Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
PLoS Biol ; 21(9): e3002302, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37733651

RESUMEN

Organ laterality of vertebrates is specified by accelerated asymmetric decay of Dand5 mRNA mediated by Bicaudal-C1 (Bicc1) on the left side, but whether binding of this or any other mRNA to Bicc1 can be regulated is unknown. Here, we found that a CRISPR-engineered truncation in ankyrin and sterile alpha motif (SAM)-containing 3 (ANKS3) leads to symmetric mRNA decay mediated by the Bicc1-interacting Dand5 3' UTR. AlphaFold structure predictions of protein complexes and their biochemical validation by in vitro reconstitution reveal a novel interaction of the C-terminal coiled coil domain of ANKS3 with Bicc1 that inhibits binding of target mRNAs, depending on the conformation of ANKS3 and its regulation by ANKS6. The dual regulation of RNA binding by mutually opposing structured protein domains in this multivalent protein network emerges as a novel mechanism linking associated laterality defects and possibly other ciliopathies to perturbed dynamics in Bicc1 ribonucleoparticle (RNP) formation.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Lateralidad Funcional , Animales , Dominios Proteicos , ARN Mensajero/genética , Ribonucleoproteínas/genética
2.
iScience ; 26(6): 106855, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37275520

RESUMEN

The growing number of diseases linked to aberrant phase transitioning of ribonucleoproteins highlights the need to uncover how the interplay between multivalent protein and RNA interactions is regulated. Cytoplasmic granules of the RNA binding protein Bicaudal-C (Bicc1) are regulated by the ciliopathy proteins ankyrin (ANK) and sterile alpha motif (SAM) domain-containing ANKS3 and ANKS6, but whether and how target mRNAs are affected is unknown. Here, we show that head-to-tail polymers of Bicc1 nucleated by its SAM domain are interconnected by K homology (KH) domains in a protein meshwork that mediates liquid-to-gel transitioning of client transcripts. Moreover, while the dispersion of these granules by ANKS3 concomitantly released bound mRNAs, co-recruitment of ANKS6 by ANKS3 reinstated Bicc1 condensation and ribonucleoparticle assembly. RNA-independent Bicc1 polymerization and its dual regulation by ANKS3 and ANKS6 represent a new mechanism to couple the reversible immobilization of client mRNAs to controlled protein phase transitioning between distinct metastable states.

3.
Front Immunol ; 14: 1335207, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38304252

RESUMEN

The transforming growth factor-ß (TGF-ß) family member activin A (hereafter Activin-A) is overexpressed in many cancer types, often correlating with cancer-associated cachexia and poor prognosis. Activin-A secretion by melanoma cells indirectly impedes CD8+ T cell-mediated anti-tumor immunity and promotes resistance to immunotherapies, even though Activin-A can be proinflammatory in other contexts. To identify underlying mechanisms, we here analyzed the effect of Activin-A on syngeneic grafts of Braf mutant YUMM3.3 mouse melanoma cells and on their microenvironment using single-cell RNA sequencing. We found that the Activin-A-induced immune evasion was accompanied by a proinflammatory interferon signature across multiple cell types, and that the associated increase in tumor growth depended at least in part on pernicious STING activity within the melanoma cells. Besides corroborating a role for proinflammatory signals in facilitating immune evasion, our results suggest that STING holds considerable potential as a therapeutic target to mitigate tumor-promoting Activin-A signaling at least in melanoma.


Asunto(s)
Activinas , Melanoma , Factor de Crecimiento Transformador beta , Escape del Tumor , Animales , Ratones , Activinas/metabolismo , Melanoma/inmunología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
4.
ACS Nano ; 15(11): 17613-17622, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34751034

RESUMEN

Nanocharacterization plays a vital role in understanding the complex nanoscale organization of cells and organelles. Understanding cellular function requires high-resolution information about how the cellular structures evolve over time. A number of techniques exist to resolve static nanoscale structure of cells in great detail (super-resolution optical microscopy, EM, AFM). However, time-resolved imaging techniques tend to either have a lower resolution, are limited to small areas, or cause damage to the cells, thereby preventing long-term time-lapse studies. Scanning probe microscopy methods such as atomic force microscopy (AFM) combine high-resolution imaging with the ability to image living cells in physiological conditions. The mechanical contact between the tip and the sample, however, deforms the cell surface, disturbs the native state, and prohibits long-term time-lapse imaging. Here, we develop a scanning ion conductance microscope (SICM) for high-speed and long-term nanoscale imaging of eukaryotic cells. By utilizing advances in nanopositioning, nanopore fabrication, microelectronics, and controls engineering, we developed a microscopy method that can resolve spatiotemporally diverse three-dimensional (3D) processes on the cell membrane at sub-5-nm axial resolution. We tracked dynamic changes in live cell morphology with nanometer details and temporal ranges of subsecond to days, imaging diverse processes ranging from endocytosis, micropinocytosis, and mitosis to bacterial infection and cell differentiation in cancer cells. This technique enables a detailed look at membrane events and may offer insights into cell-cell interactions for infection, immunology, and cancer research.


Asunto(s)
Microscopía de Sonda de Barrido , Orgánulos , Microscopía de Sonda de Barrido/métodos , Microscopía de Fuerza Atómica , Membrana Celular
5.
Nat Commun ; 12(1): 4071, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210974

RESUMEN

Molecular left-right (L-R) asymmetry is established at the node of the mouse embryo as a result of the sensing of a leftward fluid flow by immotile cilia of perinodal crown cells and the consequent degradation of Dand5 mRNA on the left side. We here examined how the fluid flow induces Dand5 mRNA decay. We found that the first 200 nucleotides in the 3' untranslated region (3'-UTR) of Dand5 mRNA are necessary and sufficient for the left-sided decay and to mediate the response of a 3'-UTR reporter transgene to Ca2+, the cation channel Pkd2, the RNA-binding protein Bicc1 and their regulation by the flow direction. We show that Bicc1 preferentially recognizes GACR and YGAC sequences, which can explain the specific binding to a conserved GACGUGAC motif located in the proximal Dand5 3'-UTR. The Cnot3 component of the Ccr4-Not deadenylase complex interacts with Bicc1 and is also required for Dand5 mRNA decay at the node. These results suggest that Ca2+ currents induced by leftward fluid flow stimulate Bicc1 and Ccr4-Not to mediate Dand5 mRNA degradation specifically on the left side of the node.


Asunto(s)
Embrión de Mamíferos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores CCR4/metabolismo , Regiones no Traducidas 3' , Animales , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Receptores CCR4/genética , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción
6.
Cell Signal ; 68: 109499, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31838063

RESUMEN

Polycystic kidneys frequently associate with mutations in individual components of cilia, basal bodies or centriolar satellites that perturb complex protein networks. In this review, we focus on the RNA-binding protein Bicaudal-C1 (BICC1) which was found mutated in renal cystic dysplasia, and on its interactions with the ankyrin repeat and sterile α motif (SAM)-containing proteins ANKS3 and ANKS6 and associated kinases and their partially overlapping ciliopathy phenotypes. After reviewing BICC1 homologs in model organisms and their functions in mRNA and cell metabolism during development and in renal tubules, we discuss recent insights from cell-based assays and from structure analysis of the SAM domains, and how SAM domain oligomerization might influence multivalent higher order complexes that are implicated in ciliary signal transduction.


Asunto(s)
Enfermedades Renales Quísticas/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Animales , Desarrollo Embrionario , Gluconeogénesis , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales Quísticas/fisiopatología , ARN/metabolismo , Proteínas de Unión al ARN/química
7.
FEBS J ; 286(22): 4597-4620, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31276291

RESUMEN

The basic proprotein convertases (PCs) furin, PC1/3, PC2, PC5/6, PACE4, PC4, and PC7 are promising drug targets for human diseases. However, developing selective inhibitors remains challenging due to overlapping substrate recognition motifs and limited structural information. Classical drug screening approaches for basic PC inhibitors involve homogeneous biochemical assays using soluble recombinant enzymes combined with fluorogenic substrate peptides that may not accurately recapitulate the complex cellular context of the basic PC-substrate interaction. Herein we report basic PC sensor (BPCS), a novel cell-based molecular sensor that allows rapid screening of candidate inhibitors and their selectivity toward individual basic PCs within mammalian cells. BPCS consists of Gaussia luciferase linked to a sortilin-1 membrane anchor via a cleavage motif that allows efficient release of luciferase specifically if individual basic PCs are provided in the same membrane. Screening of selected candidate peptidomimetic inhibitors revealed that BPCS can readily distinguish between general and selective PC inhibitors in a high-throughput screening format. The robust and cost-effective assay format of BPCS makes it suitable to identify novel specific small-molecule inhibitors against basic PCs for therapeutic application. Its cell-based nature will allow screening for drug targets in addition to the catalytically active mature enzyme, including maturation, transport, and cellular factors that modulate the enzyme's activity. This broadened 'target range' will enhance the likelihood to identify novel small-molecule compounds that inhibit basic PCs in a direct or indirect manner and represents a conceptual advantage.


Asunto(s)
Técnicas Biosensibles/métodos , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Peptidomiméticos/farmacología , Proproteína Convertasas/metabolismo , Células A549 , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Técnicas Biosensibles/normas , Descubrimiento de Drogas/normas , Inhibidores Enzimáticos/química , Genes Reporteros , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Peptidomiméticos/química , Proproteína Convertasas/antagonistas & inhibidores , Sensibilidad y Especificidad
9.
PLoS Genet ; 14(7): e1007487, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29995892

RESUMEN

Altered glucose and lipid metabolism fuel cystic growth in polycystic kidneys, but the cause of these perturbations is unclear. Renal cysts also associate with mutations in Bicaudal C1 (Bicc1) or in its self-polymerizing sterile alpha motif (SAM). Here, we found that Bicc1 maintains normoglycemia and the expression of the gluconeogenic enzymes FBP1 and PEPCK in kidneys. A proteomic screen revealed that Bicc1 interacts with the C-Terminal to Lis-Homology domain (CTLH) complex. Since the orthologous Gid complex in S. cerevisae targets FBP1 and PEPCK for degradation, we mapped the topology among CTLH subunits and found that SAM-mediated binding controls Bicc1 protein levels, whereas Bicc1 inhibited the accumulation of several CTLH subunits. Under the conditions analyzed, Bicc1 increased FBP1 protein levels independently of the CTLH complex. Besides linking Bicc1 to cell metabolism, our findings reveal new layers of complexity in the regulation of renal gluconeogenesis compared to lower eukaryotes.


Asunto(s)
Gluconeogénesis/fisiología , Glucosa/biosíntesis , Riñón/metabolismo , Enfermedades Renales Poliquísticas/patología , Proteínas de Unión al ARN/metabolismo , Animales , Animales Recién Nacidos , Glucemia , Fructosa-Bifosfatasa/metabolismo , Glucosa/análisis , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Unión Proteica/fisiología , Mapeo de Interacción de Proteínas , Multimerización de Proteína/fisiología , ARN Mensajero/metabolismo , Motivo alfa Estéril/fisiología
10.
Cell Rep ; 22(8): 2176-2189, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466742

RESUMEN

Furin trafficking, and that of related proprotein convertases (PCs), may regulate which substrates are accessible for endoproteolysis, but tools to directly test this hypothesis have been lacking. Here, we develop targeted biosensors that indicate Furin activity in endosomes is 10-fold less inhibited by decanoyl-RVKR-chloromethylketone and enriched >3-fold in endosomes compared to the trans-Golgi network (TGN). Endogenous PC7, which resists this inhibitor, was active in distinct vesicles. Only overexpressed PC7 activity reached the cell surface, endosomes, and the TGN. A PLC motif in the cytosolic tail of PC7 was dispensable for endosomal activity, but it was specifically required for TGN recycling and to rescue proActivin-A cleavage in Furin-depleted B16F1 melanoma cells. In sharp contrast, PC7 complemented Furin in cleaving Notch1 independently of PLC-mediated TGN access. Our study provides a proof in principle that compartment-specific biosensors can be used to gain insight into the regulation of PC trafficking and to map the tropism of PC-specific inhibitors.


Asunto(s)
Técnicas Biosensibles , Compartimento Celular , Furina/metabolismo , Subtilisinas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Endocitosis , Endosomas/metabolismo , Exocitosis , Transferencia Resonante de Energía de Fluorescencia , Edición Génica , Células HEK293 , Células HeLa , Humanos , Melanoma/genética , Melanoma/patología , Melanoma Experimental/patología , Proteínas Mutantes/metabolismo , Proteolisis , Fracciones Subcelulares/metabolismo , Especificidad por Sustrato , Red trans-Golgi/metabolismo
11.
Structure ; 26(2): 209-224.e6, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29290488

RESUMEN

Head-to-tail polymers of sterile alpha motifs (SAM) can scaffold large macromolecular complexes. Several SAM-domain proteins that bind each other are mutated in patients with cystic kidneys or laterality defects, including the Ankyrin (ANK) and SAM domain-containing proteins ANKS6 and ANKS3, and the RNA-binding protein Bicc1. To address how their interactions are regulated, we first determined a high-resolution crystal structure of a Bicc1-SAM polymer, revealing a canonical SAM polymer with a high degree of flexibility in the subunit interface orientations. We further mapped interactions between full-length and distinct domains of Bicc1, ANKS3, and ANKS6. Neither ANKS3 nor ANKS6 alone formed macroscopic homopolymers in vivo. However, ANKS3 recruited ANKS6 to Bicc1, and the three proteins together cooperatively generated giant macromolecular complexes. Thus, the giant assemblies are shaped by SAM domains, their flanking sequences, and SAM-independent protein-protein and protein-mRNA interactions.


Asunto(s)
Proteínas Portadoras/química , Ciliopatías/metabolismo , Proteínas Nucleares/química , Proteínas de Unión al ARN/química , Cristalografía por Rayos X , Células HEK293 , Células HeLa , Humanos , Polímeros , Conformación Proteica , Motivo alfa Estéril
12.
J Invest Dermatol ; 137(12): 2578-2587, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28844941

RESUMEN

The secreted growth factor Activin-A of the transforming growth factor ß family and its receptors can promote or inhibit several cancer hallmarks including tumor cell proliferation and differentiation, vascularization, lymphangiogenesis and inflammation. However, a role in immune evasion and its relationship with tumor-induced muscle wasting and tumor vascularization, and the relative contributions of autocrine versus paracrine Activin signaling remain to be evaluated. To address this, we compared the effects of truncated soluble Activin receptor IIB as a ligand trap, or constitutively active mutant type IB receptor versus secreted Activin-A or the related ligand Nodal in mouse and human melanoma cell lines and tumor grafts. We found that although cell-autonomous receptor activation arrested tumor cell proliferation, Activin-A secretion stimulated melanoma cell dedifferentiation and tumor vascularization by functional blood vessels, and it increased primary and metastatic tumor burden and muscle wasting. Importantly, in mice with impaired adaptive immunity, the tumor-promoting effect of Activin-A was lost despite sustained vascularization and cachexia, suggesting that Activin-A promotes melanoma progression by inhibiting antitumor immunity. Paracrine Activin-A signaling emerges as a potential target for personalized therapies, both to reduce cachexia and to enhance the efficacy of immunotherapies.


Asunto(s)
Activinas/metabolismo , Evasión Inmune , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Caquexia , Ciclo Celular , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema Inmunológico , Antígeno Ki-67/metabolismo , Melanoma/patología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Patológica , Fenotipo , Transducción de Señal , Neoplasias Cutáneas/patología , Microambiente Tumoral
13.
Dev Biol ; 418(1): 75-88, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521049

RESUMEN

Mammalian Host-Cell Factor 1 (HCF-1), a transcriptional co-regulator, plays important roles during the cell-division cycle in cell culture, embryogenesis as well as adult tissue. In mice, HCF-1 is encoded by the X-chromosome-linked Hcfc1 gene. Induced Hcfc1(cKO/+) heterozygosity with a conditional knockout (cKO) allele in the epiblast of female embryos leads to a mixture of HCF-1-positive and -deficient cells owing to random X-chromosome inactivation. These embryos survive owing to the replacement of all HCF-1-deficient cells by HCF-1-positive cells during E5.5 to E8.5 of development. In contrast, complete epiblast-specific loss of HCF-1 in male embryos, Hcfc1(epiKO/Y), leads to embryonic lethality. Here, we characterize this lethality. We show that male epiblast-specific loss of Hcfc1 leads to a developmental arrest at E6.5 with a rapid progressive cell-cycle exit and an associated failure of anterior visceral endoderm migration and primitive streak formation. Subsequently, gastrulation does not take place. We note that the pattern of Hcfc1(epiKO/Y) lethality displays many similarities to loss of ß-catenin function. These results reveal essential new roles for HCF-1 in early embryonic cell proliferation and development.


Asunto(s)
Tipificación del Cuerpo/genética , Movimiento Celular/genética , Proliferación Celular/genética , Desarrollo Embrionario/genética , Factor C1 de la Célula Huésped/genética , Animales , Ciclo Celular/genética , Endodermo/citología , Endodermo/metabolismo , Femenino , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Genes Ligados a X/genética , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Noqueados , Transducción de Señal , beta Catenina/metabolismo
15.
J Cell Biol ; 210(7): 1185-97, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26416966

RESUMEN

The first cell differentiation in mammalian embryos segregates polarized trophectoderm cells from an apolar inner cell mass (ICM). This lineage decision is specified in compacted morulae by cell polarization and adhesion acting on the Yes-associated protein in the Hippo signaling pathway, but the regulatory mechanisms are unclear. We show that morula compaction and ICM formation depend on PC7 and the related proprotein convertases (PCs) Furin and Pace4 and that these proteases jointly regulate cell-cell adhesion mediated by E-cadherin processing. We also mapped the spatiotemporal activity profiles of these proteases by live imaging of a transgenic reporter substrate in wild-type and PC mutant embryos. Differential inhibition by a common inhibitor revealed that all three PCs are active in inner and outer cells, but in partially nonoverlapping compartments. E-cadherin processing by multiple PCs emerges as a novel mechanism to modulate cell-cell adhesion and fate allocation.


Asunto(s)
Blastocisto/metabolismo , Cadherinas/metabolismo , Furina/metabolismo , Proproteína Convertasas/metabolismo , Subtilisinas/metabolismo , Animales , Blastocisto/citología , Cadherinas/genética , Adhesión Celular/fisiología , Furina/genética , Vía de Señalización Hippo , Ratones , Ratones Noqueados , Mutación , Proproteína Convertasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Subtilisinas/genética
16.
Mol Cell Biol ; 35(19): 3339-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26217012

RESUMEN

Loss of the RNA-binding protein Bicaudal-C (Bicc1) provokes renal and pancreatic cysts as well as ectopic Wnt/ß-catenin signaling during visceral left-right patterning. Renal cysts are linked to defective silencing of Bicc1 target mRNAs, including adenylate cyclase 6 (AC6). RNA binding of Bicc1 is mediated by N-terminal KH domains, whereas a C-terminal sterile alpha motif (SAM) self-polymerizes in vitro and localizes Bicc1 in cytoplasmic foci in vivo. To assess a role for multimerization in silencing, we conducted structure modeling and then mutated the SAM domain residues which in this model were predicted to polymerize Bicc1 in a left-handed helix. We show that a SAM-SAM interface concentrates Bicc1 in cytoplasmic clusters to specifically localize and silence bound mRNA. In addition, defective polymerization decreases Bicc1 stability and thus indirectly attenuates inhibition of Dishevelled 2 in the Wnt/ß-catenin pathway. Importantly, aberrant C-terminal extension of the SAM domain in bpk mutant Bicc1 phenocopied these defects. We conclude that polymerization is a novel disease-relevant mechanism both to stabilize Bicc1 and to present associated mRNAs in specific silencing platforms.


Asunto(s)
ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Riñón/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Multimerización de Proteína , Transporte de Proteínas , Interferencia de ARN , Transporte de ARN , ARN Mensajero/genética , Vía de Señalización Wnt
17.
J Transl Med ; 13: 103, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25888842

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by numerous fluid-filled cysts that frequently result in end-stage renal disease. While promising treatment options are in advanced clinical development, early diagnosis and follow-up remain a major challenge. We therefore evaluated the diagnostic value of Fetuin-A as a new biomarker of ADPKD in human urine. RESULTS: We found that renal Fetuin-A levels are upregulated in both Pkd1 and Bicc1 mouse models of ADPKD. Measurement by ELISA revealed that urinary Fetuin-A levels were significantly higher in 66 ADPKD patients (17.5 ± 12.5 µg/mmol creatinine) compared to 17 healthy volunteers (8.5 ± 3.8 µg/mmol creatinine) or 50 control patients with renal diseases of other causes (6.2 ± 2.9 µg/mmol creatinine). Receiver operating characteristics (ROC) analysis of urinary Fetuin-A levels for ADPKD rendered an optimum cut-off value of 12.2 µg/mmol creatinine, corresponding to 94% of sensitivity and 60% of specificity (area under the curve 0.74 ; p = 0.0019). Furthermore, urinary Fetuin-A levels in ADPKD patients correlated with the degree of renal insufficiency and showed a significant increase in patients with preserved renal function followed for two years. CONCLUSIONS: Our findings establish urinary Fetuin-A as a sensitive biomarker of the progression of ADPKD. Further studies are required to examine the pathogenic mechanisms of elevated renal and urinary Fetuin-A in ADPKD.


Asunto(s)
Progresión de la Enfermedad , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/orina , alfa-2-Glicoproteína-HS/orina , Adulto , Anciano , Animales , Biomarcadores/orina , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Fallo Renal Crónico/orina , Masculino , Ratones Noqueados , Persona de Mediana Edad , Proteínas de Unión al ARN/metabolismo , Curva ROC , Regulación hacia Arriba
18.
Development ; 142(5): 858-70, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25715394

RESUMEN

In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Técnica del Anticuerpo Fluorescente , Genotipo , Factor Nuclear 6 del Hepatocito/genética , Etiquetado Corte-Fin in Situ , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Células Madre/citología , Células Madre/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
19.
J Biol Chem ; 289(25): 17854-71, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24798330

RESUMEN

The TGFß family member Nodal is central to control pluripotent stem cell fate, but its use as a stem cell differentiation factor is limited by low specific activity. During development, Nodal depends on growth and differentiation factor (Gdf)-1 and on the shared co-receptor Cryptic to specify visceral left-right axis asymmetry. We therefore asked whether the functionality of Nodal can be augmented by Gdf1. Because Nodal and Gdf1 coimmunoprecipitate each other, they were predicted to form heterodimers, possibly to facilitate diffusion or to increase the affinity for signaling receptors. Here, we report that Gdf1 suppresses an unexpected dependence of Nodal on serum proteins and that it is critically required for non-autonomous signaling in cells expressing Cryptic. Nodal, Gdf1, and their cleaved propeptides copurified as a heterodimeric low molecular weight complex that stimulated Activin receptor (Acvr) signaling far more potently than Nodal alone. Although heterodimerization with Gdf1 did not increase binding of Nodal to Fc fusions of co-receptors or Acvr extracellular domains, it was essential for soluble Acvr2 to inhibit Nodal signaling. This implies that Gdf1 potentiates Nodal activity by stabilizing a low molecular weight fraction that is susceptible to neutralization by soluble Acvr2. Finally, in differentiating human ES cells, endodermal markers were more efficiently induced by Nodal·Gdf1 than by Nodal, suggesting that Nodal·Gdf1 is an attractive new reagent to direct stem cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Factor 1 de Diferenciación de Crecimiento/metabolismo , Proteína Nodal/metabolismo , Multimerización de Proteína/fisiología , Transducción de Señal/fisiología , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Endodermo/citología , Factor 1 de Diferenciación de Crecimiento/genética , Células HEK293 , Células Hep G2 , Humanos , Ratones , Ratones Noqueados , Proteína Nodal/genética , Estructura Terciaria de Proteína
20.
Semin Cell Dev Biol ; 32: 85-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24508081

RESUMEN

Secreted cytokines of the TGFß family are found in all multicellular organisms and implicated in regulating fundamental cell behaviors such as proliferation, differentiation, migration and survival. Signal transduction involves complexes of specific type I and II receptor kinases that induce the nuclear translocation of Smad transcription factors to regulate target genes. Ligands of the BMP and Nodal subgroups act at a distance to specify distinct cell fates in a concentration-dependent manner. These signaling gradients are shaped by multiple factors, including proteases of the proprotein convertase (PC) family that hydrolyze one or several peptide bonds between an N-terminal prodomain and the C-terminal domain that forms the mature ligand. This review summarizes information on the proteolytic processing of TGFß and related precursors, and its spatiotemporal regulation by PCs during development and various diseases, including cancer. Available evidence suggests that the unmasking of receptor binding epitopes of TGFß is only one (and in some cases a non-essential) function of precursor processing. Future studies should consider the impact of proteolytic maturation on protein localization, trafficking and turnover in cells and in the extracellular space.


Asunto(s)
Precursores de Proteínas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Proteínas de Unión a TGF-beta Latente/química , Proteínas de Unión a TGF-beta Latente/metabolismo , Modelos Moleculares , Proproteína Convertasas/química , Proproteína Convertasas/metabolismo , Unión Proteica , Precursores de Proteínas/química , Estructura Terciaria de Proteína , Factor de Crecimiento Transformador beta/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...