Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Neuromolecular Med ; 26(1): 34, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39167282

RESUMO

Both of exosomes derived from mesenchymal stem cells (MSCs) and glial cell line-derived neurotrophic factor (GDNF) show potential for the treatment of neuropathic pain. Here, the analgesic effects of exosomes derived from bone marrow MSCs (BMSCs) were investigated. BMSCs-derived exosomes were isolated and characterized. Chronic constriction injury (CCI) was constructed to induce neuropathic pain in rats, which were then treated with exosomes. Pain behaviors were evaluated by measuring paw withdrawal thresholds and latency. The changes of key proteins, including cytokines, were explored using Western blot and ELISA. Administration of BMSCs-derived exosomes alleviated neuropathic pain, as demonstrated by the decrease of thermal hyperalgesia and mechanical allodynia, as well as the reduced secretion of pro-inflammatory cytokines in CCI rats. These effects were comparable to the treatment of GDNF alone. Mechanically, the exosomes suppressed the CCI-induced activation of TLR2/MyD88/NF-κB signaling pathway, while GDNF knockdown impaired their analgesic effects on CCI rat. BMSCs-derived exosomes may alleviate CCI-induced neuropathic pain and inflammation in rats by transporting GDNF.


Assuntos
Modelos Animais de Doenças , Exossomos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hiperalgesia , Células-Tronco Mesenquimais , Fator 88 de Diferenciação Mieloide , NF-kappa B , Ratos Sprague-Dawley , Transdução de Sinais , Receptor 2 Toll-Like , Animais , Exossomos/transplante , Ratos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Masculino , Hiperalgesia/etiologia , Fator 88 de Diferenciação Mieloide/genética , NF-kappa B/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/biossíntese , Neuralgia/etiologia , Neuralgia/terapia , Citocinas , Transplante de Células-Tronco Mesenquimais , Células da Medula Óssea , Neuropatia Ciática , Constrição
2.
Front Endocrinol (Lausanne) ; 15: 1379830, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38803476

RESUMO

Background and objective: Psychological insulin resistance (PIR), which refers to the reluctance of diabetic patients to use insulin, is a frequently encountered clinical issue. Needle-free injection (NFI) offers advantages in terms of expediting insulin absorption and mitigating adverse reactions related to injection. To evaluate the effects of subcutaneous injection of insulin aspart 30 with NFI on PIR and insulin dosage in patients with type 2 diabetes mellitus (T2DM). Methods: Sixty-four patients with T2DM participated in this randomized, prospective, open, crossover study. Insulin aspart 30 was administered subcutaneously to each subject via QS-P NFI and Novo Pen 5 (NP) successively. The effects of NFI on PIR were analyzed. Differences in insulin dosage, glycemic variability, and injection safety were compared at similar levels of glycemic control. Results: After the administration of NFI, the insulin treatment attitude scale score decreased (53.7 ± 7.3 vs. 58.9 ± 10.7, p<0.001), the insulin treatment adherence questionnaire score increased (46.3 ± 4.9 vs. 43.8 ± 7.1, p<0.001), and the insulin treatment satisfaction questionnaire score increased (66.6 ± 10.5 vs. 62.4 ± 16.5, p<0.001). At the same blood glucose level, NFI required a smaller dosage of insulin aspart 30 compared with that of NP (30.42 ± 8.70 vs. 33.66 ± 9.13 U/d, p<0.001). There were no differences in glycemic variability indices (standard deviation, mean amplitude of glycemic excursion or coefficient of variation) between the two injection methods. Compared with NP, NFI did not increase the incidence of hypoglycemia (17.2% vs. 14.1%, p=0.774), and it decreased the incidence of induration (4.7% vs. 23.4%, p=0.002) and leakage (6.3% vs. 20.3%, p=0.022) while decreasing the pain visual analog scale score (2.30 ± 1.58 vs. 3.11 ± 1.40, p<0.001). Conclusion: NFI can improve PIR in patients with T2DM and be used with a smaller dose of insulin aspart 30 while maintaining the same hypoglycemic effect. Clinical trial registration: https://www.chictr.org.cn/, identifier ChiCTR2400083658.


Assuntos
Glicemia , Estudos Cross-Over , Diabetes Mellitus Tipo 2 , Hipoglicemiantes , Insulina Aspart , Resistência à Insulina , Insulina , Humanos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/psicologia , Masculino , Feminino , Pessoa de Meia-Idade , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Injeções Subcutâneas , Insulina Aspart/administração & dosagem , Insulina Aspart/uso terapêutico , Idoso , Estudos Prospectivos , Insulina/administração & dosagem , Insulina/uso terapêutico , Insulina/análogos & derivados , Glicemia/análise , Glicemia/efeitos dos fármacos , Adulto , Insulina Isófana/administração & dosagem , Insulina Isófana/uso terapêutico
3.
Tissue Eng Part A ; 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38818810

RESUMO

Therapy with mesenchymal stem cells (MSCs) is considered an attractive strategy for the repair or regeneration of damaged tissues. However, low survival of MSCs limits their applications clinically. Oxidized low-density lipoprotein (ox-LDL) is significantly increased in patients with hyperlipidemia and decreases the survival of MSCs. Bcl-2 is critically involved in important cell functions, including cell membrane integrity and cell survival. The present study was designed to test the hypothesis that ox-LDL attenuates the survival of MSCs through suppression of Bcl-2 expression. Bone marrow MSCs from C57BL/6 mice were cultured with ox-LDL at different concentrations (0-140 µg/mL) for 24 h with native LDL as control. Ox-LDL treatment substantially decreased the survival of MSCs dose-dependently and enhanced the release of intracellular lactate dehydrogenase (LDH) in association with a significant decrease in Bcl-2 protein level without change in BAX protein expression in MSCs. Bcl-2 overexpression effectively protected MSCs against ox-LDL-induced damages with preserved cell numbers without significant increase in LDH release. Treatment with N-acetylcysteine (NAC) (1 mM) effectively preserved Bcl-2 protein expression in MSCs and significantly attenuated ox-LDL-induced decrease of cell number and increase in the release of intracellular LDH. These data indicated that ox-LDL treatment resulted in a significant damage of cell membrane and dramatically decreased the survival of MSCs dose-dependently through inhibition of Bcl-2 expression. NAC treatment significantly protected MSCs against the damage of cell membrane by ox-LDL and promoted the survival of MSCs in association with preserved Bcl-2 expression.

4.
Cell Death Dis ; 13(3): 275, 2022 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-35347118

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is closely associated with insulin resistance (IR) and type 2 diabetes mellitus (T2DM), which are all complex metabolic disorders. Selenoprotein S (SelS) is an endoplasmic reticulum (ER) resident selenoprotein involved in regulating ER stress and has been found to participate in the occurrence and development of IR and T2DM. However, the potential role and mechanism of SelS in NAFLD remains unclear. Here, we analyzed SelS expression in the liver of high-fat diet (HFD)-fed mice and obese T2DM model (db/db) mice and generated hepatocyte-specific SelS knockout (SelSH-KO) mice using the Cre-loxP system. We showed that hepatic SelS expression levels were significantly downregulated in HFD-fed mice and db/db mice. Hepatic SelS deficiency markedly increased ER stress markers in the liver and caused hepatic steatosis via increased fatty acid uptake and reduced fatty acid oxidation. Impaired insulin signaling was detected in the liver of SelSH-KO mice with decreased phosphorylation levels of insulin receptor substrate 1 (IRS1) and protein kinase B (PKB/Akt), which ultimately led to disturbed glucose homeostasis. Meanwhile, our results showed hepatic protein kinase Cɛ (PKCɛ) activation participated in the negative regulation of insulin signaling in SelSH-KO mice. Moreover, the inhibitory effect of SelS on hepatic steatosis and IR was confirmed by SelS overexpression in primary hepatocytes in vitro. Thus, we conclude that hepatic SelS plays a key role in regulating hepatic lipid accumulation and insulin action, suggesting that SelS may be a potential intervention target for the prevention and treatment of NAFLD and T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo
5.
Food Chem Toxicol ; 160: 112771, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34920032

RESUMO

As an environmental toxicant, arsenic exposure may cause insulin resistance (IR). Previous studies have shown that pyroptosis plays an important role in the occurrence and development of IR. Although gasdermin D (GSDMD) functions as an executor of pyroptosis, the relationship between GSDMD-mediated pyroptosis and hepatic IR remains unclear. Here, we observed that sodium arsenite (NaAsO2) activated NOD-like receptors containing pyrin domain 3 (NLRP3) inflammasomes, promoted GSDMD activation, induced pyroptosis and hepatic IR, while GSDMD knockdown attenuated pyroptosis and hepatic IR caused by NaAsO2. However, GSDMD interference did not affect NLRP3 activation. Ubiquitination modification is widely involved in protein regulation and intracellular signal transduction, and whether it regulates GSDMD and affects its degradation, and exerts effects on arsenic-induced pyroptosis remain unclear. We observed that NaAsO2 reduced the K48- and K63-linked ubiquitination of GSDMD, thereby inhibiting its degradation through the ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway (ALP), causing GSDMD to accumulate and lyse into GSDMD-N, which promoted pyroptosis. In summary, we demonstrated that GSDMD participated in arsenic-induced hepatic IR. Moreover, NaAsO2 reduced GSDMD ubiquitination and decreased its intracellular degradation, aggravating pyroptosis and hepatic IR. We have revealed the molecular mechanism underpinning arsenic-induced IR, and we provide potential solutions for the prevention and treatment of type 2 diabetes (T2D).


Assuntos
Arsenitos/toxicidade , Resistência à Insulina , Fígado/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptose/efeitos dos fármacos , Compostos de Sódio/toxicidade , Animais , Linhagem Celular , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Ligação a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Ratos , Ratos Sprague-Dawley , Ubiquitinação/efeitos dos fármacos
6.
Int J Biochem Cell Biol ; 141: 106111, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34715363

RESUMO

Glucolipid metabolism disorder in diabetes mellitus (DM) causes human endothelial injury and autophagy dysfunction is an important cause of endothelial dysfunction (ED). Selenoprotein S (SelS) could protect endothelium from oxidative stress, inflammatory responses, and apoptosis. This study assessed the effect of SelS on autophagy in glucolipid metabolic disorders and protection of the resulted vascular endothelial injury. The results showed that high glucose (HG), high oxidized low-density lipoprotein (HL), and HG combined with HL (HGL) could reduce viability of human aortic endothelial cells (HAECs), induce HAECs injury and increase SelS expression in a time-dependent manner. HG, HL, and HGL also initially induced autophagy but later reduced it in HAECs, while activity of the Akt/mTOR signaling was inhibited, especially in HGL culture of HAECs. SelS overexpression reduced the endothelial injury and autophagy and activated the Akt/mTOR signaling in HG, HL and HGL-cultured HAECs, compared to the control. Conversely, knockdown of SelS expression had the opposite effects on HAECs. In conclusion, SelS demonstrated a protective effect on endothelial injury induced by high glucose and/or ox-LDL and the underlying molecular events might be related to its regulation of HAECs autophagy by activating the Akt/mTOR signaling. SelS could be a potential intervention target in prevention and treatment of diabetic vascular complications.


Assuntos
Células Endoteliais , Lipoproteínas LDL , Autofagia , Proteínas Proto-Oncogênicas c-akt
7.
Exp Gerontol ; 137: 110963, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32353414

RESUMO

Endothelial dysfunction (ED) is a critical and initiating factor in the genesis of diabetic vascular complications whose occurrence and development is closely related to the complex intravascular microenvironment. However, currently, there is no dynamic model simulating the diabetic vascular endothelial microenvironment that can be used to investigate the mechanism underlying multifactor-induced ED. Here, we developed an integrated microfluidic chip as a new methodological platform to study vascular ED. Selenoprotein S (SELENOS) was found to be involved in the defense against oxidative stress-induced vascular endothelial injury in our previous studies. However, the regulatory signaling pathway underlying this process has not been described. With this chip, we demonstrated that multifactor-induced oxidative stress injury in human aortic endothelial cells (HAECs) has a synergistic effects and upregulates SELENOS expression. Subsequently, SELENOS was found to protect HAECs against multifactor-induced oxidative stress injury by regulating the PKCα/PI3K/Akt/eNOS pathway in the diabetic vascular endothelial microenvironment. Based on these data, our diabetic vascular chip provides a promising tool for studying vascular endothelial function, and SELENOS may be a novel target for prevention and treatment of diabetic macrovascular complications.


Assuntos
Diabetes Mellitus , Células Endoteliais , Diabetes Mellitus/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/metabolismo , Selenoproteínas/metabolismo
8.
J Endocrinol ; 244(3): 431-443, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31846435

RESUMO

The induction of endoplasmic reticulum (ER) stress is associated with adipogenesis, during which the inositol-requiring enzyme 1 alpha (IRE1α)-X-box-binding protein 1 (XBP1) pathway is involved. Selenoprotein S (SelS), which is an ER resident selenoprotein, is involved in ER homeostasis regulation; however, little is known about the role of SelS in regulating adipogenesis. In vivo studies showed that SelS protein levels in white adipose tissue were increased in obese subjects and high-fat diet (HFD)-fed mice. Moreover, we identified that SelS protein levels increased in the early phase of adipogenesis and then decreased in the late phase during adipogenesis. Overexpression of SelS promoted adipogenesis. Conversely, knockdown (KD) of SelS resulted in the inhibition of adipogenesis, which was related to increasing cell death, decreased mitotic clonal expansion, and cell cycle G1 arrest. In vivo studies also showed that ER stress markers (p-IRE1α/IRE1α, XBP1s, and Grp78) were significantly increased with upregulating of SelS expression in subcutaneous and visceral adipose tissues in the obese subjects and HFD-fed mice. Furthermore, in SelS KD cells, the levels of Grp78 were increased and the levels of p-IRE1α/IRE1α were unchanged , but mRNA levels of spliced XBP1 (XBP1s) produced by IRE1α-mediated splicing were decreased, suggesting a role of SelS in the modulation of IRE1α-XBP1 pathway. Moreover, inhibition of adipogenesis by SelS suppression can be rescued by overexpression of XBP1s. Thus, SelS appears to function as a novel regulator of adipogenesis through the IRE1α-XBP1 signaling pathway.


Assuntos
Adipogenia , Proteínas de Membrana/metabolismo , Selenoproteínas/metabolismo , Transdução de Sinais , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Chaperona BiP do Retículo Endoplasmático , Endorribonucleases/genética , Endorribonucleases/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Selenoproteínas/genética , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
9.
Am J Physiol Endocrinol Metab ; 317(6): E1205-E1217, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31573846

RESUMO

Interleukin-8 (IL-8, also named CXCL8) binds to its receptors (CXCR1 and CXCR2) with subsequent recruitment of neutrophils and enhancement of their infiltration into inflamed sites, which exaggerates inflammation in many diseases. Recent studies have proposed that metabolic disorders can be attenuated by counteracting certain inflammatory signal pathways. In this study, we examined whether intervention with G31P, an antagonist of CXCL8, could attenuate tissue inflammation and development of metabolic disorders in db/db mice. The db/m and db/db mice were subcutaneously injected with G31P or equivalent normal saline once a day for 6 wk. The physical and metabolic parameters, glucose tolerance, insulin sensitivity, hepatic lipid accumulation, and inflammation markers were measured. G31P improved hepatic insulin sensitivity by modulating expression of genes related to gluconeogenesis and phosphorylated Akt levels. The expressions of several genes encoding proteins involved in de novo lipogenesis were decreased in G31P-treated db/db mice. Meanwhile, immune cell infiltration and cytokine release were attenuated in db/db mice with G31P treatment. G31P also improved the ratio of proinflammatory M1 and anti-inflammatory M2 macrophages. Furthermore, G31P ameliorates metabolic disturbances via inhibition of CXCR1 and CXCR2 pathways in db/db mice. These data suggest that the selective inhibition of CXC chemokines may have therapeutic effects on symptoms associated with obesity and diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina , Interleucina-8/antagonistas & inibidores , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Interleucina-8A/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Animais , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/metabolismo , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Insulina/metabolismo , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Fígado/metabolismo , Fígado/patologia , Macrófagos/efeitos dos fármacos , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
10.
Endocrinology ; 160(10): 2388-2394, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369074

RESUMO

N6-Methyladenosine (m6A) is the most common and abundant mRNA modification that involves regulating the RNA metabolism. However, the role of m6A in regulating the ß-cell function is unclear. Methyltransferase-like 14 (METTL14) is a key component of the m6A methyltransferase complex. To define the role of m6A in regulating the ß-cell function, we generated ß-cell METTL14-specific knockout (ßKO) mice by tamoxifen administration. Acute deletion of Mettl14 in ß-cells results in glucose intolerance as a result of a reduction in insulin secretion in ß-cells even though ß-cell mass is increased, which is related to increased ß-cell proliferation. To define the molecular mechanism, we performed RNA sequencing to detect the gene expression in ßKO islets. The genes responsible for endoplasmic reticulum stress, such as Ire1α, were among the top upregulated genes. Both mRNA and protein levels of IRE1α and spliced X-box protein binding 1 (sXBP-1) were increased in ßKO islets. The protein levels of proinsulin and insulin were decreased in ßKO islets. These results suggest that acute METTL14 deficiency in ß-cells induces glucose intolerance by increasing the IRE1α/sXBP-1 pathway.


Assuntos
Endorribonucleases/metabolismo , Intolerância à Glucose/genética , Células Secretoras de Insulina/metabolismo , Metiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Linhagem Celular , Endorribonucleases/genética , Regulação da Expressão Gênica/fisiologia , Insulinoma/metabolismo , Metiltransferases/genética , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Regulação para Cima , Proteína 1 de Ligação a X-Box/genética
11.
Int J Mol Med ; 44(2): 363-374, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31198979

RESUMO

Chronic, low­grade inflammation associated with obesity and diabetes result from the infiltration of adipose and vascular tissue by immune cells and contributes to cardiovascular complications. Despite an incomplete understanding of the mechanistic underpinnings of immune cell differentiation and inflammation, O­GlcNAcylation, the addition of O­linked N­acetylglucosamine (O­GlcNAc) to cytoplasmic, nuclear and mitochondrial proteins by the two cycling enzymes, the O­linked N­acetylglucosamine transferase (OGT) and the O­GlcNAcase (OGA), may contribute to fine­tune immunity and inflammation in both physiological and pathological conditions. Early studies have indicated that O­GlcNAcylation of proteins play a pro­inflammatory role in diabetes and insulin resistance, whereas subsequent studies have demonstrated that this post­translational modification could also be protective against acute injuries. These studies suggest that diverse types of insults result in dynamic changes to O­GlcNAcylation patterns, which fluctuate with cellular metabolism to promote or inhibit inflammation. In this review, the current understanding of O­GlcNAcylation and its adaptive modulation in immune and inflammatory responses is summarized.


Assuntos
Acetilglucosamina/imunologia , Imunidade , Inflamação/imunologia , Animais , Humanos , Inflamação/patologia , N-Acetilglucosaminiltransferases/imunologia , Proteínas/imunologia , beta-N-Acetil-Hexosaminidases/imunologia
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2138-2148, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31029827

RESUMO

Defects in the development, maintenance or expansion of ß-cell mass can result in impaired glucose metabolism and diabetes. N6-methyladenosine affects mRNA stability and translation efficiency, and impacts cell differentiation and stress response. To determine if there is a role for m6A in ß-cells, we investigated the effect of Mettl14, a key component of the m6A methyltransferase complex, on ß-cell survival and function using rat insulin-2 promoter-Cre-mediated deletion of Mettl14 mouse line (ßKO). We found that ßKO mice with normal chow exhibited glucose intolerance, lower levels of glucose-stimulated insulin secretion, increased ß-cell death and decreased ß-cell mass. In addition, HFD-fed ßKO mice developed glucose intolerance, decreased ß-cell mass and proliferation, exhibited lower body weight, increased adipose tissue mass, and enhanced insulin sensitivity due to enhanced AKT signaling and decreased gluconeogenesis in the liver. HFD-fed ßKO mice also showed a decrease in de novo lipogenesis, and an increase in lipolysis in the liver. RNA sequencing in islets revealed that Mettl14 deficiency in ß-cells altered mRNA expression levels of some genes related to cell death and inflammation. Together, we showed that Mettl14 in ß-cells plays a key role in ß-cell survival, insulin secretion and glucose homeostasis.


Assuntos
Secreção de Insulina , Metiltransferases/metabolismo , Animais , Sobrevivência Celular , Dieta Hiperlipídica , Regulação da Expressão Gênica , Gluconeogênese , Intolerância à Glucose , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Lipogênese , Litostatina/genética , Litostatina/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Metiltransferases/antagonistas & inibidores , Metiltransferases/genética , Camundongos , Camundongos Knockout , Proteínas Associadas a Pancreatite/genética , Proteínas Associadas a Pancreatite/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
13.
J Cell Biochem ; 120(5): 8661-8675, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30485531

RESUMO

Vascular endothelial apoptosis is closely associated with the pathogenesis and progression of diabetic macrovascular diseases. Selenoprotein S (SelS) participates in the protection of vascular endothelial and smooth muscle cells from oxidative and endoplasmic reticulum stress-induced injury. However, whether SelS can protect vascular endothelium from high glucose (HG)-induced apoptosis and the underlying mechanism remains unclear. The present study preliminarily analyzed aortic endothelial apoptosis and SelS expression in diabetic rats in vivo and the effects of HG on human umbilical vein endothelial cell (HUVEC) apoptosis and SelS expression in vitro. Subsequently, SelS expression was up- or downregulated in HUVECs using the pcDNA3.1-SelS recombinant plasmid and SelS-specific small interfering RNAs, and the effects of high/low SelS expression on HG-induced HUVEC apoptosis and a possible molecular mechanism were analyzed. As expected, HG induced vascular endothelial apoptosis and upregulated endothelial SelS expression in vivo and in vitro. SelS overexpression in HUVECs suppressed HG-induced increase in apoptosis and cleaved caspase3 level, accompanied by reduced protein kinase CßII (PKCßII), c-JUN N-terminal kinase (JNK), and B-cell lymphoma/leukemia-2 (Bcl-2) phosphorylation. In contrast, inhibiting SelS expression in HUVECs further aggravated HG-induced increase in apoptosis and cleaved caspase3 level, which was accompanied by increased PKCßII, JNK, and Bcl-2 phosphorylation. Pretreatment with PKC activators blocked the protective effects of SelS and increased the apoptosis and cleaved caspase3 level in HUVECs. In summary, SelS protects vascular endothelium from HG-induced apoptosis, and this was achieved through the inhibition of PKCßII/JNK/Bcl-2 pathway to eventually inhibit caspase3 activation. SelS may be a promising target for the prevention and treatment of diabetic macrovascular complications.

14.
Cell Physiol Biochem ; 51(5): 2185-2197, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30537728

RESUMO

BACKGROUND/AIMS: VCP-interacting membrane selenoprotein (VIMP), an ER resident selenoprotein, is highly expressed in ß-cells, however, the role of VIMP in ß-cells has not been characterized. In this study, we studied the relationship between VIMP deficiency and ß-cell survival in MIN6 insulinoma cells. METHODS: To determine the role of VIMP in ß-cells, lentiviral VIMP shRNAs were used to knock down (KD) expression of VIMP in MIN6 cells. Cell death was quantified by propidium iodide (PI) staining followed by flow cytometric analyses using a FACS Caliber and FlowJo software. Cell apoptosis and proliferation were determined by TUNEL assay and Ki67 staining, respectively. Cell cycle was analyzed after PI staining. RESULTS: The results show that 1) VIMP suppression induces ß-cell apoptosis, which is associated with a decrease in Bcl-xL, and the ß-cell apoptosis induced by VIMP suppression can be inhibited by overexpression of Bcl-xL; 2) VIMP knockdown (KD) decreases cell proliferation and G1 cell cycle arrest by accumulating p27 and decreasing E2F1; 3) VIMP KD suppresses unfolded protein response (UPR) activation by regulating the IRE1α and PERK pathways; 4) VIMP KD increases insulin secretion. CONCLUSION: These results suggest that VIMP may function as a novel regulator to modulate ß-cell survival, proliferation, cell cycle, UPR and insulin secretion in MIN6 cells.


Assuntos
Regulação para Baixo , Pontos de Checagem da Fase G1 do Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Células Secretoras de Insulina/patologia , Insulinoma/genética , Proteínas de Membrana/genética , Neoplasias Pancreáticas/genética , Selenoproteínas/genética , Animais , Apoptose , Morte Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Células Secretoras de Insulina/metabolismo , Insulinoma/patologia , Camundongos , Neoplasias Pancreáticas/patologia , Resposta a Proteínas não Dobradas
15.
Biochem Biophys Res Commun ; 503(4): 2866-2871, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30146262

RESUMO

As the most conserved branch of the unfolded protein response (UPR), the inositol-requiring enzyme 1a (IRE1a)/X-box binding protein 1 (XBP1) pathway plays crucial roles in cell survival and cell death by upregulating UPR-associated genes involved in protein entry into the endoplasmic reticulum (ER) and ER-associated degradation (ERAD). Selenoprotein S (SelS) is localized to the ER membrane and involved in ERAD. Although SelS plays an important role in restoring ER stress, the SelS-dependent protective mechanisms against cell death remain unclear. Here, using an inducible SelS knockdown (KD) 3T3-L1 cell model, we showed that SelS KD resulted adipocyte death, which was associated with imbalance of the Bcl-2 family members. Furthermore, SelS KD decreased spliced XBP1 (sXBP1), increased IRE1α and p-JNK, suggesting a role of SelS in the modulation of the IRE1α-sXBP1 pathway. Moreover, adipocyte death induced by SelS suppression can be inhibited by overexpression of sXBP1. Thus, it is proposed that SelS promotes cell survival through the IRE1α-XBP1 signaling pathway.


Assuntos
Adipócitos/metabolismo , Morte Celular/genética , Degradação Associada com o Retículo Endoplasmático , Endorribonucleases/genética , Proteínas Serina-Treonina Quinases/genética , Selenoproteínas/genética , Proteína 1 de Ligação a X-Box/genética , Células 3T3-L1 , Adipócitos/citologia , Animais , Diferenciação Celular , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/genética , Endorribonucleases/metabolismo , Regulação da Expressão Gênica , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Selenoproteínas/antagonistas & inibidores , Selenoproteínas/metabolismo , Transdução de Sinais , Proteína 1 de Ligação a X-Box/metabolismo
16.
Mediators Inflamm ; 2018: 1625414, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29805311

RESUMO

Endothelial dysfunction, partly induced by inflammatory mediators, is known to initiate and promote several cardiovascular diseases. Selenoprotein S (SelS) has been identified in endothelial cells and is associated with inflammation; however, its function in inflammation-induced endothelial dysfunction has not been described. We first demonstrated that the upregulation of SelS enhances the levels of nitric oxide and endothelial nitric oxide synthase in tumor necrosis factor- (TNF-) α-treated human umbilical vein endothelial cells (HUVECs). The levels of TNF-α-induced endothelin-1 and reactive oxygen species are also reduced by the upregulation of SelS. Furthermore, SelS overexpression blocks the TNF-α-induced adhesion of THP-1 cells to HUVECs and inhibits the increase in intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Moreover, SelS overexpression regulates TNF-α-induced inflammatory factors including interleukin-1ß, interleukin-6, interleukin-8, and monocyte chemotactic protein-1 and attenuates the TNF-α-induced activation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways. Conversely, the knockdown of SelS with siRNA results in an enhancement of TNF-α-induced injury in HUVECs. These findings suggest that SelS protects endothelial cells against TNF-α-induced dysfunction by inhibiting the activation of p38 MAPK and NF-κB pathways and implicates it as a possible modulator of vascular inflammatory diseases.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Selenoproteínas/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Selenoproteínas/genética
17.
J Diabetes Complications ; 31(8): 1286-1292, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28610945

RESUMO

PURPOSE: To analyze the impact of dynamic changes in inflammation on atherosclerosis in short-duration type 2 diabetes after multifactorial intervention. METHODS: In this randomized controlled study, a total of 150 type 2 diabetes patients who had a mean age of 49.8±7.3years, 51% male, with disease duration <1year and without evidence of atherosclerosis were randomized into an intensive intervention group (IG), in which patients received multiple risk factors intervention by the special project team and tried to reach the pre-determined intervention goals, and a conventional group (CG), in which patients received standard diabetes care by the clinic doctor. All patients recieved intervention study for 7 years, then underwent a 3-year observational follow-up study. The primary endpoints were occurrence of subclinical atherosclerosis, defined as the intima-media thickness of the common carotid artery (CC-IMT)≥1.0mm or formation of atherosclerosis plaques, and the occurrence of cardiovascular events. RESULTS: 68 patients in IG and 65 patients in CG completed the 10-year study. The cumulative incidence of subclinical atherosclerosis was 30.7% vs 57.3% (IG vs CG, HR 0.36, 95% CI 0.22-0.60, P<0.0001) and that of cardiovascular events was 12.0% vs 22.7% (IG vs CG, HR 0.46, 95% CI 0.21-0.98, P=0.0516). High sensitivity C-reactive protein (hs-CRP) reduction from baseline to the 10-year follow-up was -1.54mg/L (IG) and -0.67mg/L (CG) with difference (IG minus CG) of -0.87mg/L(95% CI -0.72 to -1.02, P=0.008) and the natural logarithm of serum amyloid A (SAA) reduction was -4.04 (IG) and -1.44 (CG) with difference (IG minus CG) of -2.60 (95% CI -2.30 to -2.90, P=0.002). The decrease in general score of inflammatory markers (combination of hs-CRP and SAA) was independently associated with subclinical atherosclerosis (OR=0.65, P=0.045) and cardiovascular events (OR=0.60, P=0.042). CONCLUSIONS: Dynamic changes in inflammation act as an important factor that affects the occurrence of atherosclerosis in type 2 diabetes patients. Multifactorial intensive intervention can reduce systemic low-grade inflammation and delay the occurrence of atherosclerosis in short-duration type 2 diabetes.


Assuntos
Aterosclerose/prevenção & controle , Diabetes Mellitus Tipo 2/terapia , Angiopatias Diabéticas/prevenção & controle , Estilo de Vida Saudável , Adulto , Doenças Assintomáticas/epidemiologia , Aterosclerose/complicações , Aterosclerose/epidemiologia , Aterosclerose/imunologia , Biomarcadores/sangue , Espessura Intima-Media Carotídea , China/epidemiologia , Terapia Combinada , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/imunologia , Angiopatias Diabéticas/diagnóstico por imagem , Angiopatias Diabéticas/epidemiologia , Angiopatias Diabéticas/imunologia , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Mediadores da Inflamação/sangue , Análise de Intenção de Tratamento , Perda de Seguimento , Masculino , Pessoa de Meia-Idade , Pacientes Desistentes do Tratamento , Placa Aterosclerótica/complicações , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/prevenção & controle , Fatores de Risco
18.
Cardiovasc Diabetol ; 15: 70, 2016 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-27121097

RESUMO

BACKGROUND: Selenoprotein S (SelS) is a transmembrane protein that is expressed in the liver, skeletal muscle, adipose tissue, pancreatic islets, kidney, and blood vessels. In addition to its transmembrane localization, SelS is also secreted from hepatoma HepG2 cells (but not L6 skeletal muscle cells, 3T3-L1 adipocytes, Min6 pancreatic ß cells and human embryonic kidney 293 cells) and has been detected in the serum of some human subjects, with a detection rate of 31.1 %. These findings prove that serum SelS is secreted by hepatocytes. However, whether vascularly expressed SelS can be secreted has not been reported. Transmembrane SelS has been suggested to play different roles in the pathogenesis and progression of diabetes mellitus (DM) and atherosclerosis (AS), but the association of secreted SelS with DM and macroangiopathy remains unclear. RESEARCH DESIGN AND METHODS: Supernatants were collected from human umbilical vein endothelial cells (HUVECs), human aortic vascular smooth muscle cells (HA/VSMCs) and human hepatoma HepG2 cells that were untransfected or transfected with the indicated plasmid and concentrated for western blotting. Serum samples were collected from 158 human subjects with or without type 2 DM (T2DM) and/or AS. Serum SelS levels were measured using an enzyme-linked immunosorbent assay. RESULTS: Secreted SelS was only detected in the supernatants of hepatoma HepG2 cells. The SelS detection rate among the 158 human serum samples was 100 %, and the average SelS level was 64.81 ng/dl. The serum SelS level in the isolated DM subjects was lower than the level in the healthy control subjects (52.66 ± 20.53 vs 70.40 ± 21.38 ng/dl). The serum SelS levels in the DM complicated with SAS subjects (67.73 ± 21.41 ng/dl) and AS subjects (71.69 ± 27.00 ng/dl) were significantly increased compared with the serum SelS level in the isolated DM subjects. There was a positive interaction effect between T2DM and AS on the serum SelS level (P = 0.002). Spearman correlation analysis showed that the serum SelS level was negatively correlated with fasting plasma glucose. CONCLUSIONS: Vascular endothelial and vascular smooth muscle cells could not secrete SelS. Serum SelS was primarily secreted by hepatocytes. SelS was universally detected in human serum samples, and the serum SelS level was associated with T2DM and its macrovascular complications. Thus, regulating liver and serum SelS levels might become a new strategy for the prevention and treatment of DM and its macrovascular complications.


Assuntos
Aterosclerose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Membrana/metabolismo , Selenoproteínas/metabolismo , Tecido Adiposo/metabolismo , Adulto , Idoso , Aterosclerose/complicações , Diabetes Mellitus Tipo 2/complicações , Feminino , Humanos , Ilhotas Pancreáticas/metabolismo , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade
19.
Exp Ther Med ; 10(5): 1937-1942, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26640576

RESUMO

The aim of the present study was to explore the associations among glycemic excursions, glycated hemoglobin (HbA1c) and high-sensitivity C-reactive protein (hs-CRP) in patients with poorly controlled type 2 diabetes mellitus (T2DM) using a continuous glucose monitoring system (CGMS). Sixty-three patients with T2DM whose HbA1c levels were >7% wore a CGMS device for 72 h. According to their HbA1c levels, patients were divided into three groups as follows: Group A (HbA1c ≤9.32%), group B (9.32%< HbA1c ≤11.76%) and group C (HbA1c >11.76%). Patients were also divided into two groups according to the mean amplitude of glycemic excursions (MAGE) as follows: Low glycemic excursion group (MAGE, <3.9 mmol/l) and high glycemic excursion group (MAGE, ≥3.9 mmol/l). Clinical data and the hs-CRP levels in different groups were compared. No significant difference was observed in the MAGE among groups A, B and C (P>0.05). The level of hs-CRP was significantly higher in group C compared with that in groups A and B, and in group B compared with that in group A (P<0.05). Multivariate stepwise regression analysis indicated that HbA1c correlated with hs-CRP (P<0.05). MAGE and HbA1c were independent indices for the assessment of glycemic control. In addition, HbA1c had a considerable effect on the serum hs-CRP level.

20.
Chin Med J (Engl) ; 127(5): 916-23, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24571888

RESUMO

BACKGROUND: The mechanisms underlying diabetic encephalopathy are largely unknown, and no effective treatments are available. Catalpol has received much attention due to its numerous biological effects, especially in neuroprotective studies. The aim of this study was to investigate the effects of catalpol on cognitive functions in diabetic rats and the underlying mechanisms. METHODS: A rat model of diabetes was established by streptozotocin injection, followed by intraperitoneal infusion of catalpol after 10 weeks. Two weeks later, the Morris water maze was used to test the spatial learning performance. Nissl staining was performed to evaluate the morphological changes in the hippocampus. Expression of protein kinase Cγ (PKCγ) and caveolin-1 (Cav-1) in the hippocampus were assessed by reverse transcription PCR and Western blotting. Activities of anti-oxidative enzymes such as glutathione (GSH), superoxide dismutase (SOD) and catalase (CAT) and levels of malonaldehyde (MDA) were measured using commercial kits. RESULTS: Significant hippocampal neuronal injury was observed in rats with streptozotocin-induced diabetes. Moreover, cognitive dysfunction was associated with markedly increased oxidative stress in the brain. Catalpol treatment significantly attenuated cognitive deficits, neuronal damage, and oxidative stress in the brain of diabetic rats. Biochemical analyses showed that catalpol reversed the down-regulation of PKCγ and Cav-1 expression in the diabetic rats. CONCLUSIONS: Spatial memory in diabetic rats is associated with the expression of PKCγ and Cav-1. Catalpol treatment markedly attenuated oxidative stress, reversed the alteration of PKCγ, Cav-1 and spatial memory deficits.


Assuntos
Caveolina 1/metabolismo , Diabetes Mellitus Experimental/metabolismo , Hipocampo/metabolismo , Glucosídeos Iridoides/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Proteína Quinase C/metabolismo , Memória Espacial/fisiologia , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/fisiopatologia , Hipocampo/efeitos dos fármacos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Memória Espacial/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...