Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.303
Filtrar
1.
Nat Commun ; 15(1): 7963, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261453

RESUMEN

Coagulation factor XII (FXII) conveys various functions as an active protease that promotes thrombosis and inflammation, and as a zymogen via surface receptors like urokinase-type plasminogen activator receptor (uPAR). While plasma levels of FXII are increased in diabetes mellitus and diabetic kidney disease (DKD), a pathogenic role of FXII in DKD remains unknown. Here we show that FXII is locally expressed in kidney tubular cells and that urinary FXII correlates with kidney dysfunction in DKD patients. F12-deficient mice (F12-/-) are protected from hyperglycemia-induced kidney injury. Mechanistically, FXII interacts with uPAR on tubular cells promoting integrin ß1-dependent signaling. This signaling axis induces oxidative stress, persistent DNA damage and senescence. Blocking uPAR or integrin ß1 ameliorates FXII-induced tubular cell injury. Our findings demonstrate that FXII-uPAR-integrin ß1 signaling on tubular cells drives senescence. These findings imply previously undescribed diagnostic and therapeutic approaches to detect or treat DKD and possibly other senescence-associated diseases.


Asunto(s)
Senescencia Celular , Nefropatías Diabéticas , Factor XII , Integrina beta1 , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Animales , Femenino , Humanos , Masculino , Ratones , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/genética , Factor XII/metabolismo , Factor XII/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal
2.
Sci Adv ; 10(36): eadk2252, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39231227

RESUMEN

Primordial germ cells (PGCs) are the precursors of gametes and the sole mechanism by which animals transmit genetic information across generations. In the mouse embryo, the transcriptional and epigenetic regulation of PGC specification has been extensively characterized. However, the initial event that triggers the soma-germline segregation remains poorly understood. Here, we uncover a critical role for the basement membrane in regulating germline entry. We show that PGCs arise in a region of the mouse embryo that lacks contact with the basement membrane, and the addition of exogenous extracellular matrix (ECM) inhibits both PGC and PGC-like cell (PGCLC) specification in mouse embryos and stem cell models, respectively. Mechanistically, we demonstrate that the engagement of ß1 integrin with laminin blocks PGCLC specification by preventing the Wnt signaling-dependent down-regulation of the PGC transcriptional repressor, Otx2. In this way, the physical segregation of cells away from the basement membrane acts as a morphogenetic fate switch that controls the soma-germline bifurcation.


Asunto(s)
Células Germinativas , Células Madre Pluripotentes , Animales , Ratones , Células Germinativas/metabolismo , Células Germinativas/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Integrinas/metabolismo , Integrinas/genética , Membrana Basal/metabolismo , Vía de Señalización Wnt , Diferenciación Celular , Matriz Extracelular/metabolismo , Laminina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/metabolismo , Integrina beta1/genética , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología
3.
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
4.
Exp Cell Res ; 442(1): 114194, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39127440

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the role and possible mechanism of lncRNA XIST in renal fibrosis and to provide potential endogenous targets for renal fibrosis in obstructive nephropathy (ON). METHODS: The study included 50 cases of ON with renal fibrosis (samples taken from patients undergoing nephrectomy due to ON) and 50 cases of normal renal tissue (samples taken from patients undergoing total or partial nephrectomy due to accidental injury, congenital malformations, and benign tumors). Treatment of human proximal renal tubular epithelium (HK-2) cells with TGF-ß1 simulated renal fibrosis in vitro. Cell viability and proliferation were measured by CCK-8 and EdU, and cell migration was measured by transwell. XIST, miR-124-3p, ITGB1, and epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, α-SMA, and fibronectin) were detected by PCR and immunoblot. The targeting relationship between miR-124-3p and XIST or ITGB1 was verified by starBase and dual luciferase reporter gene experiments. In addition, The left ureter was ligated in mice as a model of unilateral ureteral obstruction (UUO), and the renal histopathology was observed by HE staining and Masson staining. RESULTS: ON patients with renal fibrosis had elevated XIST and ITGB1 levels and reduced miR-124-3p levels. The administration of TGF-ß1 exhibited a dose-dependent promotion of HK-2 cell viability, proliferation, migration, and EMT. Conversely, depleting XIST or enhancing miR-124-3p hindered HK-2 cell viability, proliferation, migration, and EMT in TGF-ß1-damaged HK-2 cells HK-2 cells. XIST functioned as a miR-124-3p sponge. Additionally, miR-124-3p negatively regulated ITGB1 expression. Elevating ITGB1 weakened the impact of XIST depletion on TGF-ß1-damaged HK-2 cells. Down-regulating XIST improved renal fibrosis in UUO mice. CONCLUSION: XIST promotes renal fibrosis in ON by elevating miR-124-3p and reducing ITGB1 expressions.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibrosis , Enfermedades Renales , MicroARNs , ARN Largo no Codificante , ARN Largo no Codificante/genética , MicroARNs/genética , MicroARNs/metabolismo , Humanos , Fibrosis/genética , Fibrosis/patología , Fibrosis/metabolismo , Animales , Ratones , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Transición Epitelial-Mesenquimal/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Proliferación Celular , Masculino , Movimiento Celular/genética , Riñón/patología , Riñón/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Línea Celular , Femenino , Obstrucción Ureteral/patología , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/genética
5.
J Clin Invest ; 134(15)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-39087467

RESUMEN

The blood-brain barrier (BBB) acquires unique properties to regulate neuronal function during development. The formation of the BBB, which occurs in tandem with angiogenesis, is directed by the Wnt/ß-catenin signaling pathway. Yet the exact molecular interplay remains elusive. Our study reveals the G protein-coupled receptor GPR126 as a critical target of canonical Wnt signaling, essential for the development of the BBB's distinctive vascular characteristics and its functional integrity. Endothelial cell-specific deletion of the Gpr126 gene in mice induced aberrant vascular morphogenesis, resulting in disrupted BBB organization. Simultaneously, heightened transcytosis in vitro compromised barrier integrity, resulting in enhanced vascular permeability. Mechanistically, GPR126 enhanced endothelial cell migration, pivotal for angiogenesis, acting through an interaction between LRP1 and ß1 integrin, thereby balancing the levels of ß1 integrin activation and recycling. Overall, we identified GPR126 as a specifier of an organotypic vascular structure, which sustained angiogenesis and guaranteed the acquisition of the BBB properties during development.


Asunto(s)
Barrera Hematoencefálica , Integrina beta1 , Receptores Acoplados a Proteínas G , Animales , Ratones , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Movimiento Celular , Células Endoteliales/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones Noqueados , Neovascularización Fisiológica , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Vía de Señalización Wnt , Masculino , Femenino
6.
J Cell Biol ; 223(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-38990714

RESUMEN

Dermal fibroblasts deposit type I collagen, the dominant extracellular matrix molecule found in skin, during early postnatal development. Coincident with this biosynthetic program, fibroblasts proteolytically remodel pericellular collagen fibrils by mobilizing the membrane-anchored matrix metalloproteinase, Mmp14. Unexpectedly, dermal fibroblasts in Mmp14-/- mice commit to a large-scale apoptotic program that leaves skin tissues replete with dying cells. A requirement for Mmp14 in dermal fibroblast survival is recapitulated in vitro when cells are embedded within, but not cultured atop, three-dimensional hydrogels of crosslinked type I collagen. In the absence of Mmp14-dependent pericellular proteolysis, dermal fibroblasts fail to trigger ß1 integrin activation and instead actuate a TGF-ß1/phospho-JNK stress response that leads to apoptotic cell death in vitro as well as in vivo. Taken together, these studies identify Mmp14 as a requisite cell survival factor that maintains dermal fibroblast viability in postnatal dermal tissues.


Asunto(s)
Apoptosis , Supervivencia Celular , Fibroblastos , Metaloproteinasa 14 de la Matriz , Animales , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Dermis/metabolismo , Dermis/citología , Células Cultivadas , Matriz Extracelular/metabolismo , Ratones Endogámicos C57BL , Piel/metabolismo
7.
Drug Resist Updat ; 76: 101116, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38968684

RESUMEN

Drug resistance and tumor recurrence remain clinical challenges in the treatment of urothelial carcinoma (UC). However, the underlying mechanism is not fully understood. Here, we performed single-cell RNA sequencing and identified a subset of urothelial cells with epithelial-mesenchymal transition (EMT) features (EMT-UC), which is significantly correlated with chemotherapy resistance and cancer recurrence. To validate the clinical significance of EMT-UC, we constructed EMT-UC like cells by introducing overexpression of two markers, Zinc Finger E-Box Binding Homeobox 1 (ZEB1) and Desmin (DES), and examined their histological distribution characteristics and malignant phenotypes. EMT-UC like cells were mainly enriched in UC tissues from patients with adverse prognosis and exhibited significantly elevated EMT, migration and gemcitabine tolerance in vitro. However, EMT-UC was not specifically identified from tumorous tissues, certain proportion of them were also identified in adjacent normal tissues. Tumorous EMT-UC highly expressed genes involved in malignant behaviors and exhibited adverse prognosis. Additionally, tumorous EMT-UC was associated with remodeled tumor microenvironment (TME), which exhibited high angiogenic and immunosuppressive potentials compared with the normal counterparts. Furthermore, a specific interaction of COL4A1 and ITGB1 was identified to be highly enriched in tumorous EMT-UC, and in the endothelial component. Targeting the interaction of COL4A1 and ITGB1 with specific antibodies significantly suppressed tumorous angiogenesis and alleviated gemcitabine resistance of UC. Overall, our findings demonstrated that the driven force of chemotherapy resistance and recurrence of UC was EMT-UC mediated COL4A1-ITGB1 interaction, providing a potential target for future UC treatment.


Asunto(s)
Colágeno Tipo IV , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Integrina beta1 , Recurrencia Local de Neoplasia , Neovascularización Patológica , Neoplasias de la Vejiga Urinaria , Humanos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Gemcitabina/farmacología , Gemcitabina/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Integrina beta1/metabolismo , Integrina beta1/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neovascularización Patológica/genética , Pronóstico , Microambiente Tumoral/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/irrigación sanguínea , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Urotelio/irrigación sanguínea , Urotelio/efectos de los fármacos , Urotelio/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
8.
Signal Transduct Target Ther ; 9(1): 169, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38956074

RESUMEN

More than 90% of hepatocellular carcinoma (HCC) cases develop in the presence of fibrosis or cirrhosis, making the tumor microenvironment (TME) of HCC distinctive due to the intricate interplay between cancer-associated fibroblasts (CAFs) and cancer stem cells (CSCs), which collectively regulate HCC progression. However, the mechanisms through which CSCs orchestrate the dynamics of the tumor stroma during HCC development remain elusive. Our study unveils a significant upregulation of Sema3C in fibrotic liver, HCC tissues, peripheral blood of HCC patients, as well as sorafenib-resistant tissues and cells, with its overexpression correlating with the acquisition of stemness properties in HCC. We further identify NRP1 and ITGB1 as pivotal functional receptors of Sema3C, activating downstream AKT/Gli1/c-Myc signaling pathways to bolster HCC self-renewal and tumor initiation. Additionally, HCC cells-derived Sema3C facilitated extracellular matrix (ECM) contraction and collagen deposition in vivo, while also promoting the proliferation and activation of hepatic stellate cells (HSCs). Mechanistically, Sema3C interacted with NRP1 and ITGB1 in HSCs, activating downstream NF-kB signaling, thereby stimulating the release of IL-6 and upregulating HMGCR expression, consequently enhancing cholesterol synthesis in HSCs. Furthermore, CAF-secreted TGF-ß1 activates AP1 signaling to augment Sema3C expression in HCC cells, establishing a positive feedback loop that accelerates HCC progression. Notably, blockade of Sema3C effectively inhibits tumor growth and sensitizes HCC cells to sorafenib in vivo. In sum, our findings spotlight Sema3C as a novel biomarker facilitating the crosstalk between CSCs and stroma during hepatocarcinogenesis, thereby offering a promising avenue for enhancing treatment efficacy and overcoming drug resistance in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Semaforinas , Microambiente Tumoral , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Microambiente Tumoral/genética , Semaforinas/genética , Semaforinas/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Ratones , Transducción de Señal/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Neuropilina-1/genética , Neuropilina-1/metabolismo , Línea Celular Tumoral , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica/genética , Sorafenib/farmacología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Progresión de la Enfermedad
9.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000184

RESUMEN

Microglia migrate to the cerebral cortex during early embryonic stages. However, the precise mechanisms underlying microglia migration remain incompletely understood. As an extracellular matrix protein, Netrin-1 is involved in modulating the motility of diverse cells. In this paper, we found that Netrin-1 promoted microglial BV2 cell migration in vitro. Mechanism studies indicated that the activation of GSK3ß activity contributed to Netrin-1-mediated microglia migration. Furthermore, Integrin α6/ß1 might be the relevant receptor. Single-cell data analysis revealed the higher expression of Integrin α6 subunit and ß1 subunit in microglia in comparison with classical receptors, including Dcc, Neo1, Unc5a, Unc5b, Unc5c, Unc5d, and Dscam. Microscale thermophoresis (MST) measurement confirmed the high binding affinity between Integrin α6/ß1 and Netrin-1. Importantly, activation of Integrin α6/ß1 with IKVAV peptides mirrored the microglia migration and GSK3 activation induced by Netrin-1. Finally, conditional knockout (CKO) of Netrin-1 in radial glial cells and their progeny led to a reduction in microglia population in the cerebral cortex at early developmental stages. Together, our findings highlight the role of Netrin-1 in microglia migration and underscore its therapeutic potential in microglia-related brain diseases.


Asunto(s)
Movimiento Celular , Microglía , Netrina-1 , Netrina-1/metabolismo , Netrina-1/genética , Microglía/metabolismo , Animales , Ratones , Ratones Noqueados , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Línea Celular , Integrina beta1/metabolismo , Integrina beta1/genética
10.
Int Immunopharmacol ; 136: 112368, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38823175

RESUMEN

Silicosis is a chronic fibroproliferative lung disease caused by long-term inhalation of crystalline silica dust, characterized by the proliferation of fibroblasts and pulmonary interstitial fibrosis. Currently, there are no effective treatments available. Recent research suggests that the Integrin ß1/ILK/PI3K signaling pathway may be associated with the pathogenesis of silicosis fibrosis. In this study, we investigated the effects of Echistatin (Integrin ß1 inhibitor) and BYL-719 (PI3K inhibitor) on silicosis rats at 28 and 56 days after silica exposure. Histopathological analysis of rat lung tissue was performed using H&E staining and Masson staining. Immunohistochemistry, Western blotting, and qRT-PCR were employed to assess the expression of markers associated with epithelial-mesenchymal transition (EMT), fibrosis, and the Integrin ß1/ILK/PI3K pathway in lung tissue. The results showed that Echistatin, BYL 719 or their combination up-regulated the expression of E-cadherin and down-regulated the expression of Vimentin and extracellular matrix (ECM) components, including type I and type III collagen. The increase of Snail, AKT and ß-catenin in the downstream Integrin ß1/ILK/PI3K pathway was inhibited. These results indicate that Echistatin and BYL 719 can inhibit EMT and pulmonary fibrosis by blocking different stages of Integrinß1 /ILK/PI3K signaling pathway. This indicates that the Integrin ß1/ILK/PI3K signaling pathway is associated with silica-induced EMT and may serve as a potential therapeutic target for silicosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Integrina beta1 , Fosfatidilinositol 3-Quinasas , Proteínas Serina-Treonina Quinasas , Fibrosis Pulmonar , Transducción de Señal , Dióxido de Silicio , Silicosis , Animales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Integrina beta1/metabolismo , Integrina beta1/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Masculino , Dióxido de Silicio/toxicidad , Silicosis/metabolismo , Silicosis/patología , Silicosis/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Pulmón/patología , Pulmón/efectos de los fármacos , Ratas Sprague-Dawley
11.
Int J Biol Macromol ; 273(Pt 1): 133074, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38866293

RESUMEN

Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer, has a poor prognosis and limited access to efficient targeted treatments. Chronic unpredictable mild stress (CUMS) is highly risk factor for TNBC occurrence and development. Type X collagen (COL10A1), a crucial protein component of the extracellular matrix, ranks second among all aberrantly expressed genes in TNBC, and it is significantly up-regulated under CUMS. Nevertheless, the impact of CUMS and COL10A1 on TNBC, along with the underlying mechanisms are still unclear. In this research, we studied the effect of CUMS-induced norepinephrine (NE) elevation on TNBC, and uncovered that it notably enhanced TNBC cell proliferation, migration, and invasion in vitro, and also fostering tumor growth and lung metastasis in vivo. Additionally, our investigation found that COL10A1 directly interacted with integrin subunit beta 1 (ITGB1), then activates the downstream PI3K/AKT signaling pathway, thereby promoting TNBC growth and metastasis, while it was reversed by knocking down of COL10A1 or ITGB1. Our study demonstrated that the TNBC could respond to CUMS, and advocate for COL10A1 as a pivotal therapeutic target in TNBC treatment.


Asunto(s)
Proliferación Celular , Colágeno Tipo X , Integrina beta1 , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Femenino , Animales , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno Tipo X/metabolismo , Colágeno Tipo X/genética , Progresión de la Enfermedad , Ratones , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen
12.
BMC Biol ; 22(1): 139, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915055

RESUMEN

BACKGROUND: The intermediate filament protein vimentin is widely recognized as a molecular marker of epithelial-to-mesenchymal transition. Although vimentin expression is strongly associated with cancer metastatic potential, the exact role of vimentin in cancer metastasis and the underlying mechanism of its pro-metastatic functions remain unclear. RESULTS: This study revealed that vimentin can enhance integrin ß1 surface expression and induce integrin-dependent clustering of cells, shielding them against anoikis cell death. The increased integrin ß1 surface expression in suspended cells was caused by vimentin-mediated protection of the internal integrin ß1 pool against lysosomal degradation. Additionally, cell detachment was found to induce vimentin Ser38 phosphorylation, allowing the translocation of internal integrin ß1 to the plasma membrane. Furthermore, the use of an inhibitor of p21-activated kinase PAK1, one of the kinases responsible for vimentin Ser38 phosphorylation, significantly reduced cancer metastasis in animal models. CONCLUSIONS: These findings suggest that vimentin can act as an integrin buffer, storing internalized integrin ß1 and releasing it when needed. Overall, this study provides insights regarding the strong correlation between vimentin expression and cancer metastasis and a basis for blocking metastasis using this novel therapeutic mechanism.


Asunto(s)
Anoicis , Integrina beta1 , Vimentina , Vimentina/metabolismo , Vimentina/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Humanos , Animales , Supervivencia Celular , Ratones , Línea Celular Tumoral , Fosforilación , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/genética
13.
J Cell Sci ; 137(10)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38813860

RESUMEN

WW domain-containing transcription regulator 1 (WWTR1, referred to here as TAZ) and Yes-associated protein (YAP, also known as YAP1) are transcriptional co-activators traditionally studied together as a part of the Hippo pathway, and are best known for their roles in stem cell proliferation and differentiation. Despite their similarities, TAZ and YAP can exert divergent cellular effects by differentially interacting with other signaling pathways that regulate stem cell maintenance or differentiation. In this study, we show in mouse neural stem and progenitor cells (NPCs) that TAZ regulates astrocytic differentiation and maturation, and that TAZ mediates some, but not all, of the effects of bone morphogenetic protein (BMP) signaling on astrocytic development. By contrast, both TAZ and YAP mediate the effects on NPC fate of ß1-integrin (ITGB1) and integrin-linked kinase signaling, and these effects are dependent on extracellular matrix cues. These findings demonstrate that TAZ and YAP perform divergent functions in the regulation of astrocyte differentiation, where YAP regulates cell cycle states of astrocytic progenitors and TAZ regulates differentiation and maturation from astrocytic progenitors into astrocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Astrocitos , Diferenciación Celular , Proliferación Celular , Células-Madre Neurales , Transducción de Señal , Transactivadores , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Animales , Astrocitos/metabolismo , Astrocitos/citología , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Morfogenéticas Óseas/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteínas Serina-Treonina Quinasas
14.
Cells ; 13(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38727288

RESUMEN

Glioblastoma (GBM) is a devastating brain cancer for which new effective therapies are urgently needed. GBM, after an initial response to current treatment regimens, develops therapeutic resistance, leading to rapid patient demise. Cancer cells exhibit an inherent elevation of endoplasmic reticulum (ER) stress due to uncontrolled growth and an unfavorable microenvironment, including hypoxia and nutrient deprivation. Cancer cells utilize the unfolded protein response (UPR) to maintain ER homeostasis, and failure of this response promotes cell death. In this study, as integrins are upregulated in cancer, we have evaluated the therapeutic potential of individually targeting all αß1 integrin subunits using RNA interference. We found that GBM cells are uniquely susceptible to silencing of integrin α3. Knockdown of α3-induced proapoptotic markers such as PARP cleavage and caspase 3 and 8 activation. Remarkably, we discovered a non-canonical function for α3 in mediating the maturation of integrin ß1. In its absence, generation of full length ß1 was reduced, immature ß1 accumulated, and the cells underwent elevated ER stress with upregulation of death receptor 5 (DR5) expression. Targeting α3 sensitized TRAIL-resistant GBM cancer cells to TRAIL-mediated apoptosis and led to growth inhibition. Our findings offer key new insights into integrin α3's role in GBM survival via the regulation of ER homeostasis and its value as a therapeutic target.


Asunto(s)
Estrés del Retículo Endoplásmico , Glioblastoma , Integrina alfa3 , Integrina beta1 , Ligando Inductor de Apoptosis Relacionado con TNF , Humanos , Apoptosis/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Glioblastoma/patología , Glioblastoma/metabolismo , Glioblastoma/genética , Integrina alfa3/metabolismo , Integrina alfa3/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
15.
Cell Biol Toxicol ; 40(1): 32, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38767703

RESUMEN

BACKGROUND: Recent studies have emphasized the critical role of Telocytes (TCs)-derived exosomes in organ tissue injury and repair. Our previous research showed a significant increase in ITGB1 within TCs. Pulmonary Arterial Hypertension (PAH) is marked by a loss of microvessel regeneration and progressive vascular remodeling. This study aims to investigate whether exosomes derived from ITGB1-modified TCs (ITGB1-Exo) could mitigate PAH. METHODS: We analyzed differentially expressed microRNAs (DEmiRs) in TCs using Affymetrix Genechip miRNA 4.0 arrays. Exosomes isolated from TC culture supernatants were verified through transmission electron microscopy and Nanoparticle Tracking Analysis. The impact of miR-429-3p-enriched exosomes (Exo-ITGB1) on hypoxia-induced pulmonary arterial smooth muscle cells (PASMCs) was evaluated using CCK-8, transwell assay, and inflammatory factor analysis. A four-week hypoxia-induced mouse model of PAH was constructed, and H&E staining, along with Immunofluorescence staining, were employed to assess PAH progression. RESULTS: Forty-five miRNAs exhibited significant differential expression in TCs following ITGB1 knockdown. Mus-miR-429-3p, significantly upregulated in ITGB1-overexpressing TCs and in ITGB1-modified TC-derived exosomes, was selected for further investigation. Exo-ITGB1 notably inhibited the migration, proliferation, and inflammation of PASMCs by targeting Rac1. Overexpressing Rac1 partly counteracted Exo-ITGB1's effects. In vivo administration of Exo-ITGB1 effectively reduced pulmonary vascular remodeling and inflammation. CONCLUSIONS: Our findings reveal that ITGB1-modified TC-derived exosomes exert anti-inflammatory effects and reverse vascular remodeling through the miR-429-3p/Rac1 axis. This provides potential therapeutic strategies for PAH treatment.


Asunto(s)
Exosomas , Integrina beta1 , MicroARNs , Telocitos , Proteína de Unión al GTP rac1 , MicroARNs/genética , MicroARNs/metabolismo , Animales , Exosomas/metabolismo , Exosomas/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Ratones , Telocitos/metabolismo , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Ratones Endogámicos C57BL , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Hipoxia/metabolismo , Hipoxia/genética , Hipoxia/complicaciones , Proliferación Celular/genética , Movimiento Celular/genética , Humanos , Remodelación Vascular/genética , Neuropéptidos
16.
JCI Insight ; 9(10)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775153

RESUMEN

Small cell lung cancer (SCLC) is the most aggressive lung cancer entity with an extremely limited therapeutic outcome. Most patients are diagnosed at an extensive stage. However, the molecular mechanisms driving SCLC invasion and metastasis remain largely elusive. We used an autochthonous SCLC mouse model and matched samples from patients with primary and metastatic SCLC to investigate the molecular characteristics of tumor metastasis. We demonstrate that tumor cell invasion and liver metastasis in SCLC are triggered by an Angiopoietin-2 (ANG-2)/Integrin ß-1-dependent pathway in tumor cells, mediated by focal adhesion kinase/Src kinase signaling. Strikingly, CRISPR-Cas9 KO of Integrin ß-1 or blocking Integrin ß-1 signaling by an anti-ANG-2 treatment abrogates liver metastasis formation in vivo. Interestingly, analysis of a unique collection of matched samples from patients with primary and metastatic SCLC confirmed a strong increase of Integrin ß-1 in liver metastasis in comparison with the primary tumor. We further show that ANG-2 blockade combined with PD-1-targeted by anti-PD-1 treatment displays synergistic treatment effects in SCLC. Together, our data demonstrate a fundamental role of ANG-2/Integrin ß-1 signaling in SCLC cells for tumor cell invasion and liver metastasis and provide a potentially new effective treatment strategy for patients with SCLC.


Asunto(s)
Angiopoyetina 2 , Integrina beta1 , Neoplasias Hepáticas , Neoplasias Pulmonares , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas , Animales , Femenino , Humanos , Masculino , Ratones , Angiopoyetina 2/metabolismo , Angiopoyetina 2/genética , Línea Celular Tumoral , Integrina beta1/metabolismo , Integrina beta1/genética , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamiento farmacológico , Invasividad Neoplásica , Metástasis de la Neoplasia , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico
17.
J Cell Sci ; 137(10)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38639242

RESUMEN

WW domain-containing transcription regulator 1 (WWTR1, referred to here as TAZ) and Yes-associated protein (YAP, also known as YAP1) are transcriptional co-activators traditionally studied together as a part of the Hippo pathway, and are best known for their roles in stem cell proliferation and differentiation. Despite their similarities, TAZ and YAP can exert divergent cellular effects by differentially interacting with other signaling pathways that regulate stem cell maintenance or differentiation. In this study, we show in mouse neural stem and progenitor cells (NPCs) that TAZ regulates astrocytic differentiation and maturation, and that TAZ mediates some, but not all, of the effects of bone morphogenetic protein (BMP) signaling on astrocytic development. By contrast, both TAZ and YAP mediate the effects on NPC fate of ß1-integrin (ITGB1) and integrin-linked kinase signaling, and these effects are dependent on extracellular matrix cues. These findings demonstrate that TAZ and YAP perform divergent functions in the regulation of astrocyte differentiation, where YAP regulates cell cycle states of astrocytic progenitors and TAZ regulates differentiation and maturation from astrocytic progenitors into astrocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Astrocitos , Diferenciación Celular , Corteza Cerebral , Proteínas Señalizadoras YAP , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Astrocitos/metabolismo , Astrocitos/citología , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/genética , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteínas Señalizadoras YAP/metabolismo
18.
J Allergy Clin Immunol ; 154(3): 745-753, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38636606

RESUMEN

BACKGROUND: IgE-mediated degranulation of mast cells (MCs) provides rapid protection against environmental hazards, including animal venoms. A fraction of tissue-resident MCs intimately associates with blood vessels. These perivascular MCs were reported to extend projections into the vessel lumen and to be the first MCs to acquire intravenously injected IgE, suggesting that IgE loading of MCs depends on their vascular association. OBJECTIVE: We sought to elucidate the molecular basis of the MC-blood vessel interaction and to determine its relevance for IgE-mediated immune responses. METHODS: We selectively inactivated the Itgb1 gene, encoding the ß1 chain of integrin adhesion molecules (ITGB1), in MCs by conditional gene targeting in mice. We analyzed skin MCs for blood vessel association, surface IgE density, and capability to bind circulating antibody specific for MC surface molecules, as well as in vivo responses to antigen administered via different routes. RESULTS: Lack of ITGB1 expression severely compromised MC-blood vessel association. ITGB1-deficient MCs showed normal densities of surface IgE but reduced binding of intravenously injected antibodies. While their capacity to degranulate in response to IgE ligation in vivo was unimpaired, anaphylactic responses to antigen circulating in the vasculature were largely abolished. CONCLUSIONS: ITGB1-mediated association of MCs with blood vessels is key for MC immune surveillance of blood vessel content, but is dispensable for slow steady-state loading of endogenous IgE onto tissue-resident MCs.


Asunto(s)
Inmunoglobulina E , Integrina beta1 , Mastocitos , Animales , Mastocitos/inmunología , Integrina beta1/inmunología , Integrina beta1/metabolismo , Integrina beta1/genética , Inmunoglobulina E/inmunología , Ratones , Vasos Sanguíneos/inmunología , Ratones Noqueados , Anafilaxia/inmunología , Degranulación de la Célula/inmunología , Ratones Endogámicos C57BL , Piel/inmunología , Piel/irrigación sanguínea
19.
Cancer Res ; 84(9): 1517-1533, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38587552

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by an immunosuppressive tumor microenvironment enriched with cancer-associated fibroblasts (CAF). This study used a convergence approach to identify tumor cell and CAF interactions through the integration of single-cell data from human tumors with human organoid coculture experiments. Analysis of a comprehensive atlas of PDAC single-cell RNA sequencing data indicated that CAF density is associated with increased inflammation and epithelial-mesenchymal transition (EMT) in epithelial cells. Transfer learning using transcriptional data from patient-derived organoid and CAF cocultures provided in silico validation of CAF induction of inflammatory and EMT epithelial cell states. Further experimental validation in cocultures demonstrated integrin beta 1 (ITGB1) and vascular endothelial factor A (VEGFA) interactions with neuropilin-1 mediating CAF-epithelial cell cross-talk. Together, this study introduces transfer learning from human single-cell data to organoid coculture analyses for experimental validation of discoveries of cell-cell cross-talk and identifies fibroblast-mediated regulation of EMT and inflammation. SIGNIFICANCE: Adaptation of transfer learning to relate human single-cell RNA sequencing data to organoid-CAF cocultures facilitates discovery of human pancreatic cancer intercellular interactions and uncovers cross-talk between CAFs and tumor cells through VEGFA and ITGB1.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal , Inflamación , Integrina beta1 , Neoplasias Pancreáticas , Análisis de la Célula Individual , Microambiente Tumoral , Humanos , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/genética , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Inflamación/patología , Inflamación/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Organoides/patología , Organoides/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Neuropilina-1/metabolismo , Neuropilina-1/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Comunicación Celular
20.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38673859

RESUMEN

The dynamic relationship between heart failure and cancer poses a dual challenge. While cardiac remodeling can promote cancer growth and metastasis, tumor development can ameliorate cardiac dysfunction and suppress fibrosis. However, the precise mechanism through which cancer influences the heart and fibrosis is yet to be uncovered. To further explore the interaction between heart failure and cancer, we used the MDX mouse model, which suffers from cardiac fibrosis and cardiac dysfunction. A previous study from our lab demonstrated that tumor growth improves cardiac dysfunction and dampens fibrosis in the heart and diaphragm muscles of MDX mice. We used breast Polyoma middle T (PyMT) and Lewis lung carcinoma (LLC) cancer cell lines that developed into large tumors. To explore whether the aggressiveness of the cancer cell line is crucial for the beneficial phenotype, we employed a PyMT breast cancer cell line lacking integrin ß1, representing a less aggressive cell line compared to the original PyMT cells. In addition, we examined immortalized and primary MEF cells. The injection of integrin ß1 KO PyMT cancer cells and Mouse Embryo Fibroblasts cells (MEF) resulted in the improvement of cardiac function and decreased fibrosis in the heart, diaphragm, and skeletal muscles of MDX mice. Collectively, our data demonstrate that the cancer line aggressiveness as well as primary MEF cells are sufficient to impose the beneficial phenotype. These discoveries present potential novel clinical therapeutic approaches with beneficial outcome for patients with fibrotic diseases and cardiac dysfunction that do not require tumor growth.


Asunto(s)
Modelos Animales de Enfermedad , Fibrosis , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne , Animales , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/metabolismo , Ratones , Línea Celular Tumoral , Ratones Endogámicos C57BL , Femenino , Miocardio/patología , Miocardio/metabolismo , Integrina beta1/metabolismo , Integrina beta1/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA