Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Mol Reprod Dev ; 89(4): 202-213, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35307908

RESUMEN

Dendrobium nobile Lindl polysaccharides (DNLP) exhibited various biological functions. This study aimed to investigate the protective effects of DNLP on testicular spermatogenic function in streptozotocin (STZ)-induced diabetic rats in comparison with metformin. The blood glucose level was significantly increased and the homeostatic model assessment for insulin resistance (HOMA-IR) aggravated markedly in diabetic rats. The weight of testis and epididymis, and the sperm number and motility were decreased in the diabetic rats. The pathologic changes occurred in the spermatogenic tubules along with the decreased number of spermatogenic cells, downregulated proliferating cell nuclear antigen (PCNA) and Sirtuin 1 (SIRT1) expression and increased cell apoptosis in the testes. Compared with the model group, DNLP and metformin treatment significantly decreased the level of blood glucose, improved the HOMA-IR, and increased the weight of testis and epididymis, as well as the sperm number and sperm motility. Furthermore, the pathologic changes in the spermatogenic tubules improved significantly with increased number of spermatogenic cells, the upregulation of PCNA and SIRT1 and suppression of cell apoptosis in the testes. Collectively, our study for the first time examined the effects of DNLP on the male reproductive system of STZ-induced diabetic rats, and indicated that DNLP was protective against diabetes mellitus-induced testis injury via increasing the proliferation, inhibiting cell apoptosis and upregulating SIRT1 expression in testicular spermatogenic cells.


Asunto(s)
Dendrobium , Diabetes Mellitus Experimental , Metformina , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Masculino , Metformina/farmacología , Metformina/uso terapéutico , Polisacáridos/metabolismo , Polisacáridos/farmacología , Antígeno Nuclear de Célula en Proliferación , Ratas , Sirtuina 1/metabolismo , Motilidad Espermática , Estreptozocina/efectos adversos , Estreptozocina/metabolismo , Testículo/metabolismo
2.
Curr Mol Med ; 21(7): 539-548, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33272176

RESUMEN

Male fertility is closely related to the normal function of the hypothalamicpituitary- testicular axis. The testis is an important male reproductive organ that secretes androgen and produces sperm through spermatogenesis. Spermatogenesis refers to the process by which spermatogonial stem cells (SSCs) produce highly differentiated spermatozoa and is divided into three stages: mitosis, meiosis and spermiogenesis. Spermatogenesis requires SSCs to strike a proper balance between self-renewal and differentiation and the commitment of spermatocytes to meiosis, which involves many molecules and signalling pathways. Abnormal gene expression or signal transduction in the hypothalamus and pituitary, but particularly in the testis, may lead to spermatogenic disorders and male infertility. The phosphoinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathway is involved in many stages of male reproduction, including the regulation of the hypothalamus-pituitarygonad (HPG) axis during spermatogenesis, the proliferation and differentiation of spermatogonia and somatic cells, and the regulation of sperm autophagy and testicular endocrine function in the presence of environmental pollutants, particularly endocrinedisrupting chemicals (EDCs). In the PI3K/AKT/mTOR signalling pathway, mTOR is considered the central integrator of several signals, regulating metabolism, cell growth and proliferation. In particular, mTOR plays an important role in the maintenance and differentiation of SSCs, as well as in regulating the redox balance and metabolic activity of Sertoli cells, which play an important role in nutritional support during spermatogenesis.


Asunto(s)
Fertilidad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Espermatogénesis , Serina-Treonina Quinasas TOR/metabolismo , Humanos , Masculino , Células de Sertoli/metabolismo , Espermatogonias/metabolismo
3.
Exp Biol Med (Maywood) ; 246(2): 177-186, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33023330

