Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 144(12): 1314-1328, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-38848533

RESUMEN

ABSTRACT: The liver plays a crucial role in maintaining systemic iron homeostasis by secreting hepcidin, which is essential for coordinating iron levels in the body. Imbalances in iron homeostasis are associated with various clinical disorders related to iron deficiency or iron overload. Despite the clinical significance, the mechanisms underlying how hepatocytes sense extracellular iron levels to regulate hepcidin synthesis and iron storage are not fully understood. In this study, we identified Foxo1, a well-known regulator of macronutrient metabolism, which translocates to the nucleus of hepatocytes in response to high-iron feeding, holo-transferrin, and bone morphogenetic protein 6 (BMP6) treatment. Furthermore, Foxo1 plays a crucial role in mediating hepcidin induction in response to both iron and BMP signals by directly interacting with evolutionally conserved Foxo binding sites within the hepcidin promoter region. These binding sites were found to colocalize with Smad-binding sites. To investigate the physiological relevance of Foxo1 in iron metabolism, we generated mice with hepatocyte-specific deletion of Foxo1. These mice exhibited reduced hepatic hepcidin expression and serum hepcidin levels, accompanied by elevated serum iron and liver nonheme iron concentrations. Moreover, high-iron diet further exacerbated these abnormalities in iron metabolism in mice lacking hepatic Foxo1. Conversely, hepatocyte-specific Foxo1 overexpression increased hepatic hepcidin expression and serum hepcidin levels, thereby ameliorating iron overload in a murine model of hereditary hemochromatosis (Hfe-/- mice). In summary, our study identifies Foxo1 as a critical regulator of hepcidin and systemic iron homeostasis. Targeting Foxo1 may offer therapeutic opportunities for managing conditions associated with aberrant iron metabolism.


Asunto(s)
Proteína Forkhead Box O1 , Hepatocitos , Hepcidinas , Homeostasis , Hierro , Animales , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Hierro/metabolismo , Hepcidinas/metabolismo , Hepcidinas/genética , Ratones , Hepatocitos/metabolismo , Humanos , Ratones Noqueados , Hígado/metabolismo , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Regulación de la Expresión Génica
2.
Eur Heart J ; 45(4): 287-305, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-37992083

RESUMEN

BACKGROUND AND AIMS: Stanford type A aortic dissection (AD) is a degenerative aortic remodelling disease marked by an exceedingly high mortality without effective pharmacologic therapies. Smooth muscle cells (SMCs) lining tunica media adopt a range of states, and their transformation from contractile to synthetic phenotypes fundamentally triggers AD. However, the underlying pathomechanisms governing this population shift and subsequent AD, particularly at distinct disease temporal stages, remain elusive. METHODS: Ascending aortas from nine patients undergoing ascending aorta replacement and five individuals undergoing heart transplantation were subjected to single-cell RNA sequencing. The pathogenic targets governing the phenotypic switch of SMCs were identified by trajectory inference, functional scoring, single-cell regulatory network inference and clustering, regulon, and interactome analyses and confirmed using human ascending aortas, primary SMCs, and a ß-aminopropionitrile monofumarate-induced AD model. RESULTS: The transcriptional profiles of 93 397 cells revealed a dynamic temporal-specific phenotypic transition and marked elevation of the activator protein-1 (AP-1) complex, actively enabling synthetic SMC expansion. Mechanistically, tumour necrosis factor signalling enhanced AP-1 transcriptional activity by dampening mitochondrial oxidative phosphorylation (OXPHOS). Targeting this axis with the OXPHOS enhancer coenzyme Q10 or AP-1-specific inhibitor T-5224 impedes phenotypic transition and aortic degeneration while improving survival by 42.88% (58.3%-83.3% for coenzyme Q10 treatment), 150.15% (33.3%-83.3% for 2-week T-5224), and 175.38% (33.3%-91.7% for 3-week T-5224) in the ß-aminopropionitrile monofumarate-induced AD model. CONCLUSIONS: This cross-sectional compendium of cellular atlas of human ascending aortas during AD progression provides previously unappreciated insights into a transcriptional programme permitting aortic degeneration, highlighting a translational proof of concept for an anti-remodelling intervention as an attractive strategy to manage temporal-specific AD by modulating the tumour necrosis factor-OXPHOS-AP-1 axis.


Asunto(s)
Enfermedades de la Aorta , Disección Aórtica , Benzofenonas , Isoxazoles , Enfermedades Vasculares , Humanos , Factor de Transcripción AP-1 , Aminopropionitrilo , Estudios Transversales , Disección Aórtica/genética , Enfermedades de la Aorta/patología , Enfermedades Vasculares/patología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Factores de Necrosis Tumoral
3.
EMBO J ; 39(7): e103255, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32128839

RESUMEN

The Hippo-YAP signaling pathway plays an essential role in epithelial cells during intestinal regeneration and tumorigenesis. However, the molecular mechanism linking stromal signals to YAP-mediated intestinal regeneration and tumorigenesis is poorly defined. Here, we report a stroma-epithelium ISLR-YAP signaling axis essential for stromal cells to modulate epithelial cell growth during intestinal regeneration and tumorigenesis. Specifically, upon inflammation and in cancer, an oncogenic transcription factor ETS1 in stromal cells induces expression of a secreted protein ISLR that can inhibit Hippo signaling and activate YAP in epithelial cells. Deletion of Islr in stromal cells in mice markedly impaired intestinal regeneration and suppressed tumorigenesis in the colon. Moreover, the expression of stromal cell-specific ISLR and ETS1 significantly increased in inflamed mucosa of human IBD patients and in human colorectal adenocarcinoma, accounting for the epithelial YAP hyperactivation. Collectively, our findings provide new insights into the signaling crosstalk between stroma and epithelium during tissue regeneration and tumorigenesis.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Células HCT116 , Células HEK293 , Células HT29 , Vía de Señalización Hippo , Humanos , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Masculino , Ratones , Mutación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
4.
Int J Mol Sci ; 25(5)2024 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-38473909

RESUMEN

Mutations of the FBN1 gene lead to Marfan syndrome (MFS), which is an autosomal dominant connective tissue disorder featured by thoracic aortic aneurysm risk. There is currently no effective treatment for MFS. Here, we studied the role of mitochondrial dysfunction in the phenotypic transformation of human smooth muscle cells (SMCs) and whether a mitochondrial boosting strategy can be a potential treatment. We knocked down FBN1 in SMCs to create an MFS cell model and used rotenone to induce mitochondrial dysfunction. Furthermore, we incubated the shFBN1 SMCs with Coenzyme Q10 (CoQ10) to assess whether restoring mitochondrial function can reverse the phenotypic transformation. The results showed that shFBN1 SMCs had decreased TFAM (mitochondrial transcription factor A), mtDNA levels and mitochondrial mass, lost their contractile capacity and had increased synthetic phenotype markers. Inhibiting the mitochondrial function of SMCs can decrease the expression of contractile markers and increase the expression of synthetic genes. Imposing mitochondrial stress causes a double-hit effect on the TFAM level, oxidative phosphorylation and phenotypic transformation of FBN1-knockdown SMCs while restoring mitochondrial metabolism with CoQ10 can rapidly reverse the synthetic phenotype. Our results suggest that mitochondria function is a potential therapeutic target for the phenotypic transformation of SMCs in MFS.


Asunto(s)
Síndrome de Marfan , Enfermedades Mitocondriales , Ubiquinona/análogos & derivados , Humanos , Síndrome de Marfan/genética , Fenotipo , Miocitos del Músculo Liso/metabolismo , Enfermedades Mitocondriales/metabolismo , Fibrilina-1/metabolismo , Adipoquinas/metabolismo
5.
Cell Commun Signal ; 21(1): 4, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36604720

RESUMEN

Autophagy is a multi-step catabolic process that delivers cellular components to lysosomes for degradation and recycling. The dysregulation of this precisely controlled process disrupts cellular homeostasis and leads to many pathophysiological conditions. The mechanistic target of rapamycin (mTOR) is a central nutrient sensor that integrates growth signals with anabolism to fulfil biosynthetic and bioenergetic requirements. mTOR nucleates two distinct evolutionarily conserved complexes (mTORC1 and mTORC2). However, only mTORC1 is acutely inhibited by rapamycin. Consequently, mTORC1 is a well characterized regulator of autophagy. While less is known about mTORC2, the availability of acute small molecule inhibitors and multiple genetic models has led to increased understanding about the role of mTORC2 in autophagy. Emerging evidence suggests that the regulation of mTORC2 in autophagy is mainly through its downstream effector proteins, and is variable under different conditions and cellular contexts. Here, we review recent advances that describe a role for mTORC2 in this catabolic process, and propose that mTORC2 could be a potential clinical target for the treatment of autophagy-related diseases. Video abstract.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Serina-Treonina Quinasas TOR , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Sirolimus/farmacología , Autofagia
6.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36834896

RESUMEN

Vascular remodeling is a common pathological hallmark of many cardiovascular diseases. Vascular smooth muscle cells (VSMCs) are the predominant cell type lining the tunica media and play a crucial role in maintaining aortic morphology, integrity, contraction and elasticity. Their abnormal proliferation, migration, apoptosis and other activities are tightly associated with a spectrum of structural and functional alterations in blood vessels. Emerging evidence suggests that mitochondria, the energy center of VSMCs, participate in vascular remodeling through multiple mechanisms. For example, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α)-mediated mitochondrial biogenesis prevents VSMCs from proliferation and senescence. The imbalance between mitochondrial fusion and fission controls the abnormal proliferation, migration and phenotypic transformation of VSMCs. Guanosine triphosphate-hydrolyzing enzymes, including mitofusin 1 (MFN1), mitofusin 2 (MFN2), optic atrophy protein 1 (OPA1) and dynamin-related protein 1 (DRP1), are crucial for mitochondrial fusion and fission. In addition, abnormal mitophagy accelerates the senescence and apoptosis of VSMCs. PINK/Parkin and NIX/BINP3 pathways alleviate vascular remodeling by awakening mitophagy in VSMCs. Mitochondrial DNA (mtDNA) damage destroys the respiratory chain of VSMCs, resulting in excessive ROS production and decreased ATP levels, which are related to the proliferation, migration and apoptosis of VSMCs. Thus, maintaining mitochondrial homeostasis in VSMCs is a possible way to relieve pathologic vascular remodeling. This review aims to provide an overview of the role of mitochondria homeostasis in VSMCs during vascular remodeling and potential mitochondria-targeted therapies.


Asunto(s)
Músculo Liso Vascular , Remodelación Vascular , Humanos , Músculo Liso Vascular/metabolismo , Mitocondrias/metabolismo , ADN Mitocondrial/metabolismo , Homeostasis , Dinámicas Mitocondriales/fisiología
7.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36555691

RESUMEN

High mortality rates due to cardiovascular diseases (CVDs) have attracted worldwide attention. It has been reported that mitochondrial dysfunction is one of the most important mechanisms affecting the pathogenesis of CVDs. Mitochondrial DNA (mtDNA) mutations may result in impaired oxidative phosphorylation (OXPHOS), abnormal respiratory chains, and ATP production. In dysfunctional mitochondria, the electron transport chain (ETC) is uncoupled and the energy supply is reduced, while reactive oxygen species (ROS) production is increased. Here, we discussed and analyzed the relationship between mtDNA mutations, impaired mitophagy, decreased OXPHOS, elevated ROS, and CVDs from the perspective of mitochondrial dysfunction. Furthermore, we explored current potential therapeutic strategies for CVDs by eliminating mtDNA mutations (e.g., mtDNA editing and mitochondrial replacement), enhancing mitophagy, improving OXPHOS capacity (e.g., supplement with NAD+, nicotinamide riboside (NR), nicotinamide mononucleotide (NMN), and nano-drug delivery), and reducing ROS (e.g., supplement with Coenzyme Q10 and other antioxidants), and dissected their respective advantages and limitations. In fact, some therapeutic strategies are still a long way from achieving safe and effective clinical treatment. Although establishing effective and safe therapeutic strategies for CVDs remains challenging, starting from a mitochondrial perspective holds bright prospects.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedades Mitocondriales , Humanos , Especies Reactivas de Oxígeno/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/terapia , Enfermedades Cardiovasculares/metabolismo , Mitocondrias/metabolismo , ADN Mitocondrial/metabolismo , Transporte de Electrón , Enfermedades Mitocondriales/terapia , Enfermedades Mitocondriales/tratamiento farmacológico
8.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36142635

RESUMEN

Doxorubicin (DOXO) is a potent chemotherapeutic drug widely used to treat various cancers. However, its clinical application is limited due to serious adverse effects on dose-dependent cardiotoxicity. Although the underlying mechanism has not been fully clarified, DOXO-induced cardiotoxicity has been mainly attributed to the accumulation of reactive oxygen species (ROS) in cardiomyocytes. Fucoidan, as a kind of sulphated polysaccharide existing in numerous brown seaweed, has potent anti-oxidant, immune-regulatory, anti-tumor, anti-coagulate and anti-viral activities. Here, we explore the potential protective role and mechanism of fucoidan in DOXO-induced cardiotoxicity in mice. Our results show that oral fucoidan supplement exerts potent protective effects against DOXO-induced cardiotoxicity by reducing oxidative stress and preventing mitochondrial function injury. The improved effect of fucoidan on DOXO-induced cardiotoxicity was evaluated by echocardiography, cardiac myocytes size and cardiac fibrosis analysis, and the expression of genes related to cardiac dysfunction and remodeling. Fucoidan reduced the ROS content and the MDA levels but enhanced the activity of antioxidant enzymes GSH-PX and SOD in the mouse serum in a DOXO-induced cardiotoxicity model. In addition, fucoidan also increased the ATP production capacity and restored the levels of a mitochondrial respiratory chain complex in heart tissue. Collectively, this study highlights fucoidan as a potential polysaccharide for protecting against DOXO-induced cardiovascular diseases.


Asunto(s)
Antioxidantes , Cardiotoxicidad , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/metabolismo , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Doxorrubicina/farmacología , Ratones , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Polisacáridos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
9.
Int J Mol Sci ; 23(12)2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35743109

RESUMEN

Mitochondria, as the main site of cellular energy metabolism and the generation of oxygen free radicals, are the key switch for mitochondria-mediated endogenous apoptosis. Ca2+ is not only an important messenger for cell proliferation, but it is also an indispensable signal for cell death. Ca2+ participates in and plays a crucial role in the energy metabolism, physiology, and pathology of mitochondria. Mitochondria control the uptake and release of Ca2+ through channels/transporters, such as the mitochondrial calcium uniporter (MCU), and influence the concentration of Ca2+ in both mitochondria and cytoplasm, thereby regulating cellular Ca2+ homeostasis. Mitochondrial Ca2+ transport-related processes are involved in important biological processes of tumor cells including proliferation, metabolism, and apoptosis. In particular, MCU and its regulatory proteins represent a new era in the study of MCU-mediated mitochondrial Ca2+ homeostasis in tumors. Through an in-depth analysis of the close correlation between mitochondrial Ca2+ and energy metabolism, autophagy, and apoptosis of tumor cells, we can provide a valuable reference for further understanding of how mitochondrial Ca2+ regulation helps diagnosis and therapy.


Asunto(s)
Canales de Calcio , Calcio , Calcio/metabolismo , Canales de Calcio/metabolismo , Calcio de la Dieta , Citoplasma/metabolismo , Mitocondrias/metabolismo
10.
Int J Mol Sci ; 22(14)2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34299144

RESUMEN

Mitochondria are vital to life and provide biological energy for other organelles and cell physiological processes. On the mitochondrial double layer membrane, there are a variety of channels and transporters to transport different metal ions, such as Ca2+, K+, Na+, Mg2+, Zn2+ and Fe2+/Fe3+. Emerging evidence in recent years has shown that the metal ion transport is essential for mitochondrial function and cellular metabolism, including oxidative phosphorylation (OXPHOS), ATP production, mitochondrial integrity, mitochondrial volume, enzyme activity, signal transduction, proliferation and apoptosis. The homeostasis of mitochondrial metal ions plays an important role in maintaining mitochondria and cell functions and regulating multiple diseases. In particular, channels and transporters for transporting mitochondrial metal ions are very critical, which can be used as potential targets to treat neurodegeneration, cardiovascular diseases, cancer, diabetes and other metabolic diseases. This review summarizes the current research on several types of mitochondrial metal ion channels/transporters and their functions in cell metabolism and diseases, providing strong evidence and therapeutic strategies for further insights into related diseases.


Asunto(s)
Canales Iónicos/metabolismo , Metales/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Humanos , Transporte Iónico
11.
Proc Natl Acad Sci U S A ; 114(36): E7622-E7631, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827364

RESUMEN

The blood-brain barrier (BBB) establishes a protective interface between the central neuronal system and peripheral blood circulation and is crucial for homeostasis of the CNS. BBB formation starts when the endothelial cells (ECs) invade the CNS and pericytes are recruited to the nascent vessels during embryogenesis. Despite the essential function of pericyte-EC interaction during BBB development, the molecular mechanisms coordinating the pericyte-EC behavior and communication remain incompletely understood. Here, we report a single cell receptor, CD146, that presents dynamic expression patterns in the cerebrovasculature at the stages of BBB induction and maturation, coordinates the interplay of ECs and pericytes, and orchestrates BBB development spatiotemporally. In mouse brain, CD146 is first expressed in the cerebrovascular ECs of immature capillaries without pericyte coverage; with increased coverage of pericytes, CD146 could only be detected in pericytes, but not in cerebrovascular ECs. Specific deletion of Cd146 in mice ECs resulted in reduced brain endothelial claudin-5 expression and BBB breakdown. By analyzing mice with specific deletion of Cd146 in pericytes, which have defects in pericyte coverage and BBB integrity, we demonstrate that CD146 functions as a coreceptor of PDGF receptor-ß to mediate pericyte recruitment to cerebrovascular ECs. Moreover, we found that the attached pericytes in turn down-regulate endothelial CD146 by secreting TGF-ß1 to promote further BBB maturation. These results reveal that the dynamic expression of CD146 controls the behavior of ECs and pericytes, thereby coordinating the formation of a mature and stable BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/fisiología , Encéfalo/fisiología , Antígeno CD146/metabolismo , Regulación hacia Abajo/fisiología , Células Endoteliales/fisiología , Ratones , Ratones Noqueados , Pericitos/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
12.
Am J Pathol ; 184(5): 1604-16, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24767106

RESUMEN

Recently, enhanced CD146 expression was reported on endothelial cells in intestinal biopsies from patients with inflammatory bowel disease. However, the underlying mechanism remains unknown. Here, we found that overexpressed endothelial CD146 promoted the inflammatory responses in inflammatory bowel disease, which further potentiated the occurrence of colitis-associated colorectal carcinogenesis. Eliminating endothelial CD146 by conditional knockout significantly ameliorated the severity of inflammation in two different murine models of colitis, and decreased tumor incidence and tumor progression in a murine model of colitis-associated colorectal carcinogenesis. Mechanistic study showed that cytokine tumor necrosis factor-α (TNF-α) up-regulated the expression of endothelial CD146 through NF-κB transactivation. In turn, the enhanced endothelial CD146 expression promoted both angiogenesis and proinflammatory leukocyte extravasations, contributing to inflammation. Using an anti-CD146 antibody, AA98, alone or together with an anti-TNF-α antibody significantly attenuated colitis and prevented colitis-associated colorectal carcinogenesis in mice. Our study provides the first evidence that CD146 plays a dual role on endothelium, facilitating leukocyte extravasations and angiogenesis, thus promoting inflammation. This finding not only reveals the function and regulating mechanism of CD146 in inflammatory bowel disease, but also provides a promising therapeutic strategy for treating inflammatory bowel disease and preventing colitis-associated colorectal carcinogenesis.


Asunto(s)
Antígeno CD146/metabolismo , Carcinogénesis/patología , Colitis/patología , Colitis/prevención & control , Animales , Anticuerpos/farmacología , Comunicación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Sulfato de Dextran , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/patología , Interleucina-1beta/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neovascularización Patológica/patología , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
Proc Natl Acad Sci U S A ; 109(4): 1127-32, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22210108

RESUMEN

The epithelial-mesenchymal transition (EMT) plays an important role in breast cancer metastasis, especially in the most aggressive and lethal subtype, "triple-negative breast cancer" (TNBC). Here, we report that CD146 is a unique activator of EMTs and significantly correlates with TNBC. In epithelial breast cancer cells, overexpression of CD146 down-regulated epithelial markers and up-regulated mesenchymal markers, significantly promoted cell migration and invasion, and induced cancer stem cell-like properties. We further found that RhoA pathways positively regulated CD146-induced EMTs via the key EMT transcriptional factor Slug. An orthotopic breast tumor model demonstrated that CD146-overexpressing breast tumors showed a poorly differentiated phenotype and displayed increased tumor invasion and metastasis. We confirmed these findings by conducting an immunohistochemical analysis of 505 human primary breast tumor tissues and found that CD146 expression was significantly associated with high tumor stage, poor prognosis, and TNBC. CD146 was expressed at abnormally high levels (68.9%), and was strongly associated with E-cadherin down-regulation in TNBC samples. Taken together, these findings provide unique evidence that CD146 promotes breast cancer progression by induction of EMTs via the activation of RhoA and up-regulation of Slug. Thus, CD146 could be a therapeutic target for breast cancer, especially for TNBC.


Asunto(s)
Neoplasias de la Mama/genética , Antígeno CD146/genética , Transición Epitelial-Mesenquimal/fisiología , Adulto , Animales , Neoplasias de la Mama/metabolismo , Línea Celular , Perros , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Transfección , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/metabolismo
14.
Luminescence ; 30(6): 818-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25511847

RESUMEN

Isoenzyme c of horseradish peroxidase (HRP-C) is widely used in enzyme immunoassay combined with chemiluminescence (CL) detection. For this application, HRP-C activity measurement is usually based on luminol oxidation in the presence of hydrogen peroxide (H2O2). However, this catalysis reaction was enhancer dependent. In this study, we demonstrated that Jatropha curcas peroxidase (JcGP1) showed high efficiency in catalyzing luminol oxidation in the presence of H2O2. Compared with HRP-C, the JcGP1-induced reaction was enhancer independent, which made the enzyme-linked immunosorbent assay (ELISA) simpler. In addition, the JcGP1 catalyzed reaction showed a long-term stable CL signal. We optimized the conditions for JcGP1 catalysis and determined the favorable conditions as follows: 50 mM Tris buffer (pH 8.2) containing 10 mM H2 O2, 14 mM luminol and 0.75 M NaCl. The optimum catalysis temperature was 30°C. The detection limit of JcGP1 under optimum condition was 0.2 pM. Long-term stable CL signal combined with enhancer-independent property indicated that JcGP1 might be a valuable candidate peroxidase for clinical diagnosis and enzyme immunoassay with CL detection.


Asunto(s)
Jatropha/enzimología , Mediciones Luminiscentes/métodos , Luminol/química , Peroxidasa/química , Catálisis , Peroxidasa de Rábano Silvestre/química , Peroxidasa de Rábano Silvestre/metabolismo , Peróxido de Hidrógeno/química , Cinética , Límite de Detección , Luminiscencia , Oxidación-Reducción , Peroxidasa/metabolismo , Hojas de la Planta/enzimología , Temperatura
15.
Blood ; 120(11): 2330-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22718841

RESUMEN

CD146 is a novel endothelial biomarker and plays an essential role in angiogenesis; however, its role in the molecular mechanism underlying angiogenesis remains poorly understood. In the present study, we show that CD146 interacts directly with VEGFR-2 on endothelial cells and at the molecular level and identify the structural basis of CD146 binding to VEGFR-2. In addition, we show that CD146 is required in VEGF-induced VEGFR-2 phosphorylation, AKT/p38 MAPKs/NF-κB activation, and thus promotion of endothelial cell migration and microvascular formation. Furthermore, we show that anti-CD146 AA98 or CD146 siRNA abrogates all VEGFR-2 activation induced by VEGF. An in vivo angiogenesis assay showed that VEGF-promoted microvascular formation was impaired in the endothelial conditional knockout of CD146 (CD146(EC-KO)). Our animal experiments demonstrated that anti-CD146 (AA98) and anti-VEGF (bevacizumab) have an additive inhibitory effect on xenografted human pancreatic and melanoma tumors. The results of the present study suggest that CD146 is a new coreceptor for VEGFR-2 and is therefore a promising target for blocking tumor-related angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Neovascularización Patológica/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno CD146/química , Antígeno CD146/genética , Antígeno CD146/metabolismo , Línea Celular Tumoral , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
16.
Zhonghua Nei Ke Za Zhi ; 53(8): 631-4, 2014 Aug.
Artículo en Zh | MEDLINE | ID: mdl-25376826

RESUMEN

OBJECTIVE: To investigate the correlation between the expression of adhesion molecule CD146 and the vulnerability of carotid atherosclerotic plaque. METHODS: The plaque samples were collected from 40 patients who underwent the carotid endarterectomy and were divided into the stable plaque group and the instable plaque group by ultrasound imaging. Five carotid artery samples were taken from the healthy donors as the control. Immunohistochemistry was applied to test the CD146 expression in all samples. RESULTS: Higher expression of CD146 was observed in the atherosclerotic plaques than in the healthy control. Moreover, statistical difference was found in the expression of CD146 in the plaques between the instable plaque group and the stable plaque group (0.31 ± 0.19 vs 0.17 ± 0.07, P < 0.05). The expression of CD146 was positively correlated with the necrotic area (r = 0.471 8, P = 0.019 9) and the matrix metalloproteinase (MMP)-9 expression in the plaques (r = 0.535 6, P = 0.000 9). CONCLUSION: The CD146 expression is correlated with the vulnerability of carotid atherosclerotic plaque.


Asunto(s)
Placa Aterosclerótica/metabolismo , Antígeno CD146/metabolismo , Endarterectomía Carotidea , Humanos , Inmunohistoquímica , Metaloproteinasa 9 de la Matriz/metabolismo
17.
Zhonghua Yi Xue Za Zhi ; 94(43): 3392-6, 2014 Nov 25.
Artículo en Zh | MEDLINE | ID: mdl-25622668

RESUMEN

OBJECTIVE: To explore the relationship between circulating level of soluble CD146 (sCD146) and plaque vulnerability or inflammatory factors in patients with carotid atherosclerosis (CAS). METHODS: Forty CAS patients with carotid stenosis ( ≥ 70%) were enrolled and divided into 2 groups of stable and unstable plaque by ultrasonic imaging. And another 40 healthy subjects were enrolled for control group. Double-antibody sandwich enzyme-linked immunosorbent assay (ELISA) was employed to measure the serum levels of sCD146 and matrix metalloproteinase-9 (MMP-9), analyze the correlation of sCD146 with MMP-9 and high sensitivity CRP (hs-CRP), and evaluate whether sCD146 correlates with plaque vulnerability. RESULTS: Soluble CD146 level was elevated in CAS patients versus healthy donors [(212 ± 43) vs (173 ± 36) ng/ml, P < 0.001]. And sCD146 level significantly increased in CAS patients with unstable plaques than those with stable plaque [(218 ± 28) vs (176 ± 25) ng/ml, P < 0.001]. And sCD146 was correlated with high-sensitivity C-reactive protein (hsCRP, r = 0.370 9, P = 0.018 5), a well-known marker for CAS inflammation. Also it was an independent risk factor for plaque vulnerability (OR = 1.16, 95%CI:1.020-1.310, P = 0.019 2). And its level was not correlated with the risk factors of CAS, such as age, homocysteine, triglyceride, total cholesterol (TC), low density lipoprotein-cholesterol (LDL-C) or high density lipoprotein-cholesterol (HDL-C) (P > 0.05). But there was a good correlation with the serum level of MMP-9 in CAS patients (r = 0.677 2, P < 0.001). CONCLUSION: The concentration of soluble CD146 is positively correlated with hsCRP and MMP-9 in CAS patients. And inflammation and neovascularization may interact with each other during atherosclerotic process. The serum level of sCD146 is correlated independently with plaque vulnerability.


Asunto(s)
Enfermedades de las Arterias Carótidas , Placa Aterosclerótica , Biomarcadores , Proteína C-Reactiva , Antígeno CD146 , Estenosis Carotídea , HDL-Colesterol , LDL-Colesterol , Ensayo de Inmunoadsorción Enzimática , Humanos , Metaloproteinasa 9 de la Matriz , Factores de Riesgo , Triglicéridos
18.
Nutrients ; 16(12)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38931310

RESUMEN

BACKGROUND: Sarcopenia is an age-related condition characterized by progressive loss of muscle mass, strength, and function. The occurrence of sarcopenia has a huge impact on physical, psychological, and social health. Therefore, the prevention and treatment of sarcopenia is becoming an important public health issue. METHOD: 35 six-week-old male C57BL/6 mice were randomly divided into five groups, one of which served as a control group, while the rest of the groups were constructed as a model of sarcopenia by intraperitoneal injection of D-galactose. The intervention with lactoferrin, creatine, and their mixtures, respectively, was carried out through gavage for 8 weeks. Muscle function was assessed based on their endurance, hanging time, and grip strength. The muscle tissues were weighed to assess the changes in mass, and the muscle RNA was extracted for myogenic factor expression and transcriptome sequencing to speculate on the potential mechanism of action by GO and KEGG enrichment analysis. RESULT: The muscle mass (lean mass, GAS index), and muscle function (endurance, hanging time, and grip strength) decreased, and the size and structure of myofiber was smaller in the model group compared to the control group. The intervention with lactoferrin and creatine, either alone or combination, improved muscle mass and function, restored muscle tissue, and increased the expression of myogenic regulators. The combined group demonstrated the most significant improvement in these indexes. The RNA-seq results revealed enrichment in the longevity-regulated pathway, MAPK pathway, focal adhesion, and ECM-receptor interaction pathway in the intervention group. The intervention group may influence muscle function by affecting the proliferation, differentiation, senescence of skeletal muscle cell, and contraction of muscle fiber. The combined group also enriched the mTOR-S6K/4E-BPs signaling pathway, PI3K-Akt signaling pathway, and energy metabolism-related pathways, including Apelin signaling, insulin resistance pathway, and adipocytokine signaling pathway, which affect energy metabolism in muscle. CONCLUSIONS: Lactoferrin and creatine, either alone or in combination, were found to inhibit the progression of sarcopenia by influencing the number and cross-sectional area of muscle fibers and muscle protein synthesis. The combined intervention appears to exert a more significant effect on energy metabolism.


Asunto(s)
Creatina , Modelos Animales de Enfermedad , Lactoferrina , Ratones Endogámicos C57BL , Músculo Esquelético , Sarcopenia , Animales , Lactoferrina/farmacología , Masculino , Sarcopenia/tratamiento farmacológico , Sarcopenia/metabolismo , Creatina/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ratones , Fuerza Muscular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
19.
High Alt Med Biol ; 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38995860

RESUMEN

Yu Liu, Zhengyang Zhang, Yongting Luo, Peng An, Jingyi Qi, Xu Zhang, Shuaishuai Zhou, Yongzhi Li, Chong Xu, Junjie Luo, and Jiaping Wang. Product of traditional Chinese medicine longgui yangxinwan protects the human body from altitude sickness damage by reducing oxidative stress and preventing mitochondrial dysfunction. High Alt Med Biol. 00:00-00, 2024. Background: Plateau reaction, caused by high-altitude exposure, results in symptoms like headaches, dyspnea, palpitations, fatigue, shortness of breath, and insomnia due to reduced oxygen levels. Mitochondria are crucial for high-altitude acclimatization as they regulate oxygen metabolism and cellular energy, reducing oxidative stress and maintaining bodily functions. Methods: The study participants were randomly divided into placebo group, Rhodiola group and longgui yangxinwan (Original name: taikong yangxinwan) group, with 20 people in each group. Three groups of subjects were sampled at three time points (PI: pre-intervention; P-D1: high-altitude day 1; P-D7: high-altitude day 7), and blood pressure, blood oxygen, heart rate, hemoglobin, and red blood cell count were measured. The ATP content, mitochondrial DNA copy number, expression of mitochondria-related genes, reactive oxygen species (ROS), glutathione peroxidase (GSH-PX) and malondialdehyde (MDA) levels, and mitochondrial morphology were measured in blood at each time point. Results: Our study results demonstrate that longgui yangxinwan keeps the selected human physiological indicators stable and prevents mitochondrial dysfunction in the high altitude. Mechanically, longgui yangxinwan decreases the level of ROS in human serum, whereas increases the activity of the antioxidant enzyme GSH-PX. At high-altitude day 1 (P-D1) and high-altitude day 7 (P-D7), ROS in the placebo group were 1.5 and 2.2-fold higher than those of the longgui yangxinwan group, respectively. In addition, longgui yangxinwan enhances ATP production capacity, restores the levels of mitochondrial respiratory chain complexes, and effectively maintains mitochondrial morphology and integrity. At P-D1 and P-D7, the ATP levels in the longgui yangxinwan group were 19-fold and 26-fold higher than those in the placebo group, respectively. Conclusions: Our study highlights longgui yangxinwan as a potential drug for protecting humans from high-altitude damage by reducing oxidative stress and preventing mitochondrial dysfunction.

20.
Nutrients ; 16(14)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39064656

RESUMEN

The efficacy of functional lipids with antioxidant properties in reducing cardiovascular risk has not been consistent. Randomized controlled trials (RCTs) reporting estimates for the effects of antioxidant functional lipid supplementations on cardiometabolic risk factors were searched up to 1 May 2024. Overall, antioxidant lipid supplementations, compared with placebo, had favorable effects on systolic blood pressure (lycopene: -1.95 [-3.54, -0.36] mmHg), low-density lipoprotein cholesterol (n6 fatty acid: -0.39 [-0.71, -0.06] mmol/L; astaxanthin: -0.11 [-0.21, -0.01] mmol/L), high-density lipoprotein cholesterol (n3 fatty acid: 0.20 [0.13, 0.27] mmol/L; n6 fatty acid: 0.08 [0.01, 0.14] mmol/L; astaxanthin: 0.13 [0.05, 0.21] mmol/L), total cholesterol (n6 fatty acid: -0.24 [-0.37, -0.11] mmol/L; astaxanthin: -0.22 [-0.32, -0.12] mmol/L; beta-carotene: -0.13 [-0.23, -0.04] mmol/L), triglyceride (n3 fatty acid: -0.37 [-0.47, -0.28] mmol/L; astaxanthin: -0.46 [-0.83, -0.10] mmol/L), and fasting blood insulin (astaxanthin: -2.66 [-3.98, -1.34] pmol/L). The benefits of antioxidant lipid supplementations appeared to be most evident in blood pressure and blood lipids in participants with different cardiometabolic health statuses. Notably, n9 fatty acid increased triglyceride and hemoglobin A1C in the total population, which increases CVD risk. Antioxidant lipid supplementations ameliorate cardiometabolic risk factors, while their effect may depend on type and cardiometabolic health status. Long-term RCTs are needed to corroborate risk-benefit ratios across different antioxidant functional lipid supplementation settings.


Asunto(s)
Antioxidantes , Enfermedades Cardiovasculares , Suplementos Dietéticos , Factores de Riesgo de Enfermedad Cardiaca , Lípidos , Humanos , Antioxidantes/administración & dosificación , Enfermedades Cardiovasculares/prevención & control , Lípidos/sangre , Ensayos Clínicos Controlados Aleatorios como Asunto , Presión Sanguínea/efectos de los fármacos , Masculino , Femenino , Persona de Mediana Edad , Xantófilas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA