Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 2392, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765080

RESUMEN

After ovulation, the mitochondrial enzyme CYP11A1 cleavage the cholesterol into pregnenolone for progesterone synthesis, suggesting that mitochondrial dynamics play a vital role in the female reproductive system. The changes in the mitochondria dynamics throughout the ovarian cycle have been reported in literature, but the correlation to its role in the ovarian cycle remains unclear. In this study, mitochondrial fusion promotor, M1, was used to study the impact of mitochondria dynamics in the female reproductive system. Our results showed that M1 treatment in mice can lead to the disruptions of estrous cycles in vagina smears. The decrease in serum LH was recorded in the animal. And the inhibitions of progesterone secretion and ovulations were observed in ovarian culture. Although no significant changes in mitochondrial networks were observed in the ovaries, significant up-regulation of mitochondrial respiratory complexes was revealed in M1 treatments through transcriptomic analysis. In contrast to the estrogen and steroid biosynthesis up-regulated in M1, the molecules of extracellular matrix, remodeling enzymes, and adhesion signalings were decreased. Collectively, our study provides novel targets to regulate the ovarian cycles through the mitochondria. However, more studies are still necessary to provide the functional connections between mitochondria and the female reproductive systems.


Asunto(s)
Dinámicas Mitocondriales , Progesterona , Ratones , Femenino , Animales , Proestro , Ciclo Estral/fisiología , Ovario , Estradiol
2.
PeerJ ; 10: e13867, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35990905

RESUMEN

Aims: Studies have observed changes in autophagic flux in the adipose tissue of type 2 diabetes patients with obesity. However, the role of autophagy in obesity-induced insulin resistance is unclear. We propose to confirm the effect of a high-fat diet (HFD) on autophagy and insulin signaling transduction from adipose tissue to clarify whether altered autophagy-mediated HFD induces insulin resistance, and to elucidate the possible mechanisms in autophagy-regulated adipose insulin sensitivity. Methods: Eight-week-old male C57BL/6 mice were fed with HFD to confirm the effect of HFD on autophagy and insulin signaling transduction from adipose tissue. Differentiated 3T3-L1 adipocytes were treated with 1.2 mM fatty acids (FAs) and 50 nM Bafilomycin A1 to determine the autophagic flux. 2.5 mg/kg body weight dose of Chloroquine (CQ) in PBS was locally injected into mouse epididymal adipose (10 and 24 h) and 40 µM of CQ to 3T3-L1 adipocytes for 24 h to evaluate the role of autophagy in insulin signaling transduction. Results: The HFD treatment resulted in a significant increase in SQSTM1/p62, Rubicon expression, and C/EBP homologous protein (CHOP) expression, yet the insulin capability to induce Akt (Ser473) and GSK3ß (Ser9) phosphorylation were reduced. PHLPP1 and PTEN remain unchanged after CQ injection. In differentiated 3T3-L1 adipocytes treated with CQ, although the amount of phospho-Akt stimulated by insulin in the CQ-treated group was significantly lower, CHOP expressions and cleaved caspase-3 were increased and bafilomycin A1 induced less accumulation of LC3-II protein. Conclusion: Long-term high-fat diet promotes insulin resistance, late-stage autophagy inhibition, ER stress, and apoptosis in adipose tissue. Autophagy suppression may not affect insulin signaling transduction via phosphatase expression but indirectly causes insulin resistance through ER stress or apoptosis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Ratones , Masculino , Animales , Dieta Alta en Grasa/efectos adversos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo/metabolismo , Obesidad/tratamiento farmacológico , Insulina/farmacología , Autofagia
3.
Sci Rep ; 11(1): 15564, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330986

RESUMEN

In mammalian ovaries, mitochondria are integral sites of energy production and steroidogenesis. While shifts in cellular activities and steroidogenesis are well characterized during the differentiation of large luteal cells in folliculogenesis and luteal formation, mitochondrial dynamics during this process have not been previously evaluated. In this study, we collected ovaries containing primordial follicles, mature follicles, corpus hemorrhagicum, or corpus luteum from goats at specific times in the estrous cycle. Enzyme histochemistry, ultrastructural observations, and 3D structural analysis of serial sections of mitochondria revealed that branched mitochondrial networks were predominant in follicles, while spherical and tubular mitochondria were typical in large luteal cells. Furthermore, the average mitochondrial diameter and volume increased from folliculogenesis to luteal formation. In primordial follicles, the signals of cytochrome c oxidase and ATP synthase were undetectable in most cells, and the large luteal cells from the corpus hemorrhagicum also showed low enzyme signals and content when compared with granulosa cells in mature follicles or large luteal cells from the corpus luteum. Our findings suggest that the mitochondrial enlargement could be an event during folliculogenesis and luteal formation, while the modulation of mitochondrial morphology and respiratory enzyme expressions may be related to tissue remodeling during luteal formation.


Asunto(s)
Lipogénesis/fisiología , Células Lúteas/metabolismo , Animales , Femenino , Cabras , Dinámicas Mitocondriales/fisiología
4.
Sci Rep ; 11(1): 12561, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131222

RESUMEN

Obese men have lower circulating testosterone than men with an optimal body mass index. Elevated fatty acids (FAs) caused by obesity have been reported to suppress the steroidogenesis of Leydig cells. Recent studies have demonstrated that autophagy regulates steroidogenesis in endocrine cells; however, few studies have investigated the molecular mechanisms of FA-impaired steroidogenesis. To study FA regulation in the steroidogenesis of Leydig cells, MA-10 cells were treated with an FA mixture and co-treated with 8-Br-cAMP to stimulate the steroidogenesis capacity. We showed that FAs led to cellular lipid accumulation and decreased steroidogenesis of MA-10 cells, and FA-suppressed steroidogenesis was largely recovered by P5 treatment but not by 22R-OHC treatment, suggesting the primary defect was the deficiency of CYP11A1. To examine the involvement of autophagy in the steroidogenesis of Leydig cells, we treated MA-10 cells with autophagy regulators, including rapamycin, bafilomycin, and chloroquine. Inhibition of late-stage autophagy including FA-upregulated Rubicon suppressed the steroidogenesis of MA-10 cells. More interestingly, Rubicon played a novel regulatory role in the steroidogenesis of MA-10 cells, independent of inhibitors of late-stage autophagy. Collectively, this study provides novel targets to investigate the interaction between FAs and steroidogenesis in steroidogenic cells.


Asunto(s)
Autofagia/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Ácidos Grasos/metabolismo , Esteroides/metabolismo , Animales , Autofagia/efectos de los fármacos , Cloroquina/farmacología , Células Endocrinas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Lípidos/genética , Macrólidos/farmacología , Masculino , Ratones , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Sirolimus/farmacología , Esteroides/biosíntesis
5.
J Chin Med Assoc ; 84(4): 389-399, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33784266

RESUMEN

BACKGROUND: Kisspeptin (KISS1) and kisspeptin receptor (KISS1R) are essential gatekeepers of the reproductive system. The functions of KISS1 and KISS1R in corpus luteal cells remain ambiguous. The objective was to observe normal physiologic functions of corpus luteal cells in vivo and clarify the functions of KISS1 in vitro. METHODS: We conducted an in vivo observation of cellular patterns as well as the levels of steroidogenic enzymes and KISS1/KISS1R in corpus luteal cells obtained from female crossbred Taiwan native goats in the estrous cycle; the observation was performed using hematoxylin and eosin and immunohistochemistry staining. Subsequently, we used kisspeptin-10 (Kp-10) to stimulate temperature sensitive-caprine luteal cell line (ts-CLC-D) cells to investigate the progesterone (P4) levels, steroidogenic messenger RNA (mRNA)/protein levels, cell survival rate, intracellular Ca2+ concentration, and cell proliferation-related mRNA/protein levels in the mitogen-activated protein kinase pathway in vitro by applying immunofluorescence staining, Western blotting, 2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide assay, and real-time polymerase chain reaction. RESULTS: We observed the presence of proteins and mRNAs for STAR, CYP11A1, HSD3B, KISS1, and KISS1R in the corpus luteal cells from goats in vivo. In vitro, the addition of Kp-10 reduced the P4 levels (p < 0.01) and increased cell proliferation (p < 0.05) of the ts-CLC-D cells. Furthermore, we found that the levels of proteins and mRNA for STAR, CYP11A1, and HSD3B decreased significantly when Kp-10 was added (p < 0.05). However, adding Kp-10 did not affect the mRNA levels for PLCG2, DAG1, PRKCA, KRAS, RAF1, MAP2K1, MAP2K2, MAPK3, MAPK1, and MAPK14. CONCLUSION: We determined that KISS1 could affect the P4 levels, steroidogenesis, and cell proliferation in luteal cells. However, further research is required to clarify how KISS1 regulates proliferation and steroid production in luteal cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Kisspeptinas/farmacología , Células Lúteas/efectos de los fármacos , Animales , Supervivencia Celular , Femenino , Expresión Génica/genética , Cabras , Reacción en Cadena de la Polimerasa , ARN Mensajero , Taiwán
6.
J Chin Med Assoc ; 84(2): 203-211, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33543882

RESUMEN

BACKGROUND: Kisspeptin and its receptor KISS1R have been found to be essential regulators of reproductive function. Previous data have revealed the presence of Kiss1 and Kiss1r mRNAs in the hypothalamus and the testis of humans and rodents. However, the precise location and possible physiological role of the kisspeptin/KISS1R system in the testis remain ambiguous. METHODS: We first produced an anti-KISS1R immunoglobulin Y antibody for KISS1R identification. To detect the exact sites of KISS1R and kisspeptin expression in the testis, we conducted immunohistochemistry assays on sections of testes. We used real-time polymerase chain reactions to identify Kiss1r in mice and to determine the expression levels of testicular genes. Finally, to verify the upstream regulation on the Kisspeptin/KISS1 receptor system, we treated primary mouse Leydig cells and MA-10 cells with luteinizing hormone (LH) and Br-cAMP, respectively, and examined Kiss1 and Kiss1r mRNA expression. RESULTS: Immunohistochemistry assays revealed that kisspeptin was expressed in Leydig cells and KISS1R was localized in the seminiferous tubules. With real-time polymerase chain reactions, we found Kiss1r mRNA was constitutively expressed in the mouse testis from birth until the postnatal fourth week. Furthermore, mRNA expression of Kiss1 was synchronized with that of Insl3 and Cyp19a. However, the expression of the LH receptor-encoding gene increased 1 week earlier than did Kiss1 expression. This indicated that the kisspeptin/KISS1R system in the testis may be controlled by LH and cAMP signaling pathways. Finally, we confirmed that Kiss1 mRNA expression was increased in both LH-treated primary Leydig cells and Br-cAMP-treated MA-10 cells (p < 0.05). On the other hand, cotreatment of both cell lines with Br-cAMP and a protein kinase A inhibitor RP-cAMP significantly suppressed 50% of Br-cAMP-induced Kiss1 expression (p < 0.05). CONCLUSION: We discovered that Kiss1 expression in mouse Leydig cells was induced by LH through the cAMP/PKA pathway. Based on the presence of kisspeptin receptors on spermatids, we inferred that kisspeptin- and development-related factors have synergistic effects on spermatogenesis. Nevertheless, more studies are required to elaborate the role of the kisspeptin/KISS1R system in testicular development.


Asunto(s)
Receptores de Kisspeptina-1/metabolismo , Testículo/crecimiento & desarrollo , Animales , Humanos , Masculino , Ratones
7.
Reproduction ; 160(6): 843-852, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33112765

RESUMEN

Kisspeptin and its receptor KISS1R have been proven as pivotal regulators on controlling the hypothalamus-pituitary-gonad axis. Inactivating mutations in one of them cause idiopathic hypogonadotropic hypogonadism in human as well as rodent models. Notably, gonadotropin insensitivity, failure in hCG response, was presented in the male patients with loss-function-mutations in KISS1R gene; this reveals the essential role of KISS1R signaling in regulating testosterone production beyond the hypothalamic functions of kisspeptin. In this study, we hypothesized that the autocrine action of kisspeptin on Leydig cells may modulate steroidogenesis. Based on the mouse cell model, we first demonstrated that the cAMP/protein kinase A (PKA)/cAMP response element-binding protein (CREB) signaling pathway mediated gonadotropin-induced kisspeptin expression. By using siRNA interfering technique, knockdown of Kiss1r in MA-10 cells, a mouse Leydig tumor cell line, significantly reduced progesterone productions in both basal and hCG-treated conditions. Integrating the results from both quantitative real-time PCR and steroidogenic enzyme-activity assay, we found that this steroidogenic defect was associated with decreased luteinizing hormone/choriogonadotropin receptor (Lhcgr) and StAR protein (Star) expressions. Furthermore, exogenous expression of human LHCGR completely rescued hCG-stimulated progesterone production in the KISS1R-deficient cells. In conclusion, we proposed that the reproductive functions of KISS1R signaling in Leydig cell include modulating Lhcgr and steroidogenic gene expressions, which may shed the light on the pathophysiology of gonadotropin insensitivity.


Asunto(s)
Gonadotropina Coriónica/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/farmacología , Progesterona/biosíntesis , Receptores de Kisspeptina-1/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Receptores de Kisspeptina-1/genética , Sustancias para el Control de la Reproducción/farmacología , Transducción de Señal
8.
J Chin Med Assoc ; 2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33044408

RESUMEN

BACKGROUND: Kisspeptin and its receptor KISS1R have been found to be essential regulators of reproductive function. Previous data have revealed the presence of Kiss1 and Kiss1r mRNAs in the hypothalamus and the testis of humans and rodents. However, the precise location and possible physiological role of the kisspeptin/KISS1R system in the testis remain ambiguous. METHODS: We first produced an anti-KISS1R immunoglobulin Y antibody for KISS1R identification. To detect the exact sites of KISS1R and kisspeptin expression in the testis, we conducted immunohistochemistry assays on sections of testes. We used real-time polymerase chain reactions (PCR) to identify Kiss1r in mice and to determine the expression levels of testicular genes. Finally, to verify the upstream regulation on the Kisspeptin/KISS1 receptor system, we treated primary mouse Leydig cells and MA-10 cells with luteinizing hormone (LH) and Br-cAMP, respectively and examined Kiss1 and Kiss1r mRNA expression. RESULTS: Immunohistochemistry assays revealed that kisspeptin was expressed in Leydig cells and KISS1R was localized in the seminiferous tubules. With real-time PCR, we found Kiss1r mRNA was constitutively expressed in the mouse testis from birth until the postnatal fourth week. Furthermore, mRNA expression of Kiss1 was synchronized with that of Insl3 and Cyp19a. However, the expression of the LH receptor-encoding gene increased 1 week earlier than did Kiss1 expression. This indicated that the kisspeptin/KISS1R system in the testis may be controlled by LH and cAMP signaling pathways. Finally, we confirmed that Kiss1 mRNA expression was increased in both LH-treated primary Leydig cells and Br-cAMP-treated MA-10 cells (p < 0.05). On the other hand, cotreatment of both cell lines with Br-cAMP and a protein kinase A inhibitor RP-cAMP significantly suppressed 50% of Br-cAMP-induced Kiss1 expression (p < 0.05). CONCLUSION: We discovered that Kiss1 expression in mouse Leydig cells was induced by LH through the cAMP/PKA pathway. Based on the presence of kisspeptin receptors on spermatids, we inferred that kisspeptin and development-related factors have synergistic effects on spermatogenesis. Nevertheless, more studies are required to elaborate the role of the kisspeptin/KISS1R system in testicular development.

9.
Oncol Lett ; 20(5): 206, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32963612

RESUMEN

Patients with lung cancer harboring activating epidermal growth factor (EGFR) mutations and pre-existing diabetes have been demonstrated to exhibit poor responses to first-line EGFR-tyrosine kinase inhibitor (TKI) therapy. Strategies for the management of acquired resistance to EGFR-TKIs in patients with advanced non-small cell lung cancer (NSCLC) are urgently required. Only a limited number of studies have been published to date on the effects of insulin on EGFR-TKI resistance in NSCLC. Hence, the aim of the present study was to investigate the roles of hyperinsulinemia and hyperglycemia in mediating gefitinib resistance in NSCLC cells with activating EGFR mutations. In the present study, the HCC4006 cell line, which harbors EGFR mutations, was co-treated with gefitinib and long-acting insulin glargine. Whether hyperinsulinemia is able to mediate EGFR-TKI resistance in the NSCLC cell line harboring activating EGFR mutations was also investigated, and the possible underlying mechanisms responsible for these actions were explored. The inhibition of cell proliferation, and the potential mechanism of gefitinib resistance, were examined using an MTS proliferation assay and western blot analysis, and through the transfection of siRNAs. Whether the inhibition of AKT is able to overcome EGFR-TKI resistance induced by long-acting insulin was also investigated. The results obtained suggested that hyperinsulinemia induced by glargine upregulated NSCLC cell proliferation and survival, and induced gefitinib resistance. By contrast, the morphology and proliferation of the cells in a medium containing a 2-fold concentration of glucose were not significantly affected. Gefitinib resistance induced by hyperinsulinemia may have been mediated via the phosphoinositide 3-kinase (PI3K)/AKT pathway rather than the mitogen-activated protein kinase extracellular signal regulated kinase (MAPK/ERK) pathway. AKT serine/threonine kinase 1 knockdown by siRNA rescued the gefitinib resistance that was induced by hyperinsulinemia. In conclusion, hyperinsulinemia, but not hyperglycemia, was identified to cause the development of gefitinib resistance in NSCLC cells with activating EGFR mutations. However, additional studies are required to investigate strategies, such as co targeting hyperinsulinemia and the PI3K/AKT pathway, for overcoming EGFR-TKI resistance in patients with NSCLC.

10.
J Sci Food Agric ; 100(6): 2443-2452, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31951016

RESUMEN

BACKGROUND: Cardio-renal syndrome (CRS) is an integrative problem related to chronic malnutrition, obesity, etc. Amino acids and peptides are regarded as protective and essential for tissues. Pepsin-digested chicken liver hydrolysates (CLHs), which are made from the byproducts of the poultry industry, are amino-acid based and of animal origin, and may be protective against the myocardial and renal damage induced by a high-fat diet (HFD). RESULTS: Our results showed that CLHs contain large quantities of anserine, taurine, and branched-chain amino acids (BCAAs), and supplementing the diet with CLHs reduced (P < 0.05) weight gain, liver weight, peri-renal fat mass / adipocyte-area sizes, serum total cholesterol (TC), aspartate aminotransferase (AST), and low-density lipoprotein cholesterol (LDLC) levels in HFD-fed mice but increased (P < 0.05) serum high-density lipoprotein cholesterol (HDLC) levels. By histological analyses, CLHs alleviated (P < 0.05) renal lipid deposition and fibrosis, as well as cardiac fibrosis and inflammation of HFD-fed mice. Meanwhile, increased (P < 0.05) inflammatory and fibrotic cytokines levels in the myocardia of the HFD-fed mice were downregulated (P < 0.05) by CLH supplementation. Regarding autophagy-related protein levels, protective effects of CLHs on the myocardia against HFD feeding may result from the early blockade of the autophagy pathway to prevent autophagosome accumulation. CONCLUSION: Functional CLHs could be a novel food ingredient as a cardio-renal protective agent against a high-fat dietary habit in a niche market. © 2020 Society of Chemical Industry.


Asunto(s)
Síndrome Cardiorrenal/dietoterapia , Dieta Alta en Grasa/efectos adversos , Hígado/química , Hidrolisados de Proteína/administración & dosificación , Animales , Aspartato Aminotransferasas/sangre , Autofagia , Pollos , Colesterol/sangre , Fibrosis , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Miocardio/patología , Hidrolisados de Proteína/química
11.
Sci Rep ; 9(1): 17451, 2019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767891

RESUMEN

Previous studies have demonstrated the important role of kisspeptin in impaired glucose-stimulated insulin secretion (GSIS). In addition, it was reported that the activation of autophagy in pancreatic ß-cells decreases insulin secretion by selectively degrading insulin granules. However, it is currently unknown whether kisspeptin suppresses GSIS in ß-cells by activating autophagy. To investigate the involvement of autophagy in kisspeptin-regulated insulin secretion, we overexpressed Kiss1 in NIT-1 cells to mimic the long-term exposure of pancreatic ß-cells to kisspeptin during type 2 diabetes (T2D). Interestingly, our data showed that although kisspeptin potently decreases the intracellular proinsulin and insulin ((pro)insulin) content and insulin secretion of NIT-1 cells, autophagy inhibition using bafilomycin A1 and Atg5 siRNAs only rescues basal insulin secretion, not kisspeptin-impaired GSIS. We also generated a novel in vivo model to investigate the long-term exposure of kisspeptin by osmotic pump. The in vivo data demonstrated that kisspeptin lowers GSIS and (pro)insulin levels and also activated pancreatic autophagy in mice. Collectively, our data demonstrated that kisspeptin suppresses both GSIS and non-glucose-stimulated insulin secretion of pancreatic ß-cells, but only non-glucose-stimulated insulin secretion depends on activated autophagic degradation of (pro)insulin. Our study provides novel insights for the development of impaired insulin secretion during T2D progression.


Asunto(s)
Autofagia/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Kisspeptinas/fisiología , Animales , Línea Celular , Diabetes Mellitus Tipo 2/fisiopatología , Genes Reporteros , Glucosa/farmacología , Kisspeptinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Proinsulina/metabolismo , Proteínas Recombinantes/metabolismo , Transfección
12.
Endocr J ; 65(8): 833-840, 2018 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-29887570

RESUMEN

Although curcumin was widely applied as a functional food for different diseases, it was found to reduce serum testosterone level and fertility in male animals by unknown molecular mechanisms. Here in our study, we investigated the possible mechanisms of curcumin-suppressed testosterone production in Leydig cells. Our enzyme immunoassay results showed that curcumin cell-autonomously suppressed ovine luteinizing hormone-stimulated testosterone production in primary Leydig cells and 8-bromo-cyclic adenosine monophosphate (8-br-cAMP)-induced progesterone production in MA-10 cells. Furthermore, our real-time PCR, Western blot, and 22R-OHC/pregnenolone supplementing experiment data demonstrated that curcumin suppressed 8-br-cAMP-induced steroidogenesis in Leydig cells by inhibiting the expression of StAR and Cyp11a1. Interestingly, our Western blot data showed that although curcumin suppressed PKA activity, it did not alter the 8-br-cAMP-induced phosphorylation of CREB. On the contrary, the real-time PCR results showed that curcumin suppressed 8-br-cAMP-induced expression of Nr5a1 and Fos, which are crucial for cAMP-stimulated StAR and Cyp11a1 expression in Leydig cells. Collectively, our data demonstrated that curcumin may suppress cAMP-induced steroidogenesis in mouse Leydig cells by down-regulating Nr5a1/Fos-controlled StAR and Cyp11a1 expression independently of the PKA-CREB signaling pathway.


Asunto(s)
Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Curcumina/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Células Intersticiales del Testículo/efectos de los fármacos , Fosfoproteínas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Línea Celular , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/farmacología , Masculino , Ratones , Progesterona/biosíntesis , Transducción de Señal/fisiología , Testosterona/biosíntesis
13.
Chem Biol Interact ; 290: 12-18, 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-29753610

RESUMEN

Macrophages and inflammasome pathway are involved in high-glucose toxicity and development of insulin resistance. Silymarin (SMR) was known to modulate glucose homeostasis and reduce inflammation. However, it is still unknown whether SMR possess anti-hyperglycemic effects in diabetic-like knockout mice (Hnf-1αkin/-/Ins.cre mice) with insulin resistance and also unclear how SMR regulates LPS induced stress markers and pro-inflammatory cytokines under stresses of high glucose (HG) or NLRP3 inflammasome activation. Current results show that oral administration of SMR (100 mg/kg) reduced hyperglycemia in the mouse model of maturity-onset diabetes of the young type 3-like mice. In cultured macrophages, SMR (5-20 µg/ml) reduces high glucose (HG)-enhanced expressions of inducible nitric oxide synthase, nitric oxide generation stimulated by LPS; however, no effects on COX-2 expressions. The enhanced interleukin-1ß (ΙL-1ß) secretions in the presence of HG or palmitate were also significantly down regulated by SMR in dose-dependent manner in LPS-treated macrophages. Such observations may result from the decreased extracellular signal-regulated kinase 1/2 phosphorylation, while without affecting protein kinase C-α phosphorylation and nuclear factor-κB activation. These findings together show that SMR acts as a protector against HG-related stresses not only by lowering hyperglycemia but also suppressing HG- and inflammasome-mediated IL-1ß expressions to improve insulin resistance.


Asunto(s)
Glucosa/farmacología , Hiperglucemia/patología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Silimarina/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Factor Nuclear 1-alfa del Hepatocito/deficiencia , Factor Nuclear 1-alfa del Hepatocito/genética , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Inflamasomas/efectos de los fármacos , Resistencia a la Insulina , Interleucina-1beta/genética , Lipopolisacáridos/toxicidad , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Células RAW 264.7 , Silimarina/uso terapéutico
14.
Sci Rep ; 7(1): 13999, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-29070903

RESUMEN

Previous studies have demonstrated that saturated fatty acids (SFAs) are more lipotoxic than unsaturated fatty acids (UFAs) in inhibiting hepatic autophagy and promoting non-alcoholic steatohepatitis (NASH). However, there have been few studies have investigated the effects of carbon chain length on SFA-induced autophagy impairment and lipotoxicity. To investigate whether SFAs with shorter carbon chain lengths have differential effects on hepatic autophagy and NASH development, we partially replaced lard with coconut oil to elevate the ratio of medium-chain fatty acids (MCFAs) to long-chain fatty acids (LCFAs) in a mouse high-fat diet (HFD) and fed mice for 16 weeks. In addition, we treated HepG2 cells with different combinations of fatty acids to study the mechanisms of MCFAs-mediated hepatic protections. Our results showed that increasing dietary MCFA/LCFA ratio mitigated HFD-induced Type 2 diabetes and NASH in mice. Importantly, we demonstrated that increased MCFA ratio exerted its protective effects by restoring Rubicon-suppressed autophagy. Our study suggests that the relative amount of LCFAs and MCFAs in the diet, in addition to the amount of SFAs, can significantly contribute to autophagy impairment and hepatic lipotoxicity. Collectively, we propose that increasing dietary MCFAs could be an alternative therapeutic and prevention strategy for Type 2 diabetes and NASH.


Asunto(s)
Autofagia , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/farmacología , Ácidos Grasos/farmacología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Animales , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología
15.
Diabetol Metab Syndr ; 9: 81, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29046729

RESUMEN

BACKGROUND: During the prediabetic development, the changes in ß-cell function and tissue-specific insulin resistance have been described. However, there are conflicting views in insulin secretory capacity between early clinical observation and recent proposed mathematical model. On the basis of digestive and metabolic similarities with humans, swine have great potential as an animal model to investigate the progressive mechanisms of prediabetes. The aim of this study was to investigate the insulin secretory response and tissue-specific insulin resistance in a dietary-induced prediabetic porcine model. METHODS: Adult male Taiwan Lee-Sung miniature pigs were randomized into two groups: (1) low-fat diet and (2) high-fat plus high-fructose diet (HFHF; 20.9% crude fat and 17.8% fructose). During the 12-month dietary intervention, body weights and blood glucose levels were measured monthly. Intravenous glucose tolerance test was used for measuring glucose tolerance and insulin secretory capacity. At the end of the experiment, liver and soleus muscle specimens were collected for ex vivo insulin sensitivity testing. RESULTS: The results showed that the HFHF group had obesity, hyperinsulinemia, and dyslipidemia, but normal fasting glucose levels. The HFHF pigs exhibited enhanced first- and second-phase insulin secretion and high 2-h postload glucose levels in intravenous glucose tolerance test. Furthermore, the skeletal muscle specimens from the HFHF group were desensitized to insulin stimulation as shown by the lack of AKT Ser473 phosphorylation; however, the liver specimens remained a normal response. CONCLUSIONS: In conclusion, the HFHF diet-fed pigs developed isolated impaired glucose tolerance corresponding to prediabetes with an intense insulin secretory response and skeletal muscle insulin resistance.

16.
Environ Toxicol ; 32(6): 1792-1800, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28181416

RESUMEN

Thioacetamide (TAA), usually used as a fungicide to control the decay of citrus products, itself is not toxic to the liver, but its intermediates are able to increase oxidative stress in livers and further cause fibrosis. Ophiocordyceps sinensis mycelium (OSM) which contains 10% polysaccharides and 0.25% adenosine decreased (P < 0.05) the lipid accumulation and increased (P < 0.05) antioxidative capacity in livers of thioacetamide (TAA) injected rats. Meanwhile, the increased (P < 0.05) liver sizes, serum alanine transaminase (AST) and aspartate transaminase (ALT) values in thioacetamide (TAA)-injected rats were ameliorated (P < 0.05) by OSM supplementation. Moreover, the levels of proinflammatory cytokines, such as the tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), were also reduced (P < 0.05). The fibrosis phenomena in pathological (Masson's trichrome and H&E stainings) and immunohistochemical [α-smooth actin (αSMA) and CD86/ED1] observations in TAA-treated rats were reduced (P < 0.05) by OSM cotreatment. The protective effect of OSM against TAA-induced liver inflammation/fibrosis may be via downregulations (P < 0.05) of TGF-ß pathways and NFκB which further influenced (P < 0.05) the expressions of fibrotic and inflammatory genes (i. e., αSMA, Col1α, COX2). Therefore, OSM shows preventive effects on the development of TAA-induced hepatic fibrosis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Hypocreales/química , Cirrosis Hepática/prevención & control , Micelio/química , Tioacetamida/toxicidad , Actinas/metabolismo , Alanina Transaminasa/sangre , Animales , Antiinflamatorios/aislamiento & purificación , Antioxidantes/metabolismo , Aspartato Aminotransferasas/sangre , Citocinas/metabolismo , Interleucina-1beta/metabolismo , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Pruebas de Función Hepática , Masculino , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
17.
Biomed Res Int ; 2016: 9576503, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27034954

RESUMEN

Valproic acid (VPA) is one of the most widely used antiepilepsy drugs. However, several side effects, including weight gain and fatty liver, have been reported in patients following VPA treatment. In this study, we explored the molecular mechanisms of VPA-induced hepatic steatosis using FL83B cell line-based in vitro model. Using fluorescent lipid staining technique, we found that VPA enhanced oleic acid- (OLA-) induced lipid accumulation in a dose-dependent manner in hepatocytes; this may be due to upregulated lipid uptake, triacylglycerol (TAG) synthesis, and lipid droplet formation. Real-time PCR results showed that, following VPA treatment, the expression levels of genes encoding cluster of differentiation 36 (Cd36), low-density lipoprotein receptor-related protein 1 (Lrp1), diacylglycerol acyltransferase 2 (Dgat2), and perilipin 2 (Plin2) were increased, that of carnitine palmitoyltransferase I a (Cpt1a) was not affected, and those of acetyl-Co A carboxylase α (Acca) and fatty acid synthase (Fasn) were decreased. Furthermore, using immunofluorescence staining and flow cytometry analyses, we found that VPA also induced peroxisome proliferator-activated receptor γ (PPARγ) nuclear translocation and increased levels of cell-surface CD36. Based on these results, we propose that VPA may enhance OLA-induced hepatocyte steatosis through the upregulation of PPARγ- and CD36-dependent lipid uptake, TAG synthesis, and lipid droplet formation.


Asunto(s)
Antígenos CD36/biosíntesis , Hígado Graso/genética , Metabolismo de los Lípidos/efectos de los fármacos , PPAR gamma/biosíntesis , Ácido Valproico/efectos adversos , Antígenos CD36/genética , Diacilglicerol O-Acetiltransferasa/biosíntesis , Ácidos Grasos/biosíntesis , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado Graso/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Lipoproteínas LDL/biosíntesis , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , PPAR gamma/genética , Perilipina-2/biosíntesis , Triglicéridos/biosíntesis , Triglicéridos/metabolismo , Ácido Valproico/uso terapéutico
18.
Anim Reprod Sci ; 170: 38-50, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27102356

RESUMEN

The present study was designed to study the ultrastructure of goat corpora lutea (CL, n=10) and structural changes as related to steroidogenic functions during the estrous cycle. The reproduction status of goats was estimated by analyzing serum progesterone concentrations. The CL at various stages was surgically collected. To characterize ultrastructural features associated with steroidogenesis, tissue and cellular structures were studied. Blood supplies were examined based on features of the endothelial cells and capillary structures in the CL. Activated endothelial cells and developing vessels were observed in the early stage, whereas mature endothelial cells, accumulating extracellular matrix fibers, and stabilized vessels were observed in the middle and late stages of assessment. In the late stage of assessment, shrunken goat luteal cells scattered around the capillaries were detected and formed circular regression areas. Features of autophagy and luteal cell apoptosis were noted. In large luteal cells, steroidogenic organelles were present, including microvillar channels, endoplasmic reticulum, and mitochondria. Conformational changes in the endoplasmic reticulum and increased mitochondria with tubular cristae were observed in the early-middle CL transitions. In contrast, mitochondria swelled and the cristae transformed to the lamellar type in the late stage, suggesting that organelle plasticity could contribute to steroidogenesis in goat CL. In conclusion, results suggest angiogenesis occurs in early developing CL and programmed cell death occurred in the late stage of CL assessment in the present study. Structures and quantiles of steroidogenic organelles are correlated with the steroidogenic functions in goats.


Asunto(s)
Cuerpo Lúteo/ultraestructura , Ciclo Estral/fisiología , Cabras/fisiología , Animales , Cuerpo Lúteo/fisiología , Femenino
19.
Sleep Med Rev ; 28: 125-32, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26694311

RESUMEN

Obstructive sleep apnea (OSA) is a highly prevalent disease which carries substantial public health burden. Polysomnography is the standard procedure used to diagnose OSA. However cost, accessibility, technical requirements, and skilled interpretation needs constrain its widespread use and have a role in the under-diagnosis of sleep disordered breathing. There is a clinical need to develop expedient and widely accessible tools to detect this disorder., Several biochemical markers have recently been proposed as diagnostic tools in OSA. Numerous neurochemicals directly influence the activity of upper airway dilator motor neurons, which subsequently influence respiration during sleep. Serotonin (5-HT) is one such neurochemical that has a key role in ventilatory stimulation. Herein, we review the current evidence demonstrating relationships between multiple biomarkers and sleep disordered breathing and focus on relationships between OSA and 5-HT. We discuss the possibility of biomarker-driven detection technology in the future as a means of diagnosing and monitoring OSA. Finally, we explore the specific role 5-HT may have in the future in both the diagnosis and treatment of OSA.


Asunto(s)
Serotonina/metabolismo , Apnea Obstructiva del Sueño/metabolismo , Biomarcadores/metabolismo , Humanos , Sistema Respiratorio/fisiopatología , Sueño
20.
J Chin Med Assoc ; 78(4): 249-57, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25732868

RESUMEN

BACKGROUND: Kisspeptin, encoded by the Kiss1 gene, has many forms including kisspeptin54, kisspeptin14, kisspeptin13, and kisspeptin10, and all these peptides have the same affinity to their receptor KISS1R encoded by the Kiss1r gene. The KISS1-KISS1R system was discovered in neurons, and many reports stress on their function in the brain. However, recent studies have shown that Kiss1 and Kiss1r are expressed in the testes. The goal of this study was to demonstrate the roles of Kiss1 and Kiss1r in testicular function, especially their steroidogenic activity. METHODS: Kisspeptin10 and the kisspeptin10 antagonist peptide234 were used to determine their effect on testosterone production. Moreover, expression of steroidogenic genes in mouse testes and their gonadosomatic index (weight of the testes divided by the total body weight) and also serum testosterone level were studied between the ages of 2 weeks and 15 weeks. RESULTS: Kisspeptin10 and peptide234 did not affect testosterone production in primary Leydig cells from adult mice. Kiss1 and Esr1 expression also increased during puberty. The peak gonadosomatic index occurred at 4 weeks of age, and serum testosterone levels plateaued after the age of 4 weeks. CONCLUSION: Our results suggest that kisspeptin10 does not affect steroidogenesis in adult Leydig cells, but its pattern of expression follows the stages of testicular development. Future studies should determine if kisspeptin regulates testicular development during puberty.


Asunto(s)
Kisspeptinas/fisiología , Células Intersticiales del Testículo/fisiología , 3-Hidroxiesteroide Deshidrogenasas/genética , Factores de Edad , Animales , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Receptor alfa de Estrógeno/genética , Masculino , Ratones , Ratones Endogámicos ICR , Receptores Acoplados a Proteínas G/fisiología , Receptores de Kisspeptina-1 , Testosterona/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...