RESUMEN

Long noncoding RNAs play an important role in the occurrence, invasion, as well as metastasis of various human cancers, including hepatocellular carcinoma. Long noncoding RNAs can affect the biological functions of hepatocellular carcinoma cells by regulating various genes; however, only a small fraction of molecular mechanisms of long noncoding RNAs have been elucidated. In the present study, lnc AC010973.1 (lnc-ATG9B-4) was first identified by microarray analysis from 8 patients with hepatocellular carcinoma and confirmed by quantitative PCR in 176 patients with hepatocellular carcinoma. We demonstrated that lnc-ATG9B-4 was tightly relative to the tumorous size, TNM stages, portal vein tumor thrombus (PVTT), the tumor capsule, metastasis, degree of differentiation, and poor prognosis of hepatocellular carcinoma according to long-term follow-up data. In hepatocellular carcinoma cells, overexpression of lnc-ATG9B-4 promoted proliferation, invasion, as well as migration, while inhibiting lnc-ATG9B-4 by siRNA significantly attenuated the proliferation, invasion, as well as migration. Interestingly, lnc-ATG9B-4 increased the expression of cyclin-dependent kinase 5 (CDK5), which was closely related to the development and chemotherapy sensitivity of hepatocellular carcinoma. In summary, our results revealed that lnc-ATG9B-4 suggests an unfavorable prognosis of hepatocellular carcinoma and facilitates the proliferation, invasion, as well as migration of hepatocellular carcinoma cells by upregulating CDK5. This research suggests that lnc-ATG9B-4 may be a new biomarker for predicting the prognosis of hepatocellular carcinoma; meanwhile, targeting lnc-ATG9B-4 might serve as a potential strategy for the treatment hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/genética , Movimiento Celular/genética , Quinasa 5 Dependiente de la Ciclina/genética , Progresión de la Enfermedad , Neoplasias Hepáticas/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , Invasividad Neoplásica , ARN Largo no Codificante/genética , Análisis de Supervivencia
4.
Cytokine ; 135: 155242, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32799009

RESUMEN

Midkine (MK) is a low molecular-weight protein that was first identified as the product of a retinoic acid-responsive gene involved in embryonic development. Recent studies have indicated that MK levels are related to various diseases, including cardiovascular disease (CVD), renal disease and autoimmune disease. MK is a growth factor involved in multiple pathophysiological processes, such as inflammation, the repair of damaged tissues and cancer. The pathophysiological roles of MK are diverse. MK enhances the recruitment and migration of inflammatory cells upon inflammation directly and also through induction of chemokines, and contributes to tissue damage. In lung endothelial cells, oxidative stress increased the expression of MK, which induced angiotensin-converting enzyme (ACE) expression and the consequent conversion from Ang I to Ang II, leading to further oxidative stress. MK inhibited cholesterol efflux from macrophages by reducing ATP-binding cassette transporter A1 (ABCA1) expression, which is involved in lipid metabolism, suggesting that MK is an important positive factor involved in inflammation, oxidative stress and lipid metabolism. Furthermore, MK can regulate the expansion, differentiation and activation of T cells as well as B-cell survival; mediate angiogenic and antibacterial activity; and possess anti-apoptotic activity. In this paper, we summarize the pathophysiological roles of MK in human disease.


Asunto(s)
Midkina/metabolismo , Animales , Apoptosis/fisiología , Enfermedad , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo
5.
Life Sci ; 255: 117830, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32450172

RESUMEN

Advanced glycation end products (AGEs) are heterogeneous products of the non-enzymatic interaction between proteins and reducing sugars. Numerous studies have shown that AGEs are associated with senescence, diabetes, vascular disease, aging and kidney disease. Infertility has been affected approximately 10 to15% of couples of reproductive ages. AGEs accumulation has been shown to play a crucial role in pathogenesis of infertility-related diseases. The present review provides the generation process, mechanism and pathological significance of AGEs and the novel treatment targeting AGEs for infertility.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Infertilidad/fisiopatología , Animales , Femenino , Humanos , Infertilidad/epidemiología , Masculino
6.
Circ J ; 84(2): 217-225, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31915322

RESUMEN

BACKGROUND: Midkine (MK), a heparin-binding protein, participates in multiple cellular processes, such as immunity, cellular growth and apoptosis. Overwhelming evidence indicates that MK plays an important role in various pathological processes, including chronic inflammation, autoimmunity, cancer, and infection. Recent studies demonstrated that MK may be involved in the development of atherosclerosis, yet the mechanism has not been fully explored. Therefore, this study aims to investigate the effect and mechanism of MK on macrophage cholesterol efflux.Methods and Results:Using Oil Red O staining, NBD-cholesterol fluorescence labeling and enzymatic methods, it observed that MK markedly promoted macrophage lipid accumulation. Liquid scintillation counting (LSC) showed that MK decreased cholesterol efflux. Moreover, cell immunofluorescence, western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) showed that MK downregulated ATP-binding membrane cassette transport protein A1 (ABCA1) expression. Functional promotion of ABCA1 expression attenuated the inhibitory effects of MK on cholesterol efflux, which reduced lipid accumulation. Additionally, intervention of adenosine monophosphate activated protein (AMPK)-mammalian target of rapamycin (mTOR) signaling molecule by the AMPK activator, AICAR, increased p-AMPK and ABCA1 expression, decreased p-mTOR expression and promoted cholesterol efflux, resulting in an obvious reduction in intracellular lipid content. CONCLUSIONS: These data suggest that MK reduces the expression of ABCA1, inhibits the efflux of cholesterol and promotes the accumulation of lipids in RAW264.7 macrophages, and AMPK-mTOR signaling is involved in MK-mediated regulation of cholesterol metabolism in RAW264.7 macrophages.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Colesterol/metabolismo , Macrófagos/efectos de los fármacos , Midkina/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Animales , Regulación hacia Abajo , Activación Enzimática , Macrófagos/enzimología , Ratones , Fosforilación , Células RAW 264.7 , Transducción de Señal
7.
Clin Chim Acta ; 502: 214-221, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31733195

