Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Biochem J ; 481(7): 547-564, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38533769

RESUMEN

Activins are one of the three distinct subclasses within the greater Transforming growth factor ß (TGFß) superfamily. First discovered for their critical roles in reproductive biology, activins have since been shown to alter cellular differentiation and proliferation. At present, members of the activin subclass include activin A (ActA), ActB, ActC, ActE, and the more distant members myostatin and GDF11. While the biological roles and signaling mechanisms of most activins class members have been well-studied, the signaling potential of ActE has remained largely unknown. Here, we characterized the signaling capacity of homodimeric ActE. Molecular modeling of the ligand:receptor complexes showed that ActC and ActE shared high similarity in both the type I and type II receptor binding epitopes. ActE signaled specifically through ALK7, utilized the canonical activin type II receptors, ActRIIA and ActRIIB, and was resistant to the extracellular antagonists follistatin and WFIKKN. In mature murine adipocytes, ActE invoked a SMAD2/3 response via ALK7, like ActC. Collectively, our results establish ActE as a specific signaling ligand which activates the type I receptor, ALK7.


Asunto(s)
Proteínas Portadoras , Factor de Crecimiento Transformador beta , Ratones , Animales , Factor de Crecimiento Transformador beta/metabolismo , Ligandos , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Activinas/metabolismo
2.
J Biol Chem ; 298(7): 102076, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35643319

RESUMEN

BMPR2 is a type II Transforming Growth Factor (TGF)-ß family receptor that is fundamentally associated with pulmonary arterial hypertension (PAH) in humans. BMPR2 shares functional similarities with the type II activin receptors ACVR2A and ACVR2B, as it interacts with an overlapping group of TGF-ß family growth factors (GFs). However, how BMPR2 recognizes GFs remains poorly understood. Here, we solved crystal structures of BMPR2 in complex with the GF activin B and of ACVR2A in complex with the related GF activin A. We show that both BMPR2 and ACVR2A bind GFs with nearly identical geometry using a conserved hydrophobic hot spot, while differences in contacting residues are predominantly found in loop areas. Upon further exploration of the GF-binding spectrum of the two receptors, we found that although many GFs bind both receptors, the high-affinity BMPR2 GFs comprise BMP15, BMP10, and Nodal, whereas those of ACVR2A are activin A, activin B, and GDF11. Lastly, we evaluated GF-binding domain BMPR2 variants found in human PAH patients. We demonstrate that mutations within the GF-binding interface resulted in loss of GF binding, while mutations in loop areas allowed BMPR2 to retain the ability to bind cognate GFs with high affinity. In conclusion, the in vitro activities of BMPR2 variants and the crystal structures reported here indicate biochemically relevant complexes that explain how some GF-binding domain variants can lead to PAH.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Receptores de Activinas/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/genética , Factores de Diferenciación de Crecimiento , Humanos , Mutación , Factor de Crecimiento Transformador beta/genética
3.
Development ; 147(12)2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32439760

RESUMEN

Physical forces are important participants in the cellular dynamics that shape developing organs. During heart formation, for example, contractility and blood flow generate biomechanical cues that influence patterns of cell behavior. Here, we address the interplay between function and form during the assembly of the cardiac outflow tract (OFT), a crucial connection between the heart and vasculature that develops while circulation is under way. In zebrafish, we find that the OFT expands via accrual of both endocardial and myocardial cells. However, when cardiac function is disrupted, OFT endocardial growth ceases, accompanied by reduced proliferation and reduced addition of cells from adjacent vessels. The flow-responsive TGFß receptor Acvrl1 is required for addition of endocardial cells, but not for their proliferation, indicating distinct modes of function-dependent regulation for each of these essential cell behaviors. Together, our results indicate that cardiac function modulates OFT morphogenesis by triggering endocardial cell accumulation that induces OFT lumen expansion and shapes OFT dimensions. Moreover, these morphogenetic mechanisms provide new perspectives regarding the potential causes of cardiac birth defects.


Asunto(s)
Endocardio/metabolismo , Corazón/fisiología , Pez Cebra/metabolismo , Receptores de Activinas/antagonistas & inhibidores , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Proliferación Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endocardio/citología , Corazón/anatomía & histología , Corazón/crecimiento & desarrollo , Morfolinos/metabolismo , Troponina T/antagonistas & inhibidores , Troponina T/genética , Troponina T/metabolismo , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Angiogenesis ; 23(2): 203-220, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31828546

RESUMEN

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal-dominant vascular disorder characterized by development of high-flow arteriovenous malformations (AVMs) that can lead to stroke or high-output heart failure. HHT2 is caused by heterozygous mutations in ACVRL1, which encodes an endothelial cell bone morphogenetic protein (BMP) receptor, ALK1. BMP9 and BMP10 are established ALK1 ligands. However, the unique and overlapping roles of these ligands remain poorly understood. To define the physiologically relevant ALK1 ligand(s) required for vascular development and maintenance, we generated zebrafish harboring mutations in bmp9 and duplicate BMP10 paralogs, bmp10 and bmp10-like. bmp9 mutants survive to adulthood with no overt phenotype. In contrast, combined loss of bmp10 and bmp10-like results in embryonic lethal cranial AVMs indistinguishable from acvrl1 mutants. However, despite embryonic functional redundancy of bmp10 and bmp10-like, bmp10 encodes the only required Alk1 ligand in the juvenile-to-adult period. bmp10 mutants exhibit blood vessel abnormalities in anterior skin and liver, heart dysmorphology, and premature death, and vascular defects correlate with increased cardiac output. Together, our findings support a unique role for Bmp10 as a non-redundant Alk1 ligand required to maintain the post-embryonic vasculature and establish zebrafish bmp10 mutants as a model for AVM-associated high-output heart failure, which is an increasingly recognized complication of severe liver involvement in HHT2.


Asunto(s)
Receptores de Activinas/metabolismo , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Neovascularización Fisiológica/genética , Regeneración/genética , Proteínas de Pez Cebra/metabolismo , Receptores de Activinas/genética , Animales , Animales Modificados Genéticamente , Malformaciones Arteriovenosas/genética , Malformaciones Arteriovenosas/metabolismo , Malformaciones Arteriovenosas/patología , Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular/genética , Embrión no Mamífero , Células Endoteliales/fisiología , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal/genética , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
5.
Apoptosis ; 24(11-12): 972-989, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31641960

RESUMEN

Developmentally regulated programmed cell death (PCD) is one of the key cellular events for precise controlling of neuronal population during postembryonic development of the central nervous system. Previously we have shown that a group of corazonin-producing peptidergic neurons (vCrz) undergo apoptosis in response to ecdysone signaling via ecdysone receptor (EcR)-B isoforms and Ultraspiracle during early phase of metamorphosis. Further utilizing genetic, transgenic, and mosaic analyses, we have found that TGF-ß signaling mediated by a glia-produced ligand, Myoglianin, type-I receptor Baboon (particularly Babo-A isoform) and dSmad2, is also required autonomously for PCD of the vCrz neurons. Our studies show that TGF-ß signaling is not acting epistatically to EcR or vice versa. We also show that ectopic expression of a constitutively active phosphomimetic form of dSmad2 (dSmad2PM) is capable of inducing premature death of vCrz neurons in larva but not other larval neurons. Intriguingly, the dSmad2PM-mediated killing is completely suppressed by coexpression of a dominant-negative form of EcR (EcRDN), suggesting that EcR function is required for the proapoptotic dSmad2PM function. Based on these data, we suggest that TGF-ß and ecdysone signaling pathways act cooperatively to induce vCrz neuronal PCD. We propose that this type of two-factor authentication is a key developmental strategy to ensure the timely PCD of specific larval neurons during metamorphosis.


Asunto(s)
Receptores de Activinas/metabolismo , Apoptosis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Metamorfosis Biológica/genética , Neuronas/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Activinas/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Ecdisona/metabolismo , Ecdisona/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Larva/citología , Larva/metabolismo , Metamorfosis Biológica/fisiología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Isoformas de Proteínas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Esteroides/genética , Transducción de Señal/genética , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología
6.
Development ; 142(5): 883-92, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25633355

RESUMEN

How the numbers of niches and resident stem cells within a particular organ are determined during development and how they may be modulated or corrected is a question with significant medical implications. In the larval ovary of Drosophila melanogaster, somatic precursors for niches, and germ cells that will become germline stem cells, co-develop. Somatic precursors proliferate during the first 3 days of larval development. By mid-third instar, adult terminal filament (TF) (part of the germline stem cell niche) cells first appear, and differentiation terminates 24 h later when 16-20 TFs fully form. The developmental sequence responsible for TF cell determination and final TF numbers is only partially understood. We show that TF formation proceeds through several, hitherto uncharacterized stages, which include an early exit from the cell cycle to form TF precursors and two steps of cell shape change to form the mature TF cells. The Activin receptor Baboon (Babo) is required for somatic precursor cell proliferation and therefore determines the pool of TF precursors available for TF differentiation. During the final differentiation stage, Babo facilitates TF and germ cell differentiation, and promotes the accumulation of Broad-Z1, which is also a target of the steroid hormone ecdysone. Epistasis analysis shows that Activin controls cell proliferation in an ecdysone-independent manner and TF differentiation by affecting ecdysone targets. We propose that this mode of function allows Activin to balance proliferation and differentiation, and to equilibrate niche numbers. These results suggest a novel model for how niche numbers are corrected during development.


Asunto(s)
Activinas/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Femenino , Ovario/citología , Transducción de Señal , Nicho de Células Madre/fisiología
7.
Planta ; 247(4): 1031-1042, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29453662

RESUMEN

MAIN CONCLUSION: The overexpression of RXam1 leads to a reduction in bacterial growth of XamCIO136, suggesting that RXam1 might be implicated in strain-specific resistance. Cassava bacterial blight (CBB) caused by Xanthomonas axonopodis pv. manihotis (Xam) is a prevalent disease in all regions, where cassava is cultivated. CBB is a foliar and vascular disease usually controlled through host resistance. Previous studies have found QTLs explaining resistance to several Xam strains. Interestingly, one QTL called XM5 that explained 13% of resistance to XamCIO136 was associated with a similar fragment of the rice Xa21-resistance gene called PCR250. In this study, we aimed to further identify and characterize this fragment and its role in resistance to CBB. Screening and hybridization of a BAC library using the molecular marker PCR250 as a probe led to the identification of a receptor-like kinase similar to Xa21 and were called RXam1 (Resistance to Xam 1). Here, we report the functional characterization of susceptible cassava plants overexpressing RXam1. Our results indicated that the overexpression of RXam1 leads to a reduction in bacterial growth of XamCIO136. This suggests that RXAM1 might be implicated in strain-specific resistance to XamCIO136.


Asunto(s)
Resistencia a la Enfermedad/genética , Manihot/genética , Enfermedades de las Plantas/microbiología , Xanthomonas axonopodis , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Genes de Plantas/genética , Inmunidad de la Planta/genética , Reacción en Cadena de la Polimerasa , Sitios de Carácter Cuantitativo/genética
8.
Acta Neuropathol ; 135(6): 887-906, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29397421

RESUMEN

The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.


Asunto(s)
Receptores de Activinas/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Oligodendroglía/metabolismo , Receptores de Activinas/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Células Cultivadas , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Oligodendroglía/patología , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos , Andamios del Tejido
9.
Int J Mol Sci ; 19(9)2018 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-30142896

RESUMEN

The high cardiovascular mortality associated with chronic kidney disease (CKD) is caused in part by the CKD-mineral bone disorder (CKD-MBD) syndrome. The CKD-MBD consists of skeletal, vascular and cardiac pathology caused by metabolic derangements produced by kidney disease. The prevalence of osteopenia/osteoporosis resulting from the skeletal component of the CKD-MBD, renal osteodystrophy (ROD), in patients with CKD exceeds that of the general population and is a major public health concern. That CKD is associated with compromised bone health is widely accepted, yet the mechanisms underlying impaired bone metabolism in CKD are not fully understood. Therefore, clarification of the molecular mechanisms by which CKD produces ROD is of crucial significance. We have shown that activin A, a member of the transforming growth factor (TGF)-ß super family, is an important positive regulator of receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis with Smad-mediated signaling being crucial for inducing osteoclast development and function. Recently, we have demonstrated systemic activation of activin receptors and activin A levels in CKD mouse models, such as diabetic CKD and Alport (AL) syndrome. In these CKD mouse models, bone remodeling caused by increased osteoclast numbers and activated osteoclastic bone resorption was observed and treatment with an activin receptor ligand trap repaired CKD-induced-osteoclastic bone resorption and stimulated individual osteoblastic bone formation, irrespective of parathyroid hormone (PTH) elevation. These findings have opened a new field for exploring mechanisms of activin A-enhanced osteoclast formation and function in CKD. Activin A appears to be a strong candidate for CKD-induced high-turnover ROD. Therefore, the treatment with the decoy receptor for activin A might be a good candidate for treatment for CKD-induced osteopenia or osteoporosis, indicating that the new findings from in these studies will lead to the identification of novel therapeutic targets for CKD-related and osteopenia and osteoporosis in general. In this review, we describe the impact of CKD-induced Smad signaling in osteoclasts, osteoblasts and vascular cells in CKD.


Asunto(s)
Activinas/metabolismo , Resorción Ósea/metabolismo , Huesos/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Riñón/metabolismo , Transducción de Señal , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Activinas/genética , Animales , Remodelación Ósea , Resorción Ósea/genética , Resorción Ósea/patología , Huesos/patología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/genética , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Riñón/patología , Ratones , Osteogénesis/genética , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo
10.
Development ; 140(16): 3403-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863480

RESUMEN

Blood flow plays crucial roles in vascular development, remodeling and homeostasis, but the molecular pathways required for transducing flow signals are not well understood. In zebrafish embryos, arterial expression of activin receptor-like kinase 1 (alk1), which encodes a TGFß family type I receptor, is dependent on blood flow, and loss of alk1 mimics lack of blood flow in terms of dysregulation of a subset of flow-responsive arterial genes and increased arterial endothelial cell number. These data suggest that blood flow activates Alk1 signaling to promote a flow-responsive gene expression program that limits nascent arterial caliber. Here, we demonstrate that restoration of endothelial alk1 expression to flow-deprived arteries fails to rescue Alk1 activity or normalize arterial endothelial cell gene expression or number, implying that blood flow may play an additional role in Alk1 signaling independent of alk1 induction. To this end, we define cardiac-derived Bmp10 as the crucial ligand for endothelial Alk1 in embryonic vascular development, and provide evidence that circulating Bmp10 acts through endothelial Alk1 to limit endothelial cell number in and thereby stabilize the caliber of nascent arteries. Thus, blood flow promotes Alk1 activity by concomitantly inducing alk1 expression and distributing Bmp10, thereby reinforcing this signaling pathway, which functions to limit arterial caliber at the onset of flow. Because mutations in ALK1 cause arteriovenous malformations (AVMs), our findings suggest that an impaired flow response initiates AVM development.


Asunto(s)
Receptores de Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Arterias Carótidas/enzimología , Embrión no Mamífero/irrigación sanguínea , Endotelio Vascular/enzimología , Receptores de Activinas/genética , Animales , Malformaciones Arteriovenosas/enzimología , Malformaciones Arteriovenosas/patología , Proteínas Morfogenéticas Óseas/genética , Recuento de Células , Embrión no Mamífero/metabolismo , Endotelina-1/genética , Endotelina-1/metabolismo , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Miocardio/enzimología , Miocardio/patología , Fosforilación , Transporte de Proteínas , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Development ; 140(3): 649-59, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293296

RESUMEN

Imaginal disc development in Drosophila requires coordinated cellular proliferation and tissue patterning. In our studies of TGFß superfamily signaling components, we found that a protein null mutation of Smad2, the only Activin subfamily R-Smad in the fruit fly, produces overgrown wing discs that resemble gain of function for BMP subfamily signaling. The wing discs are expanded specifically along the anterior-posterior axis, with increased proliferation in lateral regions. The morphological defect is not observed in mutants for the TGFß receptor baboon, and epistasis tests showed that baboon is epistatic to Smad2 for disc overgrowth. Rescue experiments indicate that Baboon binding, but not canonical transcription factor activity, of Smad2 is required for normal disc growth. Smad2 mutant discs generate a P-Mad stripe that is narrower and sharper than the normal gradient, and activation targets are correspondingly expressed in narrowed domains. Repression targets of P-Mad are profoundly mis-regulated, with brinker and pentagone reporter expression eliminated in Smad2 mutants. Loss of expression requires a silencer element previously shown to be controlled by BMP signaling. Epistasis experiments show that Baboon, Mad and Schnurri are required to mediate the ectopic silencer output in the absence of Smad2. Taken together, our results show that loss of Smad2 permits promiscuous Baboon activity, which represses genes subject to control by Mad-dependent silencer elements. The absence of Brinker and Pentagone in Smad2 mutants explains the compound wing disc phenotype. Our results highlight the physiological relevance of substrate inhibition of a kinase, and reveal a novel interplay between the Activin and BMP pathways.


Asunto(s)
Receptores de Activinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Discos Imaginales/fisiología , Proteína Smad2/metabolismo , Alas de Animales/fisiología , Receptores de Activinas/genética , Alelos , Animales , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Tamaño de la Célula , Cruzamientos Genéticos , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Epistasis Genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Discos Imaginales/metabolismo , Inmunohistoquímica , Masculino , Mutagénesis Sitio-Dirigida , Mutación , Tamaño de los Órganos , Fenotipo , Interferencia de ARN , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Smad Reguladas por Receptores , Proteína Smad2/genética , Factores de Tiempo , Alas de Animales/metabolismo
12.
BMC Genet ; 16: 123, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26486459

RESUMEN

BACKGROUND: Female fertility, a fundamental trait required for animal reproduction, has gradually declined in the last 2 decades in Japanese Black cattle. To identify associated genetic variants in Japanese Black cattle, we evaluated female fertility as a metric to describe the average inverse of the number of artificial inseminations required for conception from the first through the fourth parity (ANAI4) and conducted a genome-wide association study (GWAS) using 430 animals with extreme ANAI4 values from 10,399 animals. RESULTS: We found that 2 variants, namely a single-nucleotide polymorphisms (SNP; g.48476925C > T) and a 3-bp indel (g.48476943_48476946insGGC), in the upstream region of the activin receptor IIA gene (ACVR2A) were associated with ANAI4. ACVR2A transcripts from Japanese Black cattle of the Q haplotype, defined by the SNP and the 3-bp indel, with increased ANAI4 were 1.29-1.32-fold more abundant than q-derived transcripts. In agreement, reporter assay results revealed that the activity of the ACVR2A promoter was higher in reporter constructs with the Q haplotype than in those with the q haplotype by approximately 1.2 fold. Expression of exogenous ACVR2A induced dose-dependent increases of reporter activity from the follicle-stimulating hormone, beta polypeptide (FSHB) promoter in response to activin A in a pituitary gonadotrophic cell line. The findings suggested that sequence variations in the upstream region of ACVR2A with the Q haplotype increased ACVR2A transcription, which in turn induced FSHB expression. This association was replicated using a sample population size of 1,433 animals; the frequency of the Q haplotype was 0.39, and Q-to-q haplotype substitution resulted in an increase of 0.02 in terms of ANAI4. CONCLUSIONS: This GWAS identified variants in the upstream region of ACVR2A, which were associated with female fertility in Japanese Black cattle. The variants affected the level of ACVR2A mRNA expression, which could lead to an allelic imbalance. This association was replicated with a sample population of 1,433 animals. Thus, the results suggest that the Q haplotype could serve as a useful marker to select Japanese Black cattle with superior female fertility.


Asunto(s)
Receptores de Activinas/genética , Bovinos/genética , Fertilidad/genética , Variación Genética , Estudio de Asociación del Genoma Completo , Receptores de Activinas/metabolismo , Desequilibrio Alélico/genética , Animales , Secuencia de Bases , Línea Celular , Cromosomas de los Mamíferos/genética , Femenino , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica , Gonadotrofos/citología , Gonadotrofos/metabolismo , Haplotipos/genética , Mutación INDEL/genética , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas , Sitios de Carácter Cuantitativo/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
13.
Audiol Neurootol ; 20(1): 51-61, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25428170

RESUMEN

Activin, a member of the TGF-F superfamily, was found to play an important role in the development, repair and apoptosis of different tissues and organs. Accordingly, activin signaling is involved in the development of the cochlea. Activin binds to its receptor ActRII, then dimerizes with ActRI and induces a signaling pathway resulting in gene expression. A study reported a case of fibrodysplasia ossificans progressiva with an unusual mutation in the ActRI gene leading to sensorineural hearing loss. This draws attention to the role of activin and its receptors in the developed cochlea. To date, only the expression of ActRII is known in the adult mammalian cochlea. In this study, we present for the first time the presence of activin A and ActRIB in the adult cochlea. Transgenic mice with postnatal dominant-negative ActRIB expression causing disruption of activin signaling in vivo were used for assessing cochlear morphology and hearing ability through the auditory brainstem response (ABR) threshold. Nonfunctioning ActRIB did not affect the ABR thresholds and did not alter the microscopic anatomy of the cochlea. We conclude, therefore, that activin signaling is not necessary for hearing in adult mice under physiological conditions but may be important during and after damaging events in the inner ear.


Asunto(s)
Receptores de Activinas/metabolismo , Activinas/metabolismo , Cóclea/metabolismo , Audición/fisiología , Transducción de Señal/genética , Receptores de Activinas/genética , Activinas/genética , Animales , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Ratones , Ratones Transgénicos
14.
Genome Biol Evol ; 16(5)2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701021

RESUMEN

The genomes of plant and animal species are influenced by ancestral whole-genome duplication (WGD) events, which have profound impacts on the regulation and function of gene networks. To gain insight into the consequences of WGD events, we characterized the sequence conservation and expression patterns of ohnologs in the highly duplicated activin receptor signaling pathway in rainbow trout (RBT). The RBT activin receptor signaling pathway is defined by tissue-specific expression of inhibitors and ligands and broad expression of receptors and Co-Smad signaling molecules. Signaling pathway ligands exhibited shared expression, while inhibitors and Smad signaling molecules primarily express a single dominant ohnolog. Our findings suggest that gene function influences ohnolog evolution following duplication of the activin signaling pathway in RBT.


Asunto(s)
Evolución Molecular , Duplicación de Gen , Oncorhynchus mykiss , Transducción de Señal , Animales , Oncorhynchus mykiss/genética , Genoma , Activinas/metabolismo , Activinas/genética , Receptores de Activinas/genética , Receptores de Activinas/metabolismo
15.
Mar Biotechnol (NY) ; 26(4): 766-775, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39052141

RESUMEN

Activin signaling is essential for proper embryonic, skeletal muscle, and reproductive development. Duplication of the pathway in teleost fish has enabled diversification of gene function across the pathway but how gene duplication influences the function of activin signaling in non-mammalian species is poorly understood. Full characterization of activin receptor signaling pathway expression was performed across embryonic development and during early skeletal muscle growth in rainbow trout (RBT, Oncorhynchus mykiss). Rainbow trout are a model salmonid species that have undergone two additional rounds of whole genome duplication. A small number of genes were expressed early in development and most genes increased expression throughout development. There was limited expression of activin Ab in RBT embryos despite these genes exhibiting significantly elevated expression in post-hatch skeletal muscle. CRISPR editing of the activin Aa1 ohnolog and subsequent production of meiotic gynogenetic offspring revealed that biallelic disruption of activin Aa1 did not result in developmental defects, as occurs with knockout of activin A in mammals. The majority of gynogenetic offspring exhibited homozygous activin Aa1 genotypes (wild type, in-frame, or frameshift) derived from the mosaic founder female. The research identifies mechanisms of specialization among the duplicated activin ohnologs across embryonic development and during periods of high muscle growth in larval and juvenile fish. The knowledge gained provides insights into potential viable gene-targeting approaches for engineering the activin receptor signaling pathway and establishes the feasibility of employing meiotic gynogenesis as a tool for producing homozygous F1 genome-edited fish for species with long-generation times, such as salmonids.


Asunto(s)
Músculo Esquelético , Oncorhynchus mykiss , Transducción de Señal , Animales , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/crecimiento & desarrollo , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/embriología , Músculo Esquelético/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Activinas/metabolismo , Activinas/genética , Desarrollo Embrionario/genética , Desarrollo de Músculos/genética , Edición Génica , Embrión no Mamífero/metabolismo , Sistemas CRISPR-Cas , Receptores de Activinas/metabolismo , Receptores de Activinas/genética , Proteínas de Peces/genética , Proteínas de Peces/metabolismo
16.
PLoS One ; 19(6): e0305696, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38913612

RESUMEN

In Drosophila coordinated proliferation of two neural stem cells, neuroblasts (NB) and neuroepithelial (NE) cells, is pivotal for proper larval brain growth that ultimately determines the final size and performance of an adult brain. The larval brain growth displays two phases based on behaviors of NB and NEs: the first one in early larval stages, influenced by nutritional status and the second one in the last larval stage, promoted by ecdysone signaling after critical weight checkpoint. Mutations of the baboon (babo) gene that produces three isoforms (BaboA-C), all acting as type-I receptors of Activin-type transforming growth factor ß (TGF-ß) signaling, cause a small brain phenotype due to severely reduced proliferation of the neural stem cells. In this study we show that loss of babo function severely affects proliferation of NBs and NEs as well as conversion of NEs from both phases. By analyzing babo-null and newly generated isoform-specific mutants by CRISPR mutagenesis as well as isoform-specific RNAi knockdowns in a cell- and stage-specific manner, our data support differential contributions of the isoforms for these cellular events with BaboA playing the major role. Stage-specific expression of EcR-B1 in the brain is also regulated primarily by BaboA along with function of the other isoforms. Blocking EcR function in both neural stem cells results in a small brain phenotype that is more severe than baboA-knockdown alone. In summary, our study proposes that the Babo-mediated signaling promotes proper behaviors of the neural stem cells in both phases and achieves this by acting upstream of EcR-B1 expression in the second phase.


Asunto(s)
Encéfalo , Proliferación Celular , Proteínas de Drosophila , Larva , Células-Madre Neurales , Células Neuroepiteliales , Isoformas de Proteínas , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Larva/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Encéfalo/metabolismo , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Transducción de Señal , Receptores de Activinas/metabolismo , Receptores de Activinas/genética
17.
Reprod Fertil Dev ; 25(3): 570-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23164397

RESUMEN

Activin A and its inhibitors follistatin and inhibin play key roles in development and function of the male reproductive tract. Quantitative (q) polymerase chain reaction (PCR) was used to evaluate the expression of Inhba (the gene encoding activin A subunits), Inha and Inhbb (genes encoding the inhibin B subunits), as well as the genes for follistatin (Fst) and follistatin-like 3 (Fstl3) and the activin receptor subunits, in the male mouse reproductive tract. A qPCR assay that discriminated between the two follistatin variants of Fst288 (tissue-bound form) and Fst315 (circulating) was established. Activin A protein was measured by ELISA, whereas the inhibin α-subunit and total follistatin proteins were measured by radioimmunoassay (RIA). A screen of 22 tissues demonstrated tissue-specific regulation of the follistatin variants, with Fst288 highly expressed in the vas deferens and Fst315 most highly expressed in the skin. The expression of Fst288 and Fst315 and follistatin protein levels increased progressively from the testis through to the distal vas deferens. Inhba and the activin receptors were highly expressed in the epididymis, but activin A protein was elevated in both the epididymis and vas deferens. Inhibin α-subunit mRNA and protein and Inhbb expression were highest in the testis. These results indicate a role for activin A within the epididymis, but also that activin A bioactivity may be increasingly inhibited by follistatin distally along the male reproductive tract.


Asunto(s)
Activinas/metabolismo , Epidídimo/metabolismo , Folistatina/metabolismo , Regulación de la Expresión Génica , Inhibinas/metabolismo , Conducto Deferente/metabolismo , Regiones no Traducidas 3' , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Activinas/genética , Animales , Secuencia de Bases , Exones , Folistatina/química , Folistatina/genética , Genitales Masculinos/metabolismo , Inhibinas/genética , Masculino , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Especificidad de Órganos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
18.
Biomolecules ; 13(7)2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37509165

RESUMEN

Although structurally similar to type II counterparts, type I or activin receptor-like kinases (ALKs) are set apart by a metastable helix-loop-helix (HLH) element preceding the protein kinase domain that, according to a longstanding paradigm, serves passive albeit critical roles as an inhibitor-to-substrate-binding switch. A single recurrent mutation in the codon of the penultimate residue, directly adjacent the position of a constitutively activating substitution, causes milder activation of ACVR1/ALK2 leading to sporadic heterotopic bone deposition in patients presenting with fibrodysplasia ossificans progressiva, or FOP. To determine the protein structural-functional basis for the gain of function, R206H mutant, Q207D (aspartate-substituted caALK2) and HLH subdomain-truncated (208 Ntrunc) forms were compared to one another and the wild-type enzyme through in vitro kinase and protein-protein interaction analyses that were complemented by signaling read-out (p-Smad) in primary mouse embryonic fibroblasts and Drosophila S2 cells. Contrary to the paradigm, the HLH subdomain actively suppressed the phosphotransferase activity of the enzyme, even in the absence of FKBP12. Unexpectedly, perturbation of the HLH subdomain elevated kinase activity at a distance, i.e., allosterically, at the ATP-binding and polypeptide-interacting active site cleft. Accessibility to polypeptide substrate (BMP Smad C-terminal tails) due to allosterically altered conformations of type I active sites within heterohexameric cytoplasmic signaling complexes-assembled noncanonically by activin-type II receptors extracellularly-is hypothesized to produce a gain of function of the R206H mutant protein responsible for episodic heterotopic ossification in FOP.


Asunto(s)
Receptores de Activinas Tipo I , Mutación con Ganancia de Función , Animales , Ratones , Receptores de Activinas/genética , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Fibroblastos/metabolismo , Mutación , Péptidos/genética
19.
Stroke ; 43(5): 1432-5, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22328553

RESUMEN

BACKGROUND AND PURPOSE: The lack of an appropriate animal model has been a limitation in studying hemorrhage from arteriovenous malformations (AVMs) in the central nervous system. METHODS: Novel mouse central nervous system AVM models were generated by conditionally deleting the activin receptor-like kinase (Alk1; Acvrl1) gene with the SM22-Cre transgene. All mice developed AVMs in their brain and/or spinal cord, and >80% of them showed a paralysis or lethality phenotype due to internal hemorrhages during the first 10 to 15 weeks of life. The mice that survived this early lethal period, however, showed significantly reduced lethality rates even though they carried multiple AVMs. RESULTS: The age-dependent change in hemorrhage rates allowed us to identify molecular factors uniquely upregulated in the rupture-prone AVM lesions. CONCLUSIONS: Upregulation of angiopoietin 2 and a few inflammatory genes were identified in the hemorrhage-prone lesions, which may be comparable with human pathology. These models will be an exceptional tool to study pathophysiology of AVM hemorrhage.


Asunto(s)
Receptores de Activinas/genética , Envejecimiento/patología , Malformaciones Arteriovenosas Intracraneales/genética , Malformaciones Arteriovenosas Intracraneales/mortalidad , Proteínas de Microfilamentos/genética , Modelos Animales , Proteínas Musculares/genética , Envejecimiento/metabolismo , Angiopoyetina 2/metabolismo , Animales , Malformaciones Arteriovenosas Intracraneales/complicaciones , Hemorragias Intracraneales/epidemiología , Hemorragias Intracraneales/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Prevalencia , Factores de Riesgo , Regulación hacia Arriba
20.
Mol Cell Biochem ; 363(1-2): 179-90, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22167620

RESUMEN

Bone-morphogenetic proteins (BMPs) play an important role in development and many cellular processes. However, their functional role in the development and progression of breast cancer is not clearly understood. In the present study, we performed a systematic expression analysis of the 14 types of BMPs in 10 human breast cancer cell lines. We found that bone morphogenetic protein 4 (BMP4) was one of the most frequently expressed BMPs. Furthermore, the expression level of BMP4 was maybe correlated with the metastatic potential of the cancer lines. Accordingly, overexpression of BMP4 in the breast cancer cell lines MCF-7 and MBA-MD-231 promoted the migration and invasion phenotypes of the cancer cells, whereas RNAi-mediated knockdown of BMP4 expression inhibited the migration and invasion activities of the cancer cells. To identify the important factors that may mediate the BMP4 functions in breast cancer cells, we analyzed a panel of cancer-related genes, and found that the expression of matrix metalloproteinase-1 (MMP-1) and C-X-C chemokine receptor type 4 (CXCR4) sharply increased at both the mRNA and protein levels in the breast cancer cells overexpressing BMP4. Interestingly, when breast cancer cells MDA-MB-231 or MCF-7 were co-cultured with the osteoblast-like cells MG63 to mimic a bone metastasis microenvironment, BMP4 did not exhibit any significant effect on the expression of OPG or RANKL, two important factors in bone remodeling. BMPs antagonists, Noggin, parallel inhibited breast cancer cell migration and invasion and induced bone remodeling. Taken together, our results strongly suggest that BMP4 may promote the migration and invasion of breast cancer cells, at least in part by up-regulating the expressions of MMP-1 and CXCR4. It is conceivable that novel therapeutics for breast cancer may be developed by targeting BMP4 signaling pathway and/or its important downstream mediators in breast cancer cells.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Neoplasias de la Mama/metabolismo , Movimiento Celular , Receptores de Activinas/genética , Proteína Morfogenética Ósea 4/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Invasividad Neoplásica , Osteoblastos/metabolismo , Osteoprotegerina/metabolismo , Fenotipo , Ligando RANK/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Smad/genética , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda