Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.834
Filtrar
1.
Sci Rep ; 14(1): 10340, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710764

RESUMEN

This study aims to evaluate the role of trefoil factor 3 (TFF3) peptides in type 2 diabetes mellitus (T2DM) from an inflammatory perspective. The focus was on exploring how TFF3 affects the function of T cells. TFF3 overexpression model was constructed using lentivirus in Jurkat cell lines. We evaluated the impact of TFF3 on the proliferation, apoptosis, and IL-17A levels of Jurkat cells cultured in high glucose. The T2DM model was induced in TFF3 knockout (KO) mice through streptozotocin combined with high-fat diet. The measurements included glucose tolerance, insulin tolerance, inflammation markers, Th17 cell proportion, and pancreatic pathological changes. The T2DM modeling led to splenomegaly in mice, and increased expression of TFF3 in their spleens. Overexpression of TFF3 increased the proportion of IL-17+ T cells and the levels of Th17-related cytokines in Jurkat cells. There was no difference in body weight and blood glucose levels between wild-type and TFF3 KO mice. However, T2DM mice lacking the TFF3 gene showed improved glucose utilization, ameliorated pancreatic pathology, decreased inflammation levels, and reduced Th17 cell ratio. TFF3 may be involved in the chronic inflammatory immune response in T2DM. Its mechanism may be related to the regulation of the RORγt/IL-17 signaling pathway and its impact on T cell proliferation and apoptosis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ratones Noqueados , Células Th17 , Factor Trefoil-3 , Células Th17/inmunología , Células Th17/metabolismo , Animales , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/inmunología , Ratones , Factor Trefoil-3/metabolismo , Factor Trefoil-3/genética , Células Jurkat , Interleucina-17/metabolismo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Masculino , Proliferación Celular , Apoptosis , Dieta Alta en Grasa/efectos adversos
2.
Int Immunopharmacol ; 132: 112019, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38599099

RESUMEN

OBJECTIVE: The current study aimed to assess the modulating effect of IL-2 encapsulated chitosan-nanoparticles (CSNPs) on the function of Treg cells through induction of type 1 diabetes (T1D). Treg cell function was monitored by the forkhead box P3 (FoxP3) and transforming growth factor beta (TGFß) levels, correlating them with blood glucose and serum insulin levels. MATERIALS AND METHODS: In this case-control study, a low dose of IL-2 (free and chitosan-loaded) was injected into a diabetic mice group. The levels of FoxP3 and TGF-ß 1 were assessed using Enzyme-Linked Immunosorbent Assay. In addition, blood glucose and serum insulin levels were determined. RESULTS: The mean glucose level decreased significantly after free rIL-2 or rIL-2 / CSNPs treatment. Meanwhile, the mean serum insulin level was significantly increased after treatment with free rIL-2 or rIL-2/CSNPs. The mean levels of FoxP3 and TGFß 1 were significantly increased with either free rIL-2 or rIL-2/CSNPs compared to the T1D untreated group (P < 0.001). In the treated mice group receiving free CSNPs, there was a significant negative correlation between glucose and insulin levels. Moreover, FoxP3 & TGFß 1 levels had a significant positive correlation. In treated mice groups with free rIL-2 and IL-2 CSNPs, there was a significant positive correlation between FoxP3 and glucose levels. A significant negative correlation was found after conducting a correlation between insulin level and FoxP3 in the T1D/ rIL-2 / CSNPs group. CONCLUSIONS: Low-dose IL-2 selectively modulates FoxP3 + Tregs, and TGFß 1 increases their levels. These results demonstrated that IL-2-free and chitosan-loaded nanoparticles can be therapeutic agents in T1D.


Asunto(s)
Glucemia , Quitosano , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Factores de Transcripción Forkhead , Insulina , Interleucina-2 , Nanopartículas , Linfocitos T Reguladores , Animales , Quitosano/química , Quitosano/administración & dosificación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Interleucina-2/metabolismo , Interleucina-2/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/inmunología , Glucemia/efectos de los fármacos , Ratones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/sangre , Factores de Transcripción Forkhead/metabolismo , Insulina/sangre , Masculino , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/sangre , Estreptozocina , Humanos
3.
Int Immunopharmacol ; 133: 112056, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38626546

RESUMEN

OBJECTIVES: The aim of this study was to investigate the effect of 4µ8c, an inhibitor targeting the endoplasmic reticulum stress-associated factor IRE1α, on macrophage polarization in an experimental model of diabetic periodontitis through ex vivo experiments. MATERIALS AND METHODS: Local alveolar bone parameters were evaluated using Micro-CT following intraperitoneal administration of 4µ8c in mice with experimental diabetic periodontitis. Surface markers indicating macrophage polarization were identified using immunofluorescence. In vitro experiments were performed employing bone marrow-derived macrophages and gingival fibroblasts. Macrophage polarization was determined using flow cytometry. Principal impacted signaling pathways were identified through Western blot analysis. RESULTS: Results from both in vitro and in vivo experiments demonstrated that 4µ8c mitigated alveolar bone resorption and inflammation in mice with diabetic periodontitis. Furthermore, it modulated macrophage polarization towards the M2 phenotype and augmented M2 macrophage polarization through the MAPK signaling pathway. CONCLUSIONS: These findings suggest that inhibiting IRE1α can modulate macrophage polarization and alleviate ligature-induced diabetic periodontitis via the MAPK signaling pathway. This unveils a novel mechanism, offering a scientific foundation for the treatment of experimental diabetic periodontitis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Estrés del Retículo Endoplásmico , Endorribonucleasas , Macrófagos , Ratones Endogámicos C57BL , Periodontitis , Proteínas Serina-Treonina Quinasas , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Periodontitis/inmunología , Periodontitis/metabolismo , Endorribonucleasas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Masculino , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Células Cultivadas , Pérdida de Hueso Alveolar/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Humanos
4.
Transplantation ; 108(5): 1115-1126, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38192025

RESUMEN

BACKGROUND: The utilization of islet-like cells derived from pluripotent stem cells may resolve the scarcity of islet transplantation donors. The subcutaneous space is a promising transplantation site because of its capacity for graft observation and removal, thereby ensuring safety. To guarantee subcutaneous islet transplantation, physicians should ensure ample blood supply. Numerous methodologies, including prevascularization, have been investigated to augment blood flow, but the optimal approach remains undetermined. METHODS: From C57BL/6 mice, 500 syngeneic islets were transplanted into the prevascularized subcutaneous site of recipient mice by implanting agarose rods with basic fibroblast growth factor at 1 and 2 wk. Before transplantation, the blood glucose levels, cell infiltration, and cytokine levels at the transplant site were evaluated. Furthermore, we examined the impact of the extracellular matrix capsule on graft function and the inflammatory response. RESULTS: Compared with the 1-wk group, the 2-wk group exhibited improved glycemic control, indicating that longer prevascularization enhanced transplant success. Flow cytometry analysis detected immune cells, such as neutrophils and macrophages, in the extracellular matrix capsules, whereas cytometric bead array analysis indicated the release of inflammatory and proinflammatory cytokines. Treatment with antitumor necrosis factor and anti-interleukin-6R antibodies in the 1-wk group improved graft survival, similar to the 2-wk group. CONCLUSIONS: In early prevascularization before subcutaneous transplantation, neutrophil and macrophage accumulation prevented early engraftment owing to inflammatory cytokine production.


Asunto(s)
Glucemia , Citocinas , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/inmunología , Animales , Glucemia/metabolismo , Citocinas/metabolismo , Ratones , Masculino , Factores de Tiempo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/cirugía , Tejido Subcutáneo/irrigación sanguínea , Tejido Subcutáneo/inmunología , Matriz Extracelular/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/irrigación sanguínea , Neovascularización Fisiológica
5.
Biochem Pharmacol ; 216: 115764, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37634595

RESUMEN

Development of specific therapies that target and accelerate diabetic wound repair is an urgent need to alleviate pain and suffering and the huge socioeconomic burden of this debilitating disease. C-X-C Motif Chemokine Ligand 12 (CXCL12) also know an stromal cell-derived factor 1α (SDF-1α) is a chemokine that binds the CXC chemokine receptor type 4 (CXCR4) and activates downstream signaling resulting in recruitment of hematopoietic cells to locations of tissue injury and promotes tissue repair. In diabetes, low expression of CXCL12 correlates with impaired wound healing. Activation of CXCR4 receptor signaling with agonists or positive allosteric modulators (PAMs) provides a potential for small molecule therapeutic discovery and development. We recently reported high throughput screening and identification of the CXCR4 partial agonist UCUF-728, characterization of in vitro activity and reduced wound closure time in diabetic mice at 100 µM as a proof-of-concept study. We report here, the discovery of a second chemical scaffold demonstrating increased agonist potency and represented by thiadiazine derivative, UCUF-965. UCUF-965 is a potent partial agonist of ß-arrestin recruitment in CXCR4 receptor overexpressing cell line. Furthermore, UCUF-965 potentiates the CXCL12 maximal response in cAMP signaling pathway, activates CXCL12 stimulated migration in lymphoblast cells and modulates the levels of specific microRNA involved in the complex wound repair process, specifically in mouse fibroblasts. Our results indicate that UCUF-965 acts as a PAM agonist of the CXCR4 receptor. Furthermore, UCUF-965 enhanced angiogenesis markers and reduced wound healing time by 36% at 10.0 µM in diabetic mice models compared to untreated control.


Asunto(s)
Diabetes Mellitus Experimental , Receptores CXCR4 , Cicatrización de Heridas , Animales , Ratones , Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Células Madre Hematopoyéticas , Receptores CXCR4/agonistas , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
6.
Nutr Diabetes ; 13(1): 4, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031163

RESUMEN

BACKGROUND: Lentinan (LNT) is a complex fungal component that possesses effective antitumor and immunostimulating properties. However, there is a paucity of studies regarding the effects and mechanisms of LNT on type 1 diabetes. OBJECTIVE: In the current study, we investigated whether an intraperitoneal injection of LNT can diminish the risk of developing type 1 diabetes (T1D) in non-obese diabetic (NOD) mice and further examined possible mechanisms of LNT's effects. METHODS: Pre-diabetic female NOD mice 8 weeks of age, NOD mice with 140-160 mg/dL, 200-230 mg/dL or 350-450 mg/dL blood glucose levels were randomly divided into two groups and intraperitoneally injected with 5 mg/kg LNT or PBS every other day. Then, blood sugar levels, pancreas slices, spleen, PnLN and pancreas cells from treatment mice were examined. RESULTS: Our results demonstrated that low-dosage injections (5 mg/kg) of LNT significantly suppressed immunopathology in mice with autoimmune diabetes but increased the Foxp3+ regulatory T cells (Treg cells) proportion in mice. LNT treatment induced the production of Tregs in the spleen and PnLN cells of NOD mice in vitro. Furthermore, the adoptive transfer of Treg cells extracted from LNT-treated NOD mice confirmed that LNT induced Treg function in vivo and revealed an enhanced suppressive capacity as compared to the Tregs isolated from the control group. CONCLUSION: LNT was capable of stimulating the production of Treg cells from naive CD4 + T cells, which implies that LNT exhibits therapeutic values as a tolerogenic adjuvant and may be used to reverse hyperglycaemia in the early and late stages of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Lentinano , Estado Prediabético , Linfocitos T Reguladores , Animales , Femenino , Ratones , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Inyecciones Intraperitoneales , Lentinano/administración & dosificación , Lentinano/inmunología , Lentinano/farmacología , Lentinano/uso terapéutico , Ratones Endogámicos NOD , Estado Prediabético/tratamiento farmacológico , Estado Prediabético/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
7.
Int J Mol Sci ; 23(21)2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36362050

RESUMEN

Diabetic kidney disease (DKD) frequently leads to end-stage renal disease and other life-threatening illnesses. The dysregulation of glomerular cell types, including mesangial cells, endothelial cells, and podocytes, appears to play a vital role in the development of DKD. Myeloid-derived suppressor cells (MDSCs) exhibit immunoregulatory and anti-inflammatory properties through the depletion of L-arginine that is required by T cells, through generation of oxidative stress, interference with T-cell recruitment and viability, proliferation of regulatory T cells, and through the promotion of pro-tumorigenic functions. Under hyperglycemic conditions, mouse mesangial cells reportedly produce higher levels of fibronectin and pro-inflammatory cytokines. Moreover, the number of MDSCs is noticeably decreased, weakening inhibitory immune activities, and creating an inflammatory environment. In diabetic mice, immunotherapy with MDSCs that were induced by a combination of granulocyte-macrophage colony-stimulating factor, interleukin (IL)-1ß, and IL-6, reduced kidney to body weight ratio, fibronectin expression, and fibronectin accumulation in renal glomeruli, thus ameliorating DKD. In conclusion, MDSCs exhibit anti-inflammatory activities that help improve renal fibrosis in diabetic mice. The therapeutic targeting of the proliferative or immunomodulatory pathways of MDSCs may represent an alternative immunotherapeutic strategy for DKD.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Células Supresoras de Origen Mieloide , Animales , Ratones , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/inmunología , Nefropatías Diabéticas/metabolismo , Células Endoteliales/metabolismo , Fibronectinas/metabolismo , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/inmunología
8.
Elife ; 112022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35112667

RESUMEN

Infection is a major co-morbidity that contributes to impaired healing in diabetic wounds. Although impairments in diabetic neutrophils have been blamed for this co-morbidity, what causes these impairments and whether they can be overcome, remain largely unclear. Diabetic neutrophils, isolated from diabetic individuals, exhibit chemotaxis impairment but this peculiar functional impairment has been largely ignored because it appears to contradict the clinical findings which blame excessive neutrophil influx as a major impediment to healing in chronic diabetic ulcers. Here, we report that exposure to glucose in diabetic range results in impaired chemotaxis signaling through the formyl peptide receptor (FPR) in neutrophils, culminating in reduced chemotaxis and delayed neutrophil trafficking in the wound of Leprdb (db/db) type two diabetic mice, rendering diabetic wound vulnerable to infection. We further show that at least some auxiliary receptors remain functional under diabetic conditions and their engagement by the pro-inflammatory cytokine CCL3, overrides the requirement for FPR signaling and substantially improves infection control by jumpstarting the neutrophil trafficking toward infection, and stimulates healing in diabetic wound. We posit that CCL3 may have therapeutic potential for the treatment of diabetic foot ulcers if it is applied topically after the surgical debridement process which is intended to reset chronic ulcers into acute fresh wounds.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Diabetes Mellitus Experimental/inmunología , Neutrófilos/patología , Receptores de Formil Péptido/genética , Transducción de Señal/inmunología , Cicatrización de Heridas/inmunología , Infección de Heridas/microbiología , Animales , Quimiocina CCL3/inmunología , Complicaciones de la Diabetes/microbiología , Glucosa/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores de Formil Péptido/inmunología , Infección de Heridas/tratamiento farmacológico , Infección de Heridas/etiología
9.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163121

RESUMEN

Type 1 diabetes (T1D) is caused by the destruction of ß cells in pancreatic islets by autoimmune T cells. Islet transplantation has been established as an effective treatment for T1D. However, the survival of islet grafts is often disrupted by recurrent autoimmunity. Alpha-lipoic acid (ALA) has been reported to have immunomodulatory effects and, therefore, may have therapeutic potential in the treatment of T1D. In this study, we investigated the therapeutic potential of ALA in autoimmunity inhibition. We treated non-obese diabetic (NOD) mice with spontaneous diabetes and islet-transplantation mice with ALA. The onset of diabetes was decreased and survival of the islet grafts was extended. The populations of Th1 cells decreased, and regulatory T cells (Tregs) increased in ALA-treated mice. The in vitro Treg differentiation was significantly increased by treatment with ALA. The adoptive transfer of ALA-differentiated Tregs into NOD recipients improved the outcome of the islet grafts. Our results showed that in vivo ALA treatment suppressed spontaneous diabetes and autoimmune recurrence in NOD mice by inhibiting the Th1 immune response and inducing the differentiation of Tregs. Our study also demonstrated the therapeutic potential of ALA in Treg-based cell therapies and islet transplantation used in the treatment of T1D.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/citología , Linfocitos T Reguladores/inmunología , Ácido Tióctico/farmacología , Animales , Antioxidantes/farmacología , Diferenciación Celular , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Supervivencia de Injerto , Ratones , Ratones Endogámicos NOD , Células TH1
10.
FASEB J ; 36(1): e22107, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34939700

RESUMEN

Mounting evidence has linked the metabolic disease to neurovascular disorders and cognitive decline. Using a murine model of a high-fat high-sugar diet mimicking obesity-induced type 2 diabetes mellitus (T2DM) in humans, we show that pro-inflammatory mediators and altered immune responses damage the blood-brain barrier (BBB) structure, triggering a proinflammatory metabolic phenotype. We find that disruption to tight junctions and basal lamina due to loss of control in the production of matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) causes BBB impairment. Together the disruption to the structural and functional integrity of the BBB results in enhanced transmigration of leukocytes across the BBB that could contribute to an initiation of a neuroinflammatory response through activation of microglia. Using a humanized in vitro model of the BBB and T2DM patient post-mortem brains, we show the translatable applicability of our results. We find a leaky BBB phenotype in T2DM patients can be attributed to a loss of junctional proteins through changes in inflammatory mediators and MMP/TIMP levels, resulting in increased leukocyte extravasation into the brain parenchyma. We further investigated therapeutic avenues to reduce and restore the BBB damage caused by HFHS-feeding. Pharmacological treatment with recombinant annexin A1 (hrANXA1) or reversion from a high-fat high-sugar diet to a control chow diet (dietary intervention), attenuated T2DM development, reduced inflammation, and restored BBB integrity in the animals. Given the rising incidence of diabetes worldwide, understanding metabolic-disease-associated brain microvessel damage is vital and the proposed therapeutic avenues could help alleviate the burden of these diseases.


Asunto(s)
Barrera Hematoencefálica/inmunología , Colagenasas/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 2/inmunología , Inhibidores Tisulares de Metaloproteinasas/inmunología , Animales , Anexina A1/farmacología , Barrera Hematoencefálica/patología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Humanos , Masculino , Ratones , Proteínas Recombinantes/farmacología
11.
Life Sci ; 288: 120184, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838848

RESUMEN

AIMS: Rosmarinic acid (RA) is a polyphenol that occurs in plants of the Lamiaceae family. Phenethyl ester of RA (PERA), a novel RA derivative, has been developed and evaluated in vivo in an animal model of type 1 diabetes (T1D). METHODS: T1D was induced in male C57BL/6 mice using multiple low doses of streptozotocin (STZ) administered intraperitoneally for 5 consecutive days. Intraperitoneal administration of PERA (2.5 mg/kg bw) began from the first STZ injection and continued for 20 days. KEY FINDINGS: PERA-treated mice exhibited lower incidence of T1D (monitored up to 38 days from the disease induction), and fluorescent histochemical analysis showed that their pancreatic islets expressed more insulin. PERA treatment significantly down-regulated the proportions of CD11b+ and CD11c+ myeloid cells in the immune cell infiltrates in the pancreatic islets early during T1D pathogenesis (on day 9 after T1D induction), while on day 15, PERA significantly reduced the proportions of CD11c+, CD8+, Th1 and Th17 cells. Simultaneously, it was found that the cells from the pancreatic infiltrates of PERA-treated mice produced significantly less reactive oxygen species than cells from the control group. SIGNIFICANCE: These findings suggest that PERA efficiently prevented T1D development in mice. Interestingly, PERA attenuated the inflammatory process in the islets through temporally specific interference with the innate and adaptive immune response and therefore shows great promise for further clinical evaluation as a novel T1D therapeutic.


Asunto(s)
Autoinmunidad , Cinamatos/farmacología , Depsidos/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/prevención & control , Ésteres/química , Islotes Pancreáticos/efectos de los fármacos , Animales , Cinamatos/química , Depsidos/química , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Alcohol Feniletílico/química , Ácido Rosmarínico
12.
J Mol Med (Berl) ; 100(1): 101-113, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34651203

RESUMEN

Infections are common in patients with diabetes, but increasing antibiotic resistance hampers successful bacterial clearance and calls for alternative treatment strategies. Hypoxia-inducible factor 1 (HIF-1) is known to influence the innate immune defense and could therefore serve as a possible target. However, the impact of high glucose on HIF-1 has received little attention and merits closer investigation. Here, we show that higher levels of proinflammatory cytokines and CAMP, encoding for the antimicrobial peptide cathelicidin, LL-37, correlate with HIF-1 in type 2 diabetic patients. Chemical activation of HIF-1 further enhanced LL-37, IL-1ß, and IL-8 in human uroepithelial cells exposed to high glucose. Moreover, HIF-1 activation of transurethrally infected diabetic mice resulted in lower bacterial load. Drugs activating HIF-1 could therefore in the future potentially have a therapeutic role in clearing bacteria in diabetic patients with infections where antibiotic treatment failed. KEY MESSAGES: • Mohanty et al. "HIF-1 mediated activation of antimicrobial peptide LL-37 in type 2 diabetic patients." • Our study highlights induction of the antimicrobial peptide, LL-37, and strengthening of the innate immunity through hypoxia-inducible factor 1 (HIF-1) in diabetes. • Our key observations are: 1. HIF-1 activation increased LL-37 expression in human urothelial cells treated with high glucose. In line with that, we demonstrated that patients with type 2 diabetes living at high altitude had increased levels of the LL-37. 2. HIF-1 activation increased IL-1ß and IL-8 in human uroepithelial cells treated with high glucose concentration. 3. Pharmacological activation of HIF-1 decreased bacterial load in the urinary bladder of mice with hereditary diabetes. • We conclude that enhancing HIF-1 may along with antibiotics in the future contribute to the treatment in selected patient groups where traditional therapy is not possible.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 2/inmunología , Infecciones por Escherichia coli/inmunología , Factor 1 Inducible por Hipoxia/inmunología , Infecciones Urinarias/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Citocinas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Infecciones por Escherichia coli/genética , Femenino , Humanos , Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , Persona de Mediana Edad , Infecciones Urinarias/genética , Urotelio/citología , Catelicidinas
13.
Signal Transduct Target Ther ; 6(1): 409, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34848693

RESUMEN

Natural killer (NK) cells have been suggested to be associated with type 2 diabetes by regulating systemic inflammation. However, the mechanism by which NK cells regulate insulin sensitivity remains unknown. This study shows that NK-derived exosomes from lean mice attenuate obesity-induced insulin resistance and inflammation in mice of type 2 diabetes. Moreover, lean NK-derived exosomes enhance insulin sensitivity and relieve inflammation in adipocytes and hepatocytes. MiR-1249-3p, which is significantly upregulated in lean NK-derived exosomes, can be transferred from NK cells to adipocytes and hepatocytes via exosomes. NK-derived exosomal miR-1249-3p dramatically induces cellular insulin sensitivity and relieves inflammation. Mechanistically, exosomal miR-1249-3p directly targets SKOR1 to regulate the formation of ternary complex SMAD6/MYD88/SMURF1, which mediates glucose homeostasis by suppressing the TLR4/NF-κB signaling pathway. This study reveals an emerging role for NK-derived exosomal miR-1249-3p in remission of insulin resistance, and provides a series of potential therapeutic targets in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 2/inmunología , Exosomas/inmunología , Resistencia a la Insulina/inmunología , Células Asesinas Naturales/inmunología , MicroARNs/inmunología , Animales , Inflamación/inmunología , Masculino , Ratones
14.
Mediators Inflamm ; 2021: 9940009, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712101

RESUMEN

Alloxan (ALX) and streptozotocin (STZ) are extensively used to induce type 1 diabetes (T1D) in animal models. This study is aimed at evaluating the differences in immune parameters caused by ALX and STZ. T1D was induced either with ALX or with STZ, and the animals were followed for up to 180 days. Both ALX and STZ induced a decrease in the total number of circulating leukocytes and lymphocytes, with an increase in granulocytes when compared to control mice (CT). STZ-treated mice also exhibited an increase in neutrophils and a reduction in the lymphocyte percentage in the bone marrow. In addition, while the STZ-treated group showed a decrease in total CD3+, CD4-CD8+, and CD4+CD8+ T lymphocytes in the thymus and CD19+ B lymphocytes in the pancreas and spleen, the ALX group showed an increase in CD4-CD8+ and CD19+ only in the thymus. Basal levels of splenic interleukin- (IL-) 1ß and pancreatic IL-6 in the STZ group were decreased. Both diabetic groups showed atrophy of the thymic medulla and degeneration of pancreatic islets of Langerhans composed of inflammatory infiltration and hyperemia with vasodilation. ALX-treated mice showed a decrease in reticuloendothelial cells, enhanced lymphocyte/thymocyte cell death, and increased number of Hassall's corpuscles. Reduced in vitro activation of splenic lymphocytes was found in the STZ-treated group. Furthermore, mice immunized with ovalbumin (OVA) showed a more intense antigen-specific paw edema response in the STZ-treated group, while production of anti-OVA IgG1 antibodies was similar in both groups. Thereby, important changes in immune cell parameters in vivo and in vitro were found at an early stage of T1D in the STZ-treated group, whereas alterations in the ALX-treated group were mostly found in the chronic phase of T1D, including increased mortality rates. These findings suggest that the effects of ALX and STZ influenced, at different times, lymphoid organs and their cell populations.


Asunto(s)
Aloxano/toxicidad , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Linfocitos/efectos de los fármacos , Estreptozocina/toxicidad , Animales , Glucemia/análisis , Citocinas/biosíntesis , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos , Páncreas/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Timo/efectos de los fármacos , Timo/patología
15.
Cell Rep ; 37(5): 109942, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34706272

RESUMEN

Anti-viral monoclonal antibody (mAb) treatments may provide immediate but short-term immunity from coronavirus disease 2019 (COVID-19) in high-risk populations, such as people with diabetes and the elderly; however, data on their efficacy in these populations are limited. We demonstrate that prophylactic mAb treatment blocks viral replication in both the upper and lower respiratory tracts in aged, type 2 diabetic rhesus macaques. mAb infusion dramatically curtails severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated stimulation of interferon-induced chemokines and T cell activation, significantly reducing development of interstitial pneumonia. Furthermore, mAb infusion significantly dampens the greater than 3-fold increase in SARS-CoV-2-induced effector CD4 T cell influx into the cerebrospinal fluid. Our data show that neutralizing mAbs administered preventatively to high-risk populations may mitigate the adverse inflammatory consequences of SARS-CoV-2 exposure.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , COVID-19/prevención & control , SARS-CoV-2/inmunología , Envejecimiento/inmunología , Animales , COVID-19/líquido cefalorraquídeo , COVID-19/complicaciones , COVID-19/inmunología , Complicaciones de la Diabetes/inmunología , Complicaciones de la Diabetes/virología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/inmunología , Femenino , Humanos , Activación de Linfocitos , Macaca mulatta , Masculino , Neuritis/inmunología , Neuritis/prevención & control , Profilaxis Pre-Exposición , Linfocitos T/inmunología , Replicación Viral/inmunología
16.
J Neuroinflammation ; 18(1): 244, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702288

RESUMEN

BACKGROUND: Diabetes mellitus (DM) is a common concomitant disease of late-onset myasthenia gravis (MG). However, the impacts of DM on the progression of late-onset MG were unclear. METHODS: In this study, we examined the immune response in experimental autoimmune myasthenia gravis (EAMG) rats with DM or not. The phenotype and function of the spleen and lymph nodes were determined by flow cytometry. The serum antibodies, Tfh cells, and germinal center B cells were determined by ELISA and flow cytometry. The roles of advanced glycation end products (AGEs) in regulating Tfh cells were further explored in vitro by co-culture assays. RESULTS: Our results indicated clinical scores of EAMG rats were worse in diabetes rats compared to control, which was due to the increased production of anti-R97-116 antibody and antibody-secreting cells. Furthermore, diabetes induced a significant upregulation of Tfh cells and the subtypes of Tfh1 and Tfh17 cells to provide assistance for antibody production. The total percentages of B cells were increased with an activated statue of improved expression of costimulatory molecules CD80 and CD86. We found CD4+ T-cell differentiation was shifted from Treg cells towards Th1/Th17 in the DM+EAMG group compared to the EAMG group. In addition, in innate immunity, diabetic EAMG rats displayed more CXCR5 expression on NK cells. However, the expression of CXCR5 on NKT cells was down-regulated with the increased percentages of NKT cells in the DM+EAMG group. Ex vivo studies further indicated that Tfh cells were upregulated by AGEs instead of hyperglycemia. The upregulation was mediated by the existence of B cells, the mechanism of which might be attributed the elevated molecule CD40 on B cells. CONCLUSIONS: Diabetes promoted both adaptive and innate immunity and exacerbated clinical symptoms in EAMG rats. Considering the effect of diabetes, therapy in reducing blood glucose levels in MG patients might improve clinical efficacy through suppressing the both innate and adaptive immune responses. Additional studies are needed to confirm the effect of glucose or AGEs reduction to seek treatment for MG.


Asunto(s)
Inmunidad Adaptativa/fisiología , Diabetes Mellitus Experimental/inmunología , Inmunidad Innata/fisiología , Mediadores de Inflamación/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Técnicas de Cocultivo , Diabetes Mellitus Experimental/metabolismo , Femenino , Mediadores de Inflamación/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Miastenia Gravis Autoinmune Experimental/metabolismo , Ratas , Ratas Endogámicas Lew , Células Th17/inmunología , Células Th17/metabolismo
17.
Eur J Pharmacol ; 910: 174468, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34478692

RESUMEN

Acute kidney injury (AKI) is one of the major complications with increased oxidative stress and inflammation in diabetic patients. Hyperglycemia stimulates the formation of advanced glycation end products (AGEs). However, hyperglycemia directly triggers the interaction between AGEs and transmembrane AGEs receptors (RAGE), which enhances oxidative stress and increases the production of inflammatory substances. Therefore, diabetes plays a pivotal role in kidney injury. Hydralazine, a vasodilator and antihypertensive drug, was found to have the ability to reduce ROS, oxidative stress, and inflammation. We applied Hydralazine co-culture with AGEs in rat mesangial cells (RMC) and to renal ischemia/reperfusion(I/R) injury models in streptozotocin-induced diabetic rats. Hydralazine significantly decreased AGEs-induced RAGE, iNOS, and COX-2 expressions in RMC. Compared to the diabetic with AKI group, hydralazine decreased inflammation-related protein, and JAK2, STAT3 signaling in rat kidney tissue. Our studies indicate that Hydralazine has the potential to become a beneficial drug in the treatment of diabetic acute kidney injury.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Nefropatías Diabéticas/tratamiento farmacológico , Hidralazina/farmacología , Nefritis/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Animales , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/inmunología , Nefropatías Diabéticas/inmunología , Nefropatías Diabéticas/patología , Productos Finales de Glicación Avanzada , Humanos , Hidralazina/uso terapéutico , Masculino , Células Mesangiales , Nefritis/inmunología , Nefritis/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Cultivo Primario de Células , Ratas , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Estreptozocina/administración & dosificación , Estreptozocina/toxicidad
18.
Front Immunol ; 12: 616215, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34447366

RESUMEN

Tolerogenic vaccinations using beta-cell antigens are attractive for type 1 diabetes prevention, but clinical trials have been disappointing. This is probably due to the late timing of intervention, when multiple auto-antibodies are already present. We therefore devised a strategy to introduce the initiating antigen preproinsulin (PPI) during neonatal life, when autoimmunity is still silent and central tolerance mechanisms, which remain therapeutically unexploited, are more active. This strategy employs an oral administration of PPI-Fc, i.e. PPI fused with an IgG Fc to bind the intestinal neonatal Fc receptor (FcRn) that physiologically delivers maternal antibodies to the offspring during breastfeeding. Neonatal oral PPI-Fc vaccination did not prevent diabetes development in PPI T-cell receptor-transgenic G9C8.NOD mice. However, PPI-Fc was efficiently transferred through the intestinal epithelium in an Fc- and FcRn-dependent manner, was taken up by antigen presenting cells, and reached the spleen and thymus. Although not statistically significant, neonatal oral PPI-Fc vaccination delayed diabetes onset in polyclonal Ins2-/-.NOD mice that spontaneously develop accelerated diabetes. Thus, this strategy shows promise in terms of systemic and thymic antigen delivery via the intestinal FcRn pathway, but the current PPI-Fc formulation/regimen requires further improvements to achieve diabetes prevention.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Antígenos de Histocompatibilidad Clase I/inmunología , Insulina/farmacología , Precursores de Proteínas/farmacología , Receptores Fc/inmunología , Proteínas Recombinantes de Fusión/farmacología , Timo/inmunología , Administración Oral , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Insulina/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Precursores de Proteínas/genética , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética
19.
J Exp Med ; 218(10)2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34415994

RESUMEN

Understanding mechanisms of immune regulation is key to developing immunotherapies for autoimmunity and cancer. We examined the role of mononuclear phagocytes during peripheral T cell regulation in type 1 diabetes and melanoma. MERTK expression and activity in mononuclear phagocytes in the pancreatic islets promoted islet T cell regulation, resulting in reduced sensitivity of T cell scanning for cognate antigen in prediabetic islets. MERTK-dependent regulation led to reduced T cell activation and effector function at the disease site in islets and prevented rapid progression of type 1 diabetes. In human islets, MERTK-expressing cells were increased in remaining insulin-containing islets of type 1 diabetic patients, suggesting that MERTK protects islets from autoimmune destruction. MERTK also regulated T cell arrest in melanoma tumors. These data indicate that MERTK signaling in mononuclear phagocytes drives T cell regulation at inflammatory disease sites in peripheral tissues through a mechanism that reduces the sensitivity of scanning for antigen leading to reduced responsiveness to antigen.


Asunto(s)
Autoinmunidad/fisiología , Islotes Pancreáticos/enzimología , Fagocitos/fisiología , Linfocitos T/inmunología , Tirosina Quinasa c-Mer/inmunología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos/inmunología , Antígenos/metabolismo , Antígenos CD11/metabolismo , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/patología , Femenino , Humanos , Islotes Pancreáticos/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/inmunología , Fagocitos/inmunología , Piperazinas/farmacología , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo
20.
Circulation ; 144(12): 961-982, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34255973

RESUMEN

BACKGROUND: Cardiovascular risk in diabetes remains elevated despite glucose-lowering therapies. We hypothesized that hyperglycemia induces trained immunity in macrophages, promoting persistent proatherogenic characteristics. METHODS: Bone marrow-derived macrophages from control mice and mice with diabetes were grown in physiological glucose (5 mmol/L) and subjected to RNA sequencing (n=6), assay for transposase accessible chromatin sequencing (n=6), and chromatin immunoprecipitation sequencing (n=6) for determination of hyperglycemia-induced trained immunity. Bone marrow transplantation from mice with (n=9) or without (n=6) diabetes into (normoglycemic) Ldlr-/- mice was used to assess its functional significance in vivo. Evidence of hyperglycemia-induced trained immunity was sought in human peripheral blood mononuclear cells from patients with diabetes (n=8) compared with control subjects (n=16) and in human atherosclerotic plaque macrophages excised by laser capture microdissection. RESULTS: In macrophages, high extracellular glucose promoted proinflammatory gene expression and proatherogenic functional characteristics through glycolysis-dependent mechanisms. Bone marrow-derived macrophages from diabetic mice retained these characteristics, even when cultured in physiological glucose, indicating hyperglycemia-induced trained immunity. Bone marrow transplantation from diabetic mice into (normoglycemic) Ldlr-/- mice increased aortic root atherosclerosis, confirming a disease-relevant and persistent form of trained innate immunity. Integrated assay for transposase accessible chromatin, chromatin immunoprecipitation, and RNA sequencing analyses of hematopoietic stem cells and bone marrow-derived macrophages revealed a proinflammatory priming effect in diabetes. The pattern of open chromatin implicated transcription factor Runt-related transcription factor 1 (Runx1). Similarly, transcriptomes of atherosclerotic plaque macrophages and peripheral leukocytes in patients with type 2 diabetes were enriched for Runx1 targets, consistent with a potential role in human disease. Pharmacological inhibition of Runx1 in vitro inhibited the trained phenotype. CONCLUSIONS: Hyperglycemia-induced trained immunity may explain why targeting elevated glucose is ineffective in reducing macrovascular risk in diabetes and suggests new targets for disease prevention and therapy.


Asunto(s)
Aterosclerosis/inmunología , Diabetes Mellitus Experimental/inmunología , Hiperglucemia/inmunología , Inmunidad Celular/inmunología , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Animales , Aterosclerosis/patología , Células Cultivadas , Diabetes Mellitus Experimental/patología , Endarterectomía Carotidea , Humanos , Hiperglucemia/patología , Leucocitos Mononucleares/patología , Macrófagos/patología , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...