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex and heterogeneous endocrine disease characterized by clinical or laboratorial hyperandrogenism, oligo-anovulation and metabolic abnormalities, including insulin resistance, excessive weight or obesity, type II diabetes, dyslipidemia and an increased risk of cardiovascular disease. The most significant clinical manifestation of PCOS is hyperandrogenism. Excess androgen profoundly affects granulosa cell function and follicular development via complex mechanisms that lead to obesity and insulin resistance. Most PCOS patients with hyperandrogenism have steroid secretion defects that result in abnormal folliculogenesis and failed dominant follicle selection. Hyperandrogenism induces obesity, hairy, acne, and androgenetic alopecia. These symptoms can bring great psychological stress to women. Drugs such as combined oral contraceptive pills, metformin, pioglitazone and low-dose spironolactone help improve pregnancy rates by decreasing androgen levels in vivo. Notably, PCOS is heterogeneous, and hyperandrogenism is not the only pathogenic factor. Obesity and insulin resistance aggravate the symptoms of hyperandrogenism, forming a vicious cycle that promotes PCOS development. Although numerous studies have been conducted, the definitive pathogenic mechanisms of PCOS remain uncertain. This review summarizes and discusses previous and recent findings regarding the relationship between hyperandrogenism, insulin resistance, obesity and PCOS.


Asunto(s)
Hiperandrogenismo/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Andrógenos/biosíntesis , Femenino , Humanos , Hiperandrogenismo/diagnóstico , Hiperandrogenismo/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Obesidad/diagnóstico , Obesidad/tratamiento farmacológico , Pioglitazona/uso terapéutico , Síndrome del Ovario Poliquístico/diagnóstico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Espironolactona/uso terapéutico
8.
Clin Chim Acta ; 499: 142-148, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31525346

RESUMEN

Polycystic ovary syndrome (PCOS), one of the most common endocrine diseases that causes infertility in reproductive women, is characterized by hyperandrogenemia, chronic anovulation, and polycystic ovary morphology (PCOM), and most women with PCOS have metabolic abnormalities. A reduction in plasma sex hormone-binding globulin (SHBG), a transport carrier that binds estrogen and androgens and regulates their biological activities, is often used as an indicator of hyperandrogenism in women with PCOS. Low serum SHBG levels are considered a biomarker of abnormal metabolism and are related to insulin resistance (IR), compensatory hyperinsulinemia and abnormalities in glucose and lipid metabolism in PCOS patients. SHBG is also associated with the long-term prognosis of PCOS. SHBG gene polymorphism is correlated with the risk of PCOS. As SHBG plays a vital role in the occurrence and development of PCOS, knowledge regarding its role in PCOS is helpful for further understanding the molecular mechanism of SHBG in PCOS development and providing new ideas for the treatment of female infertility. Hepatocyte nuclear factor-4α (HNF-4α) is a vital transcription factor in the SHBG synthesis process. HNF-4α binds to the cis-type element DR1 in the SHBG promoter to initiate transcription and regulates hepatic SHBG levels by modulating glucose and lipid metabolism and inflammatory factors. However, it remains unclear whether HNF-4α is indirectly involved in the pathogenesis of PCOS via regulation of hepatic SHBG synthesis. Therefore, this review discusses the interaction between SHBG and the various complications of PCOS as well as the regulatory effect of HNF-4α on SHBG expression.


Asunto(s)
Síndrome del Ovario Poliquístico/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Femenino , Humanos , Globulina de Unión a Hormona Sexual/genética
9.
Taiwan J Obstet Gynecol ; 58(4): 447-453, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31307731

RESUMEN

Polycystic ovary syndrome (PCOS) is widely accepted as the most common endocrine abnormality in women of childbearing age and may be accompanied by dyslipidemia, hyperandrogenism, hyperinsulinemia, oxidative stress and infertility. Dyslipidemia is now known to play an important role in the development of PCOS. Lipid abnormalities, including elevated low-density lipoprotein and triglyceride levels and reduced high-density lipoprotein levels, are often found in women with PCOS and play an important role in PCOS; therefore, we summarize the effect of lipid abnormalities on hyperandrogenism, insulin resistance, oxidative stress and infertility in PCOS and review the effects of common lipid-lowering drugs on patients with PCOS. The purpose of this article is to elucidate the mechanisms of lipid metabolism abnormalities in the development of PCOS.


Asunto(s)
Dislipidemias/epidemiología , Hiperandrogenismo/epidemiología , Hipolipemiantes/uso terapéutico , Resistencia a la Insulina/fisiología , Síndrome del Ovario Poliquístico/epidemiología , Adulto , Comorbilidad , Dislipidemias/diagnóstico , Dislipidemias/tratamiento farmacológico , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperandrogenismo/diagnóstico , Persona de Mediana Edad , Síndrome del Ovario Poliquístico/diagnóstico , Prevalencia , Pronóstico , Medición de Riesgo
10.
Clin Chim Acta ; 497: 54-60, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31302099

RESUMEN

Male fertility depends on the regulatory balance between germ cell self-renewal and differentiation, and the spatial and temporal patterns of this balance must be maintained throughout the life cycle. Retinoic acid and its receptors are important factors in spermatogenesis. Spermatogonia cells can self-proliferate and differentiate and have unique meiotic capabilities; they halve their genetic material and produce monomorphic sperm to pass genetic material to the next generation. A number of studies have found that the spermatogenesis process is halted in animals with vitamin A deficiency and that most germ cells are degraded, but they tend to recover after treatment with RA or vitamin A. This literature review discusses our understanding of how RA regulates sperm cell differentiation and meiosis and also reviews the functional information and details of RA.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Meiosis/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Espermatogonias/patología , Tretinoina/farmacología , Animales , Humanos , Masculino , Espermatogonias/citología , Espermatogonias/metabolismo
11.
Curr Pharm Biotechnol ; 20(5): 422-432, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30947667

RESUMEN

BACKGROUND: Tanshinone IIA (Tan IIA) and Omentin-1 have a protective role in the cardiovascular system. However, if and how Tan IIA and Omentin-1 regulate cholesterol metabolism in macrophages has not been fully elucidated. OBJECTIVE: To investigate the possible mechanisms of Tan IIA and Omentin-1 on preventing macrophage cholesterol accumulation and atherosclerosis development. METHODS: The effect of Tan IIA on the protein and mRNA levels of Omentin-1 and ATP-binding cassette transporter A1 (ABCA1) in macrophages was examined by Western blot and qRT-PCR assay, respectively. Cholesterol efflux was assessed by liquid scintillation counting (LSC). Cellular lipid droplet was measured by Oil Red O staining, and intracellular lipid content was detected by high performance liquid chromatography (HPLC). In addition, the serum lipid profile of apoE-/- mice was measured by enzymatic method. The size of atherosclerotic lesion areas and content of lipids and collagen in the aortic of apoE-/- mice were examined by Sudan IV, Oil-red O, and Masson staining, respectively. RESULTS: Tan IIA up-regulated expression of Omentin-1 and ABCA1 in THP-1 macrophages, promoting ABCA1-mediated cholesterol efflux and consequently decreasing cellular lipid content. Consistently, Tan IIA increased reverse cholesterol transport in apoE-/- mice. Plasma levels of high-density lipoprotein cholesterol (HDL-C), ABCA1 expression and atherosclerotic plaque collagen content were increased while plasma levels of low-density lipoprotein cholesterol (LDL-C) and atherosclerotic plaque sizes were reduced in Tan IIA-treated apoE-/- mice. These beneficial effects were, however, essentially blocked by knockdown of Omentin-1. CONCLUSION: Our results revealed that Tan IIA promotes cholesterol efflux and ameliorates lipid accumulation in macrophages most likely via the Omentin-1/ABCA1 pathway, reducing the development of aortic atherosclerosis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Abietanos/farmacología , Aterosclerosis/tratamiento farmacológico , Colesterol/metabolismo , Citocinas/metabolismo , Proteínas Ligadas a GPI/metabolismo , Lectinas/metabolismo , Macrófagos/efectos de los fármacos , Abietanos/uso terapéutico , Animales , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Transporte Biológico , Línea Celular Tumoral , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Regulación hacia Arriba
12.
Int J Oncol ; 53(5): 1827-1835, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30106141

RESUMEN

The novel neurite outgrowth inhibitor B (Nogo­B) receptor (NgBR) is specific for Nogo­B, which is highly expressed in various human organs and cells, including the lung, liver, kidney, smooth muscle cells, blood vessel endothelial cells and inflammatory cells. Previous studies have indicated that NgBR directly interacts with Nogo­B and is able to independently influence lipid and cholesterol homeostasis, angiogenesis, N­glycosylation, the epithelial-mesenchymal transition, the chemotaxis of endothelial cells and cellular proliferation and apoptosis. These multiple functions and actions of this receptor provide an understanding of the important roles of NgBR in various conditions, including fatty liver, atherosclerosis, intracranial microaneurysms, retinitis pigmentosa and severe neurological impairment. Furthermore, NgBR has been demonstrated to exert protean, multifunctional and enigmatic effects in cancer. The present review summarizes the latest knowledge on the suppressing and activating effects of NgBR, emphasizing its function in cancer. Further basic and medical research on this receptor may provide novel insight into its clinical implications on the prognosis of relevant human cancer types.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Neoplasias/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Estrés del Retículo Endoplásmico , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , Metabolismo de los Lípidos , Receptores X del Hígado/metabolismo , Neoplasias/patología , Neovascularización Patológica/etiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Respuesta de Proteína Desplegada/fisiología
13.
Atherosclerosis ; 270: 57-67, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29407889

RESUMEN

BACKGROUND AND AIMS: Liver scavenger receptor class B type I (SR-BI) exerts atheroprotective effects through selective lipid uptake (SLU) from high-density lipoprotein cholesterol (HDL-C). Low hepatic SR-BI expression leads to high HDL-C levels in the circulation and an increased risk of atherosclerosis. Furthermore, macrophage SR-BI mediates bidirectional cholesterol flux and may protect against atherogenesis. Previous studies have revealed that miR-24 is closely related to cardiovascular disease (CVD) progression. We aimed to investigate the molecular mechanisms by which miR-24 participates in SR-BI-mediated selective HDL cholesteryl ester (HDL-CE) uptake and further atherogenesis in apoE-/- mice. METHODS: Bioinformatic predictions and luciferase reporter assays were utilized to detect the association between miR-24 and the SR-BI 3' untranslated region (3' UTR), and RT-PCR and western blotting were used to evaluate SR-BI mRNA and protein expression, respectively. The effects of miR-24 on Dil-HDL uptake were determined by flow cytometry assay. Double-radiolabeled HDL (125I-TC-/[3H] CEt-HDL) was utilized to measure the effects of miR-24 on HDL and CE binding and SLU in HepG2 and PMA-treated THP-1 cells. In addition, total cholesterol (TC) levels in HepG2 cells were analyzed using enzymatic methods, and macrophage lipid content was evaluated by high-performance liquid chromatography (HPLC) assay. Small interfering RNA (siRNA) and pcDNA3.1(-)-hSR-BI plasmid transfection procedures were utilized to confirm the role of SR-BI in the effects of miR-24 on Dil-HDL uptake, SLU and cholesterol levels in both cell types. Hepatic SR-BI level in apoE-/- mice was measured by western blotting. Liver TC, FC and CE levels and plasma triglycerides (TG), TC and HDL-C levels were evaluated enzymatically using commercial test kits. Atherosclerotic lesion sizes were measured using Oil Red O and hematoxylin-eosin staining. RESULTS: miR-24 directly repressed SR-BI expression by targeting its 3'UTR. In addition, miR-24 decreased Dil-HDL uptake and SLU in HepG2 and THP-1 macrophages. In the presence of HDL, miR-24 decreased TC levels in HepG2 cells and TC, free cholesterol (FC) and CE levels in macrophages. Overexpression and down-regulation assays showed that SR-BI mediated the effects of miR-24 on Dil-HDL uptake, SLU and cholesterol levels. Lastly, miR-24 administration decreased hepatic SR-BI expression and promoted atheromatous plaque formation in apoE-/- mice, findings in line with those of our in vitro studies. CONCLUSIONS: These findings indicate that miR-24 accelerates atherogenesis by repressing SR-BI-mediated SLU from HDL-C.


Asunto(s)
Aterosclerosis/sangre , HDL-Colesterol/sangre , Hígado/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Receptores Depuradores de Clase B/metabolismo , Regiones no Traducidas 3' , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Sitios de Unión , Modelos Animales de Enfermedad , Células HEK293 , Células Hep G2 , Humanos , Masculino , Ratones Noqueados para ApoE , MicroARNs/genética , Procesamiento Postranscripcional del ARN , Receptores Depuradores de Clase B/genética , Células THP-1
14.
Acta Pharmacol Sin ; 39(8): 1249-1258, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29323337

RESUMEN

Atherosclerosis is an autoimmune disease caused by self- and non-self-antigens contributing to excessive activation of T and B cell immune responses. These responses further aggravate vascular infiammation and promote progression of atherosclerosis and vulnerability to plaques via releasing pro-infiammatory cytokines. Regulatory T cells (Tregs) as the major immunoregulatory cells, in particular, induce and maintain immune homeostasis and tolerance by suppressing the immune responses of various cells such as T and B cells, natural killer (NK) cells, monocytes, and dendritic cells (DCs), as well as by secreting inhibitory cytokines interleukin (IL)-10, IL-35 and transcription growth factor ß (TGF-ß) in both physiological and pathological states. Numerous evidence demonstrates that reduced numbers and dysfunction of Treg may be involveved in atherosclerosis pathogenesis. Increasing or restoring the numbers and improving the immunosuppressive capacity of Tregs may serve as a fundamental immunotherapy to treat atherosclerotic cardiovascular diseases. In this article, we briefiy present current knowledge of Treg subsets, summarize the relationship between Tregs and atherosclerosis development, and discuss the possibilities of regulating Tregs for prevention of atherosclerosis pathogenesis and enhancement of plaque stability. Although the exact molecular mechanisms of Treg-mediated protection against atherosclerosis remain to be elucidated, the strategies for targeting the regulation of Tregs may provide specific and significant approaches for the prevention and treatment of atherosclerotic cardiovascular diseases.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Colesterol/metabolismo , Células Dendríticas/metabolismo , Células Espumosas/metabolismo , Humanos , Inmunoterapia , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
15.
Mol Med Rep ; 17(1): 705-713, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115523

RESUMEN

The Sertoli cell, which is the supporting cell of spermatogenesis, has an important role in the endocrine and paracrine control of spermatogenesis. Functionally, it provides the cells of the seminiferous epithelium with nutrition, conveys mature spermatids to the lumen of seminiferous tubules, secretes androgen­binding protein and interacts with endocrine Leydig cells. In addition, the levels of cholesterol, as well as its intermediates, vary greatly between nongonadal tissues and the male reproductive system. Throughout spermatogenesis, a dynamic and constant alteration in the membrane lipid composition of Sertoli cells occurs. In several mammalian species, testis meiosis­activating sterol and desmosterol, as well as other cholesterol precursors, accumulate in the testes and spermatozoa. In addition, certain cholesterogenic genes exhibit stage­specific expression patterns during spermatogenesis, including the cytochrome P450 enzyme lanosterol 14α­demethylase. Inconsistency in the patterns of gene expression during spermatogenesis indicates a cell­type specific and complex temporary modulation of lipids and cholesterol, which also implicates the dynamic interactions between Sertoli cells and germ cells. Furthermore, in the female reproductive tract and during epididymal transit, which is a prerequisite for valid fertilization, the modulation of cholesterol occurring in spermatozoal membranes further indicates the functional importance of sterol compounds in spermatogenesis. However, the exact role of cholesterol metabolism in Sertoli cells in sperm production is unknown. The present review article describes the progress made in the research regarding the characteristics of the Sertoli cell, particularly the regulation of its cholesterol metabolism during spermatogenesis.


Asunto(s)
Colesterol/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis/genética , Esterol 14-Desmetilasa/genética , Colesterol/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Metabolismo de los Lípidos/genética , Masculino , Esterol 14-Desmetilasa/metabolismo , Testículo/metabolismo
16.
DNA Cell Biol ; 36(12): 1142-1150, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29058484

RESUMEN

Members of the reticulon protein family are predominantly distributed within the endoplasmic reticulum. The neurite outgrowth inhibitor (Nogo) has three subtypes, including Nogo-A (200 kDa), Nogo-B (55 kDa), and Nogo-C (25 kDa). Nogo-A and Nogo-C are potent Nogos that are predominantly expressed in the central nervous system. Nogo-B, the splice variant of reticulon-4, is expressed widely in multiple human organ systems, including the liver, lung, kidney, blood vessels, and inflammatory cells. Moreover, the Nogo-B receptor (NgBR) can interact with Nogo-B and can independently affect nervous system regeneration, the chemotaxis of endothelial cells, proliferation, and apoptosis. In recent years, it has been demonstrated that NgBR plays an important role in human pathophysiological processes, including lipid metabolism, angiogenesis, N-glycosylation, cell apoptosis, chemoresistance in human hepatocellular carcinoma, and epithelial-mesenchymal transition. The pathophysiologic effects of NgBR have garnered increased attention, and the detection and enhancement of NgBR expression may be a novel approach to monitor the development and to improve the prognosis of relevant human clinical diseases.


Asunto(s)
Metabolismo de los Lípidos , Proteínas Nogo/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Apoptosis , Proteínas Portadoras/metabolismo , Proliferación Celular , Fosfatos de Dolicol/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Humanos , Trastornos del Metabolismo de los Lípidos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Fisiológica , Enfermedad de Niemann-Pick Tipo C/metabolismo , Receptores Nogo/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Transporte Vesicular
17.
J. physiol. biochem ; 73(2): 287-296, mayo 2017. graf, ilus
Artículo en Inglés | IBECS | ID: ibc-168485

RESUMEN

Endothelial dysfunction plays a vital role during the initial stage of atherosclerosis. Oxidized low-density lipoprotein (ox-LDL) induces vascular endothelial injury and vessel wall inflammation. Sphingosine-1-phosphate (S1P) exerts numerous vasoprotective effects by binding to diverse S1P receptors (S1PRs; S1PR1-5). A number of studies have shown that in endothelial cells (ECs), S1PR2 acts as a pro-atherosclerotic mediator by stimulating vessel wall inflammation through the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Scavenger receptor class B member I (SR-BI), a high-affinity receptor for apolipoprotein A-I (apoA-I)/high-density lipoprotein (HDL), inhibits nuclear factor-κB (NF-κB) translocation and decreases the plasma levels of inflammatory mediators via the PI3K/Akt pathway. We hypothesized that the inflammatory effects of S1P/S1PR2 on ECs may be regulated by apoA-I/SR-BI. The results showed that ox-LDL, a pro-inflammatory factor, augmented the S1PR2 level in human umbilical vein endothelial cells (HUVECs) in a dose- and time-dependent manner. In addition, S1P/S1PR2 signaling influenced the levels of inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-10, aggravating inflammation in HUVECs. Moreover, the pro-inflammatory effects induced by S1P/S1PR2 were attenuated by SR-BI overexpression and enhanced by an SR-BI inhibitor, BLT-1. Further experiments showed that the PI3K/Akt signaling pathway was involved in this process. Taken together, these results demonstrate that apoA-I/SR-BI negatively regulates S1P/S1PR2-mediated inflammation in HUVECs by activating the PI3K/Akt signaling pathway (AU)


No disponible


Asunto(s)
Humanos , Endotelio Vascular/metabolismo , Lisofosfolípidos/metabolismo , Receptores Depuradores de Clase B/agonistas , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Transducción de Señal , Fosfatidilinositol 3-Quinasas/metabolismo , Regulación de la Expresión Génica , Transporte Activo de Núcleo Celular , Ciclopentanos/farmacología , Tiosemicarbazonas/farmacología , Factor de Necrosis Tumoral alfa , Células Endoteliales de la Vena Umbilical Humana , Lipoproteínas LDL , Proteínas Proto-Oncogénicas c-akt
18.
J Physiol Biochem ; 73(2): 287-296, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28181168

RESUMEN

Endothelial dysfunction plays a vital role during the initial stage of atherosclerosis. Oxidized low-density lipoprotein (ox-LDL) induces vascular endothelial injury and vessel wall inflammation. Sphingosine-1-phosphate (S1P) exerts numerous vasoprotective effects by binding to diverse S1P receptors (S1PRs; S1PR1-5). A number of studies have shown that in endothelial cells (ECs), S1PR2 acts as a pro-atherosclerotic mediator by stimulating vessel wall inflammation through the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Scavenger receptor class B member I (SR-BI), a high-affinity receptor for apolipoprotein A-I (apoA-I)/high-density lipoprotein (HDL), inhibits nuclear factor-κB (NF-κB) translocation and decreases the plasma levels of inflammatory mediators via the PI3K/Akt pathway. We hypothesized that the inflammatory effects of S1P/S1PR2 on ECs may be regulated by apoA-I/SR-BI. The results showed that ox-LDL, a pro-inflammatory factor, augmented the S1PR2 level in human umbilical vein endothelial cells (HUVECs) in a dose- and time-dependent manner. In addition, S1P/S1PR2 signaling influenced the levels of inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-10, aggravating inflammation in HUVECs. Moreover, the pro-inflammatory effects induced by S1P/S1PR2 were attenuated by SR-BI overexpression and enhanced by an SR-BI inhibitor, BLT-1. Further experiments showed that the PI3K/Akt signaling pathway was involved in this process. Taken together, these results demonstrate that apoA-I/SR-BI negatively regulates S1P/S1PR2-mediated inflammation in HUVECs by activating the PI3K/Akt signaling pathway.


Asunto(s)
Apolipoproteína A-I/metabolismo , Endotelio Vascular/metabolismo , Lisofosfolípidos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Receptores Depuradores de Clase B/agonistas , Transducción de Señal , Esfingosina/análogos & derivados , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apolipoproteína A-I/genética , Células Cultivadas , Ciclopentanos/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-10/agonistas , Interleucina-10/metabolismo , Interleucina-1beta/agonistas , Interleucina-1beta/metabolismo , Cinética , Lipoproteínas LDL/efectos adversos , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Receptores Depuradores de Clase B/antagonistas & inhibidores , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Tiosemicarbazonas/farmacología , Factor de Necrosis Tumoral alfa/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
19.
Lipids ; 52(2): 109-117, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28039587

RESUMEN

Apolipoprotein M (apoM) is a relatively novel apolipoprotein that plays pivotal roles in many dyslipidemia-associated diseases; however, its regulatory mechanisms are poorly understood. Many cytokines have been identified that down-regulate apoM expression in HepG2 cells, among which transforming growth factor-ß (TGF-ß) exerts the most potent effects. In addition, c-Jun, a member of the activated protein 1 (AP-1) family whose activity is modulated by c-Jun N-terminal kinase (JNK), decreases apoM expression at the transcriptional level by binding to the regulatory element in the proximal apoM promoter. In this study, we investigated the molecular mechanisms through which TGF-ß decreases the apoM level in HepG2 cells. The results revealed that TGF-ß inhibited apoM expression at both the mRNA and protein levels in a dose- and time-dependent manner and that it suppressed apoM secretion. These effects were attenuated by treatment of cells with either SP600125 (JNK inhibitor) or c-Jun siRNA. 5Z-7-oxozeaenol [(a TGF-ß-activated kinase 1 (TAK-1) inhibitor)] also attenuated the TGF-ß-mediated inhibition of apoM expression and suppressed the activation of JNK and c-Jun. These results have demonstrated that TGF-ß suppresses apoM expression through the TAK-1-JNK-c-Jun pathway in HepG2 cells.


Asunto(s)
Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Antracenos/farmacología , Apolipoproteínas M , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Lactonas/farmacología , Regiones Promotoras Genéticas , Resorcinoles/farmacología , Factores de Tiempo
20.
J. physiol. biochem ; 72(4): 657-667, dic. 2016. graf, ilus
Artículo en Inglés | IBECS | ID: ibc-168373

RESUMEN

Sphingosine-1-phosphate (S1P), which has emerged as a pivotal signaling mediator that participates in the regulation of multiple cellular processes, is derived from various cells, including vascular endothelial cells. S1P accumulates in lipoproteins, especially HDL, and the majority of free plasma S1P is bound to HDL. We hypothesized that HDL-associated S1P is released through mechanisms associated with the HDL maturation process. ApoA-I, a major HDL apolipoprotein, is a critical factor for nascent HDL formation and lipid trafficking via ABCA1. Moreover, apoA-I is capable of promoting bidirectional lipid movement through SR-BI. In the present study, we confirmed that apoA-I can facilitate the production and release of S1P by HUVECs. Furthermore, we demonstrated that ERK1/2 and SphK activation induced by apoA-I is involved in the release of S1P from HUVECs. Inhibitor and siRNA experiments showed that ABCA1 and SR-BI are required for S1P release and ERK1/2 phosphorylation induced by apoA-I. However, the effects triggered by apoA-I were not suppressed by inhibiting ABCA1/JAK2 or the SR-BI/Src pathway. S1P released due to apoA-I activation can stimulate the (ERK1/2)/SphK1 pathway through S1PR (S1P receptor) 1/3. These results indicated that apoA-I not only promotes S1P release through ABCA1 and SR-BI but also indirectly activates the (ERK1/2)/SphK1 pathway by releasing S1P to trigger their receptors. In conclusion, we suggest that release of S1P induced by apoA-I from endothelial cells through ABCA1 and SR-BI is a self-positive-feedback process: apoA-I-(ABCA1 and SR-BI)-(S1P release)-S1PR-ERK1/2-SphK1-(S1P production)-(more S1P release induced by apoA-I) (AU)


No disponible


Asunto(s)
Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apolipoproteína A-I/farmacología , Lisofosfolípidos , Receptores Depuradores de Clase B , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Esfingosina/análogos & derivados , Transportador 1 de Casete de Unión a ATP/metabolismo , Transducción de Señal , Regulación de la Expresión Génica , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica , Células Endoteliales de la Vena Umbilical Humana , ARN Interferente Pequeño
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA