Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.830
Filtrar
1.
J. physiol. biochem ; 79(2): 313-325, may. 2023.
Artículo en Inglés | IBECS | ID: ibc-222544

RESUMEN

Signaling by the transforming growth factor (TGF)-β superfamily is necessary for proper neural development and is involved in pain processing under both physiological and pathological conditions. Sensory neurons that reside in the dorsal root ganglia (DRGs) initially begin to perceive noxious signaling from their innervating peripheral target tissues and further convey pain signaling to the central nervous system. However, the transcriptional profile of the TGF-β superfamily members in DRGs during chronic inflammatory pain remains elusive. We developed a custom microarray to screen for transcriptional changes in members of the TGF-β superfamily in lumbar DRGs of rats with chronic inflammatory pain and found that the transcription of the TGF-β superfamily members tends to be downregulated. Among them, signaling of the activin/inhibin and bone morphogenetic protein/growth and differentiation factor (BMP/GDF) families dramatically decreased. In addition, peripherally pre-local administration of activins A and C worsened formalin-induced acute inflammatory pain, whereas activin C, but not activin A, improved formalin-induced persistent inflammatory pain by inhibiting the activation of astrocytes. This is the first report of the TGF-β superfamily transcriptional profiles in lumbar DRGs under chronic inflammatory pain conditions, in which transcriptional changes in cytokines or pathway components were found to contribute to, or be involved in, inflammatory pain processing. Our data will provide more targets for pain research. (AU)


Asunto(s)
Animales , Ratas , Ganglios Espinales , Factor de Crecimiento Transformador beta , Proteínas Morfogenéticas Óseas/fisiología , Grupos Diagnósticos Relacionados , Péptidos y Proteínas de Señalización Intercelular , Dolor
2.
J Physiol Biochem ; 79(2): 313-325, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36696051

RESUMEN

Signaling by the transforming growth factor (TGF)-ß superfamily is necessary for proper neural development and is involved in pain processing under both physiological and pathological conditions. Sensory neurons that reside in the dorsal root ganglia (DRGs) initially begin to perceive noxious signaling from their innervating peripheral target tissues and further convey pain signaling to the central nervous system. However, the transcriptional profile of the TGF-ß superfamily members in DRGs during chronic inflammatory pain remains elusive. We developed a custom microarray to screen for transcriptional changes in members of the TGF-ß superfamily in lumbar DRGs of rats with chronic inflammatory pain and found that the transcription of the TGF-ß superfamily members tends to be downregulated. Among them, signaling of the activin/inhibin and bone morphogenetic protein/growth and differentiation factor (BMP/GDF) families dramatically decreased. In addition, peripherally pre-local administration of activins A and C worsened formalin-induced acute inflammatory pain, whereas activin C, but not activin A, improved formalin-induced persistent inflammatory pain by inhibiting the activation of astrocytes. This is the first report of the TGF-ß superfamily transcriptional profiles in lumbar DRGs under chronic inflammatory pain conditions, in which transcriptional changes in cytokines or pathway components were found to contribute to, or be involved in, inflammatory pain processing. Our data will provide more targets for pain research.


Asunto(s)
Ganglios Espinales , Factor de Crecimiento Transformador beta , Ratas , Animales , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Péptidos y Proteínas de Señalización Intercelular , Dolor , Grupos Diagnósticos Relacionados
3.
Injury ; 53(12): 4165-4168, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36261312

RESUMEN

The repair of the tendon-bone interface, which is composed of tendon, fibrocartilage, and bony attachment, remains a clinical challenge. The application of mesenchymal stem cells (MSCs), collagen-rich extracellular matrix (ECMs), as well as growth factors, has the potential to regenerate this special multiple-tissue structure through the so-called biological augmentation. We present here an in vitro tendon regeneration model with C3H10T1/2 cells cultured on Collagen I matrix and evaluated the lineage determination effects of Growth Differentiation Factor 7 (GDF-7). We found that besides tenogenic effect, GDF-7 also stimulates the expression of osteoblastic as well as adipocytic genes. Our results indicate that GDF-7 might be a promising growth factor for regeneration of the tendon-bone interface due to its multiple-lineage stimulating effects. However, the side effect on adipogenic differentiation should be of concern, as it is a known risk factor for repair failures.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Células Madre Mesenquimatosas , Proteínas Morfogenéticas Óseas/fisiología , Colágeno/metabolismo , Animales , Ratones , Línea Celular , Tendones , Regeneración
4.
Reprod Biol Endocrinol ; 20(1): 34, 2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35183204

RESUMEN

BACKGROUND: Growth differentiation factor-11 (GDF-11) belongs to the transforming growth factor-ß (TGF-ß) superfamily. To date, the expression of GDF-11 in the ovary and its role in regulating ovarian function are completely unknown. Ovarian granulosa cell-mediated steroidogenesis plays a pivotal role in maintaining normal female reproductive function. GDF-11 and GDF-8 share high sequence similarity and exhibit many similar features and functions. Steroidogenic acute regulatory protein (StAR) regulates the rate-limiting step in steroidogenesis and its expression can be downregulated by GDF-8. Polycystic ovary syndrome (PCOS) is the most common cause of female infertility. The expression levels of GDF-8 are upregulated in the human follicular fluid and granulosa-lutein (hGL) cells of PCOS patients. However, whether similar results can be observed for the GDF-11 needs to be determined. METHODS: The effect of GDF-11 on StAR expression and the underlying molecular mechanisms were explored by a series of in vitro experiments in a primary culture of hGL cells obtained from patients undergoing in vitro fertilization (IVF) treatment. Human follicular fluid samples were obtained from 36 non-PCOS patients and 36 PCOS patients. GDF-11 levels in follicular fluid were measured by ELISA. RESULTS: GDF-11 downregulates StAR expression, whereas the expression levels of the P450 side-chain cleavage enzyme (P450scc) and 3ß-hydroxysteroid dehydrogenase (3ß-HSD) are not affected by GDF-11 in hGL cells. Using pharmacological inhibitors and a siRNA-mediated approach, we reveal that ALK5 but not ALK4 mediates the suppressive effect of GDF-11 on StAR expression. Although GDF-11 activates both SMAD2 and SMAD3 signaling pathways, only SMAD3 is involved in the GDF-11-induced downregulation of StAR expression. In addition, we show that SMAD1/5/8, ERK1/2, and PI3K/AKT signaling pathways are not activated by GDF-11 in hGL cells. RT-qPCR and ELISA detect GDF-11 mRNA expression in hGL cells and GDF-11 protein expression in human follicular fluid, respectively. Interestingly, unlike GDF-8, the expression levels of GDF-11 are not varied in hGL cells and follicular fluid between non-PCOS and PCOS patients. CONCLUSIONS: This study increases the understanding of the biological function of GDF-11 and provides important insights into the regulation of ovarian steroidogenesis.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Factores de Diferenciación de Crecimiento/fisiología , Células Lúteas/metabolismo , Fosfoproteínas/genética , Adulto , Células Cultivadas , Regulación hacia Abajo/genética , Femenino , Líquido Folicular/metabolismo , Células de la Granulosa/metabolismo , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Fosfoproteínas/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/fisiología , Proteína smad3/metabolismo
5.
Mol Brain ; 14(1): 134, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488822

RESUMEN

Growth differentiation factor 11 (GDF11) is a transforming factor-ß superfamily member that functions as a negative regulator of neurogenesis during embryonic development. However, when recombinant GDF11 (rGDF11) is administered systemically in aged mice, it promotes neurogenesis, the opposite of its role during development. The goal of the present study was to reconcile this apparent discrepancy by performing the first detailed investigation into the expression of endogenous GDF11 in the adult brain and its effects on neurogenesis. Using quantitative histological analysis, we observed that Gdf11 is most highly expressed in adult neurogenic niches and non-neurogenic regions within the hippocampus, choroid plexus, thalamus, habenula, and cerebellum. To investigate the role of endogenous GDF11 during adult hippocampal neurogenesis, we generated a tamoxifen inducible mouse that allowed us to reduce GDF11 levels. Depletion of Gdf11 during adulthood increased proliferation of neural progenitors and decreased the number of newborn neurons in the hippocampus, suggesting that endogenous GDF11 remains a negative regulator of hippocampal neurogenesis in adult mice. These findings further support the idea that circulating systemic GDF11 and endogenously expressed GDF11 in the adult brain have different target cells or mechanisms of action. Our data describe a role for GDF11-dependent signaling in adult neurogenesis that has implications for how GDF11 may be used to treat CNS disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Factores de Diferenciación de Crecimiento/fisiología , Hipocampo/citología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , Envejecimiento/metabolismo , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , División Celular , Cruzamientos Genéticos , Femenino , Factores de Diferenciación de Crecimiento/biosíntesis , Factores de Diferenciación de Crecimiento/deficiencia , Factores de Diferenciación de Crecimiento/genética , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Especificidad de Órganos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Nicho de Células Madre
6.
Metabolism ; 123: 154837, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34331962

RESUMEN

The current worldwide obesity pandemic highlights a need to better understand the regulation of energy balance and metabolism, including the role of the nervous system in controlling energy intake and energy expenditure. Neural plasticity in the hypothalamus of the adult brain has been implicated in full-body metabolic health, however, the mechanisms surrounding hypothalamic plasticity are incompletely understood. Bone morphogenetic proteins (BMPs) control metabolic health through actions in the brain as well as in peripheral tissues such as adipose, together regulating both energy intake and energy expenditure. BMP ligands, receptors, and inhibitors are found throughout plastic adult brain regions and have been demonstrated to modulate neurogenesis and gliogenesis, as well as synaptic and dendritic plasticity. This role for BMPs in adult neural plasticity is distinct from their roles in brain development. Existing evidence suggests that BMPs induce weight loss through hypothalamic pathways, and part of the mechanism of action may be through inducing neural plasticity. In this review, we summarize the data regarding how BMPs affect neural plasticity in the adult mammalian brain, as well as the relationship between central BMP signaling and metabolic health.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Metabolismo Energético/fisiología , Plasticidad Neuronal/fisiología , Adulto , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Humanos , Neurogénesis/fisiología , Transducción de Señal
7.
Dev Biol ; 477: 251-261, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34102166

RESUMEN

BMP signaling plays iterative roles during vertebrate neural crest development from induction through craniofacial morphogenesis. However, far less is known about the role of BMP activity in cranial neural crest epithelial-to-mesenchymal transition and delamination. By measuring canonical BMP signaling activity as a function of time from specification through early migration of avian midbrain neural crest cells, we found elevated BMP signaling during delamination stages. Moreover, inhibition of canonical BMP activity via a dominant negative mutant Type I BMP receptor showed that BMP signaling is required for neural crest migration from the midbrain, independent from an effect on EMT and delamination. Transcriptome profiling on control compared to BMP-inhibited cranial neural crest cells identified novel BMP targets during neural crest delamination and early migration including targets of the Notch pathway that are upregulated following BMP inhibition. These results suggest potential crosstalk between the BMP and Notch pathways in early migrating cranial neural crest and provide novel insight into mechanisms regulated by BMP signaling during early craniofacial development.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Mesencéfalo/embriología , Cresta Neural/metabolismo , Transducción de Señal , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Embrión de Pollo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Mesencéfalo/metabolismo , Cresta Neural/embriología , Cráneo/embriología , Cráneo/metabolismo , Técnicas de Cultivo de Tejidos
8.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946990

RESUMEN

Ubiquitination and deubiquitination are protein post-translational modification processes that have been recognized as crucial mediators of many complex cellular networks, including maintaining ubiquitin homeostasis, controlling protein stability, and regulating several signaling pathways. Therefore, some of the enzymes involved in ubiquitination and deubiquitination, particularly E3 ligases and deubiquitinases, have attracted attention for drug discovery. Here, we review recent findings on USP15, one of the deubiquitinases, which regulates diverse signaling pathways by deubiquitinating vital target proteins. Even though several basic previous studies have uncovered the versatile roles of USP15 in different signaling networks, those have not yet been systematically and specifically reviewed, which can provide important information about possible disease markers and clinical applications. This review will provide a comprehensive overview of our current understanding of the regulatory mechanisms of USP15 on different signaling pathways for which dynamic reverse ubiquitination is a key regulator.


Asunto(s)
Procesamiento Proteico-Postraduccional/fisiología , Transducción de Señal/fisiología , Proteasas Ubiquitina-Específicas/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Complejo del Señalosoma COP9/fisiología , Humanos , Inmunidad Innata , Masculino , Ratones , FN-kappa B/metabolismo , Neoplasias de la Próstata/metabolismo , Dominios Proteicos , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Precursores del ARN/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteasas Ubiquitina-Específicas/química , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación , Vía de Señalización Wnt/fisiología , Proteínas de Xenopus/metabolismo
9.
Theranostics ; 11(13): 6573-6591, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995677

RESUMEN

Mesenchymal stem cells (MSCs) have been identified in many adult tissues. MSCs can regenerate through cell division or differentiate into adipocytes, osteoblasts and chondrocytes. As a result, MSCs have become an important source of cells in tissue engineering and regenerative medicine for bone tissue and cartilage. Several epigenetic factors are believed to play a role in MSCs differentiation. Among these, microRNA (miRNA) regulation is involved in the fine modulation of gene expression during osteogenic/chondrogenic differentiation. It has been reported that miRNAs are involved in bone homeostasis by modulating osteoblast gene expression. In addition, countless evidence has demonstrated that miRNAs dysregulation is involved in the development of osteoporosis and bone fractures. The deregulation of miRNAs expression has also been associated with several malignancies including bone cancer. In this context, bone-associated circulating miRNAs may be useful biomarkers for determining the predisposition, onset and development of osteoporosis, as well as in clinical applications to improve the diagnosis, follow-up and treatment of cancer and metastases. Overall, this review will provide an overview of how miRNAs activities participate in osteogenic/chondrogenic differentiation, while addressing the role of miRNA regulatory effects on target genes. Finally, the role of miRNAs in pathologies and therapies will be presented.


Asunto(s)
Enfermedades Óseas/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Osteogénesis/genética , Proteínas Morfogenéticas Óseas/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Sistemas de Liberación de Medicamentos , Fracturas Óseas/metabolismo , Histona Desacetilasas/fisiología , Humanos , Metaloproteinasa 13 de la Matriz/fisiología , Proteínas Represoras/fisiología , Transducción de Señal , Proteínas Smad/fisiología , Factor de Transcripción Sp7/fisiología , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
10.
Curr Treat Options Oncol ; 22(6): 48, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33866453

RESUMEN

OPINION STATEMENT: Malignant melanoma is a deadly form of skin cancer caused by neoplastic transformation of melanocytic cells. Despite recent progress in melanoma therapy, by inhibition of activated oncogenes or immunotherapy, survival rate for metastatic melanoma patients remains low. The remarkable phenotypic plasticity of melanoma cells allows for rapid development of invasive properties and metastatic tumors, the main cause of mortality in melanoma patients. Phenotypic and molecular analyses of developing tumors revealed that epithelial-mesenchymal transition (EMT), a cellular and molecular mechanism, controls transition from mature melanocyte to less differentiated melanocyte lineage progenitor cells forming melanoma tumors. This transition is facilitated by persistence of transcriptional regulatory circuit characteristic of embryonic stage in mature melanocytes. Switching of the developmental program of mature melanocyte to EMT is induced by accumulated mutations, especially targeting BRAF, N-RAS, or MEK1/2 signaling pathways, and further promoted by dynamic stimuli from local environment including hypoxia, interactions with extracellular matrix and growth factors or cytokines. Recent reports demonstrate that signaling mediated by transforming growth factor-ß (TGF-ß) and bone morphogenic proteins (BMPs) play critical roles in inducing EMT by controlling expression of critical transcription factors. BMPs are essential modulators of differentiation, proliferation, apoptosis, invasiveness, and metastases in developing melanoma tumors. They control transcription and epigenetic landscape of melanoma cells. Better understanding of the role of BMPs may lead to new strategies to control EMT processes in melanocyte cell lineage and to achieve clinical benefits for the patients.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Melanoma/etiología , Neoplasias Cutáneas/etiología , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal , Humanos , Melanoma/patología , Transducción de Señal/fisiología , Neoplasias Cutáneas/patología
11.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925390

RESUMEN

Decussation of axonal tracts is an important hallmark of vertebrate neuroanatomy resulting in one brain hemisphere controlling the contralateral side of the body and also computing the sensory information originating from that respective side. Here, we show that BMP interferes with optic chiasm formation and RGC pathfinding in zebrafish. Experimental induction of BMP4 at 15 hpf results in a complete ipsilateral projection of RGC axons and failure of commissural connections of the forebrain, in part as the result of an interaction with shh signaling, transcriptional regulation of midline guidance cues and an affected optic stalk morphogenesis. Experimental induction of BMP4 at 24 hpf, resulting in only a mild repression of forebrain shh ligand expression but in a broad expression of pax2a in the diencephalon, does not per se prevent RGC axons from crossing the midline. It nevertheless shows severe pathologies of RGC projections e.g., the fasciculation of RGC axons with the ipsilateral optic tract resulting in the innervation of one tectum by two eyes or the projection of RGC axons in the direction of the contralateral eye.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Quiasma Óptico/embriología , Células Ganglionares de la Retina/metabolismo , Animales , Axones/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Quiasma Óptico/metabolismo , Quiasma Óptico/fisiología , Nervio Óptico/fisiología , Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Vías Visuales/fisiología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
12.
Commun Biol ; 4(1): 90, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469151

RESUMEN

Leucine-rich repeats and immunoglobulin-like domains (LRIG) proteins have been implicated as regulators of growth factor signaling; however, the possible redundancy among mammalian LRIG1, LRIG2, and LRIG3 has hindered detailed elucidation of their physiological functions. Here, we show that Lrig-null mouse embryonic fibroblasts (MEFs) are deficient in adipogenesis and bone morphogenetic protein (BMP) signaling. In contrast, transforming growth factor-beta (TGF-ß) and receptor tyrosine kinase (RTK) signaling appeared unaltered in Lrig-null cells. The BMP signaling defect was rescued by ectopic expression of LRIG1 or LRIG3 but not by expression of LRIG2. Caenorhabditis elegans with mutant LRIG/sma-10 variants also exhibited a lipid storage defect. Human LRIG1 variants were strongly associated with increased body mass index (BMI) yet protected against type 2 diabetes; these effects were likely mediated by altered adipocyte morphology. These results demonstrate that LRIG proteins function as evolutionarily conserved regulators of lipid metabolism and BMP signaling and have implications for human disease.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Adipogénesis/fisiología , Adulto , Anciano , Animales , Índice de Masa Corporal , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Caenorhabditis elegans , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Ratones , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Transducción de Señal/fisiología
13.
Surg Today ; 51(3): 340-349, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32754843

RESUMEN

Organ liver transplantation and hepatocyte transplantation are not performed to their full potential because of donor shortage, which could be resolved by identifying new donor sources for the development of hepatocyte-like cells (HLCs). HLCs have been differentiated from some stem cell sources as alternative primary hepatocytes throughout the world; however, the currently available techniques cannot differentiate HLCs to the level of normal adult primary hepatocytes. The outstanding questions are as follows: which stem cells are the best cell sources? which protocol is the best way to differentiate them into HLCs? what is the definition of differentiated HLCs? how can we enforce the function of HLCs? what is the difference between HLCs and primary hepatocytes? what are the problems with HLC transplantation? This review summarizes the current status of HLCs, focusing on stem cell sources, the differentiation protocol for HLCs, the general characterization of HLCs, the generation of more functional HLCs, comparison with primary hepatocytes, and HLCs in cell-transplantation-based liver regeneration.


Asunto(s)
Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Técnicas Citológicas/métodos , Hepatocitos/trasplante , Hepatopatías/terapia , Células Madre/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Factores de Crecimiento de Fibroblastos/fisiología , Factor Nuclear 4 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Regeneración Hepática/fisiología , Factores de Transcripción SOXF/fisiología , Factores de Transcripción/fisiología
14.
Genes Dev ; 34(23-24): 1713-1734, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184221

RESUMEN

Through recurrent bouts synchronous with the hair cycle, quiescent melanocyte stem cells (McSCs) become activated to generate proliferative progeny that differentiate into pigment-producing melanocytes. The signaling factors orchestrating these events remain incompletely understood. Here, we use single-cell RNA sequencing with comparative gene expression analysis to elucidate the transcriptional dynamics of McSCs through quiescence, activation, and melanocyte maturation. Unearthing converging signs of increased WNT and BMP signaling along this progression, we endeavored to understand how these pathways are integrated. Employing conditional lineage-specific genetic ablation studies in mice, we found that loss of BMP signaling in the lineage leads to hair graying due to a block in melanocyte maturation. We show that interestingly, BMP signaling functions downstream from activated McSCs and maintains WNT effector, transcription factor LEF1. Employing pseudotime analysis, genetics, and chromatin landscaping, we show that following WNT-mediated activation of McSCs, BMP and WNT pathways collaborate to trigger the commitment of proliferative progeny by fueling LEF1- and MITF-dependent differentiation. Our findings shed light upon the signaling interplay and timing of cues that orchestrate melanocyte lineage progression in the hair follicle and underscore a key role for BMP signaling in driving complete differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Melanocitos/citología , Transducción de Señal/genética , Células Madre/citología , Animales , Linaje de la Célula/genética , Perfilación de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Análisis de la Célula Individual
15.
Biomed Pharmacother ; 131: 110725, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32927254

RESUMEN

INTRODUCTION: Pulmonary emphysema is characterized by destruction of alveoli leading to inadequate oxygenation, disability and frequently death. This destruction was understood so far as irreversible. Published data has shown that ATRA (All Trans Retinoic Acid) reverses elastase-induced emphysema in rats. However, the molecular mechanisms governing regeneration process are so far unknown. OBJECTIVE: To examine the therapeutic potential of ATRA on various molecular pathways and their coordination towards governance of alveolar epithelial regeneration in emphysematous rats. METHODS: Emphysema was induced by elastase versus saline in Sprague-Dawley rats. On days 26-37, rats received daily intraperitoneal injections with ATRA (500 µg/kg b.w.) versus olive-oil. Lungs were removed at day 38 for histopathology and investigation of relative mRNA and protein expressions. RESULTS: Histopathological analysis has shown that losses of alveoli were recovered in therapy (EA) group. Moreover, expressions of markers genes for alveolar cell proliferation, differentiation and EMT events at mRNA and protein levels were significantly increased in EA group than emphysema group (ES). Upon validation at genomics level, expressions of components of Notch, Hedgehog, Wnt, BMP and TGFß pathways were significantly attenuated in EA group when compared with ES and were well comparable with the healthy group. CONCLUSION: Therapeutic supplementation of ATRA rectifies the deregulated Notch, Hedgehog, Wnt, BMP and TGFß pathways in emphysema condition, resulting in alveolar epithelium regeneration. Hence, ATRA may prove to be a potential drug in the treatment of emphysema. Nevertheless, elaborated studies are to be conducted.


Asunto(s)
Alveolos Pulmonares/efectos de los fármacos , Enfisema Pulmonar/tratamiento farmacológico , Regeneración/efectos de los fármacos , Tretinoina/uso terapéutico , Animales , Acuaporina 4/genética , Peso Corporal/efectos de los fármacos , Proteínas Morfogenéticas Óseas/fisiología , Epitelio/fisiología , Masculino , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiología , Enfisema Pulmonar/patología , Enfisema Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Tretinoina/farmacología , Vimentina/genética
16.
Int J Mol Sci ; 21(18)2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32899497

