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

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Life Sci ; 267: 118451, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-32956667

RESUMEN

AIMS: We investigated the in vitro differentiation of adult rat PDESCs into ß-like cells through supplementation of different combinations of GABA, BMP7, and Activin A in basic culture media. MATERIALS AND METHODS: The PDESCs were cultured using different inducement combinations for 28 days and microscopy, dithizone (DTZ) staining, immunohistochemical staining, real-time PCR, and glucose-stimulated insulin secretion (GSIS) assay were used to delineate the differentiation inducement potential of these combinations. KEY FINDINGS: The results show that after 28 days, the PDESCs were differentiated into ICCs containing insulin-secreting ß-like cells in different groups treated with A + B, A + G, B + G, and A + B + G but not in the control group. Upon DTZ staining the cells in ICCs were stained crimson red, demonstrating the presence of ß-like cells in ICCs and the immunohistochemistry showed the expression of Pdx1 and insulin in ICCs. Further, on 28 d the expression of Pdx1 and insulin mRNA was high in inducement groups as compared to the control group and ß-like cells in ICCs also secreted insulin and C-peptide upon glucose stimulation. Thus, the supplementation of GABA, BMP7, and Activin A in different combinations in basic culture media can induce the in vitro differentiation of PDESCs into ICCs containing ß-like cells. SIGNIFICANCE: The in vitro development of ß-like cells is a herald for cell therapy of diabetic patients and our results are a step closer towards finding the cure for diabetes.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Madre/metabolismo , Activinas/metabolismo , Activinas/farmacología , Animales , Proteína Morfogenética Ósea 7/metabolismo , Proteína Morfogenética Ósea 7/farmacología , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Cultivadas , Medios de Cultivo/química , Insulina/metabolismo , Secreción de Insulina , Conductos Pancreáticos/citología , Conductos Pancreáticos/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
2.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33179113

RESUMEN

Rheumatic heart disease (RHD) is an autoimmune disease caused by rheumatic fever following group A hemolytic streptococcal infection and primarily affects the mitral valve. RHD is currently a major global health problem. However, the exact pathological mechanisms associated with RHD­induced cardiac valve damage remain to be elucidated. The endothelial­mesenchymal transition (EndMT) serves a key role in a number of diseases with an important role in cardiac fibrosis and the activin/Smad2 and 3 signaling pathway is involved in regulating the EndMT. Nevertheless, there are no studies to date, to the best of the authors' knowledge, investigating the association between RHD and EndMT. Thus, the aim of the current study was to investigate the potential role of EndMT in cardiac valve damage and assess whether activin/Smad2 and 3 signaling was activated during RHD­induced valvular injury in a rat model of RHD induced by inactivated Group A streptococci and complete Freund's adjuvant. Inflammation and fibrosis were assessed by hematoxylin and eosin and Sirius red staining. Serum cytokine and rheumatoid factor levels were measured using ELISA kits. Expression levels of activin/Smad2 and 3 signaling pathway­related factors [activin A, Smad2, Smad3, phosphorylated (p­)Smad2 and p­Smad3], EndMT­related factors [lymphoid enhancer factor­1 (LEF­1), Snail1, TWIST, zinc finger E­box­binding homeobox (ZEB)1, ZEB2, α smooth muscle actin (α­SMA) and type I collagen α 1 (COL1A1)], apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation markers (NF­κB and p­NF­κB) were detected using reverse transcription­quantitative PCR and western blot analyses. Compared with the control group, the degree of valvular inflammation and fibrosis, serum levels of IL­6, IL­17, TNF­α and expression of apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation marker (p­NF­κB), activin/Smad2 and 3 signaling pathway­related factors (activin A, p­Smad2 and p­Smad3), EndMT­related factors (LEF­1, Snail1, TWIST, ZEB 1, ZEB2, α­SMA and COL1A1) were significantly increased in the RHD group. These results suggested that the activin/Smad2 and 3 signaling pathway was activated during the development of valvular damage caused by RHD and that the EndMT is involved in RHD­induced cardiac valve damage.


Asunto(s)
Activinas/metabolismo , Válvula Mitral/patología , Cardiopatía Reumática/patología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Adyuvante de Freund/efectos adversos , Válvula Mitral/metabolismo , Ratas , Cardiopatía Reumática/etiología , Cardiopatía Reumática/metabolismo , Transducción de Señal , Streptococcus pyogenes/patogenicidad
3.
Clin Transl Gastroenterol ; 11(5): e00152, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32358238

RESUMEN

INTRODUCTION: Acute pancreatitis (AP) is a healthcare challenge with considerable mortality. Treatment is limited to supportive care, highlighting the need to investigate disease drivers and prognostic markers. Activin A is an established mediator of inflammatory responses, and its serum levels correlate with AP severity. We hypothesized that activin A is independent of body mass index (BMI) and is a targetable promoter of the AP inflammatory response. METHODS: We assessed whether BMI and serum activin A levels are independent markers to determine disease severity in a cohort of patients with AP. To evaluate activin A inhibition as a therapeutic, we used a cerulein-induced murine model of AP and treated mice with activin A-specific neutralizing antibody or immunoglobulin G control, both before and during the development of AP. We measured the production and release of activin A by pancreas and macrophage cell lines and observed the activation of macrophages after activin A treatment. RESULTS: BMI and activin A independently predicted severe AP in patients. Inhibiting activin A in AP mice reduced disease severity and local immune cell infiltration. Inflammatory stimulation led to activin A production and release by pancreas cells but not by macrophages. Macrophages were activated by activin A, suggesting activin A might promote inflammation in the pancreas in response to injury. DISCUSSION: Activin A provides a promising therapeutic target to interrupt the cycle of inflammation and tissue damage in AP progression. Moreover, assessing activin A and BMI in patients on hospital admission could provide important predictive measures for screening patients likely to develop severe disease.


Asunto(s)
Activinas/metabolismo , Antiinflamatorios/farmacología , Páncreas/patología , Pancreatitis/diagnóstico , Índice de Severidad de la Enfermedad , Activinas/antagonistas & inhibidores , Activinas/sangre , Activinas/inmunología , Animales , Antiinflamatorios/uso terapéutico , Biomarcadores/sangre , Biomarcadores/metabolismo , Índice de Masa Corporal , Línea Celular , Ceruletida/administración & dosificación , Ceruletida/toxicidad , Estudios de Cohortes , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Activación de Macrófagos/inmunología , Macrófagos , Ratones , Páncreas/efectos de los fármacos , Páncreas/inmunología , Pancreatitis/sangre , Pancreatitis/tratamiento farmacológico , Pancreatitis/inmunología , Admisión del Paciente , Valor Predictivo de las Pruebas
4.
Kidney Int ; 96(5): 1134-1149, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31492508

RESUMEN

Glomerular matrix protein accumulation, mediated largely by mesangial cells, is central to the pathogenesis of diabetic kidney disease. Our previous studies showed that the membrane microdomains caveolae and their marker protein caveolin-1 regulate matrix protein synthesis in mesangial cells in response to diabetogenic stimuli, and that caveolin-1 knockout mice are protected against diabetic kidney disease. In a screen to identify the molecular mechanism underlying this protection, we also established that secreted antifibrotic glycoprotein follistatin is significantly upregulated by caveolin-1 deletion. Follistatin potently neutralizes activins, members of the transforming growth factor-ß superfamily. A role for activins in diabetic kidney disease has not yet been established. Therefore, in vitro, we confirmed the regulation of follistatin by caveolin-1 in primary mesangial cells and showed that follistatin controls both basal and glucose-induced matrix production through activin inhibition. In vivo, we found activin A upregulation by immunohistochemistry in both mouse and human diabetic kidney disease. Importantly, administration of follistatin to type 1 diabetic Akita mice attenuated early diabetic kidney disease, characterized by albuminuria, hyperfiltration, basement membrane thickening, loss of endothelial glycocalyx and podocyte nephrin, and glomerular matrix accumulation. Thus, activin A is an important mediator of high glucose-induced profibrotic responses in mesangial cells, and follistatin may be a potential novel therapy for the prevention of diabetic kidney disease.


Asunto(s)
Activinas/metabolismo , Caveolina 1/metabolismo , Nefropatías Diabéticas/prevención & control , Folistatina/uso terapéutico , Animales , Nefropatías Diabéticas/metabolismo , Evaluación Preclínica de Medicamentos , Proteínas de la Matriz Extracelular/biosíntesis , Folistatina/metabolismo , Masculino , Células Mesangiales/metabolismo , Ratones Noqueados
5.
J Cell Physiol ; 233(9): 7143-7156, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29574773

RESUMEN

Uterine leiomyomas (fibroids or myomas) are the most common benign tumors of premenopausal women and new medical treatments are needed. This study aimed to determine the effects of omega-3 fatty acids on the lipid profile, membrane architecture and gene expression patterns of extracellular matrix components (collagen1A1, fibronectin, versican, or activin A), mechanical signaling (integrin ß1, FAK, and AKAP13), sterol regulatory molecules (ABCG1, ABCA1, CAV1, and SREBF2), and mitochondrial enzyme (CYP11A1) in myometrial and leiomyoma cells. Myometrial tissues had a higher amount of arachidonic acid than leiomyoma tissues while leiomyoma tissues had a higher level of linoleic acid than myometrial tissues. Treatment of primary myometrial and leiomyoma cells with eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA) reduced the monounsaturated fatty acid (MUFA) content and increased the polyunsaturated fatty acid (PUFA) content in both cell types. Myometrial and leiomyoma cell membranes were in the liquid-crystalline phase, but EPA- and DHA-treated cells had decreased membrane fluidity. While we found no changes in the mRNA expression of ECM components, EPA and DHA treatment reduced levels of ABCG1, ABCA1, and AKAP13 in both cell types. EPA and DHA also reduced FAK and CYP11A1 expression in myometrial cells. The ability of omega-3 fatty acids to remodel membrane architecture and downregulate the expression of genes involved in mechanical signaling and lipid accumulation in leiomyoma cells offers to further investigate this compound as preventive and/or therapeutic option.


