Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.178
Filtrar
1.
Cells ; 13(19)2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39404385

RESUMEN

CD154, also known as CD40 ligand, is a costimulatory molecule involved in humoral and adaptive immune responses upon pairing with its classical receptor, CD40. The CD154/CD40 dyad is a key participant in the pathogenesis of many autoimmune diseases, including systemic lupus erythematosus (SLE). In SLE, the major cells at play, T and B lymphocytes, are shown to overexpress CD154 and CD40, respectively. Subsequently, these cells and other CD40-positive cells engage in numerous effector functions contributing to SLE development. With the recent identification of additional receptors for CD154, all belonging to the integrin family, the role of CD154 in SLE is more complex and calls for deeper investigation into its biological significance. Many therapeutic strategies directed against the CD154/CD40 couple have been deployed for the treatment of SLE and proved efficient in animal models and human studies. However, the incidence of thromboembolic complications in patients treated with these anti-CD154/CD40 antibodies halted their further clinical assessments and called for another class of therapies targeting these molecules. Second-generation antibodies directed against CD154 or CD40 are showing promising results in the advanced stages of clinical testing. Our review presents a thorough description of CD154 and its receptors, CD40 and the integrin family members in SLE pathogenesis. All these elements of the CD154 system represent important therapeutic targets for the treatment of SLE.


Asunto(s)
Ligando de CD40 , Lupus Eritematoso Sistémico , Animales , Humanos , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Ligando de CD40/inmunología , Integrinas/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo
2.
Nat Commun ; 15(1): 8691, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375335

RESUMEN

Morphogenesis requires building stable macromolecular structures from highly dynamic proteins. Muscles are anchored by long-lasting integrin adhesions to resist contractile force. However, the mechanisms governing integrin diffusion, immobilization, and activation within developing tissues remain elusive. Here, we show that actin polymerization-driven membrane protrusions form nanotopographies that enable strong adhesion at Drosophila muscle attachment sites (MASs). Super-resolution microscopy reveals that integrins assemble adhesive belts around Arp2/3-dependent actin protrusions, forming invadosome-like structures with membrane nanotopographies. Single protein tracking shows that, during MAS development, integrins become immobile and confined within diffusion traps formed by the membrane nanotopographies. Actin filaments also display restricted motion and confinement, indicating strong mechanical connection with integrins. Using isolated muscle cells, we show that substrate nanotopography, rather than rigidity, drives adhesion maturation by regulating actin protrusion, integrin diffusion and immobilization. These results thus demonstrate that actin-polymerization-driven membrane protrusions are essential for the formation of strong integrin adhesions sites in the developing embryo, and highlight the important contribution of geometry to morphogenesis.


Asunto(s)
Actinas , Adhesión Celular , Proteínas de Drosophila , Drosophila melanogaster , Integrinas , Animales , Actinas/metabolismo , Integrinas/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/embriología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Morfogénesis , Citoesqueleto de Actina/metabolismo , Embrión no Mamífero/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Músculos/metabolismo
3.
Nat Commun ; 15(1): 8751, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39384749

RESUMEN

The extracellular matrix (ECM) is a protein polymer network that physically supports cells within a tissue. It acts as an important physical and biochemical stimulus directing cell behaviors. For fibronectin (Fn), a predominant component of the ECM, these physical and biochemical activities are inextricably linked as physical forces trigger conformational changes that impact its biochemical activity. Here, we analyze whether oxidative post-translational modifications, specifically glutathionylation, alter Fn's mechano-chemical characteristics through stretch-dependent protein modification. ECM post-translational modifications represent a potential for time- or stimulus-dependent changes in ECM structure-function relationships that could persist over time with potentially significant impacts on cell and tissue behaviors. In this study, we show evidence that glutathionylation of Fn ECM fibers is stretch-dependent and alters Fn fiber mechanical properties with implications on the selectivity of engaging integrin receptors. These data demonstrate the existence of multimodal post-translational modification mechanisms within the ECM with high relevance to the microenvironmental regulation of downstream cell behaviors.


Asunto(s)
Matriz Extracelular , Fibronectinas , Glutatión , Integrinas , Procesamiento Proteico-Postraduccional , Fibronectinas/metabolismo , Matriz Extracelular/metabolismo , Humanos , Integrinas/metabolismo , Glutatión/metabolismo , Animales
4.
Nat Commun ; 15(1): 8824, 2024 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-39394209

RESUMEN

Mesenchymal stem cells (MSCs) interact with their surroundings via integrins, which link to the actin cytoskeleton and translate physical cues into biochemical signals through mechanotransduction. N-cadherins enable cell-cell communication and are also linked to the cytoskeleton. This crosstalk between integrins and cadherins modulates MSC mechanotransduction and fate. Here we show the role of this crosstalk in the mechanosensing of viscosity using supported lipid bilayers as substrates of varying viscosity. We functionalize these lipid bilayers with adhesion peptides for integrins (RGD) and N-cadherins (HAVDI), to demonstrate that integrins and cadherins compete for the actin cytoskeleton, leading to an altered MSC mechanosensing response. This response is characterised by a weaker integrin adhesion to the environment when cadherin ligation occurs. We model this competition via a modified molecular clutch model, which drives the integrin/cadherin crosstalk in response to surface viscosity, ultimately controlling MSC lineage commitment.


Asunto(s)
Cadherinas , Integrinas , Células Madre Mesenquimatosas , Cadherinas/metabolismo , Cadherinas/química , Cadherinas/genética , Integrinas/metabolismo , Viscosidad , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Humanos , Mecanotransducción Celular , Membrana Dobles de Lípidos/metabolismo , Membrana Dobles de Lípidos/química , Adhesión Celular , Citoesqueleto de Actina/metabolismo , Comunicación Celular , Animales , Oligopéptidos/metabolismo , Oligopéptidos/química
5.
Discov Med ; 36(189): 1998-2012, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39463220

RESUMEN

This review predominantly acquaints the role of focal adhesion kinase (FAK) and cellular-Src (c-Src) in cell adhesion. Cell adhesion is a crucial phenomenon that causes the cells to interact with the extracellular matrix (ECM) or with each other. There are different proteins involved in cell adhesion including cell adhesion molecules (CAMs)/receptors that are present on the cell surface and various cytoplasmic proteins. FAK and c-Src are two proteins in the cytoplasm, which serve as regulators of different proteins involved in cell adhesion. They activate talin, vinculin and paxillin in turn connect the integrins with the cytoskeleton and in this way strengthen the integrin interaction with ECM. FAK-Src signalling also modulates cell-cell adhesion by regulating actin interactions. Being a key modulator of cell adhesion, FAK and c-Src signalling are linked with different pathological conditions like cancer, cardiovascular diseases, and embryonic developmental disorders. Thus, comprehensive research into FAK-Src signalling is of great importance in the exploration of different signalling targets for therapeutic interpretations. Different inhibitors and antibodies against various cell adhesion proteins, such as FAK, c-Src, and integrins, have already been used in preclinical and clinical trials to treat a variety of diseases, including cancer and chronic inflammatory conditions. Furthermore, this review presents different challenges to FAK-Src and cell adhesion signalling targeted drug development, which include, cytotoxicity and cell resistance to the drug. Finally, this review remarks that FAK and c-Src are important regulators of cell adhesion and are linked to various pathologies, nevertheless, more comprehensive research on these proteins would be a significant step forward in the development of effective therapies for the diseases associated with them.


Asunto(s)
Adhesión Celular , Proteína-Tirosina Quinasas de Adhesión Focal , Transducción de Señal , Humanos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Animales , Proteína Tirosina Quinasa CSK/metabolismo , Integrinas/metabolismo
6.
ACS Nano ; 18(43): 29585-29601, 2024 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-39400273

RESUMEN

Extracellular vesicles (EVs) are a diverse population of membrane structures produced and released by cells into the extracellular space for the intercellular trafficking of cargo molecules. They are implicated in various biological processes, including angiogenesis, immunomodulation, and cancer cell signaling. While much research has focused on their biogenesis or their effects on recipient cells, less is understood about how EVs are capable of traversing diverse tissue environments and crossing biological barriers. Their interactions with extracellular matrix components are of particular interest, as such interactions govern diffusivity and mobility, providing a potential basis for organotropism. To start to untangle how EV-matrix interactions affect diffusivity, we use high speed epifluorescence microscopy, single particle tracking, and confocal reflectance microscopy to analyze particle mobility and localization in extracellular matrix-mimicking hydrogels composed of collagen I. EVs are compared with synthetic liposomes and extruded plasma membrane vesicles to better understand the importance of membrane composition on these interactions. By treating EVs with trypsin to digest surface proteins, we determine that proteins are primarily responsible for EV immobilization in collagen I hydrogels. We next use a synthetic peptide competitive inhibitor to narrow down the identity of the proteins involved to argynylglycylaspartic acid (RGD) motif-binding integrins, which interact with unincorporated or denatured nonfibrillar collagen. Moreover, the effect of integrin inhibition with RGD peptides has strong implications for the use of RGD-peptide-based drugs to treat certain cancers, as integrin inhibition appears to increase EV mobility, improving their ability to infiltrate tissue-like environments.


Asunto(s)
Colágeno Tipo I , Matriz Extracelular , Vesículas Extracelulares , Hidrogeles , Integrinas , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/química , Hidrogeles/química , Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Integrinas/metabolismo , Integrinas/química , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Humanos , Oligopéptidos/química , Oligopéptidos/farmacología , Oligopéptidos/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-39436262

RESUMEN

Integrin genes are widely involved in tumorigenesis. Yet, a comprehensive characterization of integrin family members and their interactome at the pan-cancer level is lacking. Here, we systematically analyzed integrin family in approximately 10,000 tumors across 32 cancer types. Globally, integrins represent a frequently altered and misexpressed pathway, with alteration and dysregulation overall being protumorigenic. Expression dysregulation, better than mutational landscape, of integrin family successfully identifies a subgroup of aggressive tumors with a high level of proliferation and stemness. The results reveal that several molecular mechanisms collectively regulate integrin expression in a context-dependent manner. For potential clinical usage, we constructed a weighted scoring system, integrinScore, to measure integrin signaling patterns in individual tumors. Remarkably, integrinScore was consistently correlated with predefined molecular subtypes in multiple cancers, with integrinScore-high tumors being more aggressive. Importantly, integrinScore was cancer-dependent and closely associated with proliferation, stemness, tumor microenvironment, metastasis, and immune signatures. IntegrinScore also predicted patients' response to immunotherapy. By mining drug databases, we unraveled an array of compounds that may modulate integrin signaling. Finally, we built a user-friendly database, Pan-cancer Integrin Explorer (PIExplorer; http://computationalbiology.cn/PIExplorer), to facilitate researchers to explore integrin-related knowledge. Collectively, we provide a comprehensive characterization of integrins across cancers and offer gene-specific and cancer-specific rationales for developing integrin-targeted therapy.


Asunto(s)
Integrinas , Neoplasias , Humanos , Integrinas/genética , Integrinas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal/genética , Regulación Neoplásica de la Expresión Génica , Microambiente Tumoral/genética
8.
Clin Nucl Med ; 49(11): e619-e621, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39365084

RESUMEN

ABSTRACT: Peritoneal cancer index is an integral parameter for preoperative assessment of peritoneal disease. Current imaging modalities including 18 F-FDG PET/CT, though superior to CT, underestimate the disease extent, mainly due to its high physiological background activity. Novel "cancer integrin" targeting imaging with 68 Ga-trivehexin demonstrates limited physiological activity in the abdomen with superior target-to-background ratio than 18 F-FDG PET/CT. Thus, we describe the first case demonstrating potential utility of novel "cancer integrin" αvß6 imaging agent 68 Ga-trivehexin for assessment of disease burden in advanced epithelial ovarian cancer.


Asunto(s)
Neoplasias Ováricas , Tomografía Computarizada por Tomografía de Emisión de Positrones , Humanos , Femenino , Neoplasias Ováricas/diagnóstico por imagen , Antígenos de Neoplasias/metabolismo , Integrinas/metabolismo , Radioisótopos de Galio , Persona de Mediana Edad , Tomografía Computarizada por Rayos X
9.
Eur J Pharmacol ; 983: 176983, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39243926

RESUMEN

Chronic kidney disease (CKD) is a global public health problem, involving about 10% of the global population. Unfortunately, there are currently no effective drugs. Kidney fibrosis is the main pathology of CKD, where integrins play crucial roles in renal fibrogenesis. Recently, Bexotegrast (PLN-74809) as a dual integrin αvß1/αvß6 inhibitor could reduce the degree of lung fibrosis in patients with idiopathic pulmonary fibrosis. However, the role of PLN-74809 remains unclear in fibrotic kidney disease. Here, we have revealed that PLN-74809 administration dose-dependently delayed the progression of renal fibrosis in both adenine diet- and unilateral ureteral obstruction (UUO)-induced mice. Mechanistically, PLN-74809 targeted integrin αvß1/αvß6 to inhibit FAK/Src/Akt/ß-catenin cascade in fibrotic kidneys. In summary, our results for the first time highlighted the αvß1/αvß6 inhibitor PLN-74809 exerted potential therapeutic against kidney fibrosis.


Asunto(s)
Fibrosis , Integrinas , Animales , Masculino , Ratones , Adenina/análogos & derivados , Adenina/farmacología , Antígenos de Neoplasias/metabolismo , beta Catenina/metabolismo , beta Catenina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Riñón/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Obstrucción Ureteral/patología , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico
10.
APMIS ; 132(10): 688-705, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39246244

RESUMEN

Bacterial biofilm plays a vital role in influencing several diseases, infections, metabolic pathways and communication channels. Biofilm influence over colorectal cancer (CRC) has been a booming area of research interest. The virulence factors of bacterial pathogen have a high tendency to induce metabolic pathway to accelerate CRC. The bacterial species biofilm may induce cancer through regulating the major signalling pathways responsible for cell proliferation, differentiation, survival and growth. Activation of cancer signals may get initiated from the chronic infections through bacterial biofilm species. Integrin mediates in the activation of major pathway promoting cancer. Integrin-mediated signals are expected to be greatly influenced by biofilm. Integrins are identified as an important dimer, whose dysfunction may alter the signalling cascade specially focusing on TGF-ß, PI3K/Akt/mToR, MAPK and Wnt pathway. Along with biofilm shield, the tumour gains greater resistance from radiation, chemotherapy and also from other antibiotics. The biofilm barrier is known to cause challenges for CRC patients undergoing treatment.


Asunto(s)
Biopelículas , Neoplasias Colorrectales , Integrinas , Transducción de Señal , Biopelículas/crecimiento & desarrollo , Neoplasias Colorrectales/microbiología , Humanos , Integrinas/metabolismo
11.
Biomolecules ; 14(9)2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39334945

RESUMEN

Various strategies have been employed to improve the reliability of 2D, 3D, and co-culture in vitro models of nonalcoholic fatty liver disease, including using extracellular matrix proteins such as collagen I to promote cell adhesion. While studies have demonstrated the significant benefits of culturing cells on collagen I, its effects on the HepG2 cell line after exposure to palmitate (PA) have not been investigated. Therefore, this study aimed to assess the effects of PA-induced lipotoxicity in HepG2 cultured in the absence or presence of collagen I. HepG2 cultured in the absence or presence of collagen I was exposed to PA, followed by analyses that assessed cell proliferation, viability, adhesion, cell death, mitochondrial respiration, reactive oxygen species production, gene and protein expression, and triacylglycerol accumulation. Culturing HepG2 on collagen I was associated with increased cell proliferation, adhesion, and expression of integrin receptors, and improved cellular spreading compared to culturing them in the absence of collagen I. However, PA-induced lipotoxicity was greater in collagen I-cultured HepG2 than in those cultured in the absence of collagen I and was associated with increased α2ß1 receptors. In summary, the present study demonstrated for the first time that collagen I-cultured HepG2 exhibited exacerbated cell death following exposure to PA through integrin-mediated death. The findings from this study may serve as a caution to those using 2D models or 3D scaffold-based models of HepG2 in the presence of collagen I.


Asunto(s)
Adhesión Celular , Proliferación Celular , Supervivencia Celular , Colágeno Tipo I , Humanos , Células Hep G2 , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Proliferación Celular/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Palmitatos/toxicidad , Palmitatos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Muerte Celular/efectos de los fármacos , Integrina alfa2beta1/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Integrinas/metabolismo , Integrinas/genética
12.
Int Immunopharmacol ; 142(Pt B): 113218, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39317053

RESUMEN

BACKGROUND: Integrins are upregulated on endothelial cells and T-lymphocytes in autoimmune thyroid disease (AITD), potentially contributing to immune response localization. The role of integrins on B-cells in AITD remains unclear. METHODS: Peripheral blood samples were collected from healthy controls (n = 56), patients with Graves' disease (GD) (n = 37) and Hashimoto's thyroiditis (HT) (n = 52). Ultrasound-guided fine-needle aspiration (FNA) of the thyroid was performed in patients with non-autoimmune thyroid disease (nAITD) (n = 19), GD (n = 11), and HT (n = 40). Integrins α4ß7, α4ß1, and αEß7 in B cells were measured by flow cytometry. Serum zonulin levels were quantified via ELISA. Associations of integrins on B cells with thyroid hormones, thyroid autoantibodies, AITD duration, and zonulin were analyzed. RESULTS: HT patients exhibited lower α4ß7 and higher α4ß1 expression on B cells compared to healthy controls and GD patients. While α4ß7 was predominant on circulating B cells, the dominant integrin expressed on intrathyroidal B cells varied with specific thyroid diseases. In GD patients, α4ß7 and α4ß1 expression on circulating B cells correlated positively and negatively with thyroid function and thyroid stimulating immunoglobulins (TSI) levels, respectively. Intrathyroidal α4ß1+ B cells positively correlated with TSH levels in HT patients. Additionally, serum zonulin was elevated in HT patients, and intrathyroidal α4ß7+ B cells and α4ß1+ B cells correlated negatively and positively with zonulin levels, respectively. Integrin αEß7 on B cells showed no significant association with AITD. CONCLUSION: Integrins expressed on B cells potentially play a role in the pathogenesis of AITD and might serve as immune biomarkers for the disease.


Asunto(s)
Linfocitos B , Enfermedad de Graves , Integrinas , Humanos , Enfermedad de Graves/inmunología , Enfermedad de Graves/sangre , Masculino , Femenino , Linfocitos B/inmunología , Linfocitos B/metabolismo , Adulto , Persona de Mediana Edad , Integrinas/metabolismo , Integrina alfa4beta1/metabolismo , Integrina alfa4beta1/inmunología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Enfermedad de Hashimoto/inmunología , Enfermedad de Hashimoto/sangre , Enfermedad de Hashimoto/metabolismo , Haptoglobinas/metabolismo , Precursores de Proteínas/metabolismo , Toxina del Cólera/inmunología , Enterotoxinas/inmunología , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/patología
13.
J Med Chem ; 67(19): 17497-17519, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39269712

RESUMEN

A series of 3-aryl((S)-3-fluoropyrrolidin-1-yl)butanoic acids were developed as potent orally bioavailable αvß6 integrin inhibitors. Starting from a zwitterionic peptidomimetic series optimized for inhaled administration, the balancing of potency and passive permeability to achieve suitable oral agents through modification and exploration of aryl substituents and pKa of the central cyclic amine is described. (S)-4-((S)-3-Fluoro-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)-3-(3-(2-methoxyethoxy)phenyl)butanoic acid was found to have highly desirable oral pharmacokinetic profiles in rat, dog, and minipig, with low to moderate clearance (26%, 7%, and 18% liver blood flow, respectively), moderate volumes of distribution (3.6, 1.4, and 0.9 L/kg, respectively), high to complete oral bioavailabilities, high αvß6 integrin potency of pIC50 of 8.0, and high solubility in physiological media (>2 mg/mL). Equating to the estimated human dose range of 10-75 mg b.i.d. to achieve 90% αvß6 target engagement at Cmin, it was selected for further investigation as a potential therapeutic agent for the treatment of idiopathic pulmonary fibrosis.


Asunto(s)
Antígenos de Neoplasias , Disponibilidad Biológica , Integrinas , Animales , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Perros , Ratas , Administración Oral , Humanos , Porcinos , Antígenos de Neoplasias/metabolismo , Relación Estructura-Actividad , Descubrimiento de Drogas , Porcinos Enanos , Masculino , Ratas Sprague-Dawley
14.
Connect Tissue Res ; 65(5): 343-363, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39297793

RESUMEN

Features of the extracellular matrix, along with biochemical factors, have a momentous impress in making genes on and/or off. The interaction of cells and the extracellular matrix is mediated by integrins. Therefore, these molecules have pivotal roles in regulating cell behaviors. Integrins include a group of molecules with a variety of characteristics that can affect different molecular cascades. Considering the importance of these molecules in tissue regeneration after injury, it is necessary to know well the integrins involved in the process of connecting cells to the extracellular matrix in each tissue.With the increase in life expectancy, bone tissue engineering has received more attention from researchers. Integrins are critical components in osteoblast differentiation, survival, and bone mechanotransduction. During osteogenic differentiation in stem cells, specific integrins facilitate multiple signaling pathways through their cytoplasmic domain, leading to the induction of osteogenic differentiation. Also, due to the importance of using biomaterials in bone tissue engineering, efforts have been made to design and use biomaterials with maximum interaction with integrins. Notably, the use of RGD peptide or fibronectin for surface modification is a well-established and commonly employed approach to manipulate integrin activity.This review article looks into integrins' role in bone development and regeneration. It then goes on to explore the complex mechanisms by which integrins contribute to these processes. In addition, this review discusses the use of natural and synthetic biomaterials that target integrins to promote bone regeneration.


Asunto(s)
Materiales Biocompatibles , Regeneración Ósea , Integrinas , Humanos , Regeneración Ósea/efectos de los fármacos , Integrinas/metabolismo , Materiales Biocompatibles/farmacología , Animales , Osteogénesis/efectos de los fármacos , Ingeniería de Tejidos
15.
Cancer Lett ; 604: 217196, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39222676

RESUMEN

Hepatocyte growth factor (HGF) plays a critical role in promoting tumor migration, invasion, and metastasis, partly by upregulating integrins. The molecular mechanisms behind how HGF facilitates integrin-mediated tumorigenesis are not fully understood. In this study, we demonstrate that the ubiquitin-specific peptidase 22 (USP22) is essential for HGF-induced melanoma metastasis. HGF treatment dramatically increased the expression of both USP22 and multiple integrin family members in particular ITGAV, ITGB3, and ITGA1. An unbiased analysis of the TCGA database reveals integrins as common downstream targets of both USP22 and HGF across multiple human cancer types. Notably, CRISPR-mediated deletion of USP22 completely eliminates HGF-induced integrin expression in melanoma cells. At the molecular level, USP22 acts as a bona fide deubiquitinase for Sp1, a transcription factor for the ITGAV, ITGB3, and ITGA1 genes. USP22 interacts with and inhibits Sp1 ubiquitination, protecting against Sp1 proteasomal degradation. Supporting this, immunohistology analysis detects a positive correlation among USP22, Sp1, and integrin αv in human melanoma tissues. This study identifies the death from the signature gene USP22 as a critical positive regulator for HGF-induced integrin expression by deubiquitinating the Sp1 transcription factor during melanoma metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito , Melanoma , Factor de Transcripción Sp1 , Ubiquitina Tiolesterasa , Regulación hacia Arriba , Humanos , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/genética , Melanoma/genética , Melanoma/patología , Melanoma/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp1/genética , Línea Celular Tumoral , Animales , Integrinas/metabolismo , Integrinas/genética , Ubiquitinación , Ratones , Movimiento Celular , Metástasis de la Neoplasia , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
16.
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
17.
PLoS Biol ; 22(9): e3002783, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39226305

RESUMEN

Cell shape remodeling is a principal driver of epithelial tissue morphogenesis. While progress continues to be made in our understanding of the pathways that control the apical (top) geometry of epithelial cells, we know comparatively little about those that control cell basal (bottom) geometry. To examine this, we used the Drosophila ommatidium, which is the basic visual unit of the compound eye. The ommatidium is shaped as a hexagonal prism, and generating this 3D structure requires ommatidial cells to adopt specific apical and basal polygonal geometries. Using this model system, we find that generating cell type-specific basal geometries starts with patterning of the basal extracellular matrix, whereby Laminin accumulates at discrete locations across the basal surface of the retina. We find the Dystroglycan receptor complex (DGC) is required for this patterning by promoting localized Laminin accumulation at the basal surface of cells. Moreover, our results reveal that localized accumulation of Laminin and the DGC are required for directing Integrin adhesion. This induces cell basal geometry remodeling by anchoring the basal surface of cells to the extracellular matrix at specific, Laminin-rich locations. We propose that patterning of a basal extracellular matrix by generating discrete Laminin domains can direct Integrin adhesion to induce cell shape remodeling in epithelial morphogenesis.


Asunto(s)
Forma de la Célula , Proteínas de Drosophila , Drosophila melanogaster , Distroglicanos , Matriz Extracelular , Integrinas , Laminina , Retina , Animales , Distroglicanos/metabolismo , Laminina/metabolismo , Integrinas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Matriz Extracelular/metabolismo , Retina/metabolismo , Retina/crecimiento & desarrollo , Retina/citología , Retina/embriología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Morfogénesis , Adhesión Celular , Drosophila/metabolismo , Drosophila/crecimiento & desarrollo
18.
Nat Commun ; 15(1): 7965, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261465

RESUMEN

Current adeno-associated virus (AAV) gene therapy using nature-derived AAVs is limited by non-optimal tissue targeting. In the treatment of muscular diseases (MD), high doses are often required but can lead to severe adverse effects. Here, we rationally design an AAV capsid that specifically targets skeletal muscle to lower treatment doses. We computationally integrate binding motifs of human integrin alphaV beta6, a skeletal muscle receptor, into a liver-detargeting capsid. Designed AAVs show higher productivity and superior muscle transduction compared to their parent. One variant, LICA1, demonstrates comparable muscle transduction to other myotropic AAVs with reduced liver targeting. LICA1's myotropic properties are observed across species, including non-human primate. Consequently, LICA1, but not AAV9, effectively delivers therapeutic transgenes and improved muscle functionality in two mouse MD models (male mice) at a low dose (5E12 vg/kg). These results underline the potential of our design method for AAV engineering and LICA1 variant for MD gene therapy.


Asunto(s)
Dependovirus , Terapia Genética , Músculo Esquelético , Dependovirus/genética , Animales , Humanos , Músculo Esquelético/metabolismo , Ratones , Terapia Genética/métodos , Masculino , Vectores Genéticos/genética , Integrinas/metabolismo , Integrinas/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Enfermedades Musculares/terapia , Enfermedades Musculares/genética , Transducción Genética , Hígado/metabolismo , Cápside/metabolismo , Receptores de Vitronectina/metabolismo , Receptores de Vitronectina/genética , Modelos Animales de Enfermedad , Células HEK293 , Transgenes , Ratones Endogámicos C57BL , Antígenos de Neoplasias
19.
J Immunol ; 213(7): 988-997, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39140892

RESUMEN

Immune cells survey their microenvironment by forming dynamic cellular protrusions that enable chemotaxis, contacts with other cells, and phagocytosis. Podosomes are a unique type of protrusion structured by an adhesive ring of active integrins that surround an F-actin-rich core harboring degradative proteases. Although the features of podosomes, once-established, have been well defined, the steps that lead to podosome formation remain poorly understood by comparison. In this study, we report that spleen tyrosine kinase (Syk) is a critical regulator of podosome formation. Deletion of Syk or targeting its kinase activity eliminated the ability for murine macrophages to form podosomes. We found that the kinase activity of Syk was important for the phosphorylation of its substrates, HS1 and Pyk2, both of which regulate podosome formation. Additionally, before podosomes form, we report that the tandem Src homology 2 domains of Syk afforded multivalent clustering of ITAM-containing adaptors that associated with integrins to structure platforms that initiate podosomes. We therefore propose that Syk has a dual role in regulating podosomes: first, by facilitating the assembly of multivalent signaling hubs that nucleate their formation and second, by sustaining tyrosine kinase activity of the podosomes once they form against their substrates. In cells expressing recently identified gain-of-function variants of SYK, podosomes were dysregulated. These results implicate SYK in the (patho)physiological functions of podosomes in macrophages.


Asunto(s)
Macrófagos , Podosomas , Quinasa Syk , Quinasa Syk/metabolismo , Animales , Ratones , Podosomas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Fosforilación , Quinasa 2 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/genética , Ratones Noqueados , Integrinas/metabolismo , Transducción de Señal , Humanos , Ratones Endogámicos C57BL , Quinasa 1 de Adhesión Focal
20.
Oncogene ; 43(39): 2951-2969, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39174859

RESUMEN

High invasive capacity and acquired tyrosine kinase inhibitors (TKI) resistance of kidney renal clear cell carcinoma (KIRC) cells remain obstacles to prolonging the survival time of patients with advanced KIRC. In the present study, we reported that sine oculis homeobox 1 (SIX1) was upregulated in sunitinib-resistant KIRC cells and metastatic KIRC tissues. Subsequently, we found that SIX1 mediated metastasis and sunitinib resistance via Focal adhesion (FA) signaling, and knockdown of SIX1 enhanced the antitumor efficiency of sunitinib in KIRC. Mechanistically, Integrin subunit beta 1 (ITGB1), an upstream gene of FA signaling, was a direct transcriptional target of SIX1. In addition, we showed that DExH-box helicase 9 (DHX9) was an important mediator for SIX1-induced ITGB1 transcription, and silencing the subunits of SIX1/DHX9 complex significantly reduced transcription of ITGB1. Downregulation of SIX1 attenuated nuclear translocation of DHX9 and abrogated the binding of DHX9 to ITGB1 promoter. Collectively, our results unveiled a new signal axis SIX1/ITGB1/FAK in KIRC and identified a novel therapeutic strategy for metastatic KIRC patients.


Asunto(s)
Carcinoma de Células Renales , ARN Helicasas DEAD-box , Resistencia a Antineoplásicos , Adhesiones Focales , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio , Integrina beta1 , Neoplasias Renales , Metástasis de la Neoplasia , Transducción de Señal , Sunitinib , Humanos , Resistencia a Antineoplásicos/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Sunitinib/farmacología , Sunitinib/uso terapéutico , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Línea Celular Tumoral , Integrina beta1/genética , Integrina beta1/metabolismo , Animales , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Ratones , Transcripción Genética , Integrinas/metabolismo , Integrinas/genética , Quinasa 1 de Adhesión Focal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...