RESUMEN

Activins transduce the TGF-ß pathway through a heteromeric signaling complex consisting of type I and type II receptors, and activins also inhibit bone morphogenetic protein (BMP) signaling mediated by type I receptor ALK2. Recent studies indicated that activin A cross-activates the BMP pathway through ALK2R206H, a mutation associated with Fibrodysplasia Ossificans Progressiva (FOP). How activin A inhibits ALK2WT-mediated BMP signaling but activates ALK2R206H-mediated BMP signaling is not well understood, and here we offer some insights into its molecular mechanism. We first demonstrated that among four BMP type I receptors, ALK2 is the only subtype able to mediate the activin A-induced BMP signaling upon the dissociation of FKBP12. We further showed that BMP4 does not cross-signal TGF-ß pathway upon FKBP12 inhibition. In addition, although the roles of type II receptors in the ligand-independent BMP signaling activated by FOP-associated mutant ALK2 have been reported, their roles in activin A-induced BMP signaling remains unclear. We demonstrated in this study that the known type II BMP receptors contribute to activin A-induced BMP signaling through their kinase activity. Together, the current study provided important mechanistic insights at the molecular level into further understanding physiological and pathophysiological BMP signaling.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/fisiología , Línea Celular , Regulación de la Expresión Génica/genética , Células HEK293 , Células Hep G2 , Humanos , Ratones , Osificación Heterotópica/genética , Fosforilación , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
17.
Biochem Biophys Res Commun ; 529(1): 7-14, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32560821

RESUMEN

This study aimed to investigate the role of truncated growth differentiation factor 11 (GDF11), in which the recognition site of Furin from wild-type GDF11 was deleted to enhance the cellular stability, in skin wound healing in the setting of diabetes mellitus (DM) and the underlying mechanisms. Our study found that both truncated and natural GDF11s effectively accelerated wound healing processes in both T1DM and T2DM mice with a potency compatible to PDGF, bFGF, and EGF, but being much higher than GDF8. At the cellular level, GDF11 stimulated the proliferation and suppressed HG-induced apoptosis of HSFs. Further study revealed that GDF11 activated the YAP-Smad2/3-CTGF fibrotic signaling pathway by reversing HG-induced upregulation of phosphorylated form of YAP (p-YAP), increases p-Smad2/3 levels, and restoring HG-induced repression of CTGF expression by GDF11. Overall, the study shows that both natural and truncated GDF11s promote the healing process of skin wound in mice of both T1DM and T2DM partly via stimulating dermal fibrosis via the YAP-Smad2/3-CTGF pathway, suggesting it a potential agent for treating skin wound in diabetic population.


Asunto(s)
Proteínas Morfogenéticas Óseas/administración & dosificación , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Factores de Diferenciación de Crecimiento/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Administración Tópica , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Factores de Diferenciación de Crecimiento/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Piel/efectos de los fármacos , Piel/lesiones , Piel/patología , Cicatrización de Heridas/fisiología
18.
Behav Brain Res ; 392: 112711, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32461130

RESUMEN

Bone morphogenetic protein (BMP) signaling in the hippocampus regulates psychiatric behaviors and hippocampal neurogenesis in non-stress conditions; however, stress-induced changes in hippocampal BMP signaling have not yet been reported. Therefore, we sought to examine whether psychosocial stress, which induces psychiatric symptoms, affects hippocampal BMP signaling. A total of 32 male Sprague-Dawley rats were exposed to a psychosocial stress using a Resident/Intruder paradigm for ten consecutive days. Subsequently, rats were subjected to a battery of behavioral tests (novelty-suppressed feeding test, sucrose preference test, and forced swimming test) for the evaluation of adult neurogenesis and activity of BMP signaling in the dorsal and ventral hippocampus. Repeated social defeat promoted anxiety-like behaviors, but neither anhedonia nor behavioral despair. Socially defeated rats exhibited an increase in the number of Ki-67-positive cells, decrease in the number of doublecortin (DCX)-positive cells, and decrease only in the dorsal hippocampus of the ratio of DCX-positive to Ki-67-positive cells, a proxy for newly-born cell maturation speed and survival. In contrast, no differences were observed in the number of 5-Bromo-2'-deoxyuridine (BrdU)-positive cells, indicating survival of newly-born cells both in the dorsal and ventral hippocampus. Furthermore, psychosocial stress significantly increased the BMP-4 and phosphorylated Smad1/5/9 expression levels specifically in the dorsal hippocampus. Our findings suggest that repeated psychosocial stress activates BMP signaling and differently affects cell proliferation and neurogenesis exclusively in the dorsal hippocampus, potentially exacerbating anxiety-related symptoms. Targeting BMP signaling is a potential therapeutic strategy for psychiatric disorders.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Estrés Psicológico/metabolismo , Anhedonia/efectos de los fármacos , Anhedonia/fisiología , Animales , Ansiedad/tratamiento farmacológico , Proteína Morfogenética Ósea 4/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Depresión/tratamiento farmacológico , Proteína Doblecortina , Hipocampo/metabolismo , Masculino , Neurogénesis/fisiología , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Invest Ophthalmol Vis Sci ; 61(5): 62, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32462202

RESUMEN

Purpose: Fibrosis or scarring is a pathological outcome of wound healing and is characterized by terminally differentiated myofibroblasts. Heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3) is a unique matrix component purified from amniotic membrane that exerts an anti-inflammatory effect. Herein, we investigate whether HC-HA/PTX3 can also exert an antiscarring effect. Methods: Human corneal fibroblasts and myofibroblasts were seeded on plastic, immobilized HA or HC-HA/PTX3 or on plastic with or without soluble HA and HC-HA/PTX3 in DMEM+10% FBS, with or without AMD3100 or SB431542 in DMEM+ITS with or without transforming growth factor-ß1 (TGF-ß1). Transcript expression of keratocyte and signaling markers was determined by RT-qPCR. Immunostaining was performed to monitor cytolocalization of signaling markers and α-SMA. Western blotting was used to measure relative protein level. Results: Human corneal fibroblasts and myofibroblasts cultured in or on HC-HA/PTX3, but not HA, were refrained from cytoplasmic expression of αSMA and nuclear translocation of pSMAD2/3 when challenged with exogenous TGF-ß1. Such an antiscarring action by suppressing canonical TGF-ß1 signaling was surprisingly accompanied by phenotypic reversal to keratocan-expressing keratocytes through activation of BMP signaling. Further investigation disclosed that such phenotypic reversal was initiated by cell aggregation mediated by SDF1-CXCR4 signaling highlighted by nuclear translocation of CXCR4 and upregulation of CXCR4 transcript and protein followed by activation of canonical BMP signaling. Conclusions: These findings collectively provide mechanistic understanding explaining how amniotic membrane transplantation exerts an antiscarring action. In addition, HC-HA/PTX3 and derivatives may be developed into a new biologic to treat corneal blindness caused by stromal scar or opacity in the future.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Proteína C-Reactiva/aislamiento & purificación , Proteína C-Reactiva/fisiología , Diferenciación Celular , Córnea/citología , Queratocitos de la Córnea/citología , Fibroblastos/citología , Ácido Hialurónico/fisiología , Miofibroblastos/citología , Componente Amiloide P Sérico/aislamiento & purificación , Componente Amiloide P Sérico/fisiología , Amnios/química , Humanos , Transducción de Señal
20.
Bone ; 137: 115416, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32422297

RESUMEN

Bone Morphogenetic Proteins (BMPs) are extracellular multifunctional signalling cytokines and members of the TGFß super family. These pleiotropic growth factors crucially promote bone formation, remodeling and healing after injury. Additionally, bone homeostasis is systematically regulated by mechanical inputs from the environment, which are incorporated into the bone cells' biochemical response. These inputs range from compression and tension induced by the movement of neighboring muscle, to fluid shear stress induced by interstitial fluid flow in the canaliculi and in the vascular system. Although BMPs are widely applied in a clinic context to promote fracture healing, it is still elusive how mechanical inputs modulate this signalling pathway, hindering an efficient and side-effect free application of these ligands in bone healing. This review aims to summarize the current understanding in how mechanical cues (tension, compression, shear force and hydrostatic pressure) and substrate stiffness modulate BMP signalling. We highlight the time-dependent effects in modulating immediate early up to long-term effects of mechano-BMP crosstalk during bone formation and remodeling, considering the interplay with other already established mechanosensitive pathways, such as MRTF/SRF and Hippo signalling.


Asunto(s)
Proteínas Morfogenéticas Óseas , Huesos/fisiología , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...