Asunto(s)
Membrana Celular/metabolismo , Ácidos Grasos Omega-3/farmacología , Regulación Neoplásica de la Expresión Génica , Leiomioma/genética , Leiomioma/patología , Lípidos/química , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología , Activinas/genética , Activinas/metabolismo , Adulto , Membrana Celular/efectos de los fármacos , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Persona de Mediana Edad , Miometrio/efectos de los fármacos , Miometrio/metabolismo , Miometrio/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroles/metabolismo
6.
BMC Complement Altern Med ; 18(1): 32, 2018 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-29378560

RESUMEN

BACKGROUND: Sheng-ji Hua-yu(SJHY) formula is one of the most useful Traditional Chinese medicine (TCM) in the treatment of the delayed diabetic wound. However, elucidating the related molecular biological mechanism of how the SJHY Formula affects excessive inflammation in the process of re-epithelialization of diabetic wound healing is a task urgently needed to be fulfilled. The objectives of this study is to evaluate the effect of antagonisic expression of pro-/anti-inflammatory factors on transforming growth factor-ß(TGF-ß) superfamily (activin and follistatin) in the process of re-epithelialization of diabetic wound healing in vivo, and to characterize the involvement of the activin/follistatin protein expression regulation, phospho-Smad (pSmad2), and Nuclear factor kappa B p50 (NF-kB) p50 in the diabetic wound healing effects of SJHY formula. METHODS: SJHY Formula was prepared by pharmaceutical preparation room of Yueyang Hospital of Integrated Traditional Chinese and Western Medicine. Diabetic wound healing activity was evaluated by circular excision wound models. Wound healing activity was examined by macroscopic evaluation. Activin/follistatin expression regulation, protein expression of pSmad2 and NF-kB p50 in skin tissue of wounds were analyzed by Real Time PCR, Western blot, immunohistochemistry and hematoxylin and eosin (H&E) staining. RESULTS: Macroscopic evaluation analysis showed that wound healing of diabetic mice was delayed, and SJHY Formula accelerated wound healing time of diabetic mice. Real Time PCR analysis showed higher mRNA expression of activin/follistatin in diabetic delayed wound versus the wound in normal mice. Western Blot immunoassay analysis showed reduction of activin/follistatin proteins levels by SJHY Formula treatment 15 days after injury. Immunohistochemistry investigated the reduction of pSmad2 and NF-kB p50 nuclear staining in the epidermis of diabetic SJHY versus diabetic control mice on day 15 after wounding. H&E staining revealed that SJHY Formula accelerated re-epithelialization of diabetic wound healing. CONCLUSION: The present study found that diabetic delayed wound healing time is closely related to the high expression level of activin/follistatin, which leads to excessive inflammation in the process of re-epithelization. SJHY Formula accelerates re-epithelialization and healing time of diabetic wounds through decreasing the high expression of activin/follistatin.


Asunto(s)
Activinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Medicamentos Herbarios Chinos/farmacología , Folistatina/metabolismo , Repitelización/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/complicaciones , Medicamentos Herbarios Chinos/uso terapéutico , Femenino , Ratones , Ratones Endogámicos C57BL , Úlcera/tratamiento farmacológico
7.
Oncotarget ; 8(14): 23575-23587, 2017 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-28212568

RESUMEN

Uterine leiomyomas are highly prevalent benign tumors in reproductive aged women. Unfortunately, medical treatments are still limited and no preventive therapies have been developed. In the present study, we investigated the therapeutic effects of strawberry extract on uterine leiomyoma cells. Leiomyoma and myometrial cells were treated with strawberry (cultivar Alba) extract (250 µg/ml) for 48 h to measure apoptosis, reactive oxygen species (ROS), oxidative phosphorylation (OCR, oxygen consumption rate) and glycolysis (ECAR, extracellular acidification rate) as well as fibrosis associated gene and/or protein expression. In leiomyoma cells, strawberry increased the percentage of apoptotic and dead cells. Strawberry significantly increased ROS concentration in leiomyoma cells, while decreased it in myometrial cells. After strawberry treatment, leiomyoma cells showed a significant decreased rate of ECAR, while OCR was unchanged in both myometrial and leiomyoma cells. Strawberry significantly decreased collagen1A1, fibronectin and versican mRNA expression in leiomyoma cells. The reduced protein expression of fibronectin was observed by strawberry extract in leiomyoma cells as well. Furthermore, strawberry was able to reduce activin A induced fibronectin, collagen1A1, and versican as well as activin A and PAI-1 mRNA expression in leiomyoma cells. This study suggests that strawberry can be developed as therapeutic and/or preventive agent for uterine leiomyomas.


Asunto(s)
Antocianinas/farmacología , Apoptosis/efectos de los fármacos , Fragaria/química , Glucólisis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Activinas/genética , Activinas/metabolismo , Western Blotting , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Femenino , Fibrosis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Leiomioma/patología , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Extractos Vegetales/farmacología , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Versicanos/genética , Versicanos/metabolismo
8.
Sci Rep ; 6: 23790, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27030385

RESUMEN

Anti-Müllerian hormone (AMH) contributes to male sexual differentiation and acts on gonads of both sexes. Identification of AMH receptivity in both pituitary and brain has led to the intriguing idea that AMH participates to the hypothalamic-pituitary control of reproduction, however in vivo experimental evidence is still lacking. We show that AMH stimulates secretion and pituitary gene expression of the gonadotropin FSH in vivo in rats. AMH action is sex-dependent, being restricted to females and occurring before puberty. Accordingly, we report higher levels of pituitary AMH receptor transcripts in immature females. We show that AMH is functionally coupled to the Smad pathway in LßT2 gonadotrope cells and dose-dependently increases Fshb transcript levels. Furthermore, AMH was shown to establish complex interrelations with canonical FSH regulators as it cooperates with activin to induce Fshb expression whereas it reduces BMP2 action. We report that GnRH interferes with AMH by decreasing AMH receptivity in vivo in females. Moreover, AMH specifically regulates FSH and not LH, indicating that AMH is a factor contributing to the differential regulation of gonadotropins. Overall, our study uncovers a new role for AMH in regulating gonadotrope function and suggests that AMH participates in the postnatal elevation of FSH secretion in females.


Asunto(s)
Hormona Antimülleriana/genética , Hormona Folículo Estimulante/genética , Gonadotrofos/metabolismo , Adenohipófisis/metabolismo , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Caracteres Sexuales , Activinas/genética , Activinas/metabolismo , Animales , Animales Recién Nacidos , Hormona Antimülleriana/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Línea Celular , Femenino , Hormona Folículo Estimulante/metabolismo , Regulación del Desarrollo de la Expresión Génica , Gonadotrofos/citología , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Masculino , Adenohipófisis/citología , Adenohipófisis/crecimiento & desarrollo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Maduración Sexual , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo
9.
Clin Interv Aging ; 10: 1909-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26664104

RESUMEN

The scope of this review is to revise recent advances of the cell-based therapies of liver diseases with an emphasis on cell donor's and patient's age. Regenerative medicine with cell-based technologies as its integral part is focused on the structural and functional restoration of tissues impaired by sickness or aging. Unlike drug-based medicine directed primarily at alleviation of symptoms, regenerative medicine offers a more holistic approach to disease and senescence management aimed to achieve restoration of homeostasis. Hepatocyte transplantation and organ engineering are very probable forthcoming options of liver disease treatment in people of different ages and vigorous research and technological innovations in this area are in progress. Accordingly, availability of sufficient amounts of functional human hepatocytes is crucial. Direct isolation of autologous hepatocytes from liver biopsy is problematic due to related discomfort and difficulties with further expansion of cells, particularly those derived from aging people. Allogeneic primary human hepatocytes meeting quality standards are also in short supply. Alternatively, autologous hepatocytes can be produced by reprogramming of differentiated cells through the stage of induced pluripotent stem cells. In addition, fibroblasts and mesenchymal stromal cells can be directly induced to undergo advanced stage hepatogenic differentiation. Reprogramming of cells derived from elderly people is accompanied by the reversal of age-associated changes at the cellular level manifesting itself by telomere elongation and the U-turn of DNA methylation. Cell reprogramming can provide high quality rejuvenated hepatocytes for cell therapy and liver tissue engineering. Further technological advancements and establishment of national and global registries of induced pluripotent stem cell lines homozygous for HLA haplotypes can allow industry-style production of livers for immunosuppression-free transplantation.


Asunto(s)
Envejecimiento/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Hepatopatías/terapia , Regeneración Hepática/fisiología , Ingeniería de Tejidos/métodos , Activinas/metabolismo , Factores de Edad , Técnicas de Reprogramación Celular/métodos , Metilación de ADN , Fibroblastos/metabolismo , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ligandos de Señalización Nodal/metabolismo , Homeostasis del Telómero , Proteínas Wnt/metabolismo
10.
J Med Chem ; 58(14): 5637-48, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26098096

RESUMEN

Activin belongs to the TGFß superfamily, which is associated with several disease conditions, including cancer-related cachexia, preterm labor with delivery, and osteoporosis. Targeting activin and its related signaling pathways holds promise as a therapeutic approach to these diseases. A small-molecule ligand-binding groove was identified in the interface between the two activin ßA subunits and was used for a virtual high-throughput in silico screening of the ZINC database to identify hits. Thirty-nine compounds without significant toxicity were tested in two well-established activin assays: FSHß transcription and HepG2 cell apoptosis. This screening workflow resulted in two lead compounds: NUCC-474 and NUCC-555. These potential activin antagonists were then shown to inhibit activin A-mediated cell proliferation in ex vivo ovary cultures. In vivo testing showed that our most potent compound (NUCC-555) caused a dose-dependent decrease in FSH levels in ovariectomized mice. The Blitz competition binding assay confirmed target binding of NUCC-555 to the activin A:ActRII that disrupts the activin A:ActRII complex's binding with ALK4-ECD-Fc in a dose-dependent manner. The NUCC-555 also specifically binds to activin A compared with other TGFß superfamily member myostatin (GDF8). These data demonstrate a new in silico-based strategy for identifying small-molecule activin antagonists. Our approach is the first to identify a first-in-class small-molecule antagonist of activin binding to ALK4, which opens a completely new approach to inhibiting the activity of TGFß receptor superfamily members. in addition, the lead compound can serve as a starting point for lead optimization toward the goal of a compound that may be effective in activin-mediated diseases.


Asunto(s)
Activinas/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Interfaz Usuario-Computador , Activinas/química , Activinas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Femenino , Hormona Folículo Estimulante/antagonistas & inhibidores , Células Hep G2 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Ovario/citología , Ovario/efectos de los fármacos , Conformación Proteica , Transducción de Señal/efectos de los fármacos
11.
J Matern Fetal Neonatal Med ; 25 Suppl 4: 101-4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22958034

RESUMEN

OBJECTIVE: There is growing evidence on the usefulness of biomarkers in the early detection of preterm infants at risk for brain damage. However, among different tools Activin A, S100B protein and adrenomedullin assessment offer the possibility to investigate brain/multiorgan function and development. This could be especially useful in perinatal medicine that requires even more non-invasive techniques in order to fulfill the minimal handling in diagnostic and therapeutic strategy performance. MATERIALS AND METHODS: The concept of Unconventional Biological Fluid (UBF: urine and saliva) is becoming even stronger and regards the assessment in non-invasive biological fluids of biochemical markers involved in the cascade of events leading to brain damage. RESULTS: Activin A, S100B protein and adrenomedullin in UBF were increased in preterm newborns developing brain damage and/or ominous outcome. CONCLUSIONS: The present manuscript offers an update on the usefulness of Activin A, S100B protein an adrenomedullin in UBF as brain damage markers. The findings open a new cue on the use of these markers in daily neonatal intensive care unit (NICU) activities.


Asunto(s)
Biomarcadores/análisis , Lesiones Encefálicas/diagnóstico , Enfermedades del Prematuro/diagnóstico , Recien Nacido Prematuro , Activinas/análisis , Activinas/genética , Activinas/metabolismo , Adrenomedulina/análisis , Adrenomedulina/genética , Adrenomedulina/metabolismo , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/metabolismo , Biomarcadores/orina , Lesiones Encefálicas/líquido cefalorraquídeo , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/orina , Humanos , Recién Nacido , Recien Nacido Prematuro/líquido cefalorraquídeo , Recien Nacido Prematuro/metabolismo , Recien Nacido Prematuro/orina , Enfermedades del Prematuro/líquido cefalorraquídeo , Enfermedades del Prematuro/metabolismo , Enfermedades del Prematuro/orina , Factores de Crecimiento Nervioso/análisis , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/análisis , Proteínas S100/genética , Proteínas S100/metabolismo , Saliva/química , Saliva/metabolismo
12.
PLoS One ; 7(12): e52405, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300662

RESUMEN

Timed exposure of pluripotent stem cell cultures to exogenous molecules is widely used to drive differentiation towards desired cell lineages. However, screening differentiation conditions in conventional static cultures can become impractical in large parameter spaces, and is intrinsically limited by poor spatiotemporal control of the microenvironment that also makes it impossible to determine whether exogenous factors act directly or through paracrine-dependent mechanisms. We detail here the development of a continuous flow microbioreactor array platform that combines full-factorial multiplexing of input factors with progressive accumulation of paracrine factors through serially-connected culture chambers, and further, the use of this system to explore the combinatorial parameter space of both exogenous and paracrine factors involved in human embryonic stem cell (hESC) differentiation to a MIXL1-GFP(+) primitive streak-like population. We show that well known inducers of primitive streak (BMP, Activin and Wnt signals) do not simply act directly on hESC to induce MIXL1 expression, but that this requires accumulation of surplus, endogenous factors; and, that conditioned medium or FGF-2 supplementation is able to offset this. Our approach further reveals the presence of a paracrine, negative feedback loop to the MIXL1-GFP(+) population, which can be overcome with GSK-3ß inhibitors (BIO or CHIR99021), implicating secreted Wnt inhibitory signals such as DKKs and sFRPs as candidate effectors. Importantly, modulating paracrine effects identified in microbioreactor arrays by supplementing FGF-2 and CHIR in conventional static culture vessels resulted in improved differentiation outcomes. We therefore demonstrate that this microbioreactor array platform uniquely enables the identification and decoding of complex soluble factor signalling hierarchies, and that this not only challenges prevailing strategies for extrinsic control of hESC differentiation, but also is translatable to conventional culture systems.


Asunto(s)
Reactores Biológicos , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Análisis por Micromatrices/métodos , Comunicación Paracrina/efectos de los fármacos , Activinas/metabolismo , Activinas/farmacología , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Medios de Cultivo Condicionados/metabolismo , Evaluación Preclínica de Medicamentos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/metabolismo , Proteínas Wnt/farmacología
13.
J Neuroendocrinol ; 19(5): 390-6, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17425614

RESUMEN

GABA plays a pivotal role in reproduction by regulating luteinising hormone (LH) release from the anterior pituitary. Current evidence indicates that there is a prominent stimulatory effect of GABA on LH release in teleost fish which results from enhanced gonadotrophin-releasing hormone (GnRH) release and decreased dopamine turnover in the brain and pituitary. We hypothesised that there may be additional mechanisms underlying LH release in goldfish and investigated the relative mRNA levels of GABA synthesising enzymes (GAD65 and GAD67), degrading enzyme (GABA-T), activin betaa and betab, salmon GnRH (sGnRH), and tyrosine hydroxylase (TH) with the real-time reverse transcriptase-polymerase chain reaction after GABA agonist treatment. Sexually regressed female goldfish were i.p. injected with either the GABA(A) agonist muscimol (1 microg/g body weight) or the GABA(B) agonist baclofen (10 microg/g body weight). Both agonists significantly increased serum LH after 6 h. Muscimol decreased GAD65 (approximately ten-fold), GABA-T (approximately 15-fold) and TH (approximately three-fold) mRNA in the telencephalon. Baclofen significantly reduced GAD67 (approximately two-fold) and GABA-T (approximately two-fold) mRNA levels in the hypothalamus. Activin betaa, but not activin betab, steady-state mRNA was increased approximately three- to four-fold in both the hypothalamus and telencephalon after baclofen treatment. There was no change in sGnRH mRNA levels in either tissue after GABA agonist treatment. We show that the GABA(A) and GABA(B) receptor agonists have differing and rapid effects on gene transcription in the goldfish neuroendocrine brain and, by affecting specific targets, we identify putative genomic mechanisms underlying GABA-stimulated LH release in fish.


Asunto(s)
Agonistas del GABA/farmacología , Carpa Dorada/metabolismo , Hormona Luteinizante/metabolismo , Ácido gamma-Aminobutírico/fisiología , 4-Aminobutirato Transaminasa/genética , 4-Aminobutirato Transaminasa/metabolismo , Activinas/genética , Activinas/metabolismo , Animales , Baclofeno/farmacología , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/efectos de los fármacos , Muscimol/farmacología , ARN Mensajero/análisis , Receptores de GABA/efectos de los fármacos , Receptores de GABA/metabolismo , Telencéfalo/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
14.
Oncol Rep ; 16(1): 73-7, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16786125

RESUMEN

The mechanisms of estrogenic endocrine disruption on the male reproductive tract are poorly understood. In order to examine estrogenic properties of xenobiotic chemicals on male tissues, we have developed a mouse Leydig cell line (TM3-SF) that self-proliferates under serum-free conditions. This cell line was derived from ATCC's cell line, TM3. The development of TM3-SF was accomplished over a 4-month period by a progressive serum starvation of the original TM3 cells. The newly established cell line was maintained under serum-free conditions for 20 passages prior to testing. Sensitivity of the TM3-SF cells to estrogens was assayed by cell proliferation studies. A total of four compounds, diethylstilbestrol (DES), 17beta-estradiol, 17alpha-estradiol, and Bis-phenol A, were tested. Significant increases in cell proliferation occurred at various concentrations ranging from 1 pg/ml to 100 ng/ml for all four compounds. The order of potency observed was DES > Bis-phenol A > 17beta-estradiol and > 17alpha-estradiol. In addition, we investigated the mechanism for the self-proliferative properties of TM3-SF. The results of these trials indicate that either inhibin or activin is a primary growth factor for this cell line as a 50% inhibition of growth was noted when cell cultures were exposed to the anti-betaa subunit of inhibin/activin. Furthermore, the addition of the anti-betaa subunit of inhibin/activin blocked the DES-induced proliferation of TM3-SF. We conclude that the growth of TM3-SF cells is estrogen sensitive and that either inhibin or activin is involved in the self-regulation of growth.


Asunto(s)
Línea Celular , Células Intersticiales del Testículo/citología , Activinas/metabolismo , Animales , Proliferación Celular , Medio de Cultivo Libre de Suero/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Estrógenos/metabolismo , Inhibinas/metabolismo , Masculino , Ratones , Sensibilidad y Especificidad
15.
Med Oncol ; 23(4): 553-62, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17303915

RESUMEN

Excessive and inappropriate action of transforming growth factor (TGF)-beta has been implicated in the pathogenesis of several disease processes, especially cancer and fibrosis. To identify antagonists of the TGF- beta ligand-binding domain that may have therapeutic potential, we screened the National Cancer Institute open access chemical repository for molecules that inhibited binding of TGF-beta to the type II receptor (TbetaRII). About 30,000 molecules were screened resulting in the identification of five structurally related molecules that reduced binding of TGF-beta1 to soluble TbetaRII with an ED50 of approx 10 microM. The chemicals blocked inhibition of Mv1Lu cell growth by TGF-beta, TGF-beta - induced expression of luciferase driven by the TGF-beta response element, and induction of plasminogen inhibitor mRNA detected by Northern blot. In contrast, the chemicals did not block activin-induced inhibition of cell growth. Our results identify a novel chemical group that blocks binding of TGF-beta to its receptor and may result in novel treatment for disease.


Asunto(s)
Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Activinas/metabolismo , Animales , Northern Blotting , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Expresión Génica/efectos de los fármacos , Humanos , Unión Proteica/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/aislamiento & purificación , Factor de Crecimiento Transformador beta1/aislamiento & purificación
16.
Semin Reprod Med ; 22(3): 253-67, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15319828

RESUMEN

Inhibin, activin, and follistatin were first identified as gonadal hormones that could exert selective effects on follicle-stimulating hormone (FSH) secretion without affecting luteinizing hormone (LH). Although the primary source of inhibin remains the gonad, both activin and follistatin are produced in extragonadal tissues and can exert effects on FSH through an autocrine-paracrine mechanism. These proteins can effect the regulation of the gonadotropins at many levels. First, activin can directly stimulate FSH biosynthesis and release from the gonadotrope cells of the pituitary gland. Second, activin up-regulates gonadotropin-releasing hormone receptor (GnRHR) gene expression, leading to alterations in the synthesis and release of both gonadotropins in response to GnRH. Third, activin can stimulate GnRH release from GnRH neurons in the hypothalamus and thereby affect FSH and LH secretion. Both inhibin and follistatin can negatively regulate these effects by preventing activin binding to the activin receptor at the cell membrane and blocking activation of downstream signal transduction pathways. This review concentrates on the mechanisms through which inhibin, activin, and follistatin regulate the gonadotropins. We discuss the expression of inhibin/activin subunits and receptors throughout the hypothalamus and pituitary and their role in the regulation of FSH and LH. The mechanisms of inhibin and activin signaling are also reported, with particular attention to developments in our understanding of inhibin receptor action and activin-induced transcriptional regulation of the FSHbeta gene promoter. Finally, we present recent findings that other members of the transforming growth factor beta superfamily may also play an important role in transcriptional regulation of the pituitary gonadotropins.


Asunto(s)
Activinas/fisiología , Gonadotropinas/metabolismo , Inhibinas/fisiología , Activinas/metabolismo , Animales , Hormona Folículo Estimulante/metabolismo , Folistatina/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Inhibinas/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo
17.
Mol Cell Endocrinol ; 212(1-2): 19-27, 2003 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-14654247

RESUMEN

To examine in vivo, the local effects of inhibins and activins within the anterior pituitary, independent of their endocrine effects exerted from the gonad, in mediating FSH homeostasis, we used castrated knockout mice lacking either inhibin alpha or activin receptor II (ACVR2) alone or in combination. Compared to castrated wild-type (WT) mice, FSHbeta mRNA levels in the pituitaries of Acvr2 null mice were significantly downregulated in the absence of gonadal feedback. FSHbeta mRNA levels were not significantly higher in the pituitaries of castrated inhibin alpha null mice compared to those in Acvr2 null mice and remained the same in the pituitaries of castrated double mutant mice lacking both inhibin and ACVR2. In contrast to FSHbeta mRNA expression changes, pituitary FSH content was significantly reduced in Acvr2 null mice whereas it was only slightly upregulated in inhibin alpha null mice. Combined absence of both ACVR2 signaling and inhibins caused a decrease in FSH content compared to that in the absence of inhibins alone. These changes in pituitary content were in parallel to those in serum FSH levels in these three groups of castrated mice, suggesting that the unopposed actions of locally produced inhibins are dominant over those effects mediated by ACVR2 signaling to regulate FSH biosynthesis and secretion. Thus, our in vivo results demonstrate that within the pituitary, locally produced activins and inhibins exert their actions at distinct phases of FSH homeostasis. In an independent set of experiments, we tested whether in vivo signaling via ACVR2 is necessary for hypothalamic GnRH biosynthesis and for GnRH receptor expression. Our results demonstrate that in contrast to previous in vitro studies, signaling through ACVR2 is neither required for hypothalamic synthesis of GnRH peptide nor for expression of GnRH receptors in the anterior pituitary. We conclude that within the hypothalamic-pituitary short loop, ACVR2 signaling is critical only for FSH homeostasis and not for GnRH biosynthesis or induction of pituitary GnRH receptor expression. Our studies confirm the importance of using in vivo genetic models for studying regulation of the hypothalamic-pituitary-gonadal axis.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica , Receptores LHRH/metabolismo , Receptores de Activinas Tipo II/genética , Activinas/metabolismo , Animales , Castración , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Liberadora de Gonadotropina/metabolismo , Homeostasis , Hipotálamo/metabolismo , Inhibinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipófisis/citología , Hipófisis/metabolismo , Receptores LHRH/genética , Transducción de Señal/fisiología
18.
J Vet Med Sci ; 65(2): 237-42, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12655120

RESUMEN

The expression of activin and inhibin has been demonstrated in the hypothalamus, but their physiological roles in the brain remain to be elucidated. In the present study, involvement of activin and inhibin in the regulation of food and water intake was examined. Male rats were deprived of food or water for 12 and 60 hr, and mRNA levels of activin/inhibin alpha, betaA and betaB subunits in the hypothalamus were estimated by RT-PCR. Gene expression of alpha subunit transiently decreased at 12 hr of food deprivation, while it did not change during water deprivation. Food and water deprivation for 60 hr increased mRNA levels of betaA and betaB subunits, respectively. These results indicated that gene expression of each subunit was independently regulated. Injection of activin A (0.5 and 4.0 microg) into the third ventricle decreased food intake. Water intake was suppressed by 4.0 microg, but not 0.5 microg, of activin A. Intracerebroventricular injection of inhibin A (0.5 and 4.0 microg) decreased water intake in a dose dependent manner without affecting food intake, suggesting that inhibin could act independently of activin. Taken together, it is suggested that activin and inhibin take part in the central regulation of nutrient and fluid balance, though further study is needed to determine precise molecular species involved.


Asunto(s)
Activinas/metabolismo , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Inhibinas/metabolismo , Activinas/genética , Activinas/farmacología , Animales , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Privación de Alimentos , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Inhibinas/genética , Inhibinas/farmacología , Masculino , ARN Mensajero/análisis , Ratas , Factores de Tiempo , Privación de Agua
19.
Biol Reprod ; 66(5): 1395-402, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11967203

RESUMEN

The objective of the present study was to determine to what extent activin participates in setting the level of FSH secretion and if this regulation includes mediation via changes in GnRH secretion. We administered follistatin, the high-affinity binding protein for activin, to five ovariectomized sheep; we reasoned that the resultant binding of follistatin to activin should lower activin bioavailability and FSH secretion. Hypophyseal portal and peripheral blood samples were collected simultaneously at 10-min intervals for 18 h to measure GnRH, LH, FSH, and both activin-free and total follistatin. Six hours into collection, each ewe received 150 microg/kg i.v. of recombinant human follistatin-288. A week later, the same ewes were subjected to a second series of blood collections of similar length (time control). The FSH levels in pituitary portal blood were approximately 8-fold higher than those in the peripheral circulation. The FSH secretory patterns changed minimally during the time-control period. In contrast, follistatin had profound suppressive effects on FSH secretion. Maximal FSH suppression after FS-288 administration occurred at 5-6 h in the pituitary portal (65% suppression) and 9-10 h in the peripheral (48% suppression) circulation. Follistatin had no effect on GnRH or LH secretory patterns. Disappearance of total follistatin (i.e., free follistatin plus activin-bound follistatin) from the circulation was slower (P < 0.05) than that of free follistatin alone, suggesting that some of the follistatin was complexed with circulating activin, thus reducing the bioavailability of activin. The slower clearance of total follistatin and the lack of follistatin effects on GnRH secretion suggest that changes in activin bioavailability dictate the level of pituitary FSH secretion and that this is a pituitary-specific effect.


Asunto(s)
Activinas/farmacología , Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Sistemas Neurosecretores/fisiología , Activinas/metabolismo , Animales , Femenino , Folistatina , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Ovariectomía , Neurohipófisis/efectos de los fármacos , Neurohipófisis/metabolismo , Radioinmunoensayo , Ovinos
20.
Mol Cell Endocrinol ; 180(1-2): 163-7, 2001 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-11451587

RESUMEN

The findings that bone marrow fibroblastoid stromal cells are important for activin A production prompted our investigation of activin A expression in fibroblast-like synoviocytes in joint capsule in this and previous studies. In the proliferative reactive synovial membrane obtained from rheumatoid arthritis patients, activin A is detected prominently in the fibroblastoid synovial cells, as well as in the smooth muscle and the endothelial layer of the arteries in these vascularized proliferative tissues. The concentration of activin A in the rheumatoid arthritis synovial fluid was 33.6+/-5.0 ng/ml, much higher than the activin A content of osteoarthritis fluid (10.0+/-1.1 ng/ml). Furthermore, our previous studies also showed that inflammatory cytokines, such as interleukin (IL)-1, transforming growth factor (TGF)-beta, interferon (IFN)-gamma, IL-8, and IL-10 markedly enhance the expression of activin A mRNA in synoviocytes. These findings are consistent with our studies in regard to the regulatory control of activin A production in bone marrow stroma and monocytes. In addition, the relationship of activin A to IL-6-induced biological activities in various cell types was also investigated. Although activin A has not been directly associated with inflammatory processes, future studies are needed to investigate its production in response to the accumulated levels of inflammatory cytokines in the synovium of the patients, as well as the quantitative differences in activin A concentrations in many patients with other inflammatory diseases.


Asunto(s)
Activinas/metabolismo , Artritis/metabolismo , Subunidades beta de Inhibinas/metabolismo , Activinas/efectos de los fármacos , Activinas/farmacología , Animales , Artritis/etiología , Artritis/patología , Citocinas/efectos de los fármacos , Citocinas/farmacología , Humanos , Subunidades beta de Inhibinas/efectos de los fármacos , Subunidades beta de Inhibinas/farmacología , Líquido Sinovial/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA