Miocárdio , Músculo Esquelético/metabolismo
4.
Eur J Nutr ; 62(6): 2399-2413, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37106253

RESUMO

PURPOSE: To study the effects of feeding docosahexaenoic acid (DHA, derived from novel canola oil), with same amount of arachidonic acid (ARA), supplemented diet to lactating dams on the immune system development of suckled offspring using a T helper type-2 (Th2)-dominant BALB/c mouse. METHODS: Dams received nutritionally complete control (no ARA or DHA) or DHA + ARA diet (1% DHA and 1% ARA of total fatty acids) from 5 days pre-parturition to the end of 3-week suckling period. After euthanization, relevant tissues were collected to study fatty acids, splenocyte phenotype and function (ex vivo cytokines with/without lipopolysaccharide (LPS, bacterial challenge) or phorbol myristate acetate + ionomycin (PMAi) stimulation). RESULTS: Feeding dams a DHA diet significantly increased the mammary gland milk phospholipid concentration of DHA and ARA. This resulted in 60% higher DHA levels in splenocyte phospholipids of the pups although ARA levels showed no difference. In dams fed DHA diet, significantly higher proportion of CD27+ cytotoxic T cell (CTL) and CXCR3+ CCR6- Th (enriched in Th1) were observed than control, but there were no differences in the splenocyte function upon PMAi (non-specific lymphocyte stimulant) stimulation. Pups from DHA-fed dams showed significantly higher IL-1ß, IFN-γ and TNF-α (inflammatory cytokines) by LPS-stimulated splenocytes. This may be due to higher proportion of CD86+ macrophages and B cells (all p's < 0.05) in these pups, which may influence T cell polarization. CONCLUSION: Plant-based source of DHA in maternal diet resulted in higher ex vivo production of inflammatory cytokines by splenocytes due to change in their phenotype, and this can skew T cell towards Th1 response in a Th2-dominant BALB/c mouse.


Assuntos
Ácidos Docosa-Hexaenoicos , Hipersensibilidade , Animais , Feminino , Camundongos , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Araquidônico , Óleo de Brassica napus , Lactação , Lipopolissacarídeos/farmacologia , Suplementos Nutricionais , Dieta , Citocinas , Ácidos Graxos/farmacologia , Fosfolipídeos , Sistema Imunitário
5.
Front Endocrinol (Lausanne) ; 14: 1128622, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36992811

RESUMO

The signaling pathways downstream of the insulin receptor (InsR) are some of the most evolutionarily conserved pathways that regulate organism longevity and metabolism. InsR signaling is well characterized in metabolic tissues, such as liver, muscle, and fat, actively orchestrating cellular processes, including growth, survival, and nutrient metabolism. However, cells of the immune system also express the InsR and downstream signaling machinery, and there is increasing appreciation for the involvement of InsR signaling in shaping the immune response. Here, we summarize current understanding of InsR signaling pathways in different immune cell subsets and their impact on cellular metabolism, differentiation, and effector versus regulatory function. We also discuss mechanistic links between altered InsR signaling and immune dysfunction in various disease settings and conditions, with a focus on age related conditions, such as type 2 diabetes, cancer and infection vulnerability.


Assuntos
Diabetes Mellitus Tipo 2 , Receptor de Insulina , Humanos , Receptor de Insulina/metabolismo , Insulina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Transdução de Sinais , Sistema Imunitário/metabolismo
6.
Eur J Nutr ; 62(2): 699-711, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36197467

RESUMO

PURPOSE: To understand the effects of consuming high-fat and low-fat dairy products on postprandial cardiometabolic risk factors and intestinal immune function, we used an established low birthweight (LBW) swine model of diet-induced insulin resistance. METHODS: LBW piglets were randomized to consume one of the 3 experimental high fat diets and were fed for a total of 7 weeks: (1) Control high fat (LBW-CHF), (2) CHF diet supplemented with 3 servings of high-fat dairy (LBW-HFDairy) and (3) CHF diet supplemented with 3 servings of low-fat dairy (LBW-LFDairy). As comparison groups, normal birthweight (NBW) piglets were fed a CHF (NBW-CHF) or standard pig grower diet (NBW-Chow). At 11 weeks of age, all piglets underwent an established modified oral glucose and fat tolerance test. At 12 weeks of age, piglets were euthanized and ex vivo cytokine production by cells isolated from mesenteric lymph node (MLN) stimulated with mitogens was assessed. RESULTS: Dairy consumption did not modulate postprandial plasma lipid, inflammatory markers and glucose concentrations. A lower production of IL-2 and TNF-α after pokeweed mitogen (PWM) stimulation was observed in LBW-CHF vs NBW-Chow (P < 0.05), suggesting impaired MLN T cell function. While feeding high-fat dairy had minimal effects, feeding low-fat dairy significantly improved the production of IL-2 and TNF-α after PWM stimulation (P < 0.05). CONCLUSIONS: Irrespective of fat content, dairy had a neutral effect on postprandial cardiometabolic risk factors. Low-fat dairy products improved intestinal T cell function to a greater extent than high-fat dairy in this swine model of obesity and insulin resistance.


Assuntos
Resistência à Insulina , Animais , Peso ao Nascer , Dieta com Restrição de Gorduras , Glucose , Imunidade , Resistência à Insulina/fisiologia , Interleucina-2 , Suínos , Fator de Necrose Tumoral alfa
7.
Front Cell Dev Biol ; 10: 1044729, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36467420

RESUMO

Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.

8.
Cell Rep ; 34(2): 108609, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33440149

RESUMO

Stiffness in the tissue microenvironment changes in most diseases and immunological conditions, but its direct influence on the immune system is poorly understood. Here, we show that static tension impacts immune cell function, maturation, and metabolism. Bone-marrow-derived and/or splenic dendritic cells (DCs) grown in vitro at physiological resting stiffness have reduced proliferation, activation, and cytokine production compared with cells grown under higher stiffness, mimicking fibro-inflammatory disease. Consistently, DCs grown under higher stiffness show increased activation and flux of major glucose metabolic pathways. In DC models of autoimmune diabetes and tumor immunotherapy, tension primes DCs to elicit an adaptive immune response. Mechanistic workup identifies the Hippo-signaling molecule, TAZ, as well as Ca2+-related ion channels, including potentially PIEZO1, as important effectors impacting DC metabolism and function under tension. Tension also directs the phenotypes of monocyte-derived DCs in humans. Thus, mechanical stiffness is a critical environmental cue of DCs and innate immunity.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata/imunologia , Imunoterapia/métodos , Rigidez Vascular/imunologia , Humanos , Transdução de Sinais
9.
Nat Commun ; 10(1): 3650, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409776

RESUMO

The intestinal immune system is emerging as an important contributor to obesity-related insulin resistance, but the role of intestinal B cells in this context is unclear. Here, we show that high fat diet (HFD) feeding alters intestinal IgA+ immune cells and that IgA is a critical immune regulator of glucose homeostasis. Obese mice have fewer IgA+ immune cells and less secretory IgA and IgA-promoting immune mediators. HFD-fed IgA-deficient mice have dysfunctional glucose metabolism, a phenotype that can be recapitulated by adoptive transfer of intestinal-associated pan-B cells. Mechanistically, IgA is a crucial link that controls intestinal and adipose tissue inflammation, intestinal permeability, microbial encroachment and the composition of the intestinal microbiome during HFD. Current glucose-lowering therapies, including metformin, affect intestinal-related IgA+ B cell populations in mice, while bariatric surgery regimen alters the level of fecal secretory IgA in humans. These findings identify intestinal IgA+ immune cells as mucosal mediators of whole-body glucose regulation in diet-induced metabolic disease.


Assuntos
Imunoglobulina A/imunologia , Resistência à Insulina , Obesidade/imunologia , Tecido Adiposo/imunologia , Animais , Linfócitos B/imunologia , Estudos de Coortes , Fezes/microbiologia , Microbioma Gastrointestinal , Glucose/metabolismo , Humanos , Intestinos/imunologia , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/microbiologia
10.
Cell Rep ; 16(3): 717-30, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27373163

RESUMO

Obesity-related inflammation of metabolic tissues, including visceral adipose tissue (VAT) and liver, are key factors in the development of insulin resistance (IR), though many of the contributing mechanisms remain unclear. We show that nucleic-acid-targeting pathways downstream of extracellular trap (ET) formation, unmethylated CpG DNA, or ribonucleic acids drive inflammation in IR. High-fat diet (HFD)-fed mice show increased release of ETs in VAT, decreased systemic clearance of ETs, and increased autoantibodies against conserved nuclear antigens. In HFD-fed mice, this excess of nucleic acids and related protein antigens worsens metabolic parameters through a number of mechanisms, including activation of VAT macrophages and expansion of plasmacytoid dendritic cells (pDCs) in the liver. Consistently, HFD-fed mice lacking critical responders of nucleic acid pathways, Toll-like receptors (TLR)7 and TLR9, show reduced metabolic inflammation and improved glucose homeostasis. Treatment of HFD-fed mice with inhibitors of ET formation or a TLR7/9 antagonist improves metabolic disease. These findings reveal a pathogenic role for nucleic acid targeting as a driver of metabolic inflammation in IR.


Assuntos
Inflamação/metabolismo , Inflamação/patologia , Resistência à Insulina/fisiologia , Ácidos Nucleicos/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Adulto , Animais , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Dieta Hiperlipídica/métodos , Glucose/metabolismo , Homeostase/fisiologia , Humanos , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Fígado/metabolismo , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores Toll-Like/metabolismo
11.
Diabetes ; 64(6): 1886-97, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25999531

RESUMO

Obesity and associated insulin resistance predispose individuals to develop chronic metabolic diseases, such as type 2 diabetes and cardiovascular disease. Although these disorders affect a significant proportion of the global population, the underlying mechanisms of disease remain poorly understood. The discovery of elevated tumor necrosis factor-α in adipose tissue as an inducer of obesity-associated insulin resistance marked a new era of understanding that a subclinical inflammatory process underlies the insulin resistance and metabolic dysfunction that precedes type 2 diabetes. Advances in the field identified components of both the innate and adaptive immune response as key players in regulating such inflammatory processes. As antigen specificity is a hallmark of an adaptive immune response, its role in modulating the chronic inflammation that accompanies obesity and type 2 diabetes begs the question of whether insulin resistance and type 2 diabetes can have autoimmune components. In this Perspective, we summarize current data that pertain to the activation and perpetuation of adaptive immune responses during obesity and discuss key missing links and potential mechanisms for obesity-related insulin resistance and type 2 diabetes to be considered as potential autoimmune diseases.


Assuntos
Doenças Autoimunes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Animais , Humanos
12.
Diabetes ; 64(1): 90-103, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25048196

RESUMO

Obesity-related insulin resistance is associated with an influx of pathogenic T cells into visceral adipose tissue (VAT), but the mechanisms regulating lymphocyte balance in such tissues are unknown. Here we describe an important role for the immune cytotoxic effector molecule perforin in regulating this process. Perforin-deficient mice (Prf1(null)) show early increased body weight and adiposity, glucose intolerance, and insulin resistance when placed on high-fat diet (HFD). Regulatory effects of perforin on glucose tolerance are mechanistically linked to the control of T-cell proliferation and cytokine production in inflamed VAT. HFD-fed Prf1(null) mice have increased accumulation of proinflammatory IFN-γ-producing CD4(+) and CD8(+) T cells and M1-polarized macrophages in VAT. CD8(+) T cells from the VAT of Prf1(null) mice have increased proliferation and impaired early apoptosis, suggesting a role for perforin in the regulation of T-cell turnover during HFD feeding. Transfer of CD8(+) T cells from Prf1(null) mice into CD8-deficient mice (CD8(null)) resulted in worsening of metabolic parameters compared with wild-type donors. Improved metabolic parameters in HFD natural killer (NK) cell-deficient mice (NK(null)) ruled out a role for NK cells as a single source of perforin in regulating glucose homeostasis. The findings support the importance of T-cell function in insulin resistance and suggest that modulation of lymphocyte homeostasis in inflamed VAT is one possible avenue for therapeutic intervention.


Assuntos
Intolerância à Glucose/imunologia , Resistência à Insulina/imunologia , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Paniculite/imunologia , Perforina/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Dieta Hiperlipídica , Células-Tronco Embrionárias/citologia , Feminino , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Insulina/metabolismo , Gordura Intra-Abdominal/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Paniculite/genética , Paniculite/metabolismo , Perforina/genética , Perforina/metabolismo , Baço/citologia , Baço/imunologia
13.
ACS Nano ; 9(1): 16-30, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25469470

RESUMO

The design of nanovaccines capable of triggering effective antitumor immunity requires an understanding of how the immune system senses and responds to threats, including pathogens and tumors. Equally important is an understanding of the mechanisms employed by tumor cells to evade immunity and an appreciation of the deleterious effects that antitumor immune responses can have on tumor growth, such as by skewing tumor cell composition toward immunologically silent tumor cell variants. The immune system and tumors engage in a tug-of-war driven by competition where promoting antitumor immunity or tumor cell death alone may be therapeutically insufficient. Nanotechnology affords a unique opportunity to develop therapeutic compounds than can simultaneously tackle both aspects, favoring tumor eradication. Here, we review the current status of nanoparticle-based immunotherapeutic strategies for the treatment of cancer, ranging from antigen/adjuvant delivery vehicles (to professional antigen-presenting cell types of the immune system) to direct tumor antigen-specific T-lymphocyte-targeting compounds and their combinations thereof.


Assuntos
Imunoterapia/métodos , Nanomedicina/métodos , Nanopartículas , Neoplasias/imunologia , Neoplasias/terapia , Animais , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Humanos , Vigilância Imunológica
14.
Nat Med ; 20(5): 484-92, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24747746

RESUMO

Inflammation has a critical role in the development of insulin resistance. Recent evidence points to a contribution by the central nervous system in the modulation of peripheral inflammation through the anti-inflammatory reflex. However, the importance of this phenomenon remains elusive in type 2 diabetes pathogenesis. Here we show that rat insulin-2 promoter (Rip)-mediated deletion of Pten, a gene encoding a negative regulator of PI3K signaling, led to activation of the cholinergic anti-inflammatory pathway that is mediated by M2 activated macrophages in peripheral tissues. As such, Rip-cre(+) Pten(flox/flox) mice showed lower systemic inflammation and greater insulin sensitivity under basal conditions compared to littermate controls, which were abolished when the mice were treated with an acetylcholine receptor antagonist or when macrophages were depleted. After feeding with a high-fat diet, the Pten-deleted mice remained markedly insulin sensitive, which correlated with massive subcutaneous fat expansion. They also exhibited more adipogenesis with M2 macrophage infiltration, both of which were abolished after disruption of the anti-inflammatory efferent pathway by left vagotomy. In summary, we show that Pten expression in Rip(+) neurons has a critical role in diabetes pathogenesis through mediating the anti-inflammatory reflex.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Inflamação/metabolismo , Insulina/genética , PTEN Fosfo-Hidrolase/genética , Animais , Anti-Inflamatórios/administração & dosagem , Sistema Nervoso Central/metabolismo , Diabetes Mellitus Tipo 2/complicações , Dieta Hiperlipídica , Humanos , Inflamação/complicações , Inflamação/tratamento farmacológico , Insulina/metabolismo , Resistência à Insulina/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , PTEN Fosfo-Hidrolase/metabolismo , Regiões Promotoras Genéticas , Ratos , Receptores Muscarínicos/administração & dosagem , Deleção de Sequência , Transdução de Sinais
15.
Cold Spring Harb Perspect Med ; 2(2): a007773, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22355799

RESUMO

Development of strategies capable of specifically curbing pathogenic autoimmune responses in a disease- and organ-specific manner without impairing foreign or tumor antigen-specific immune responses represents a long sought-after goal in autoimmune disease research. Unfortunately, our current understanding of the intricate details of the different autoimmune diseases that affect mankind, including type 1 diabetes, is rudimentary. As a result, progress in the development of the so-called "antigen-specific" therapies for autoimmunity has been slow and fraught with limitations that interfere with bench-to-bedside translation. Absent or incomplete understanding of mechanisms of action and lack of adequate immunological biomarkers, for example, preclude the rational design of effective drug development programs. Here, we provide an overview of antigen-specific approaches that have been tested in preclinical models of T1D and, in some cases, human subjects. The evidence suggests that effective translation of these approaches through clinical trials and into patients will continue to meet with failure unless detailed mechanisms of action at the level of the organism are defined.


Assuntos
Antígenos/uso terapêutico , Diabetes Mellitus Tipo 1/terapia , Imunoterapia/métodos , Animais , Chaperonina 60/uso terapêutico , Ensaios Clínicos como Assunto , Células Dendríticas/imunologia , Modelos Animais de Doenças , Avaliação de Medicamentos , Inibidores Enzimáticos/uso terapêutico , Glutamato Descarboxilase/antagonistas & inibidores , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Vacinas de DNA/uso terapêutico
16.
J Mol Med (Berl) ; 89(8): 733-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21499734

RESUMO

Nanotechnology offers enormous potential in drug delivery and in vivo imaging. Nanoparticles (NPs), for example, are being extensively tested as scaffolds to deliver anti-cancer therapeutics or imaging tags. Our recent work, discussed herein, indicates that an opportunity exists to use NPs to deliver ligands for, and trigger, cognate receptors on T lymphocytes as a way to induce therapeutic immune responses in vivo. Specifically, systemic delivery of NPs coated with Type 1 diabetes (T1D)-relevant peptide-major histocompatibility complex molecules triggered the expansion of cognate memory autoregulatory (disease-suppressing) T cells, suppressed the progression of autoimmune attack against insulin-producing beta cells, and restored glucose homeostasis. This therapeutic avenue exploits a new paradigm in the progression of chronic autoimmune responses that enables the rational design of disease-specific "nanovaccines" capable of blunting autoimmunity without impairing systemic immunity, a long sought-after goal in the therapy of these disorders. Here, we discuss the research paths that led to the discovery of this therapeutic avenue and highlight the features that make it an attractive approach for the treatment, in an antigen-specific manner, of a whole host of autoimmune diseases.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Autoimunidade/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Nanopartículas/uso terapêutico , Peptídeos/imunologia , Vacinas/imunologia , Animais , Doenças Autoimunes/imunologia , Humanos
17.
J Immunol ; 181(1): 243-55, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18566390

RESUMO

Engagement of peptide-MHC by the TCR induces a conformational change in CD3epsilon that exposes a proline-rich sequence (PRS) and recruits the cytoskeletal adaptor Nck. This event, which precedes phosphorylation of the CD3epsilon ITAM, has been implicated in synapse formation and T cell function. However, there is compelling evidence that responsiveness to TCR ligation is CD3epsilon PRS independent. In this study, we show that the CD3epsilon PRS is necessary for peptide-MHC-induced phosphorylation of CD3epsilon and for recruitment of protein kinase Ctheta to the immune synapse in differentiated CD8+ T lymphocytes. However, whereas these two events are dispensable for functional T cell responsiveness to high-avidity ligands, they are required for responsiveness to low-avidity ones. Thus, in at least certain T cell clonotypes, the CD3epsilon PRS amplifies weak TCR signals by promoting synapse formation and CD3epsilon phosphorylation.


Assuntos
Complexo CD3/imunologia , Complexo CD3/metabolismo , Amplificação de Genes/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Sequência de Aminoácidos , Animais , Complexo CD3/química , Complexo CD3/genética , Diferenciação Celular/imunologia , Células Cultivadas , Antígenos de Histocompatibilidade/imunologia , Imunidade Inata/imunologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Peptídeos/farmacologia , Fosforilação , Prolina/genética , Prolina/metabolismo , Ligação Proteica , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/citologia , Linfócitos T/imunologia
18.
Endocrinology ; 148(11): 5136-44, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17673522

RESUMO

Immune modulators can arrest loss of insulin secretion in type 1 diabetes mellitus (T1DM), but they have not caused permanent disease remission or restored normal insulin secretion. We tested whether exendin-4, a glucagon-like peptide-1 receptor agonist, would enhance remission of T1DM in NOD mice treated with anti-CD3 monoclonal antibody (mAb) and studied the effects of exendin-4 treatment on cellular and metabolic responses of beta-cells. Diabetic NOD mice treated with anti-CD3 mAb and exendin-4 had a higher rate of remission (44%) than mice treated with anti-CD3 mAb alone (37%) or exendin-4 (0%) or insulin or IgG alone (0%) (P < 0.01). The effect of exendin-4 on reversal of diabetes after anti-CD3 mAb was greatest in mice with a glucose level of less than 350 mg/dl at diagnosis (63 vs. 39%, P < 0.05). Exendin-4 did not affect beta-cell area, replication, or apoptosis or reduce the frequency of diabetogenic or regulatory T cells or modulate the antigenicity of islet cells. Reversal of T1DM with anti-CD3 mAb was associated with recovery of insulin in glucose transporter-2(+)/insulin(-) islet cells that were identified at diagnosis. Glucose tolerance and insulin responses improved in mice treated with combination therapy, and exendin-4 increased insulin content and insulin release from beta-cells. We conclude that treatment with glucagon-like peptide-1 receptor agonist enhances remission of T1DM in NOD mice treated with anti-CD3 mAb by enhancing the recovery of the residual islets. This combinatorial approach may be useful in treatment of patients with new-onset T1DM.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complexo CD3/imunologia , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Peçonhas/farmacologia , Peçonhas/uso terapêutico , Animais , Células Cultivadas , Terapia Combinada , Diabetes Mellitus Tipo 1/patologia , Exenatida , Feminino , Receptor do Peptídeo Semelhante ao Glucagon 1 , Imunoterapia , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Glucagon/agonistas , Indução de Remissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA