Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Med ; 30(1): 23, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38317106

RESUMEN

BACKGROUND: Fluvoxamine is one of the selective serotonin reuptake inhibitors (SSRIs) that are regarded as the first-line drugs to manage mental disorders. It has been also recognized with the potential to treat inflammatory diseases and viral infection. However, the effect of fluvoxamine on autoimmune diseases, particularly type 1 diabetes (T1D) and the related cellular and molecular mechanisms, are yet to be addressed. METHOD: Herein in this report, we treated NOD mice with fluvoxamine for 2 weeks starting from 10-week of age to dissect the impact of fluvoxamine on the prevention of type 1 diabetes. We compared the differences of immune cells between 12-week-old control and fluvoxamine-treated mice by flow cytometry analysis. To study the mechanism involved, we extensively examined the characteristics of CD4+ T cells with fluvoxamine stimulation using RNA-seq analysis, real-time PCR, Western blot, and seahorse assay. Furthermore, we investigated the relevance of our data to human autoimmune diabetes. RESULT: Fluvoxamine not only delayed T1D onset, but also decreased T1D incidence. Moreover, fluvoxamine-treated NOD mice showed significantly attenuated insulitis coupled with well-preserved ß cell function, and decreased Th1 and Th17 cells in the peripheral blood, pancreatic lymph nodes (PLNs), and spleen. Mechanistic studies revealed that fluvoxamine downregulated glycolytic process by inhibiting phosphatidylinositol 3-kinase (PI3K)-AKT signaling, by which it restrained effector T (Teff) cell differentiation and production of proinflammatory cytokines. CONCLUSION: Collectively, our study supports that fluvoxamine could be a viable therapeutic drug against autoimmunity in T1D setting.


Asunto(s)
Enfermedades Autoinmunes , Diabetes Mellitus Tipo 1 , Ratones , Humanos , Animales , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Ratones Endogámicos NOD , Fluvoxamina/farmacología , Fluvoxamina/uso terapéutico , Células Th17 , Fosfatidilinositol 3-Quinasas , Células TH1
2.
Brain Behav Immun ; 108: 32-44, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36343753

RESUMEN

BACKGROUND: Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons (DA) and the accumulation of Lewy body deposits composed of alpha-Synuclein (α-Syn), which act as antigenic epitopes to drive cytotoxic T-cell responses in PD. Increased T helper 17 (Th17) cells and dysfunctional regulatory T cells (Tregs) have been reported to be associated with the loss of DA in PD. However, the mechanism underlying the Th17/Treg imbalance remains unknown. METHODS: Here, we examined the percentage of Th17 cells, the percentage of Tregs and the α-Syn level and analysed their correlations in the peripheral blood of PD patients and in the substantia nigra pars compacta (SNpc) and spleen of MPTP-treated mice and A53 transgenic mice. We assessed the effect of α-Syn on the stability and function of Tregs and the differentiation of Th17 cells and evaluated the role of retinoid-related orphan nuclear receptor (RORγt) upregulation in α-Syn stimulation in vivo and in vitro. RESULTS: We found that the α-Syn level and severity of motor symptoms were positively correlated with the increase in Th17 cells and decrease in Tregs in PD patients. Moreover, α-Syn stimulation led to the loss of Forkhead box protein P3 (FOXP3) expression in Tregs, accompanied by the acquisition of IL-17A expression. Increased Th17 differentiation was detected upon α-Syn stimulation when naïve CD4+ T cells were cultured under Th17-polarizing conditions. Mechanistically, α-Syn promotes the transcription of RORC, encoding RORγt, in Tregs and Th17 cells, leading to increased Th17 differentiation and loss of Treg function. Intriguingly, the increase in Th17 cells, decrease in Tregs and apoptosis of DA were suppressed by a RORγt inhibitor (GSK805) in MPTP-treated mice. CONCLUSION: Together, our data suggest that α-Syn promotes the transcription of RORC in circulating CD4+ T cells, including Tregs and Th17 cells, to impair the stability of Tregs and promote the differentiation of Th17 cells in PD. Inhibition of RORγt attenuated the apoptosis of DA and alleviated the increase in Th17 cells and decrease in Tregs in PD.


Asunto(s)
Enfermedad de Parkinson , Ratones , Animales , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores , Diferenciación Celular , Ratones Transgénicos , Células Th17/metabolismo
3.
Neurobiol Dis ; 163: 105605, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973450

RESUMEN

Parkinson's disease (PD) is characterized by impaired mitochondrial function and decreased ATP levels. Aerobic glycolysis and lactate production have been shown to be upregulated in dopaminergic neurons to sustain ATP levels, but the effect of upregulated glycolysis on dopaminergic neurons remains unknown. Since lactate promotes apoptosis and α-synuclein accumulation in neurons, we hypothesized that the lactate produced upon upregulated glycolysis is involved in the apoptosis of dopaminergic neurons in PD. In this study, we examined the expression of hexokinase 2 (HK2) and lactate dehydrogenase (LDH), the key enzymes in glycolysis, and lactate levels in the substantia nigra pars compacta (SNpc) of a MPTP-induced mouse model of PD and in MPP+-treated SH-SY5Y cells. We found that the expression of HK2 and LDHA and the lactate levels were markedly increased in the SNpc of MPTP-treated mice and in MPP+-treated SH-SY5Y cells. Exogenous lactate treatment led to the apoptosis of SH-SY5Y cells. Intriguingly, lactate production and the apoptosis of dopaminergic neurons were suppressed by the application of 3-bromopyruvic acid (3-Brpa), a HK2 inhibitor, or siRNA both in vivo and in vitro. 3-Brpa treatment markedly improved the motor behaviour of MPTP-treated mice in pole test and rotarod test. Mechanistically, lactate increases the activity of adenosine monophosphate-activated protein kinase (AMPK) and suppresses the phosphorylation of serine/threonine kinase 1 (Akt) and mammalian target of rapamycin (mTOR). Together, our data suggest that upregulated HK2 and LDHA and increased lactate levels prompt the apoptosis of dopaminergic neurons in PD. Inhibition of HK2 expression attenuated the apoptosis of dopaminergic neurons by downregulating lactate production and AMPK/Akt/mTOR pathway in PD.


Asunto(s)
Apoptosis/fisiología , Neuronas Dopaminérgicas/metabolismo , Hexoquinasa/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/metabolismo , Trastornos Parkinsonianos/metabolismo , Porción Compacta de la Sustancia Negra/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Hexoquinasa/genética , Humanos , L-Lactato Deshidrogenasa/genética , Ratones , Actividad Motora/efectos de los fármacos , Trastornos Parkinsonianos/genética , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Piruvatos/farmacología , Regulación hacia Arriba
4.
Eur Respir J ; 60(3)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35086828

RESUMEN

Although DNA methylation has been recognised in the pathogenesis of idiopathic pulmonary fibrosis (IPF), the exact mechanisms are yet to be fully addressed. Herein, we demonstrate that lungs originated from IPF patients and mice after bleomycin (BLM)-induced pulmonary fibrosis are characterised by altered DNA methylation along with overexpression in myofibroblasts of methyl-CpG-binding domain 2 (MBD2), a reader responsible for interpreting DNA methylome-encoded information. Specifically, depletion of Mbd2 in fibroblasts or myofibroblasts protected mice from BLM-induced pulmonary fibrosis coupled with a significant reduction of fibroblast differentiation. Mechanistically, transforming growth factor (TGF)-ß1 induced a positive feedback regulatory loop between TGF-ß receptor I (TßRI), Smad3 and Mbd2, and erythroid differentiation regulator 1 (Erdr1). TGF-ß1 induced fibroblasts to undergo a global DNA hypermethylation along with Mbd2 overexpression in a TßRI/Smad3 dependent manner, and Mbd2 selectively bound to the methylated CpG DNA within the Erdr1 promoter to repress its expression, through which it enhanced TGF-ß/Smad signalling to promote differentiation of fibroblast into myofibroblast and exacerbate pulmonary fibrosis. Therefore, enhancing Erdr1 expression strikingly reversed established pulmonary fibrosis. Collectively, our data support that strategies aimed at silencing Mbd2 or increasing Erdr1 could be viable therapeutic approaches for prevention and treatment of pulmonary fibrosis in clinical settings.


Asunto(s)
Fibrosis Pulmonar Idiopática , Miofibroblastos , Animales , Bleomicina/efectos adversos , Diferenciación Celular , ADN , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Ratones , Miofibroblastos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Crecimiento Transformadores/efectos adversos , Factores de Crecimiento Transformadores/metabolismo
5.
Diabetologia ; 63(5): 987-1001, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32072192

RESUMEN

AIMS/HYPOTHESIS: High-mobility group box 1 (HMGB1), an evolutionarily conserved chromosomal protein, was rediscovered to be a 'danger signal' (alarmin) that alerts the immune system once released extracellularly. Therefore, it has been recognised contributing to the pathogenesis of autoimmune diabetes, but its exact impact on the initiation and progression of type 1 diabetes, as well as the related molecular mechanisms, are yet to be fully characterised. METHODS: In the current report, we employed NOD mice as a model to dissect the impact of blocking HMGB1 on the prevention, treatment and reversal of type 1 diabetes. To study the mechanism involved, we extensively examined the characteristics of regulatory T cells (Tregs) and their related signalling pathways upon HMGB1 stimulation. Furthermore, we investigated the relevance of our data to human autoimmune diabetes. RESULTS: Neutralising HMGB1 both delayed diabetes onset and, of particular relevance, reversed diabetes in 13 out of 20 new-onset diabetic NOD mice. Consistently, blockade of HMGB1 prevented islet isografts from autoimmune attack in diabetic NOD mice. Using transgenic reporter mice that carry a Foxp3 lineage reporter construct, we found that administration of HMGB1 impairs Treg stability and function. Mechanistic studies revealed that HMGB1 activates receptor for AGE (RAGE) and toll-like receptor (TLR)4 to enhance phosphatidylinositol 3-kinase (PI3K)-Akt-mechanistic target of rapamycin (mTOR) signalling, thereby impairing Treg stability and functionality. Indeed, high circulating levels of HMGB1 in human participants with type 1 diabetes contribute to Treg instability, suggesting that blockade of HMGB1 could be an effective therapy against type 1 diabetes in clinical settings. CONCLUSIONS/INTERPRETATION: The present data support the possibility that HMGB1 could be a viable therapeutic target to prevent the initiation, progression and recurrence of autoimmunity in the setting of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Proteína HMGB1/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Western Blotting , Células Cultivadas , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Diabetes Mellitus Tipo 1/patología , Femenino , Proteína HMGB1/antagonistas & inhibidores , Humanos , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Fosfatidilinositol 3-Quinasas/metabolismo
6.
Int J Nanomedicine ; 19: 4411-4427, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774028

RESUMEN

Background: Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by synovial inflammation and joint destruction. Despite progress in RA therapy, it remains difficult to achieve long-term remission in RA patients. Phosphodiesterase 3B (Pde3b) is a member of the phosphohydrolyase family that are involved in many signal transduction pathways. However, its role in RA is yet to be fully addressed. Methods: Studies were conducted in arthritic DBA/1 mice, a suitable mouse strain for collagen-induced rheumatoid arthritis (CIA), to dissect the role of Pde3b in RA pathogenesis. Next, RNAi-based therapy with Pde3b siRNA-loaded liposomes was assessed in a CIA model. To study the mechanism involved, we investigated the effect of Pde3b knockdown on macrophage polarization and related signaling pathway. Results: We demonstrated that mice with CIA exhibited upregulated Pde3b expression in macrophages. Notably, intravenous administration of liposomes loaded with Pde3b siRNA promoted the macrophage anti-inflammatory program and alleviated CIA in mice, as indicated by the reduced inflammatory response, synoviocyte infiltration, and bone and cartilage erosion. Mechanistic study revealed that depletion of Pde3b increased cAMP levels, by which it enhanced PKA-CREB-C/EBPß pathway to transcribe the expression of anti-inflammatory program-related genes. Conclusion: Our results support that Pde3b is involved in the pathogenesis of RA, and Pde3b siRNA-loaded liposomes might serve as a promising therapeutic approach against RA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Terapia Genética , Liposomas , Macrófagos , Ratones Endogámicos DBA , ARN Interferente Pequeño , Animales , Liposomas/química , Liposomas/administración & dosificación , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/terapia , Artritis Reumatoide/inducido químicamente , Ratones , Artritis Experimental/genética , Artritis Experimental/prevención & control , Artritis Experimental/terapia , Macrófagos/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/administración & dosificación , Terapia Genética/métodos , Masculino , Transducción de Señal/efectos de los fármacos
7.
Cell Death Dis ; 14(11): 737, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957143

RESUMEN

SUMOylation is an evolutionary conserved regulatory mechanism, in which Ubc9 is the only E2 conjugating enzyme. Previous studies demonstrated that SUMOylation is involved in multiple biological processes, but its role in dendritic cells (DCs) remains to be fully addressed. Herein in this report, we found that DCs deficient in Ubc9 protected mice from dextran sulfate sodium (DSS)-induced colitis, as evidenced by the ameliorated weight loss, colon length, and disrupted colon structure. Mechanistically, Ubc9 mediated SUMOylation of RBPJ, by which it stabilized RBPJ from ubiquitin-mediated degradation to enhance its transcriptional activity, while Ciita, a critical transcription factor, is a direct target downstream of RBPJ, which forms an enhanceosome complex to transcribe the expression of MHC II genes. Therefore, loss of Ubc9 abolished RBPJ SUMOylation, which was coupled with reduced Ciita transcription, thereby attenuating the expression of MHC class II genes. As a consequence of defective MHC II expression, Ubc9-/- DCs were featured by the impaired capability to process antigen and to prime effector CD4+ T cells, thereby protecting mice from DSS-induced colitis. Together, our results shed novel insight into the understanding of SUMOylation in the regulation of DC functions in pathological conditions.


Asunto(s)
Colitis , Sumoilación , Enzimas Ubiquitina-Conjugadoras , Animales , Ratones , Antígenos , Colitis/inducido químicamente , Colitis/genética , Células Dendríticas , Regulación de la Expresión Génica , Linfocitos T , Enzimas Ubiquitina-Conjugadoras/genética , Genes MHC Clase II
8.
Nat Commun ; 14(1): 5977, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37749140

RESUMEN

The regulation of autoimmunity against pancreatic islet ß cells for type 1 diabetes (T1D) onset is still unclear. NOD/ShiLtJ (NOD) mice are prone to the onset of autoimmune diabetes, but its congenic strain, ALR/Lt (ALR), is not. Here we show that dendritic cells (DC) in ALR mice have impaired migratory and T-cell priming capability. Genomic comparative analysis maps a 33-bp deletion in the ALR Myosin IXb (Myo9b) gene when compared with NOD genome; meanwhile, data from knock-in models show that this ALR Myo9b allele impairs phenotypic and functional maturation of DCs, and prevents the development and progression of spontaneous autoimmune diabetes in NOD mice. In parallel, while the ALR 33-bp deletion of Myo9b is not conserved in human, we find a MYO9B R133Q polymorphism associating with increased risk of T1D and enhanced DC function in patients with T1D. Our results thus hint that alterations in Myo9b may contribute to altered DC function and autoimmune diabetes onset.


Asunto(s)
Células Dendríticas , Diabetes Mellitus Tipo 1 , Miosinas , Animales , Humanos , Ratones , Alelos , Diabetes Mellitus Tipo 1/genética , Ratones Endogámicos NOD , Mutación , Miosinas/genética
9.
Front Immunol ; 13: 902318, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757774

RESUMEN

Asthma is a multifactorial disorder characterized by the airway chronic inflammation, hyper-responsiveness (AHR), remodeling, and reversible obstruction. Although asthma is known as a heterogeneous group of diseases with various clinical manifestations, recent studies suggest that more than half of the clinical cases are ''T helper type 2 (Th2)-high'' type, whose pathogenesis is driven by Th2 responses to an inhaled allergen from the environmental exposures. The intensity and duration of inflammatory responses to inhaled allergens largely depend on the balance between effector and regulatory cells, but many questions regarding the mechanisms by which the relative magnitudes of these opposing forces are remained unanswered. Regulatory T cells (Tregs), which comprise diverse subtypes with suppressive function, have long been attracted extensive attention owing to their capability to limit the development and progression of allergic diseases. In this review we seek to update the recent advances that support an essential role for Tregs in the induction of allergen tolerance and attenuation of asthma progression once allergic airway inflammation established. We also discuss the current concepts about Treg induction and Treg-expressed mediators relevant to controlling asthma, and the therapies designed based on these novel insights against asthma in clinical settings.


Asunto(s)
Asma , Linfocitos T Reguladores , Alérgenos/uso terapéutico , Asma/patología , Humanos , Tolerancia Inmunológica , Inflamación
10.
Chin Med J (Engl) ; 134(9): 1031-1042, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33813508

RESUMEN

ABSTRACT: Type 1 diabetes (T1D) is an autoimmune disease that resulted from the severe destruction of the insulin-producing ß cells in the pancreases of individuals with a genetic predisposition. Genome-wide studies have identified HLA and other risk genes associated with T1D susceptibility in humans. However, evidence obtained from the incomplete concordance of diabetes incidence among monozygotic twins suggests that environmental factors also play critical roles in T1D pathogenesis. Epigenetics is a rapidly growing field that serves as a bridge to link T1D risk genes and environmental exposures, thereby modulating the expression of critical genes relevant to T1D development beyond the changes of DNA sequences. Indeed, there is compelling evidence that epigenetic changes induced by environmental insults are implicated in T1D pathogenesis. Herein, we sought to summarize the recent progress in terms of epigenetic mechanisms in T1D initiation and progression, and discuss their potential as biomarkers and therapeutic targets in the T1D setting.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 1/genética , Epigénesis Genética/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Incidencia , Gemelos Monocigóticos
11.
Cell Death Differ ; 28(6): 1880-1899, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33462408

RESUMEN

Kdm2a catalyzes H3K36me2 demethylation to play an intriguing epigenetic regulatory role in cell proliferation, differentiation, and apoptosis. Herein we found that myeloid-specific knockout of Kdm2a (LysM-Cre-Kdm2af/f, Kdm2a-/-) promoted macrophage M2 program by reprograming metabolic homeostasis through enhancing fatty acid uptake and lipolysis. Kdm2a-/- increased H3K36me2 levels at the Pparg locus along with augmented chromatin accessibility and Stat6 recruitment, which rendered macrophages with preferential M2 polarization. Therefore, the Kdm2a-/- mice were highly protected from high-fat diet (HFD)-induced obesity, insulin resistance, and hepatic steatosis, and featured by the reduced accumulation of adipose tissue macrophages and repressed chronic inflammation following HFD challenge. Particularly, Kdm2a-/- macrophages provided a microenvironment in favor of thermogenesis. Upon HFD or cold challenge, the Kdm2a-/- mice manifested higher capacity for inducing adipose browning and beiging to promote energy expenditure. Collectively, our findings demonstrate the importance of Kdm2a-mediated H3K36 demethylation in orchestrating macrophage polarization, providing novel insight that targeting Kdm2a in macrophages could be a viable therapeutic approach against obesity and insulin resistance.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/deficiencia , Macrófagos/metabolismo , Obesidad/genética , PPAR gamma/metabolismo , Animales , Dieta Alta en Grasa , Humanos , Masculino , Ratones , Obesidad/patología , Termogénesis , Transfección
12.
Sci Adv ; 7(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33277324

RESUMEN

Despite past extensive studies, the mechanisms underlying pulmonary fibrosis (PF) still remain poorly understood. Here, we demonstrated that lungs originating from different types of patients with PF, including coronavirus disease 2019, systemic sclerosis-associated interstitial lung disease, and idiopathic PF, and from mice following bleomycin (BLM)-induced PF are characterized by the altered methyl-CpG-binding domain 2 (MBD2) expression in macrophages. Depletion of Mbd2 in macrophages protected mice against BLM-induced PF. Mbd2 deficiency significantly attenuated transforming growth factor-ß1 (TGF-ß1) production and reduced M2 macrophage accumulation in the lung following BLM induction. Mechanistically, Mbd2 selectively bound to the Ship promoter in macrophages, by which it repressed Ship expression and enhanced PI3K/Akt signaling to promote the macrophage M2 program. Therefore, intratracheal administration of liposomes loaded with Mbd2 siRNA protected mice from BLM-induced lung injuries and fibrosis. Together, our data support the possibility that MBD2 could be a viable target against PF in clinical settings.


Asunto(s)
COVID-19/metabolismo , Proteínas de Unión al ADN/metabolismo , Macrófagos/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Bleomicina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Liposomas/química , Enfermedades Pulmonares Intersticiales/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/virología , ARN Interferente Pequeño/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
13.
Cell Death Differ ; 28(4): 1270-1283, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33144678

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is the most common type of idiopathic interstitial pneumonia and has one of the poorest prognosis. However, the molecular mechanisms underlying IPF progression remain largely unknown. In this study, we determined that IL-24, an IL-20 subfamily cytokine member, was increased both in the serum of IPF patients and the bronchoalveolar lavage fluid (BALF) of mice following bleomycin (BLM)-induced pulmonary fibrosis. As a result, IL-24 deficiency protected mice from BLM-induced lung injury and fibrosis. Specifically, loss of IL-24 significantly attenuated transforming growth factor ß1 (TGF-ß1) production and reduced M2 macrophage infiltration in the lung of BLM-induced mice. Mechanistically, IL-24 alone did not show a perceptible impact on the induction of M2 macrophages, but it synergized with IL-4 to promote M2 program in macrophages. IL-24 suppressed IL-4-induced expression of suppressor of cytokine signaling 1 (SOCS1) and SOCS3, through which it enhanced signal transducer and activator of transcription 6/peroxisome proliferator-activated receptor gamma (STAT6/PPARγ) signaling, thereby promoting IL-4-induced production of M2 macrophages. Collectively, our data support that IL-24 synergizes with IL-4 to promote macrophage M2 program contributing to the development of pulmonary fibrosis.


Asunto(s)
Bleomicina/efectos adversos , Interleucina-4/metabolismo , Interleucinas/deficiencia , Macrófagos/metabolismo , Fibrosis Pulmonar/prevención & control , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
14.
Mol Immunol ; 109: 43-50, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30852245

RESUMEN

Autophagy is an evolutionarily conserved catabolic process that directs cytoplasmic proteins, organelles and microbes to lysosomes for degradation. It not only represents an essential cell-intrinsic mechanism to protect against internal and external stresses but also shapes both innate and adaptive immunity. Regulatory T cells (Tregs) are a developmentally and functionally distinct T cell subpopulation engaged in sustaining immunological self-tolerance and homeostasis. There is compelling evidence that autophagy is actively regulated in Tregs and serves as a central signal-dependent controller for Tregs by restraining excessive apoptotic and metabolic activities. In this review, we discuss how autophagy modulates the stability and functionality of Tregs in different disease settings, and provide a perspective on how manipulation of autophagy enables better control of immune response by targeting the generation of Tregs and the maintenance of their stability.


Asunto(s)
Autofagia , Enfermedad , Linfocitos T Reguladores/inmunología , Animales , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Modelos Biológicos , Transducción de Señal
15.
Mol Immunol ; 114: 314-322, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31442915

RESUMEN

Hematopoietic development occurs in the bone marrow, and this process begins with hematopoietic stem cells (HSCs). Ubc9 is a unique E2-conjugating enzyme required for SUMOylation, an evolutionarily conserved post-translational modification system. We herein show that a conditional Ubc9 deletion in the hematopoietic system caused decreased thymus weight and reduced lymphocyte to myeloid cell ratio. Importantly, Ubc9 deletion in the hematopoietic system only selectively impaired the development of common lymphoid progenitors (CLPs) in the bone marrow and perturbed their potential to differentiate into lymphocytes, thereby decreasing the number of T/B cells in the periphery. Ubc9 was found to be required for CLP viability, and therefore, Ubc9 deficiency rendered CLPs to undergo apoptosis and attenuated their proliferation. Thus, Ubc9 plays a critical role in the regulation of CLP function during hematopoietic development in the bone marrow.


Asunto(s)
Médula Ósea/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/inmunología , Animales , Apoptosis/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Masculino , Ratones , Células Progenitoras Mieloides/inmunología , Linfocitos T/inmunología
16.
Cell Death Dis ; 10(12): 892, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31767832

RESUMEN

Type 1 diabetes (T1D) is characterized by the selective autoimmune destruction of the islet ß cells, and macrophages play a significant role in this process. Small ubiquitin-like modification (SUMOylation) is an important posttranslational modification involved in T1D pathogenesis, but its function in macrophages remains unexplored. We presently developed and used macrophage-specific ubiquitin-conjugating enzyme E2 (Ubc9) knockout (LyzM-Cre-Ubc9fl/fl, KO) mice to address the impact of SUMOylation on macrophage function in a T1D model. We observed that blocking Ubc9 in macrophages exacerbated multiple-low dose streptozotocin (MLD-STZ)-induced diabetes. Specifically, after STZ treatment, blood glucose levels were consistently elevated in the KO mice. The KO mice exhibited a higher diabetes incidence than WT controls (85% vs. 55%, P < 0.01) along with a higher insulitis severity. The loss of Ubc9 impaired macrophage energy metabolism and attenuated macrophage M2 program, thereby enhancing T cell activation. Pancreas-resident macrophages, rather than migrant macrophages, played a predominant role in MLD-STZ-induced diabetes. Mechanistically, Ubc9-mediated SUMOylation of interferon regulator factor 4 (IRF4) enhanced its nuclear localization and stability, thereby transcribing IL-4 and arginase 1 (Arg1) to promote the macrophage M2 program. Ubc9-mediated SUMOylation modulates T1D risk at least in part by regulating macrophage function. Modulation of disturbed SUMOylation process in macrophages, either through cell adoptive transfer or targeted drug-delivery, could help to establish a tolerant pancreatic microenvironment and promote inflammation resolution in early insulitis stage, thus hindering T1D progression.


Asunto(s)
Polaridad Celular , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Progresión de la Enfermedad , Macrófagos/enzimología , Macrófagos/patología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Animales , Antígenos/metabolismo , Arginasa/genética , Arginasa/metabolismo , Células de la Médula Ósea/metabolismo , Movimiento Celular , Núcleo Celular/metabolismo , Respiración de la Célula , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/patología , Glucólisis , Inflamación/metabolismo , Inflamación/patología , Factores Reguladores del Interferón/metabolismo , Macrófagos/inmunología , Ratones Noqueados , Mitocondrias/metabolismo , Páncreas/metabolismo , Páncreas/patología , Regiones Promotoras Genéticas/genética , Estabilidad Proteica , Estreptozocina , Sumoilación , Linfocitos T Reguladores/inmunología , Enzimas Ubiquitina-Conjugadoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA