Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Rep ; 37: 101607, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38178924

RESUMEN

Renal artery stenosis-induced chronic renal ischemia is an important cause of renal dysfunction, especially in older adults, and its incidence is currently increasing. To elucidate the mechanisms underlying chronic renal hypoperfusion-induced kidney damage, we developed a novel mouse model of renal artery coiling-based chronic hypoperfusion-related kidney injury. This model exhibits decreased renal blood flow and function, atrophy, and parenchymal injury in the coiled kidney, along with compensatory hypertrophy in the non-coiled kidney, without chronic hypertension. The availability of this mouse model, which can develop renal ischemia without genetic modification, will enhance kidney disease research by serving as a new tool to investigate the effects of acquired factors (e.g., obesity and aging) and genetic factors on renal artery stenosis-related renal parenchymal damage.

2.
Exp Anim ; 73(1): 93-100, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37661429

RESUMEN

Exocyst is an octameric protein complex implicated in exocytosis. The exocyst complex is highly conserved among mammalian species, but the physiological function of each subunit in exocyst remains unclear. Previously, we identified exocyst complex component 3-like (Exoc3l) as a gene abundantly expressed in embryonic endothelial cells and implicated in the process of angiogenesis in human umbilical cord endothelial cells. Here, to reveal the physiological roles of Exoc3l during development, we generated Exoc3l knockout (KO) mice by genome editing with CRISPR/Cas9. Exoc3l KO mice were viable and showed no significant phenotype in embryonic angiogenesis or postnatal retinal angiogenesis. Exoc3l KO mice also showed no significant alteration in cholesterol homeostasis or insulin secretion, although several reports suggest an association of Exoc3l with these processes. Despite the implied roles, Exoc3l KO mice exhibited no apparent phenotype in vascular development, cholesterol homeostasis, or insulin secretion.


Asunto(s)
Mutación con Pérdida de Función , Proteínas de Transporte Vesicular , Animales , Ratones , Humanos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Células Endoteliales/metabolismo , Secreción de Insulina , Colesterol , Mamíferos/metabolismo
3.
Biochem Pharmacol ; 218: 115899, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37907138

RESUMEN

Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.


Asunto(s)
Sistema Cardiovascular , Quinasas Asociadas a rho , Actinas/metabolismo , Sistema Cardiovascular/metabolismo , Proteínas de Unión al GTP rho , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Transducción de Señal/fisiología , Humanos
4.
Sci Rep ; 13(1): 15649, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37730951

RESUMEN

Familial hypercholesterolemia (FH) is an inherited autosomal dominant disorder that is associated with a high plasma level of low-density lipoprotein (LDL) cholesterol, leading to an increased risk of cardiovascular diseases. To develop basic and translational research on FH, we here generated an FH model in a non-human primate (cynomolgus monkeys) by deleting the LDL receptor (LDLR) gene using the genome editing technique. Six LDLR knockout (KO) monkeys were produced, all of which were confirmed to have mutations in the LDLR gene by sequence analysis. The levels of plasma cholesterol and triglyceride were quite high in the monkeys, and were similar to those in FH patients with homozygous mutations in the LDLR gene. In addition, periocular xanthoma was observed only 1 year after birth. Lipoprotein profile analysis showed that the plasma very low-density lipoprotein and LDL were elevated, while the plasma high density lipoprotein was decreased in LDLR KO monkeys. The LDLR KO monkeys were also strongly resistant to medications for hypercholesterolemia. Taken together, we successfully generated a non-human primate model of hypercholesterolemia in which the phenotype is similar to that of homozygous FH patients.


Asunto(s)
Traumatismos Craneocerebrales , Hipercolesterolemia , Hiperlipoproteinemia Tipo II , Animales , Humanos , Primates , Hiperlipoproteinemia Tipo II/genética , Lipoproteínas LDL , Macaca fascicularis
5.
J Biol Chem ; 299(3): 102993, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36758801

RESUMEN

Heart failure is one of the leading causes of death worldwide. RhoA, a small GTPase, governs actin dynamics in various tissue and cell types, including cardiomyocytes; however, its involvement in cardiac function has not been fully elucidated. Here, we generated cardiomyocyte-specific RhoA conditional knockout (cKO) mice, which demonstrated a significantly shorter lifespan with left ventricular dilation and severely impaired ejection fraction. We found that the cardiac tissues of the cKO mice exhibited structural disorganization with fibrosis and also exhibited enhanced senescence compared with control mice. In addition, we show that cardiomyocyte mitochondria were structurally abnormal in the aged cKO hearts. Clearance of damaged mitochondria by mitophagy was remarkably inhibited in both cKO cardiomyocytes and RhoA-knockdown HL-1 cultured cardiomyocytes. In RhoA-depleted cardiomyocytes, we reveal that the expression of Parkin, an E3 ubiquitin ligase that plays a crucial role in mitophagy, was reduced, and expression of N-Myc, a negative regulator of Parkin, was increased. We further reveal that the RhoA-Rho kinase axis induced N-Myc phosphorylation, which led to N-Myc degradation and Parkin upregulation. Re-expression of Parkin in RhoA-depleted cardiomyocytes restored mitophagy, reduced mitochondrial damage, attenuated cardiomyocyte senescence, and rescued cardiac function both in vitro and in vivo. Finally, we found that patients with idiopathic dilated cardiomyopathy without causal mutations for dilated cardiomyopathy showed reduced cardiac expression of RhoA and Parkin. These results suggest that RhoA promotes Parkin-mediated mitophagy as an indispensable mechanism contributing to cardioprotection in the aging heart.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Animales , Ratones , Cardiomiopatía Dilatada/metabolismo , Insuficiencia Cardíaca/metabolismo , Mitocondrias/metabolismo , Mitofagia/genética , Miocitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Hum Lact ; 39(2): 315-324, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35658699

RESUMEN

BACKGROUND: Glutamic acid, an amino acid that exhibits umami taste, is utilized in Japanese food and is abundant in human milk. We examined the influence of maternal habitual eating behavior on glutamic acid concentration in human milk. RESEARCH AIM: To determine the association between maternal dietary behaviors at the end of pregnancy and the 1st month postpartum and glutamic acid concentration in colostrum and mature milk. METHOD: This was a prospective, correlational, one-group longitudinal study. Women aged 20-30 years during the third trimester of pregnancy (N = 30) consented to participate and completed the data collection. Dietary history questionnaires were used to measure food intake. Glutamic acid levels in whey from colostrum and mature milk and in plasma during late pregnancy and the first month postpartum were measured. Data were considered significant at p < .05. Basic statistics, correlation coefficients analysis, unpaired t test, and one-way analysis of variance were performed. RESULTS: Glutamic acid concentrations in human milk and plasma were found to be significantly associated with the consumption of several different foods. There was no association between glutamic acid concentrations in human milk and plasma or between glutamic acid concentrations in colostrum and mature milk. The glutamic acid content of mature milk differed by physical activity level (mild and moderate) during the first month postpartum (t [46] = 2.87, p < .01). CONCLUSION: There was no clear association between habitual dietary behavior and glutamic acid concentration in human milk. However, maternal factors other than diet may be important and require additional research.


Asunto(s)
Ácido Glutámico , Leche Humana , Embarazo , Femenino , Humanos , Leche Humana/química , Ácido Glutámico/análisis , Ácido Glutámico/metabolismo , Estudios Prospectivos , Estudios Longitudinales , Lactancia Materna , Calostro/química , Dieta , Lactancia/metabolismo
7.
Commun Biol ; 5(1): 1071, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207400

RESUMEN

Whether a small GTPase RhoA plays a role in the pathology of abdominal aortic aneurysm (AAA) has not been determined. We show here that RhoA expression is reduced in human AAA lesions, compared with normal areas. Furthermore, incidence of AAA formation is increased in vascular smooth muscle cell (VSMC)-specific RhoA conditional knockout (cKO) mice. The contractility of the aortic rings and VSMCs from RhoA cKO mice is reduced, and expression of genes related to the VSMC contractility is attenuated by loss of RhoA. RhoA depletion activates the mitogen-activated protein (MAP) kinase signaling, including MAP4K4, in the aorta and VSMCs. Inhibition of MAP4K4 activity by DMX-5804 decreases AAA formation. Set, a binding protein to active RhoA, functions as an activator of MAP4K4 by sequestering PP2A, an inhibitor of MAP4K4, in the absence of RhoA. In conclusion, RhoA counteracts AAA formation through inhibition of MAP4K4 in cooperation with Set.


Asunto(s)
Aneurisma de la Aorta Abdominal , Músculo Liso Vascular , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/prevención & control , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitógenos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteína de Unión al GTP rhoA/genética , Quinasa de Factor Nuclear kappa B
8.
Cancer Sci ; 112(10): 4005-4012, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34420253

RESUMEN

Tumor growth and progression are complex processes mediated by mutual interactions between cancer cells and their surrounding stroma that include diverse cell types and acellular components, which form the tumor microenvironment. In this environment, direct intercellular communications play important roles in the regulation of the biological behaviors of tumors. However, the underlying molecular mechanisms are insufficiently defined. We used an in vitro coculture system to identify genes that were specifically expressed at higher levels in cancer cells associated with stromal cells. Major examples included epithelial membrane protein 1 (EMP1) and stomatin, which positively and negatively regulate tumor progression, respectively. EMP1 promotes tumor cell migration and metastasis via activation of the small GTPase Rac1, while stomatin strongly suppresses cell proliferation and induces apoptosis of cancer cells via inhibition of Akt signaling. Here we highlight important aspects of EMP1, stomatin, and their family members in cancer biology. Furthermore, we consider the molecules that participate in intercellular communications and signaling transduction between cancer cells and stromal cells, which may affect the phenotypes of cancer cells in the tumor microenvironment.


Asunto(s)
Comunicación Celular/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Células del Estroma/fisiología , Microambiente Tumoral/fisiología , Apoptosis/fisiología , Cadherinas/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Técnicas de Cocultivo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/genética , Proteína de Unión al GTP rac1/metabolismo
9.
J Biochem ; 170(5): 577-585, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34086898

RESUMEN

Transmembrane protein 168 (TMEM168) was found to be localized on the nuclear membrane. A heterozygous mutation (c.1616G>A, p. R539Q) in TMEM168 was identified in patients with Brugada syndrome. This mutation reduced expression of cardiomyocyte sodium channel Nav1.5 via Nedd4-2 E3 ubiquitin ligase-induced ubiquitination and degradation. However, the detailed molecular mechanism provoked by the TMEM168 mutant remains unclear. Here, we demonstrated that small heat shock protein αB-crystallin, which can bind to Nav1.5 and Nedd4-2 and interfere with the association of both proteins, was strongly recruited from the cell surface to the perinuclear region because of the much higher affinity of αB-crystallin with the TMEM168 mutant than with wild-type TMEM168. Following knockdown of αB-crystallin in HL-1 cardiomyocytes, the interaction of Nav1.5 with Nedd4-2 was increased, despite the reduced expression of Nav1.5. Moreover, reduction of Nav1.5 expression by αB-crystallin knockdown was rescued in the presence of a proteasome inhibitor MG-132, suggesting the importance of the αB-crystallin-modulated ubiquitin-proteasome system for the stability of Nav1.5 expression. Collectively, the balance of molecular interactions among Nav1.5, Nedd4-2 and αB-crystallin plays a role in the regulation of cardiomyocyte cell surface expression of Nav1.5, and the TMEM168 mutant disturbs this balance, resulting in a decrease in Nav1.5 expression.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Animales , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/patología , Línea Celular , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología
10.
J Biol Chem ; 296: 100761, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33971198

RESUMEN

Diabetes mellitus (DM) causes injury to tissues and organs, including to the heart and kidney, resulting in increased morbidity and mortality. Thus, novel potential therapeutics are continuously required to minimize DM-related organ damage. We have previously shown that dipeptidyl peptidase III (DPPIII) has beneficial roles in a hypertensive mouse model, but it is unknown whether DPPIII has any effects on DM. In this study, we found that intravenous administration of recombinant DPPIII in diabetic db/db mice for 8 weeks suppressed the DM-induced cardiac diastolic dysfunctions and renal injury without alteration of the blood glucose level. This treatment inhibited inflammatory cell infiltration and fibrosis in the heart and blocked the increase in albuminuria by attenuating the disruption of the glomerular microvasculature and inhibiting the effacement of podocyte foot processes in the kidney. The beneficial role of DPPIII was, at least in part, mediated by the cleavage of a cytotoxic peptide, named Peptide 2, which was increased in db/db mice compared with normal mice. This peptide consisted of nine amino acids, was a digested fragment of complement component 3 (C3), and had an anaphylatoxin-like effect determined by the Miles assay and chemoattractant analysis. The effect was dependent on its interaction with the C3a receptor and protein kinase C-mediated RhoA activation downstream of the receptor in endothelial cells. In conclusion, DPPIII plays a protective role in the heart and kidney in a DM animal model through cleavage of a peptide that is a part of C3.


Asunto(s)
Cardiomiopatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/tratamiento farmacológico , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/uso terapéutico , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Sustancias Protectoras/uso terapéutico , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Terapia Enzimática , Corazón/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Sustancias Protectoras/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico
11.
Cancer Res ; 81(9): 2318-2331, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33757977

RESUMEN

The growth and progression of cancers are crucially regulated by the tumor microenvironment where tumor cells and stromal cells are mutually associated. In this study, we found that stomatin expression was markedly upregulated by the interaction between prostate cancer cells and stromal cells. Stomatin suppressed cancer cell proliferation and enhanced apoptosis in vitro and inhibited xenograft tumor growth in vivo. Stomatin inhibited Akt activation, which is mediated by phosphoinositide-dependent protein kinase 1 (PDPK1). PDPK1 protein stability was maintained by its binding to HSP90. Stomatin interacted with PDPK1 and interfered with the PDPK1-HSP90 complex formation, resulting in decreased PDPK1 expression. Knockdown of stomatin in cancer cells elevated Akt activation and promoted cell increase by promoting the interaction between PDPK1 and HSP90. Clinically, stomatin expression levels were significantly decreased in human prostate cancer samples with high Gleason scores, and lower expression of stomatin was associated with higher recurrence of prostate cancer after the operation. Collectively, these findings demonstrate the tumor-suppressive effect of stromal-induced stomatin on cancer cells. SIGNIFICANCE: These findings reveal that interactions with stromal cells induce expression of stomatin in prostate cancer cells, which suppresses tumor growth via attenuation of the Akt signaling axis.


Asunto(s)
Proliferación Celular/genética , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Anciano , Animales , Apoptosis/genética , Comunicación Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Hep G2 , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias de la Próstata/patología , Células del Estroma/metabolismo , Transfección , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int Heart J ; 62(2): 390-395, 2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33731531

RESUMEN

Perivascular adipose tissue (PVAT) secretes large amounts of inflammatory mediators and plays a certain role in atherosclerosis formation from the exterior of the vessel. In the present study, we examined the expression level of inflammation-related mediators using adipose tissue samples harvested from patients with and without coronary artery disease (CAD). The subjects were 23 patients who underwent elective coronary bypass surgery (CAD group) and 17 patients who underwent elective mitral valve surgery (non-CAD group) between January 2017 and March 2018. The adipose tissue was harvested from three sites: the ascending aorta (AO), subcutaneous fat (SC), and pericoronary artery (CO) for the measurement of the expression levels of interleukin (IL) -1ß, IL-6, IL-10, tumor necrosis factor (TNF) -α, interferon (INF) -γ, and arginase (Arg) -1. In both the non-CAD and CAD groups, the expression levels of all mediators, except Agr-1, which showed a tendency to have higher levels in the SC than in the AO and CO, tended to upregulate in the AO than in the SC and CO. The CAD group had higher values of almost all mediators, except Arg-1. Most importantly, the expression levels of IL-1ß, IL-6, and IL-10 in the coronary artery were significantly higher in the CAD group. The expression levels of inflammatory mediators in the pericoronary adipose tissue were significantly higher in the CAD than in the non-CAD group. The adipose tissue appears to influence atherosclerosis formation from the exterior of the coronary artery.


Asunto(s)
Tejido Adiposo/metabolismo , Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Mediadores de Inflamación/metabolismo , Anciano , Aterosclerosis/diagnóstico , Biomarcadores/metabolismo , Vasos Coronarios , Femenino , Humanos , Masculino
13.
Cell Metab ; 32(3): 404-419.e6, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32726607

RESUMEN

SGLT2 inhibitors offer strong renoprotection in subjects with diabetic kidney disease (DKD). But the mechanism for such protection is not clear. Here, we report that in damaged proximal tubules of high-fat diet-fed ApoE-knockout mice, a model of non-proteinuric DKD, ATP production shifted from lipolysis to ketolysis dependent due to hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). We further found that empagliflozin raised endogenous ketone body (KB) levels, and thus its use or treatment with 1,3-butanediol, a KB precursor, prevented decreases in renal ATP levels and organ damage in the mice. The renoprotective effect of empagliflozin was abolished by gene deletion of Hmgcs2, a rate-limiting enzyme of ketogenesis. Furthermore, KBs attenuated mTORC1-associated podocyte damage and proteinuria in diabetic db/db mice. Our findings show that SGLT2 inhibition-associated renoprotection is mediated by an elevation of KBs that in turn corrects mTORC1 hyperactivation that occurs in non-proteinuric and proteinuric DKD.


Asunto(s)
Nefropatías Diabéticas/prevención & control , Cuerpos Cetónicos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Nefropatías Diabéticas/metabolismo , Femenino , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
14.
FASEB J ; 34(5): 6399-6417, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32175648

RESUMEN

Brugada syndrome (BrS) is an inherited channelopathy responsible for almost 20% of sudden cardiac deaths in patients with nonstructural cardiac diseases. Approximately 70% of BrS patients, the causative gene mutation(s) remains unknown. In this study, we used whole exome sequencing to investigate candidate mutations in a family clinically diagnosed with BrS. A heterozygous 1616G>A substitution (R539Q mutation) was identified in the transmembrane protein 168 (TMEM168) gene of symptomatic individuals. Similar to endogenous TMEM168, both TMEM168 wild-type (WT) and mutant proteins that were ectopically induced in HL-1 cells showed nuclear membrane localization. A significant decrease in Na+ current and Nav 1.5 protein expression was observed in HL-1 cardiomyocytes expressing mutant TMEM168. Ventricular tachyarrhythmias and conduction disorders were induced in the heterozygous Tmem168 1616G>A knock-in mice by pharmacological stimulation, but not in WT mice. Na+ current was reduced in ventricular cardiomyocytes isolated from the Tmem168 knock-in heart, and Nav 1.5 expression was also impaired. This impairment was dependent on increased Nedd4-2 binding to Nav 1.5 and subsequent ubiquitination. Collectively, our results show an association between the TMEM168 1616G>A mutation and arrhythmogenesis in a family with BrS.


Asunto(s)
Síndrome de Brugada/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Mutación , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Adulto , Animales , Síndrome de Brugada/patología , Femenino , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Linaje , Adulto Joven
15.
Sci Rep ; 10(1): 188, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31932617

RESUMEN

Anosmin-1 is a secreted glycoprotein encoded by the ANOS1 gene, and its loss of function causes Kallmann syndrome (KS), which is characterized by anosmia and hypogonadism due to olfactory bulb (OB) dysfunction. However, the physiological function of anosmin-1 remains to be elucidated. In KS, disordered angiogenesis is observed in OB, resulting in its hypoplasia. In this study, we examined the involvement of anosmin-1 in angiogenic processes. Anosmin-1 was detected on the vessel-like structure in OB of chick embryos, and promoted the outgrowth of vascular sprouts as shown by assays of OB tissue culture. Cell migration, proliferation, and tube formation of endothelial cells were induced by treatment with anosmin-1 as well as vascular endothelial growth factor-A (VEGF-A), and further enhanced by treatment with both of them. We newly identified that anosmin-1 activated VEGF receptor-2 (VEGFR2) by binding directly to it, and its downstream signaling molecules, phospholipase Cγ1 (PLCγ1) and protein kinase C (PKC). These results suggest that anosmin-1 plays a key role in the angiogenesis of developing OB through the VEGFR2-PLCγ1-PKC axis by enhancing the VEGF function.


Asunto(s)
Endotelio Vascular/citología , Proteínas de la Matriz Extracelular/metabolismo , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Bulbo Olfatorio/irrigación sanguínea , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Embrión de Pollo , Proteínas de la Matriz Extracelular/genética , Humanos , Morfogénesis , Proteínas del Tejido Nervioso/genética , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética
16.
Cancers (Basel) ; 11(11)2019 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-31652725

RESUMEN

The members of the family of epithelial membrane proteins (EMPs), EMP1, EMP2, and EMP3, possess four putative transmembrane domain structures and are composed of approximately 160 amino acid residues. EMPs are encoded by the growth arrest-specific 3 (GAS3)/peripheral myelin protein 22 kDa (PMP22) gene family. The GAS3/PMP22 family members play roles in cell migration, growth, and differentiation. Evidence indicates an association of these molecules with cancer progression and metastasis. Each EMP has pro- and anti-metastatic functions that are likely involved in the complex mechanisms of cancer progression. We have recently demonstrated that the upregulation of EMP1 expression facilitates cancer cell migration and invasion through the activation of a small GTPase, Rac1. The inoculation of prostate cancer cells overexpressing EMP1 into nude mice leads to metastasis to the lymph nodes and lungs, indicating that EMP1 contributes to metastasis. Pro-metastatic properties of EMP2 and EMP3 have also been proposed. Thus, targeting EMPs may provide new insights into their clinical utility. Here, we highlight the important aspects of EMPs in cancer biology, particularly invasiveness and metastasis, and describe recent therapeutic approaches.

17.
Circulation ; 139(18): 2157-2169, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30764634

RESUMEN

BACKGROUND: Bradyarrhythmia is a common clinical manifestation. Although the majority of cases are acquired, genetic analysis of families with bradyarrhythmia has identified a growing number of causative gene mutations. Because the only ultimate treatment for symptomatic bradyarrhythmia has been invasive surgical implantation of a pacemaker, the discovery of novel therapeutic molecular targets is necessary to improve prognosis and quality of life. METHODS: We investigated a family containing 7 individuals with autosomal dominant bradyarrhythmias of sinus node dysfunction, atrial fibrillation with slow ventricular response, and atrioventricular block. To identify the causative mutation, we conducted the family-based whole exome sequencing and genome-wide linkage analysis. We characterized the mutation-related mechanisms based on the pathophysiology in vitro. After generating a transgenic animal model to confirm the human phenotypes of bradyarrhythmia, we also evaluated the efficacy of a newly identified molecular-targeted compound to upregulate heart rate in bradyarrhythmias by using the animal model. RESULTS: We identified one heterozygous mutation, KCNJ3 c.247A>C, p.N83H, as a novel cause of hereditary bradyarrhythmias in this family. KCNJ3 encodes the inwardly rectifying potassium channel Kir3.1, which combines with Kir3.4 (encoded by KCNJ5) to form the acetylcholine-activated potassium channel ( IKACh channel) with specific expression in the atrium. An additional study using a genome cohort of 2185 patients with sporadic atrial fibrillation revealed another 5 rare mutations in KCNJ3 and KCNJ5, suggesting the relevance of both genes to these arrhythmias. Cellular electrophysiological studies revealed that the KCNJ3 p.N83H mutation caused a gain of IKACh channel function by increasing the basal current, even in the absence of m2 muscarinic receptor stimulation. We generated transgenic zebrafish expressing mutant human KCNJ3 in the atrium specifically. It is interesting to note that the selective IKACh channel blocker NIP-151 repressed the increased current and improved bradyarrhythmia phenotypes in the mutant zebrafish. CONCLUSIONS: The IKACh channel is associated with the pathophysiology of bradyarrhythmia and atrial fibrillation, and the mutant IKACh channel ( KCNJ3 p.N83H) can be effectively inhibited by NIP-151, a selective IKACh channel blocker. Thus, the IKACh channel might be considered to be a suitable pharmacological target for patients who have bradyarrhythmia with a gain-of-function mutation in the IKACh channel.


Asunto(s)
Fibrilación Atrial , Bloqueo Atrioventricular , Bradicardia , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Enfermedades Genéticas Congénitas , Mutación Missense , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Bloqueo Atrioventricular/genética , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/patología , Bloqueo Atrioventricular/fisiopatología , Benzopiranos/farmacología , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/patología , Bradicardia/fisiopatología , Técnicas Electrofisiológicas Cardíacas , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Enfermedades Genéticas Congénitas/fisiopatología , Humanos , Masculino , Xenopus laevis , Pez Cebra
18.
Sci Rep ; 8(1): 11507, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065301

RESUMEN

Blue cone monochromacy (BCM) is characterized by loss of function of both OPN1LW (the first) and OPN1MW (the downstream) genes on the X chromosome. The purpose of this study was to investigate the first and downstream genes in the OPN1LW/OPN1MW array in four unrelated Japanese males with BCM. In Case 1, only one gene was present. Abnormalities were found in the promoter, which had a mixed unique profile of first and downstream gene promoters and a -71A > C substitution. As the promoter was active in the reporter assay, the cause of BCM remains unclear. In Case 2, the same novel mutation, M273K, was present in exon 5 of both genes in a two-gene array. The mutant pigments showed no absorbance at any of the wavelengths tested, suggesting that the mutation causes pigment dysfunction. Case 3 had a large deletion including the locus control region and entire first gene. Case 4 also had a large deletion involving exons 2-6 of the first gene. As an intact LCR was present upstream and one apparently normal downstream gene was present, BCM in Case 4 was not ascribed solely to the deletion. The deletions in Cases 3 and 4 were considered to have been caused by non-homologous recombination.


Asunto(s)
Defectos de la Visión Cromática/genética , Opsinas de Bastones/genética , Adulto , Exones/genética , Genotipo , Humanos , Japón , Masculino , Mutación/genética , Linaje , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Conos , Opsinas de Bastones/metabolismo , Eliminación de Secuencia/genética
19.
Oncogene ; 37(40): 5416-5434, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29867202

RESUMEN

Tumor metastasis is the most common cause of cancer death. Elucidation of the mechanism of tumor metastasis is therefore important in the development of novel, effective anti-cancer therapies to reduce cancer mortality. Interaction between cancer cells and surrounding stromal cells in the tumor microenvironment is a key factor in tumor metastasis. Using a co-culture assay system with human prostate cancer LNCaP cells and primary human prostate stromal cells, we identified epithelial membrane protein 1 (EMP1) as a gene with elevated expression in the cancer cells. The orthotopic injection of LNCaP cells overexpressing EMP1 (EMP1-LNCaP cells) into the prostate of nude mice induced lymph node and lung metastases, while that of control LNCaP cells did not. EMP1-LNCaP cells had higher cell motility and Rac1 activity than control LNCaP cells. These results were also observed in other lines of cancer cells. We newly identified copine-III as an intracellular binding partner of EMP1. Knockdown of copine-III attenuated the increased cell motility and Rac1 activity in EMP1-LNCaP cells. Reduced cell motility and Rac1 activity following knockdown of copine-III in EMP1-LNCaP cells were recovered by re-expression of wild-type copine-III, but not of a copine-III mutant incapable of interacting with EMP1, suggesting the importance of the EMP1-copine-III interaction. Phosphorylated and activated Src and a Rac guanine nucleotide exchange factor Vav2 were found to be involved in the EMP1-induced enhancement of cell motility and Rac1 activation. Moreover, EMP1 was highly expressed in prostate cancer samples obtained from patients with higher Gleason score. These results demonstrate that upregulation of EMP1 significantly increases cancer cell migration that leads to tumor metastasis, suggesting that EMP1 may play an essential role as a positive regulator of tumor metastasis.


Asunto(s)
Movimiento Celular , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Células del Estroma/patología , Regulación hacia Arriba
20.
Oncotarget ; 9(30): 21022-21035, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29765517

RESUMEN

Dysregulation of cell metabolism is a hallmark of cancer. The mevalonate pathway in lipid metabolism has been implicated as a potential target of cancer therapy for hepatocellular carcinoma (HCC). The role of the Forkhead Box M1 (FoxM1) transcription factor in HCC development has been well documented, however, its involvement in cancer metabolism of HCC has not been fully determined. Here, we hypothesized that FoxM1 is involved in the mevalonate pathway of cholesterol biosynthesis in HCC. Inhibition of the mevalonate pathway by statins, inhibitors of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR), resulted in reduced expression of FoxM1 and increased cell death in human hepatoma cells. Re-exposure of mevalonate, a product of HMGCR, restored these effects. Likewise, knockdown of HMGCR reduced FoxM1 expression, indicating that FoxM1 expression was regulated by the mevalonate pathway in HCC. Mechanistically, protein geranylgeranylation was found to be responsible for FoxM1 expression and geranylgeranylated proteins, including RhoA, Rac1 or Cdc42, were shown to be involved in this process. In surgically resected human HCC tissues, the gene expression of FoxM1 had a positive correlation with that of the mevalonate pathway-related genes, such as HMGCR or sterol regulatory element-binding protein 2 (SREBP2). Furthermore, the gene expression of FoxM1 along with that of HMGCR or SREBP2 defined prognosis of HCC patients, suggesting the clinical significance of the mevalonate-FoxM1 pathway in human HCC. Our data indicate that FoxM1 links the mevalonate pathway to oncogenic signals in HCC. Thus, we propose a novel therapeutic approach to inhibit FoxM1 by targeting the mevalonate pathway for HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...