Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Inflamm Res ; 17: 1643-1658, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38504697

RESUMO

Background: Intracerebral hemorrhage (ICH), a devastating form of stroke, is characterized by elevated morbidity and mortality rates. Neuroinflammation is a common occurrence following ICH. Mesenchymal stem cells (MSCs) have exhibited potential in treating brain diseases due to their anti-inflammatory properties. However, the therapeutic efficacy of MSCs is limited by the intense inflammatory response at the transplantation site in ICH. Hence, enhancing the function of transplanted MSCs holds considerable promise as a therapeutic strategy for ICH. Notably, the iron-quercetin complex (IronQ), a metal-quercetin complex synthesized through coordination chemistry, has garnered significant attention for its biomedical applications. In our previous studies, we have observed that IronQ exerts stimulatory effects on cell growth, notably enhancing the survival and viability of peripheral blood mononuclear cells (PBMCs) and MSCs. This study aimed to evaluate the effects of pretreated MSCs with IronQ on neuroinflammation and elucidate its underlying mechanisms. Methods: The ICH mice were induced by injecting the collagenase I solution into the right brain caudate nucleus. After 24 hours, the ICH mice were randomly divided into four subgroups, the model group (Model), quercetin group (Quercetin), MSCs group (MSCs), and pretreated MSCs with IronQ group (MSCs+IronQ). Neurological deficits were re-evaluated on day 3, and brain samples were collected for further analysis. TUNEL staining was performed to assess cell DNA damage, and the protein expression levels of inflammatory factors and the cGAS-STING signaling pathway were investigated and analyzed. Results: Pretreated MSCs with IronQ effectively mitigate neurological deficits and reduce neuronal inflammation by modulating the microglial polarization. Moreover, the pretreated MSCs with IronQ suppress the protein expression levels of the cGAS-STING signaling pathway. Conclusion: These findings suggest that pretreated MSCs with IronQ demonstrate a synergistic effect in alleviating neuroinflammation, thereby improving neurological function, which is achieved through the inhibition of the cGAS-STING signaling pathway.

2.
Int J Biol Sci ; 20(1): 200-217, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38164169

RESUMO

TGF-ß/Smad3 signaling plays a critical role in type 2 diabetes (T2D) and type 2 diabetic nephropathy (T2DN), but treatment by specifically targeting Smad3 remains unexplored. To develop a new Smad3-targeted therapy for T2D and T2DN, we treated db/db mice at the pre-diabetic or established diabetic stage with a pharmacological Smad3 inhibitor SIS3. The therapeutic effect and mechanisms of anti-Smad3 treatment on T2D and T2DN were investigated. We found that anti-Smad3 treatment on pre-diabetic db/db mice largely attenuated both T2D and T2DN by markedly reducing blood glucose levels, and inhibiting the elevated serum creatinine, microalbuminuria, and renal fibrosis and inflammation. Unexpectedly, although SIS3 treatment on the established diabetic db/db mice inhibited T2DN but did not significantly improve T2D. Mechanistically, we uncovered that inhibition of T2DN in SIS3-treated db/db mice was associated with effectively restoring the balance of TGF-ß/Smad signaling by inhibiting Smad3 while increasing Smad7, thereby suppressing Smad3-mediated renal fibrosis and NF-κB-driven renal inflammation via lncRNA Erbb4-IR and LRN9884-dependent mechanisms. We also revealed that inhibition of islet ß cell injury by preventing the loss of islet Pax 6 could be the mechanism through which the pre-diabetic treatment, rather than the late SIS3 treatment on db/db mice significantly improved the T2D phenotype.


Assuntos
Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Estado Pré-Diabético , Camundongos , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Diabetes Mellitus Tipo 2/genética , Estado Pré-Diabético/complicações , Estado Pré-Diabético/patologia , Inflamação , Fator de Crescimento Transformador beta/metabolismo , Fibrose , Proteína Smad3/genética , Proteína Smad3/metabolismo , Rim/patologia
3.
Adv Sci (Weinh) ; 10(32): e2304360, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37749872

RESUMO

Podocyte injury plays a critical role in the progression of focal segmental glomerulosclerosis (FSGS). Here, it is reported that B-cell translocation gene 2 (Btg2) promotes Adriamycin (ADR)-induced FSGS via Smad3-dependent podocyte-mesenchymal transition. It is found that in FSGS patients and animal models, Btg2 is markedly upregulated by podocytes and correlated with progressive renal injury. Podocyte-specific deletion of Btg2 protected against the onset of proteinuria and glomerulosclerosis in ADR-treated mice along with inhibition of EMT markers such as α-SMA and vimentin while restoring epithelial marker E-cadherin. In cultured MPC5 podocytes, overexpression of Btg2 largely promoted ADR and TGF-ß1-induced EMT and fibrosis, which is further enhanced by overexpressing Btg2 but blocked by disrupting Btg2. Mechanistically, Btg2 is rapidly induced by TGF-ß1 and then bound Smad3 but not Smad2 to promote Smad3 signaling and podocyte EMT, which is again exacerbated by overexpressing Btg2 but blocked by deleting Btg2 in MPC5 podocytes. Interestingly, blockade of Smad3 signaling with a Smad3 inhibitor SIS3 is also capable of inhibiting Btg2 expression and Btg2-mediated podocyte EMT, revealing a TGF-ß/Smad3-Btg2 circuit mechanism in Btg2-mediated podocyte injury in FSGS. In conclusion, Btg2 is pathogenic in FSGS and promotes podocyte injury via a Smad3-dependent EMT pathway.


Assuntos
Glomerulosclerose Segmentar e Focal , Podócitos , Animais , Humanos , Camundongos , Doxorrubicina/farmacologia , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Rim/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
4.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37511155

RESUMO

Transforming growth factor-ß (TGF-ß)/Smad3 signaling has been shown to play important roles in fibrotic and inflammatory diseases. However, the role of Smad3 in dyslipidemia and non-alcoholic fatty liver disease (NAFLD) in type 2 diabetes remains unclear, and whether targeting Smad3 has a therapeutic effect on these metabolic abnormalities remains unexplored. These topics were investigated in this study in Smad3 knockout (KO)-db/db mice and by treating db/db mice with a Smad3-specific inhibitor SIS3. Compared to Smad3 wild-type (WT)-db/db mice, Smad3 KO-db/db mice were protected against dyslipidemia and NAFLD. Similarly, treatment of db/db mice with SIS3 at week 4 before the onset of type 2 diabetes until week 12 was capable of lowering blood glucose levels and improving diabetic dyslipidemia and NAFLD. In addition, using RNA-sequencing, the potential Smad3-target genes related to lipid metabolism was identified in the liver tissues of Smad3 KO/WT mice, and the regulatory mechanisms were investigated. Mechanistically, we uncovered that Smad3 targeted peroxisome proliferator-activated receptor delta (PPARδ) to induce dyslipidemia and NAFLD in db/db mice, which was improved by genetically deleting and pharmacologically inhibiting Smad3.


Assuntos
Diabetes Mellitus Tipo 2 , Hepatopatia Gordurosa não Alcoólica , PPAR delta , Proteína Smad3 , Animais , Camundongos , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/genética , PPAR delta/metabolismo , Proteína Smad3/metabolismo
5.
Mol Ther Nucleic Acids ; 33: 180-190, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37449045

RESUMO

Transforming growth factor ß (TGF-ß)/Smad3 plays a vital role in hypertensive cardiac fibrosis. The long non-coding RNA (lncRNA) Erbb4-IR is a novel Smad3-dependent lncRNA that mediates kidney fibrosis. However, the role of Erbb4-IR in hypertensive heart disease remains unexplored and was investigated in the present study by ultrasound-microbubble-mediated silencing of cardiac Erbb4-IR in hypertensive mice induced by angiotensin II. We found that chronic angiotensin II infusion induced hypertension and upregulated cardiac Erbb4-IR, which was associated with cardiac dysfunction, including a decrease in left ventricle ejection fraction (LVEF) and LV fractional shortening (LVFS) and an increase in LV mass. Knockdown of cardiac Erbb4-IR by Erbb4-IR short hairpin RNA (shRNA) gene transfer effectively improved the angiotensin II-induced deterioration of cardiac function, although blood pressure was not altered. Furthermore, silencing cardiac Erbb4-IR also inhibited angiotensin II-induced progressive cardiac fibrosis, as evidenced by reduced collagen I and III, alpha-smooth muscle actin (α-SMA), and fibronectin accumulation. Mechanistically, improved hypertensive cardiac injury by specifically silencing cardiac Erbb4-IR was associated with increased myocardial Smad7 and miR-29b, revealing that Erbb4-IR may target Smad7 and miR-29b to mediate angiotensin II-induced hypertensive cardiac fibrosis. In conclusion, Erbb4-IR is pathogenic in angiotensin II (Ang II)-induced cardiac remodeling, and targeting Erbb4-IR may be a novel therapy for hypertensive cardiovascular diseases.

6.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166755, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37196860

RESUMO

Renal fibrosis (RF) is a common pathway leading to chronic kidney disease (CKD), which lacks effective treatment. While estrogen receptor beta (ERß) is known to be present in the kidney, its role in RF remains unclear. The present study aimed to investigate the role and underlying mechanism of ERß during RF progression in patients and animal models with CKD. We found that ERß was highly expressed in the proximal tubular epithelial cells (PTECs) in healthy kidneys but its expression was largely lost in patients with immunoglobin A nephropathy (IgAN) and in mice with unilateral ureter obstruction (UUO) and subtotal nephrectomy (5/6Nx). ERß deficiency markedly exacerbated, whereas ERß activation by WAY200070 and DPN attenuated RF in both UUO and 5/6Nx mouse models, suggesting a protective role of ERß in RF. In addition, ERß activation inhibited TGF-ß1/Smad3 signaling, while loss of renal ERß was associated with overactivation of the TGF-ß1/Smad3 pathway. Furthermore, deletion or pharmacological inhibition of Smad3 prevented the loss of ERß and RF. Mechanistically, activation of ERß competitively inhibited the association of Smad3 with the Smad-binding element, thereby downregulating the transcription of the fibrosis-related genes without altering Smad3 phosphorylation in vivo and in vitro. In conclusion, ERß exerts a renoprotective role in CKD by blocking the Smad3 signaling pathway. Thus, ERß may represent as a promising therapeutic agent for RF.


Assuntos
Insuficiência Renal Crônica , Obstrução Ureteral , Animais , Camundongos , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Fibrose , Rim/patologia , Insuficiência Renal Crônica/tratamento farmacológico , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo
7.
Stem Cell Res Ther ; 14(1): 131, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37189208

RESUMO

BACKGROUND: Intracerebral hemorrhage (ICH) is a severe brain-injured disease accompanied by cerebral edema, inflammation, and subsequent neurological deficits. Mesenchymal stem cells (MSCs) transplantation has been used as a neuroprotective therapy in nervous system diseases because of its anti-inflammatory effect. Nevertheless, the biological characteristics of transplanted MSCs, including the survival rate, viability, and effectiveness, are restricted because of the severe inflammatory response after ICH. Therefore, improving the survival and viability of MSCs will provide a hopeful therapeutic efficacy for ICH. Notably, the biomedical applications of coordination chemistry-mediated metal-quercetin complex have been verified positively and studied extensively, including growth-promoting and imaging probes. Previous studies have shown that the iron-quercetin complex (IronQ) possesses extraordinary dual capabilities with a stimulating agent for cell growth and an imaging probe by magnetic resonance imaging (MRI). Therefore, we hypothesized that IronQ could improve the survival and viability of MSCs, displaying the anti-inflammation function in the treatment of ICH while also labeling MSCs for their tracking by MRI. This study aimed to explore the effects of MSCs with IronQ in regulating inflammation and further clarify their potential mechanisms. METHODS: C57BL/6 male mice were utilized in this research. A collagenase I-induced ICH mice model was established and randomly separated into the model group (Model), quercetin gavage group (Quercetin), MSCs transplantation group (MSCs), and MSCs transplantation combined with IronQ group (MSCs + IronQ) after 24 h. Then, the neurological deficits score, brain water content (BWC), and protein expression, such as TNF-α, IL-6, NeuN, MBP, as well as GFAP, were investigated. We further measured the protein expression of Mincle and its downstream targets. Furthermore, the lipopolysaccharide (LPS)-induced BV2 cells were utilized to investigate the neuroprotection of conditioned medium of MSCs co-cultured with IronQ in vitro. RESULTS: We found that the combined treatment of MSCs with IronQ improved the inflammation-induced neurological deficits and BWC in vivo by inhibiting the Mincle/syk signaling pathway. Conditioned medium derived from MSCs co-cultured with IronQ decreased inflammation, Mincle, and its downstream targets in the LPS-induced BV2 cell line. CONCLUSIONS: These data suggested that the combined treatment exerts a collaborative effect in alleviating ICH-induced inflammatory response through the downregulation of the Mincle/syk signaling pathway following ICH, further improving the neurologic deficits and brain edema.


Assuntos
Edema Encefálico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Camundongos , Animais , Masculino , Ratos Sprague-Dawley , Quercetina/efeitos adversos , Meios de Cultivo Condicionados/metabolismo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Hemorragia Cerebral , Transdução de Sinais , Inflamação/terapia , Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo
8.
Regen Ther ; 22: 181-190, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36860266

RESUMO

Intracerebral hemorrhage (ICH) is the second largest type of stroke with high mortality and morbidity. The vast majority of survivors suffer from serious neurological defects. Despite the well-established etiology and diagnose, there is still some controversy over the ideal treatment strategy. MSC-based therapy has become an attractive and promising strategy for the treatment of ICH through immune regulation and tissue regeneration. However, accumulating studies have revealed that MSC-based therapeutic effects are mainly attributed to the paracrine properties of MSC, especially small extracellular vesicles/exosome (EVs/exo) which are considered to be the key mediators of the protective efficacy from MSCs. Moreover, some papers reported that MSC-EVs/exo have better therapeutic effects than MSCs. Therefore, EVs/exo has become a new choice for the treatment of ICH stroke in recent years. In this review, we mainly concentrate on the current research progress on the use of MSC-EVs/exo in the treatment of ICH and the existing challenges in their transplation from lab to clinical practice.

9.
J Healthc Eng ; 2023: 9260747, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36824406

RESUMO

Methods: Observational study on 47 adult hospitalized cancer patients including 27 males and 20 females who received proton beam radiotherapy during December 2021 and August 2022. Nutritional assessments, 24 h dietary survey, handgrip strength (HGS) test, anthropometrical measurements, and hematological parameters were conducted or collected at the beginning and the completion of treatment. Results: The rate of nutritional risk and malnutrition among the total of 47 enrolled patients was 4.3% and 12.8% at the onset of proton radiation and raised up to 6.4% and 27.7% at the end of the treatment. 42.6% of patients experienced weight loss during the proton radiotherapy, and 1 of them had weight loss over 5%, and in general, the average body weight was stable over radiotherapy. The changes in patients' 24 h dietary intakes, HGS, and anthropometrical parameters, including triceps skinfold thickness (TSF), midupper arm circumference (MUAC), and midupper arm muscle circumference (MAMC), were statistically insignificant over the treatment (all p values > 0.05). The changes in patients' hematological parameters, including total protein (TP) and serum albumin (ALB), were not statistically significant over the treatment (all p values >0.05), and the level of hemoglobin (HGB) at the end of treatment was higher than that at the onset (p < 0.05). Conclusion: The results of this study demonstrated that proton radiotherapy might have a lighter effect on the nutritional status of cancer patients.


Assuntos
Neoplasias , Estado Nutricional , Masculino , Adulto , Feminino , Humanos , Prótons , Índice de Massa Corporal , Força da Mão , Neoplasias/radioterapia , Redução de Peso
10.
Int J Biol Sci ; 19(2): 521-536, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36632461

RESUMO

Neuropeptide Y (NPY) is produced by the nerve system and may contribute to the progression of CKD. The present study found the new protective role for NPY in AKI in both patients and animal models. Interestingly, NPY was constitutively expressed in blood and resident kidney macrophages by co-expressing NPY and CD68+ markers, which was lost in patients and mice with AKI-induced by cisplatin. Unexpectedly, NPY was renoprotective in AKI as mice lacking NPY developed worse renal necroinflammation and renal dysfunction in cisplatin and ischemic-induced AKI. Importantly, NPY was also a therapeutic agent for AKI because treatment with exogenous NPY dose-dependently inhibited cisplatin-induced AKI. Mechanistically, NPY protected kidney from AKI by inactivating M1 macrophages via the Y1R-NF-κB-Mincle-dependent mechanism as deleting or silencing NPY decreased Y1R but increased NF-κB-Mincle-mediated M1macrophage activation and renal necroinflammation, which were reversed by addition of NPY or by silencing Mincle but promoted by blocking Y1R with BIBP 3226. Thus, NPY is renoprotective and may be a novel therapeutic agent for AKI. NPY may act via Y1R to protect kidney from AKI by blocking NF-κB-Mincle-mediated M1 macrophage activation and renal necroinflammation.


Assuntos
Injúria Renal Aguda , NF-kappa B , Neuropeptídeo Y , Receptores de Neuropeptídeo Y , Animais , Camundongos , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/induzido quimicamente , Cisplatino/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/farmacologia , Neuropeptídeo Y/uso terapêutico , Receptores de Neuropeptídeo Y/metabolismo
11.
J Pharmacol Sci ; 151(2): 72-83, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36707181

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) show antifibrotic activity in various chronic kidney diseases. Here, we aimed to investigate whether Calycosin (CA), a phytoestrogen, could enhance the antifibrotic activity of MSCs in primary tubular epithelial cells (PTECs) induced by TGF-ß1 and in a mouse model of unilateral ureteral obstruction (UUO). We found that MSCs treatment significantly inhibited fibrosis, and CA pretreatment enhanced the effects of MSCs on fibrosis in vitro. Consistent with the in vitro studies, MSCs alleviated tubular injury and renal fibrosis in mice after UUO, and CA-pretreated MSCs resulted in more significant improvements in tubular injury and renal fibrosis than MSCs after UUO. Moreover, MSCs treatment significantly inhibited necroptosis by repressing the elevation of MLKL, RIPK1, and RIPK3 in PTECs treated by TGF-ß1and in mice after UUO, and CA-pretreated MSCs were superior to MSCs in alleviating necroptosis. MSCs significantly reduced TNF-α and TNFR1 expression induced by TGF-ß1 in PTECs and inhibited TGF-ß1, TNF-α, and TNFR1 expression induced by UUO in mice. These effects of MSCs were significantly enhanced after CA pretreatment. Therefore, our results suggest that CA pretreatment enhances the antifibrotic activity of MSCs by inhibiting TGF-ß1/TNF-α/TNFR1 signaling-induced necroptosis.


Assuntos
Células-Tronco Mesenquimais , Insuficiência Renal Crônica , Obstrução Ureteral , Camundongos , Animais , Obstrução Ureteral/complicações , Obstrução Ureteral/terapia , Fator de Crescimento Transformador beta1/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Necroptose , Insuficiência Renal Crônica/metabolismo , Fibrose , Rim/patologia
12.
J Nat Med ; 77(1): 73-86, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36115008

RESUMO

Excessive intrahepatocellular lipid accumulation or steatosis is caused by abnormal lipid metabolism and a common character of nonalcoholic fatty liver disease (NAFLD), which may progress into cirrhosis and hepatocellular cancer. Andrographolide (Andro) is the primary active ingredient extracted from Andrographis paniculata, showing a protective role against dietary steatosis with the mechanism not fully understood. In this study, we showed that administration of Andro (50, 100, and 200 mg/kg/day for 8 weeks, respectively) attenuated obesity and metabolic syndrome in high-fat diet (HFD)-fed mice with improved glucose tolerance, insulin sensitivity, and reduced hyperinsulinemia, hyperglycemia, and hyperlipidemia. HFD-fed mice presented hepatic steatosis, which was significantly prevented by Andro. In vitro, Andro decreased the intracellular lipid droplets in oleic acid-treated LO2 cells. The selected RT-PCR array revealed a robust expression suppression of the fatty acid transport proteins (FATPs) by Andro treatment. Most importantly, we found that Andro consistently reduced the expression of FATP2 in both the oleic acid-treated LO2 cells and liver tissues of HFD-fed mice. Overexpression of FATP2 abolished the lipid-lowering effect of Andro in oleic acid-treated LO2 cells. Andro treatment also reduced the fatty acid uptake in oleic acid-treated LO2 cells, which was blunted by FATP2 overexpression. Collectively, our findings reveal a novel mechanism underlying the anti-steatosis effect of Andro by suppressing FATP2-mediated fatty acid uptake, suggesting the potential therapeutic application of Andro in the treatment of NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Coenzima A Ligases/metabolismo , Coenzima A Ligases/farmacologia , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Ácidos Graxos/uso terapêutico , Metabolismo dos Lipídeos , Fígado , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico
13.
Medicine (Baltimore) ; 101(46): e31946, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36401452

RESUMO

The incidence and mortality rates of stroke in China are higher than the world average, seriously endangering the public's health and quality of life. It is important to predict the incidence of stroke, identify the high-risk factors in the region, and raise the risk awareness of high-risk groups. This study sought to investigate and analyze the distribution of stroke population and the main risk factors for stroke occurrence in a Chinese population, and to predict the probability of stroke occurrence in high-risk groups with risk factors, so as to provide a scientific basis for the comprehensive prevention and treatment of stroke. A whole-group sampling method was used to investigate 1009 participants in Jingzhou city in central China, and a uniform questionnaire survey and related medical examinations were conducted. The risk factors for stroke in the area were analyzed by univariate analysis, and a multifactorial logistic regression prediction model was established based on the results of univariate analysis. The results of univariate and multifactorial logistic regression analyses suggested that gender, age, family history of stroke, hypertension, atrial fibrillation, diabetes, and sedentary lifestyle were significantly associated with an increased risk of stroke in the local population (all P < .05). The top 5 risk factors for stroke were atrial fibrillation (odds ratio [OR] = 5.225, 95% confidence interval [CI]: 2.826-9.663), sedentary lifestyle (OR = 2.701, 95% CI: 1.667-4.376), age (≥65 years) (OR = 2.593, 95% CI: 1.680-4.004), hypertension (OR = 2.106, 95% CI: 1.380-3.216), and gender (male) (OR = 2.099, 95% CI: 1.270-3.471). This study effectively identifies the high risk factors for stroke and provides scientific insights for risk assessment, intervention of risk factors, and decision making of health management departments in the central region of China. The modifable risk factors for stroke such as smoking, hypertension, atrial fibrillation, diabetes mellitus, and sedentary lifestyle were also observed. Our findings further highlight the significant of the primary and secondary prevention for stroke and reveal the potential targets to reduce the heavy stroke burden in China around the world.


Assuntos
Fibrilação Atrial , Diabetes Mellitus , Hipertensão , Acidente Vascular Cerebral , Humanos , Masculino , Idoso , Estudos Transversais , Fibrilação Atrial/complicações , Fibrilação Atrial/epidemiologia , Qualidade de Vida , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/complicações , Fatores de Risco , Hipertensão/complicações , Hipertensão/epidemiologia , Diabetes Mellitus/epidemiologia
14.
Front Physiol ; 13: 937878, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091385

RESUMO

Renal fibrosis is a common feature of all types of chronic kidney disease (CKD) and is tightly regulated by the TGF-ß/Smad3 pathway. Let-7i-5p belongs to the let-7 microRNA family with diverse biological functions. It has been reported that let-7i-5p suppresses fibrotic disease in the heart, lungs, and blood vessels, while the role of let-7i-5p in renal fibrosis remains limited. In this study, we aimed to investigate the role of let-7i-5p in renal fibrosis in a mouse model of unilateral ureteral obstruction (UUO) and TGF-ß1-stimulated renal tubular cell line TCMK1. The RNA-targeting CRISPR/Cas13d system was used to knock down let-7i-5p. Renal injury and fibrosis were determined by histological analysis, RT-PCR, Western blot, and immunostaining. Our results have shown that in the kidneys after UUO, the expression of let-7i-5p was significantly increased along with notable tubular injury and interstitial fibrosis. Electroporation of let-7i-targeting Cas13d plasmid efficiently knocked down let-7i-5p in kidneys after UUO with reduced tubular injury, fibrotic area, and expression of fibrotic marker genes α-SMA, fibronectin, and Col1a1. In TGF-ß1-stimulated TCMK1 cells, knockdown of let-7i-5p by Cas13d plasmid transfection also blunted the expression of fibrotic marker genes. Most importantly, the genomic locus of let-7i showed enriched binding of Smad3 as revealed by chromatin immunoprecipitation. In TCMK1 cells, the overexpression of Smad3 can directly induce the expression of let-7i-5p. However, the deletion of Smad3 abolished TGF-ß1-stimulated let-7i-5p expression. Collectively, these findings suggest that let-7i-5p is a Smad3-dependent microRNA that plays a pathogenic role in renal fibrosis. Let-7i-5p could be a promising target for the treatment of CKD-associated renal fibrosis.

15.
Biomolecules ; 12(3)2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-35327565

RESUMO

Beta (ß) cell dysfunction or loss is the common pathological feature in all types of diabetes mellitus (diabetes). Resolving the underlying mechanism may facilitate the treatment of diabetes by preserving the ß cell population and function. It is known that TGF-ß signaling plays diverse roles in ß cell development, function, proliferation, apoptosis, and dedifferentiation. Inhibition of TGF-ß signaling expands ß cell lineage in the development. However, deletion of Tgfbr1 has no influence on insulin demand-induced but abolishes inflammation-induced ß cell proliferation. Among canonical TGF-ß signaling, Smad3 but not Smad2 is the predominant repressor of ß cell proliferation in response to systemic insulin demand. Deletion of Smad3 simultaneously improves ß cell function, apoptosis, and systemic insulin resistance with the consequence of eliminated overt diabetes in diabetic mouse models, revealing Smad3 as a key mediator and ideal therapeutic target for type-2 diabetes. However, Smad7 shows controversial effects on ß cell proliferation and glucose homeostasis in animal studies. On the other hand, overexpression of Tgfb1 prevents ß cells from autoimmune destruction without influence on ß cell function. All these findings reveal the diverse regulatory roles of TGF-ß signaling in ß cell biology.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Insulinas , Animais , Proliferação de Células , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinas/metabolismo , Camundongos , Fator de Crescimento Transformador beta/metabolismo
16.
Nat Commun ; 13(1): 427, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058428

RESUMO

Epididymal white adipose tissue (eWAT) secretes an array of cytokines to regulate the metabolism of organs and tissues in high-fat diet (HFD)-induced obesity, but its effects on bone metabolism are not well understood. Here, we report that macrophages in eWAT are a main source of osteopontin, which selectively circulates to the bone marrow and promotes the degradation of the bone matrix by activating osteoclasts, as well as modulating bone marrow-derived macrophages (BMDMs) to engulf the lipid droplets released from adipocytes in the bone marrow of mice. However, the lactate accumulation induced by osteopontin regulation blocks both lipolysis and osteoclastogenesis in BMDMs by limiting the energy regeneration by ATP6V0d2 in lysosomes. Both surgical removal of eWAT and local injection of either clodronate liposomes (for depleting macrophages) or osteopontin-neutralizing antibody show comparable amelioration of HFD-induced bone loss in mice. These results provide an avenue for developing therapeutic strategies to mitigate obesity-related bone disorders.


Assuntos
Tecido Adiposo/metabolismo , Osso e Ossos/metabolismo , Epididimo/metabolismo , Homeostase , Macrófagos/metabolismo , Osteopontina/metabolismo , Tecido Adiposo/diagnóstico por imagem , Tecido Adiposo Branco/diagnóstico por imagem , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Reabsorção Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Antígeno CD11b/metabolismo , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/metabolismo , Dieta Hiperlipídica , Inflamação/patologia , Metabolismo dos Lipídeos , Lisossomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Tamanho do Órgão , Subunidades Proteicas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Microtomografia por Raio-X
17.
Theranostics ; 12(1): 379-395, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34987651

RESUMO

Rationale: Poor ß cell proliferation is one of the detrimental factors hindering islet cell replacement therapy for patients with diabetes. Smad3 is an important transcriptional factor of TGF-ß signaling and has been shown to promote diabetes by inhibiting ß cell proliferation. Therefore, we hypothesize that Smad3-deficient islets may be a novel cell replacement therapy for diabetes. Methods: We examined this hypothesis in streptozocin-induced type-1 diabetic mice and type-2 diabetic db/db mice by transplanting Smad3 knockout (KO) and wild type (WT) islets under the renal capsule, respectively. The effects of Smad3KO versus WT islet replacement therapy on diabetes and diabetic kidney injury were examined. In addition, RNA-seq was applied to identify the downstream target gene underlying Smad3-regulated ß cell proliferation in Smad3KO-db/db versus Smad3WT-db/db mouse islets. Results: Compared to Smad3WT islet therapy, treatment with Smad3KO islets produced a much better therapeutic effect on both type-1 and type-2 diabetes by significantly lowering serum levels of blood glucose and HbA1c and protected against diabetic kidney injuries by preventing an increase in serum creatinine and the development of proteinuria, mesangial matrix expansion, and fibrosis. These were associated with a significant increase in grafted ß cell proliferation and blood insulin levels, resulting in improved glucose intolerance. Mechanistically, RNA-seq revealed that compared with Smad3WT-db/db mouse islets, deletion of Smad3 from db/db mouse islets markedly upregulated E2F3, a pivotal regulator of cell cycle G1/S entry. Further studies found that Smad3 could bind to the promoter of E2F3, and thus inhibit ß cell proliferation via an E2F3-dependent mechanism as silencing E2F3 abrogated the proliferative effect on Smad3KO ß cells. Conclusion: Smad3-deficient islet replacement therapy can significantly improve both type-1 and type-2 diabetes and protect against diabetic kidney injury, which is mediated by a novel mechanism of E2F3-dependent ß cell proliferation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Fator de Transcrição E2F3/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Smad3/metabolismo , Animais , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Int J Mol Sci ; 22(15)2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34360646

RESUMO

Diabetic nephropathy (DN) is one of the most common complications in diabetes mellitus and the leading cause of end-stage renal disease. TGF-ß is a pleiotropic cytokine and has been recognized as a key mediator of DN. However, anti-TGF-ß treatment for DN remains controversial due to the diverse role of TGF-ß1 in DN. Thus, understanding the regulatory role and mechanisms of TGF-ß in the pathogenesis of DN is the initial step towards the development of anti-TGF-ß treatment for DN. In this review, we first discuss the diverse roles and signaling mechanisms of TGF-ß in DN by focusing on the latent versus active TGF-ß1, the TGF-ß receptors, and the downstream individual Smad signaling molecules including Smad2, Smad3, Smad4, and Smad7. Then, we dissect the regulatory mechanisms of TGF-ß/Smad signaling in the development of DN by emphasizing Smad-dependent non-coding RNAs including microRNAs and long-non-coding RNAs. Finally, the potential therapeutic strategies for DN by targeting TGF-ß signaling with various therapeutic approaches are discussed.


Assuntos
Nefropatias Diabéticas/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Nefropatias Diabéticas/metabolismo , Humanos , Transdução de Sinais
19.
Phytomedicine ; 90: 153627, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34247115

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder prevalent in the aged population. Tetrandrine is a natural metabolite isolated from herbal medicine Stephania tetrandra with various activities. PURPOSE: In this study, we investigated the therapeutic role of tetrandrine in 5XFAD mouse, a transgenic model of AD. METHODS: 5XFAD mice were intraperitoneally injected with saline or different doses of tetrandrine (10, 20, and 40 mg/kg per 2 days) from the age of 5 months to 7 months followed by the determination of cognitive ability, amyloid plaque load, cell apoptosis, and inflammation in the brain. In vitro, the protective roles of tetrandrine against inflammatory activation of microglia and the resulting neurotoxicity were studied in BV2 cells and differentiated PC12 cells, respectively. RESULTS: Morris water maze test showed that two months of tetrandrine treatment dose-dependently improved the cognitive ability of 5XFAD mice. Immunostaining against Aß 1-42 demonstrated reduced amyloid plaque deposition in the brain of tetrandrine-treated 5XFAD mice. TUNEL assay revealed decreased cell apoptosis in the hippocampus after tetrandrine treatment. Further, RT-PCR showed that the ectopic transcription of inflammation-associated genes including TNFα, IL-1ß, IL-6, COX-2, iNOS, and p65 was reversed in 5XFAD mice treated with tetrandrine. In vitro, Aß 1-42 stimulated the secretion of inflammatory cytokines TNFα and IL-1ß in microglial BV2 cells as determined by ELISA, which was suppressed by tetrandrine pre-treatment. Tetrandrine pre-treatment also inhibited the expression of TLR4, p65, iNOS, and COX-2 in BV2 cells induced by Aß 1-42. Most importantly, treatment of PC12-derived neuron-like cells with conditional medium from Aß 1-42-stimulated BV2 cells remarkably impaired cell viability and promoted cell apoptosis, which was attenuated by the conditional medium from BV2 cells with tetrandrine pre-treatment. CONCLUSION: Collectively, findings in this study demonstrated that tetrandrine ameliorates AD by suppressing microglia-mediated inflammation and neurotoxicity.


Assuntos
Doença de Alzheimer , Benzilisoquinolinas/farmacologia , Microglia/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos
20.
Artigo em Inglês | MEDLINE | ID: mdl-34257680

RESUMO

Chronic kidney disease (CKD) has become a global health issue, and there is increasing evidence showing the beneficial roles of traditional Chinese medicine (TCM) in CKD treatment. Here, we studied the renoprotective role of Mahuang decoction, a famous TCM prescription, in a rat CKD model induced with the combination of doxorubicin and adenine. Our data showed that intragastric administration of Mahuang decoction inhibited the loss of bodyweight and attenuated proteinuria, serum creatinine, and blood urea nitrogen in CKD rats. Kidney histological analysis revealed decreased tubulointerstitial injury and fibrosis in CKD rats treated with Mahuang decoction accompanied with suppressed expression of TGF-ß1 and phosphorylated NF-κB/P65 (p-P65) as indicated by immunohistochemistry. ELISA analysis demonstrated reduced serum levels of proinflammatory cytokines TNFα and IL-6. Most importantly, intestinal microbiota analysis by 16s rRNA-seq showed that Mahuang decoction restored the impaired richness and diversity of intestinal microflora and recovered the disrupted microbial community through reducing the abundance of deleterious microbes and promoting the expansion of beneficial microbes in CKD rats. Collectively, our findings demonstrated that Mahuang decoction mitigated kidney functional and structural impairment in CKD rats which were associated with the restoration of dysbiosis of intestinal microbiota, implying its potential in clinical CKD treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...