Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26.094
Filtrar
1.
Sci Rep ; 14(1): 21412, 2024 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-39271758

RESUMEN

Hearing loss affects around 5% of the global population. Two preliminary studies have described genetic variants in sporadic individuals with hearing loss from Pakistan. Here we extend these studies to determine the spectrum of variants in a cohort of individuals with no previous history of hearing loss. Individuals with hearing loss born to consanguineous couples were identified from special schools. Audiograms were assessed. DNA from participants negative for GJB2 pathogenic variants was subjected to exome sequencing. Data were filtered to include variants with frequencies < 0.01 in the public databases. The effects of the missense variants on respective amino acids were analyzed by using PyMol software. Among the 44 participants, hearing loss was moderate for two individuals; 14 exhibited moderately-severe hearing loss while 25 had a severe degree of hearing loss. Hearing loss was reported to have been progressive in four participants and was currently profound in three participants. Variants were unambiguously identified in 17 genes, of which the majority affected SLC26A4. CDH23, MYO15A and OTOF were other significant contributors. Deleterious variants detected in two genes suggest new associations for hearing loss. Molecular characterization of hearing loss in our cohort revealed high genetic heterogeneity with a 75% diagnostic rate.


Asunto(s)
Secuenciación del Exoma , Pérdida Auditiva , Transportadores de Sulfato , Humanos , Masculino , Niño , Femenino , Pérdida Auditiva/genética , Transportadores de Sulfato/genética , Adolescente , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Proteínas de la Membrana/genética , Conexina 26/genética , Predisposición Genética a la Enfermedad , Pakistán , Consanguinidad , Conexinas/genética , Mutación Missense , Miosinas
2.
Proc Natl Acad Sci U S A ; 121(37): e2405560121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39231206

RESUMEN

Collective cell migration is crucial in various physiological processes, including wound healing, morphogenesis, and cancer metastasis. Adherens Junctions (AJs) play a pivotal role in regulating cell cohesion and migration dynamics during tissue remodeling. While the role and origin of the junctional mechanical tension at AJs have been extensively studied, the influence of the actin cortex structure and dynamics on junction plasticity remains incompletely understood. Moreover, the mechanisms underlying stress dissipation at junctions are not well elucidated. Here, we found that the ligand-independent phosphorylation of epithelial growth factor receptor (EGFR) downstream of de novo E-cadherin adhesion orchestrates a feedback loop, governing intercellular viscosity via the Rac pathway regulating actin dynamics. Our findings highlight how the E-cadherin-dependent EGFR activity controls the migration mode of collective cell movements independently of intercellular tension. This modulation of effective viscosity coordinates cellular movements within the expanding monolayer, inducing a transition from swirling to laminar flow patterns while maintaining a constant migration front speed. Additionally, we propose a vertex model with adjustable junctional viscosity, capable of replicating all observed cellular flow phenotypes experimentally.


Asunto(s)
Cadherinas , Movimiento Celular , Receptores ErbB , Fosforilación , Movimiento Celular/fisiología , Cadherinas/metabolismo , Receptores ErbB/metabolismo , Viscosidad , Humanos , Animales , Uniones Adherentes/metabolismo , Perros
3.
Virulence ; 15(1): 2399217, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39221673

RESUMEN

Fusobacterium nucleatum (F. nucleatum), an anaerobic resident of the oral cavity, is increasingly recognized as a contributing factor to ulcerative colitis (UC). The adhesive properties of F. nucleatum are mediated by its key virulence protein, FadA adhesin. However, further investigations are needed to understand the pathogenic mechanisms of this oral pathogen in UC. The present study aimed to explore the role of the FadA adhesin in the colonization and invasion of oral F. nucleatum in dextran sulphate sodium (DSS)-induced colitis mice via molecular techniques. In this study, we found that oral inoculation of F. nucleatum strain carrying the FadA adhesin further exacerbated DSS-induced colitis, leading to elevated alveolar bone loss, disease severity, and mortality. Additionally, CDH1 gene knockout mice treated with DSS presented increases in body weight and alveolar bone density, as well as a reduction in disease severity. Furthermore, FadA adhesin adhered to its mucosal receptor E-cadherin, leading to the phosphorylation of ß-catenin and the degradation of IκBα, the activation of the NF-κB signalling pathway and the upregulation of downstream cytokines. In conclusion, this research revealed that oral inoculation with F. nucleatum facilitates experimental colitis via the secretion of the virulence adhesin FadA. Targeting the oral pathogen F. nucleatum and its virulence factor FadA may represent a promising therapeutic approach for a portion of UC patients.


Asunto(s)
Adhesinas Bacterianas , Colitis Ulcerosa , Infecciones por Fusobacterium , Fusobacterium nucleatum , Animales , Humanos , Ratones , Adhesinas Bacterianas/metabolismo , Adhesinas Bacterianas/genética , Adhesión Bacteriana , Cadherinas/metabolismo , Colitis Ulcerosa/microbiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Infecciones por Fusobacterium/microbiología , Fusobacterium nucleatum/patogenicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1459-1466, 2024 Aug 20.
Artículo en Chino | MEDLINE | ID: mdl-39276041

RESUMEN

OBJECTIVE: To investigate the mechanism by which conbercept reverses transforming growth factor-ß2 (TGF-ß2)-induced epithelial-mesenchymal transition (EMT) in human lens epithelial cells (HLECs). METHODS: Cultured HLEC SRA01/04 cells were treated with TGF-ß2, conbercept, or both, and the changes in cell proliferation, apoptosis, and migration were observed using MTT assay, flow cytometry, scratch assay, and Transwell assay. Western blotting and qRT-PCR were used to detect the changes in the expression of EMT-related epithelial cell markers (E-Cadherin, α-SMA, and Snail), extracellular matrix components, and genes related to the TGF-ß/Smad signaling pathway. RESULTS: Conbercept significantly reduced TGF-ß2-induced EMT of SRA01/04 cells, decreased the expression levels of mesenchymal and extracellular matrix markers α-SMA, Snail, collagen I, collagen IV, and FN1, and upregulated the protein and mRNA expressions of E-cadherin (P <0.05). Transwell assay showed significantly lower cell migration ability in TGF-ß2+conbercept group than in TGF-ß2 group (P <0.05). Conbercept also inhibited the increase in Smad2/3 phosphorylation levels in HLEC-SRA01/04 cells with TGF-ß2-induced EMT (P <0.01). CONCLUSION: Conbercept inhibits TGF-ß2 induced EMT by downregulating the expression of pSmad2/3 in TGF-ß/Smad signaling pathway, indicating a potential therapeutic strategy against visual loss induced by posterior capsule opacification.


Asunto(s)
Proliferación Celular , Células Epiteliales , Transición Epitelial-Mesenquimal , Cristalino , Transducción de Señal , Proteínas Smad , Factor de Crecimiento Transformador beta2 , Humanos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Factor de Crecimiento Transformador beta2/metabolismo , Cristalino/citología , Cristalino/metabolismo , Proteínas Smad/metabolismo , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Cadherinas/metabolismo , Apoptosis/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular , Proteína Smad2/metabolismo
6.
Nat Cardiovasc Res ; 3(9): 1035-1048, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39232138

RESUMEN

Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide. Laminar shear stress from blood flow, sensed by vascular endothelial cells, protects from ASCVD by upregulating the transcription factors KLF2 and KLF4, which induces an anti-inflammatory program that promotes vascular resilience. Here we identify clustered γ-protocadherins as therapeutically targetable, potent KLF2 and KLF4 suppressors whose upregulation contributes to ASCVD. Mechanistic studies show that γ-protocadherin cleavage results in translocation of the conserved intracellular domain to the nucleus where it physically associates with and suppresses signaling by the Notch intracellular domain. γ-Protocadherins are elevated in human ASCVD endothelium; their genetic deletion or antibody blockade protects from ASCVD in mice without detectably compromising host defense against bacterial or viral infection. These results elucidate a fundamental mechanism of vascular inflammation and reveal a method to target the endothelium rather than the immune system as a protective strategy in ASCVD.


Asunto(s)
Aterosclerosis , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , Aterosclerosis/metabolismo , Aterosclerosis/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Humanos , Modelos Animales de Enfermedad , Transducción de Señal , Cadherinas/metabolismo , Cadherinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Masculino , Receptores Notch/metabolismo , Receptores Notch/genética , Proteínas Relacionadas con las Cadherinas , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología
7.
Nat Commun ; 15(1): 7734, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232006

RESUMEN

The adhesion receptor vascular endothelial (VE)-cadherin transduces an array of signals that modulate crucial lymphatic cell behaviors including permeability and cytoskeletal remodeling. Consequently, VE-cadherin must interact with a multitude of intracellular proteins to exert these functions. Yet, the full protein interactome of VE-cadherin in endothelial cells remains a mystery. Here, we use proximity proteomics to illuminate how the VE-cadherin interactome changes during junctional reorganization from dis-continuous to continuous junctions, triggered by the lymphangiogenic factor adrenomedullin. These analyses identified interactors that reveal roles for ADP ribosylation factor 6 (ARF6) and the exocyst complex in VE-cadherin trafficking and recycling. We also identify a requisite role for VE-cadherin in the in vitro and in vivo control of secretion of reelin-a lymphangiocrine glycoprotein with recently appreciated roles in governing heart development and injury repair. This VE-cadherin protein interactome shines light on mechanisms that control adherens junction remodeling and secretion from lymphatic endothelial cells.


Asunto(s)
Uniones Adherentes , Antígenos CD , Cadherinas , Células Endoteliales , Proteína Reelina , Animales , Humanos , Ratones , Uniones Adherentes/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/genética , Antígenos CD/metabolismo , Antígenos CD/genética , Cadherinas/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Uniones Intercelulares/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas , Proteómica/métodos , Serina Endopeptidasas/metabolismo
8.
Proc Natl Acad Sci U S A ; 121(37): e2400654121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39236238

RESUMEN

The Caenorhabditis elegans HMP-2/HMP-1 complex, akin to the mammalian [Formula: see text]-catenin-[Formula: see text]-catenin complex, serves as a critical mechanosensor at cell-cell adherens junctions, transducing tension between HMR-1 (also known as cadherin in mammals) and the actin cytoskeleton. Essential for embryonic development and tissue integrity in C. elegans, this complex experiences tension from both internal actomyosin contractility and external mechanical microenvironmental perturbations. While offering a valuable evolutionary comparison to its mammalian counterpart, the impact of tension on the mechanical stability of HMP-1 and HMP-2/HMP-1 interactions remains unexplored. In this study, we directly quantified the mechanical stability of full-length HMP-1 and its force-bearing modulation domains (M1-M3), as well as the HMP-2/HMP-1 interface. Notably, the M1 domain in HMP-1 exhibits significantly higher mechanical stability than its mammalian analog, attributable to interdomain interactions with M2-M3. Introducing salt bridge mutations in the M3 domain weakens the mechanical stability of the M1 domain. Moreover, the intermolecular HMP-2/HMP-1 interface surpasses its mammalian counterpart in mechanical stability, enabling it to support the mechanical activation of the autoinhibited M1 domain for mechanotransduction. Additionally, the phosphomimetic mutation Y69E in HMP-2 weakens the mechanical stability of the HMP-2/HMP-1 interface, compromising the force-transmission molecular linkage and its associated mechanosensing functions. Collectively, these findings provide mechanobiological insights into the C. elegans HMP-2/HMP-1 complex, highlighting the impact of salt bridges on mechanical stability in [Formula: see text]-catenin and demonstrating the evolutionary conservation of the mechanical switch mechanism activating the HMP-1 modulation domain for protein binding at the single-molecule level.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Mecanotransducción Celular , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Animales , Caenorhabditis elegans/metabolismo , Mecanotransducción Celular/fisiología , Imagen Individual de Molécula , Unión Proteica , Cadherinas/metabolismo , Cadherinas/química , Cadherinas/genética , Uniones Adherentes/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/química , Proteínas del Citoesqueleto , alfa Catenina
9.
Sci Rep ; 14(1): 21130, 2024 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256509

RESUMEN

Morphine has been suggested to affect cancer cell dynamics and decrease survival rates in lung cancer patients at specific doses, but the precise mechanisms poorly understood. In this study, we aimed to investigate the molecular mechanisms by which morphine modulates the malignant characteristics of non-small cell lung cancer. Cell proliferation was assessed via the Cell Counting Kit-8 assay, and cell migration and invasion were examined via wound healing and Transwell assays. We employed immunofluorescence staining to evaluate E-cadherin expression in A549 and Lewis lung cancer (LLC) cell lines and immunohistochemistry to evaluate E-cadherin expression in nude mice tumours. Additionally, the in vivo effects of morphine on lung cancer progression were explored in a xenograft tumour experiments, in which naloxone was used as a morphine antagonist. Western blot analysis was performed to detect E-cadherin, phosphorylated mTOR (p-mTOR), mTOR, phosphorylated AKT (p-AKT), AKT, phosphorylated PI3K (p-PI3K), and PI3K protein levels in A549 and LLC cells as well as in tumour samples. Morphine (10 µM) significantly increased the proliferation of A549 and LLC cells in vitro (p < 0.05). It also enhanced the migratory and invasive capacities of these cell lines (p < 0.01). Mechanistically, morphine treatment (10 µM) led to a reduction in the expression of E-cadherin, and an increase in the phosphorylation of PI3K, AKT, and mTOR in A549 and LLC cells (p < 0.01). Morphine treatment (1.5 mg/kg) also reduced E-cadherin expression in xenograft tumours and promoted tumour growth in vivo (p < 0.05). This effect was reversed by naloxone (0.1 mg/kg). The results demonstrated that morphine stimulates the malignant proliferation of A549 and LLC cell lines and promotes xenograft tumour growth. Perhaps by specifically targeting MOR, morphine triggers a signalling cascade that activates the PI3K/AKT/mTOR pathway while inhibiting the EMT marker E-cadherin, which may consequently promote the progression of lung cancer.


Asunto(s)
Cadherinas , Carcinoma de Pulmón de Células no Pequeñas , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares , Morfina , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Cadherinas/metabolismo , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Morfina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Ratones , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ratones Desnudos , Células A549 , Progresión de la Enfermedad , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación hacia Abajo/efectos de los fármacos , Naloxona/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino
10.
Breast Cancer Res ; 26(1): 131, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256827

RESUMEN

BACKGROUND: Breast cancer is the second leading cause of death in women, with invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) as the two most common forms of invasive breast cancer. While estrogen receptor positive (ER+) IDC and ILC are treated similarly, the multifocality of ILC presents challenges in detection and treatment, worsening long-term clinical outcomes in patients. With increasing documentation of chemoresistance in ILC, additional treatment options are needed. Oncolytic adenoviral therapy may be a promising option, but cancer cells must express the coxsackievirus & adenovirus receptor (CAR) for adenoviral therapy to be effective. The present study aims to evaluate the extent to which CAR expression is observed in ILC in comparison to IDC, and how the levels of CAR expression correlate with adenovirus transduction efficiency. The effect of liposome encapsulation on transduction efficiency is also assessed. METHODS: To characterize CAR expression in invasive breast carcinoma, 36 formalin-fixed paraffin-embedded (FFPE) human breast tumor samples were assayed by CAR immunohistochemistry (IHC). Localization of CAR in comparison to other junctional proteins was performed using a multiplex immunofluorescence panel consisting of CAR, p120-catenin, and E-cadherin. ILC and IDC primary tumors and cell lines were transduced with E1- and E3-deleted adenovirus type 5 inserted with a GFP transgene (Ad-GFP) and DOTAP liposome encapsulated Ad-GFP (DfAd-GFP) at various multiplicities of infection (MOIs). Transduction efficiency was measured using a fluorescence plate reader. CAR expression in the human primary breast carcinomas and cell lines was also evaluated by IHC. RESULTS: We observed membranous CAR, p120-catenin and E-cadherin expression in IDC. In ILC, we observed cytoplasmic expression of CAR and p120-catenin, with absent E-cadherin. Adenovirus effectively transduced high-CAR IDC cell lines, at MOIs as low as 12.5. Ad-GFP showed similar transduction as DfAd-GFP in high-CAR IDC cell lines. Conversely, Ad-GFP transduction of ILC cell lines was observed only at MOIs of 50 and 100. Furthermore, Ad-GFP did not transduce CAR-negative IDC cell lines even at MOIs greater than 100. Liposome encapsulation (DfAd-GFP) improved transduction efficiency 4-fold in ILC and 17-fold in CAR-negative IDC cell lines. CONCLUSION: The present study demonstrates that oncolytic adenoviral therapy is less effective in ILC than IDC due to differences in spatial CAR expression. Liposome-enhanced delivery may be beneficial for patients with ILC and tumors with low or negative CAR expression to improve adenoviral therapeutic effectiveness.


Asunto(s)
Adenoviridae , Neoplasias de la Mama , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Transducción Genética , Humanos , Femenino , Neoplasias de la Mama/terapia , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adenoviridae/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Línea Celular Tumoral , Carcinoma Lobular/metabolismo , Carcinoma Lobular/terapia , Carcinoma Lobular/genética , Carcinoma Lobular/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patología , Carcinoma Ductal de Mama/terapia , Cadherinas/metabolismo , Cadherinas/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Liposomas
11.
Braz J Med Biol Res ; 57: e13486, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39258668

RESUMEN

Silicosis is a systemic disease caused by long-term exposure to high concentrations of free silica dust particles in the workplace. It is characterized by a persistent inflammatory response, fibroblast proliferation, and excessive collagen deposition, leading to pulmonary interstitial fibrosis. Epithelial interstitial transformation (EMT) can cause epithelial cells to lose their tight junctions, cell polarity, and epithelial properties, thereby enhancing the properties of interstitial cells, which can lead to the progression of fibrosis and the formation of scar tissue. Integrin 1 (ITGB1) is considered an important factor for promoting EMT and tumor invasion in a variety of tumors and also plays an important role in the progression of fibrotic diseases. Therefore, ITGB1 can be used as a potential target for the treatment of silicosis. In this study, we found that silica exposure induced epithelial-mesenchymal transformation in rats and that the expression of integrin ITGB1 was elevated along with the EMT. We used CRISPR/Cas9 technology to construct integrin ITGB1 knockdown cell lines for in vitro experiments. We compared the expression of the EMT key proteins E-cadherin and vimentin in the ITGB1 knockdown cells and wild-type cells simultaneously stimulated by silica and detected the aggregation point distribution of E-cadherin and vimentin in the cells using laser confocal microscopy. Our results showed that ITGB1 knockout inhibited the ITGB1/ILK/Snail signaling pathway and attenuated the EMT occurrence compared to control cells. These results suggested that ITGB1 is associated with silica-induced EMT and may be a potential target for the treatment of silicosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Integrina beta1 , Fibrosis Pulmonar , Dióxido de Silicio , Animales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Dióxido de Silicio/toxicidad , Dióxido de Silicio/efectos adversos , Integrina beta1/genética , Integrina beta1/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Ratas , Silicosis/patología , Silicosis/genética , Masculino , Cadherinas/metabolismo , Cadherinas/genética
12.
Nat Commun ; 15(1): 7865, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256406

RESUMEN

Hair cell bundles consist of stereocilia arranged in rows of increasing heights, connected by tip links that transmit sound-induced forces to shorter stereocilia tips. Auditory mechanotransduction channel complexes, composed of proteins TMC1/2, TMIE, CIB2, and LHFPL5, are located at the tips of shorter stereocilia. While most components can interact with the tip link in vitro, their ability to maintain the channel complexes at the tip link in vivo is uncertain. Return, using mouse models, we show that an additional component, LOXHD1, is essential for keeping TMC1-pore forming subunits at the tip link but is dispensable for TMC2. Using SUB-immunogold-SEM, we showed that TMC1 localizes near the tip link but mislocalizes without LOXHD1. LOXHD1 selectively interacts with TMC1, CIB2, LHFPL5, and tip-link protein PCDH15. Our results demonstrate that TMC1-driven mature auditory channels require LOXHD1 to stay connected to the tip link and remain functional, while TMC2-driven developmental channels do not.


Asunto(s)
Mecanotransducción Celular , Proteínas de la Membrana , Animales , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiología , Estereocilios/metabolismo , Cadherinas/metabolismo , Cadherinas/genética , Proteínas Relacionadas con las Cadherinas , Ratones Noqueados , Femenino , Masculino , Ratones Endogámicos C57BL , Precursores de Proteínas
13.
Biol Direct ; 19(1): 76, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39238003

RESUMEN

Moyamoya disease, characterized by basal cerebral artery obstruction, was studied for differential protein expression to elucidate its pathogenesis. Proteomic analysis of cerebrospinal fluid from 10 patients, categorized by postoperative angiography into good and poor prognosis groups, revealed 46 differentially expressed proteins. Notably, cadherin 18 (CDH18) was the most significantly upregulated in the good prognosis group. In addition, the expression of cadherin 18 (CDH18) and phenotypic transformation-related proteins were measured by qRT-PCR and western blot. The effects of CDH18 in vascular smooth muscle cells were detected by CCK-8, EdU, transwell and wound healing assays. The overexpression of CDH18 in vascular smooth muscle cells (VSMCs) was found to inhibit proliferation, migration, and phenotypic transformation. These findings suggest CDH18 as a potential therapeutic target in moyamoya disease.


Asunto(s)
Angiografía de Substracción Digital , Cadherinas , Enfermedad de Moyamoya , Proteómica , Enfermedad de Moyamoya/genética , Enfermedad de Moyamoya/metabolismo , Humanos , Proteómica/métodos , Cadherinas/metabolismo , Cadherinas/genética , Masculino , Proliferación Celular , Femenino , Movimiento Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Adulto , Persona de Mediana Edad
14.
Pathol Res Pract ; 262: 155562, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39182448

RESUMEN

Mesothelin (MSLN) is expressed in the mesothelium in normal tissues but is overexpressed in various malignant tumors. In this study, we searched for genes that were more frequently expressed in cases of endometrioid carcinoma (EC) with the MELF (microcystic, elongated, and fragmented) pattern using laser microdissection and RNA sequencing, and found that MSLN was predominantly expressed in cases with the MELF pattern. The role of MSLN in EC was analyzed by generating MSLN-knockout and -knockdown EC cell lines. MSLN promoted migration and epithelial-mesenchymal transition (EMT). Moreover, we found that cadherin-6 (CDH6) expression was regulated by MSLN. MSLN is known to bind to cancer antigen 125 (CA125), and we found that CA125 can regulate CDH6 expression via MSLN. Immunohistochemical investigations showed that MSLN, CA125, and CDH6 expression levels were considerably elevated in EC with the MELF pattern. The expression of CA125 was similar to that of MSLN not only in terms of immunohistochemical staining intensity but also the blood level of CA125. Our results showed that MSLN contributes to the migration and EMT of EC cells through upstream CA125 and downstream CDH6. Therefore, MSLN has potential as a therapeutic target for EC with the MELF pattern.


Asunto(s)
Cadherinas , Carcinoma Endometrioide , Movimiento Celular , Transición Epitelial-Mesenquimal , Proteínas Ligadas a GPI , Mesotelina , Humanos , Femenino , Proteínas Ligadas a GPI/metabolismo , Carcinoma Endometrioide/patología , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/genética , Transición Epitelial-Mesenquimal/fisiología , Cadherinas/metabolismo , Antígeno Ca-125/metabolismo , Neoplasias Endometriales/patología , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/genética , Línea Celular Tumoral , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Persona de Mediana Edad , Proteínas de la Membrana
15.
PLoS One ; 19(8): e0309536, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39208202

RESUMEN

Biomarkers for classifying and grading gliomas have been extensively explored, whereas populations in public databases were mostly Western/European. Based on public databases cannot accurately represent Chinese population. To identify molecular characteristics associated with clinical outcomes of lower-grade glioma (LGG) and glioblastoma (GBM) in the Chinese population, we performed whole-exome sequencing (WES) in 16 LGG and 35 GBM tumor tissues. TP53 (36/51), TERT (31/51), ATRX (16/51), EFGLAM (14/51), and IDH1 (13/51) were the most common genes harboring mutations. IDH1 mutation (c.G395A; p.R132H) was significantly enriched in LGG, whereas PCDHGA10 mutation (c.A265G; p.I89V) in GBM. IDH1-wildtype and PCDHGA10 mutation were significantly related to poor prognosis. IDH1 is an important biomarker in gliomas, whereas PCDHGA10 mutation has not been reported to correlate with gliomas. Different copy number variations (CNVs) and oncogenic signaling pathways were identified between LGG and GBM. Differential genomic landscapes between LGG and GBM were revealed in the Chinese population, and PCDHGA10, for the first time, was identified as the prognostic factor of gliomas. Our results might provide a basis for molecular classification and identification of diagnostic biomarkers and even potential therapeutic targets for gliomas.


Asunto(s)
Neoplasias Encefálicas , Variaciones en el Número de Copia de ADN , Glioblastoma , Glioma , Isocitrato Deshidrogenasa , Mutación , Humanos , Glioblastoma/genética , Glioblastoma/patología , Masculino , Femenino , Isocitrato Deshidrogenasa/genética , Glioma/genética , Glioma/patología , Persona de Mediana Edad , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Secuenciación del Exoma , Telomerasa/genética , Biomarcadores de Tumor/genética , Clasificación del Tumor , Cadherinas/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína p53 Supresora de Tumor/genética , Anciano , Pronóstico , Genómica/métodos , Protocadherinas , Adulto Joven
16.
Nat Cardiovasc Res ; 3(7): 799-818, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39196173

RESUMEN

Pulmonary veno-occlusive disease (PVOD) is a rare form of pulmonary hypertension arising from EIF2AK4 gene mutations or mitomycin C (MMC) administration. The lack of effective PVOD therapies is compounded by a limited understanding of the mechanisms driving vascular remodeling in PVOD. Here we show that administration of MMC in rats mediates activation of protein kinase R (PKR) and the integrated stress response (ISR), which leads to the release of the endothelial adhesion molecule vascular endothelial (VE) cadherin (VE-Cad) in complex with RAD51 to the circulation, disruption of endothelial barrier and vascular remodeling. Pharmacological inhibition of PKR or ISR attenuates VE-Cad depletion, elevation of vascular permeability and vascular remodeling instigated by MMC, suggesting potential clinical intervention for PVOD. Finally, the severity of PVOD phenotypes was increased by a heterozygous BMPR2 mutation that truncates the carboxyl tail of the receptor BMPR2, underscoring the role of deregulated bone morphogenetic protein signaling in the development of PVOD.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Modelos Animales de Enfermedad , Fenotipo , Enfermedad Veno-Oclusiva Pulmonar , Animales , Enfermedad Veno-Oclusiva Pulmonar/genética , Enfermedad Veno-Oclusiva Pulmonar/tratamiento farmacológico , Enfermedad Veno-Oclusiva Pulmonar/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Remodelación Vascular/efectos de los fármacos , Cadherinas/genética , Cadherinas/metabolismo , Humanos , Masculino , Antígenos CD/metabolismo , Antígenos CD/genética , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Mutación , Permeabilidad Capilar/efectos de los fármacos , Ratas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
17.
Int J Mol Sci ; 25(16)2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39201647

RESUMEN

Invasive lobular carcinoma exhibits unique morphological features frequently associated with alterations in CDH1. Although some studies have identified abnormalities in adhesion factors other than E-cadherin, the molecular mechanisms underlying E-cadherin abnormalities in CDH1-unaltered invasive lobular carcinoma remain poorly understood. In this study, we investigated the molecular underpinnings of E-cadherin dysregulation in invasive lobular carcinoma in the absence of CDH1 gene alterations, using comprehensive bioinformatic analyses. We conducted a comparative study of CDH1-mutated and non-mutated invasive lobular carcinoma and evaluated the differences in mRNA levels, reverse-phase protein array, methylation, and miRNAs. We observed that invasive lobular carcinoma cases without CDH1 alterations exhibited a significantly higher incidence of the Claudin-low subtype (p < 0.01). The results of the reverse-phase protein array indicate no significant difference in E-cadherin expression between CDH1-mutated and non-mutated cases. Therefore, abnormalities in E-cadherin production also exist in CDH1 non-mutated invasive lobular carcinoma. Considering that there are no differences in mRNA levels and methylation status, post-translational modifications are the most plausible explanation for the same. Hence, future studies should focus on elucidating the mechanism underlying E-cadherin inactivation via post-translational modifications in CDH1 non-mutated invasive lobular carcinoma.


Asunto(s)
Antígenos CD , Neoplasias de la Mama , Cadherinas , Carcinoma Lobular , Biología Computacional , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma Lobular/genética , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patología , Humanos , Antígenos CD/metabolismo , Antígenos CD/genética , Biología Computacional/métodos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Mutación , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Invasividad Neoplásica
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(4): 987-992, 2024 Aug.
Artículo en Chino | MEDLINE | ID: mdl-39192387

RESUMEN

OBJECTIVE: To investigate the changes in serum homeobox A9 (HOXA9 ), soluble E-cadherin (SE-CAD) and type Ⅲ procollagen (PCⅢ) levels in acute myeloid leukemia (AML) patients after chemotherapy with DCAG regimen and their relationship with prognosis. METHODS: The clinical data of 80 patients with relapsed/refractory AML diagnosed and treated in our hospital from March 2018 to December 2021 were retrospectively analyzed. According to different treatment regimen, the patients were divided into DCAG group (n=40) and CAG group (n=40). The clinical efficacy and changes of HOXA9 , SE-CAD and PCⅢ levels before and after treatment were compared. In addition, all patients were divided into remission group (n=58) and non-remission group (n=22) according to the clinical efficacy. Univariate and multivariate analyses were performed to analyze the risk factors affecting the prognosis of AML patients. The predictive efficacy of the three single indicators, HOXA9 , SE-CAD, and PC III, and their combination on prognosis was analyzed. RESULTS: Compared with before treatment, the levels of HOXA9 , SE-CAD and PCⅢ in both the DCAG and CAG groups were decreased after treatment, and the improvement of each indicator and the clinical efficacy in the DCAG group were significantly better than those in the CAG group (all P < 0.05). Multivariate analysis showed that increased bone marrow blast count, HOXA9 mRNA, SE-CAD and PCⅢ levels were independent risk factors affecting the efficacy of chemotherapy in AML patients (all P < 0.05). ROC curves showed that the combination of HOXA9 mRNA, SE-CAD and PCIII could effectively predict the prognosis of AML patients, with a sensitivity of 84.80% and a specificity of 88.20%. CONCLUSION: DCAG regimen can significantly improve the levels of HOXA9 mRNA, SE-CAD and PCⅢ in AML patients, these three indicators are all independent risk factors affecting the prognosis of AML patients, and the combination of the three indicators can effectively predict the prognosis of the patients.


Asunto(s)
Cadherinas , Proteínas de Homeodominio , Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Pronóstico , Estudios Retrospectivos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Masculino , Femenino
19.
Mar Drugs ; 22(8)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39195457

RESUMEN

Tight junctional complexes (TJCs) between cerebral microvascular endothelial cells (CMECs) are essential parts of the blood-brain barrier (BBB), whose regulation closely correlates to the BBB's integrity and function. hCMEC/D3 is the typical cell line used to imitate and investigate the barrier function of the BBB via the construction of an in vitro model. This study aims to investigate the protective effect of the deep-sea-derived fibrinolytic compound FGFC1 against H2O2-induced dysfunction of TJCs and to elucidate the underlying mechanism. The barrier function was shown to decline following exposure to 1 mM H2O2 in an in vitro model of hCMEC/D3 cells, with a decreasing temperature-corrected transendothelial electrical resistance (tcTEER) value. The decrease in the tcTEER value was significantly inhibited by 80 or 100 µM FGFC1, which suggested it efficiently protected the barrier integrity, allowing it to maintain its function against the H2O2-induced dysfunction. According to immunofluorescence microscopy (IFM) and quantitative real-time polymerase chain reaction (qRT-PCR), compared to the H2O2-treated group, 80~100 µM FGFC1 enhanced the expression of claudin-5 (CLDN-5) and VE-cadherin (VE-cad). And this enhancement was indicated to be mainly achieved by both up-regulation of CLDN-5 and inhibition of the down-regulation by H2O2 of VE-cad at the transcriptional level. Supported by FGFC1's molecular docking to these proteins with reasonable binding energy, FGFC1 was proved to exert a positive effect on TJCs' barrier function in hCMEC/D3 cells via targeting CLDN-5 and VE-cad. This is the first report on the protection against H2O2-induced barrier dysfunction by FGFC1 in addition to its thrombolytic effect. With CLDN-5 and VE-cad as the potential target proteins of FGFC1, this study provides evidence at the cellular and molecular levels for FGFC1's reducing the risk of bleeding transformation following its application in thrombolytic therapy for cerebral thrombosis.


Asunto(s)
Cadherinas , Células Endoteliales , Peróxido de Hidrógeno , Uniones Estrechas , Humanos , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Línea Celular , Peróxido de Hidrógeno/toxicidad , Peróxido de Hidrógeno/farmacología , Cadherinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Fibrinolíticos/farmacología , Claudina-5/metabolismo , Antígenos CD/metabolismo , Simulación del Acoplamiento Molecular , Factores de Crecimiento de Fibroblastos/farmacología
20.
BMC Ophthalmol ; 24(1): 373, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39187782

RESUMEN

BACKGROUND: Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal diseases. However, it is still not well understand about the relationship between PCDH15 variants and RP. METHODS: In this study, we enrolled a Chinese autosomal recessive retinitis pigmentosa (arRP) pedigree and identified the causative gene in the proband by targeted whole exome sequencing (WES). The variants were validated in the family members by Sanger sequencing and co-segregation analysis. RESULTS: Novel compound heterozygous, Frame shift variants of the PCDH15 gene, NM_001384140.1:c.4368 - 2147_4368-2131del and NM_001384140.1:c exon19:c.2505del: p. T836Lfs*6 were identified in the arRP pedigree, which co-segregated with the clinical RP phenotypes. The PCDH15 protein is highly conserved among species. CONCLUSION: This is the first study to identify novel compound heterozygous variants c.4368 - 2147_4368-2131del and c.2505del(p.T836Lfs*6) in the PCDH15 gene which might be disease-causing variants, and extending the variant spectra. All above findings may be contribute to genetic counseling, molecular diagnosis and clinical management of arRP disease.


Asunto(s)
Proteínas Relacionadas con las Cadherinas , Cadherinas , Heterocigoto , Linaje , Retinitis Pigmentosa , Humanos , Masculino , Femenino , Cadherinas/genética , Retinitis Pigmentosa/genética , Adulto , China/epidemiología , Secuenciación del Exoma , Análisis Mutacional de ADN , Pueblo Asiatico/genética , Fenotipo , Persona de Mediana Edad , Pueblos del Este de Asia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA