Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.597
Filtrar
1.
Gene ; 815: 146137, 2022 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-35007686

RESUMEN

The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.


Asunto(s)
Biomarcadores de Tumor , Proteínas de la Matriz Extracelular/fisiología , Neoplasias/etiología , Neoplasias/terapia , Moléculas de Adhesión Celular/fisiología , Proteínas de la Matriz Extracelular/análisis , Humanos , Metaloproteinasas de la Matriz/fisiología , Osteonectina/análisis , Osteonectina/fisiología , Osteopontina/fisiología , Tenascina/fisiología , Trombospondina 1/fisiología , Resultado del Tratamiento
2.
Pediatr Neurol ; 126: 65-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34740135

RESUMEN

BACKGROUND: Semaphorins and plexins are ligands and cell surface receptors that regulate multiple neurodevelopmental processes such as axonal growth and guidance. PLXNA3 is a plexin gene located on the X chromosome that encodes the most widely expressed plexin receptor in fetal brain, plexin-A3. Plexin-A3 knockout mice demonstrate its role in semaphorin signaling in vivo. The clinical manifestations of semaphorin/plexin neurodevelopmental disorders have been less widely explored. This study describes the neurological and neurodevelopmental phenotypes of boys with maternally inherited hemizygous PLXNA3 variants. METHODS: Data-sharing through GeneDx and GeneMatcher allowed identification of individuals with autism or intellectual disabilities (autism/ID) and hemizygous PLXNA3 variants in collaboration with their physicians and genetic counselors, who completed questionnaires about their patients. In silico analyses predicted pathogenicity for each PLXNA3 variant. RESULTS: We assessed 14 boys (mean age, 10.7 [range 2 to 25] years) with maternally inherited hemizygous PLXNA3 variants and autism/ID ranging from mild to severe. Other findings included fine motor dyspraxia (92%), attention-deficit/hyperactivity traits, and aggressive behaviors (63%). Six patients (43%) had seizures. Thirteen boys (93%) with PLXNA3 variants showed novel or very low allele frequencies and probable damaging/disease-causing pathogenicity in one or more predictors. We found a genotype-phenotype correlation between PLXNA3 cytoplasmic domain variants (exons 22 to 32) and more severe neurodevelopmental disorder phenotypes (P < 0.05). CONCLUSIONS: We report 14 boys with maternally inherited, hemizygous PLXNA3 variants and a range of neurodevelopmental disorders suggesting a novel X-linked intellectual disability syndrome. Greater understanding of PLXNA3 variant pathogenicity in humans will require additional clinical, computational, and experimental validation.


Asunto(s)
Trastorno del Espectro Autista/genética , Moléculas de Adhesión Celular/fisiología , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/fisiología , Receptores de Superficie Celular/genética , Semaforinas/fisiología , Adolescente , Adulto , Trastorno del Espectro Autista/fisiopatología , Niño , Preescolar , Estudios de Asociación Genética , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Transducción de Señal/fisiología , Adulto Joven
3.
Clin Transl Oncol ; 24(2): 244-253, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34236615

RESUMEN

BACKGROUND: Bacterial-mediated cancer immunotherapy (BCI) elicits a more robust initial immune response than conventional immunotherapy, but does not prevent tumor recurrence and metastasis. BCI is associated with recruitment of tumor-infiltrating neutrophils, which could suppress the therapeutic efficacy of this modality. Development endothelial locus 1 (Del-1), a potent inhibitor of neutrophil recruitment, antagonizes lymphocyte function-associated antigen-1 on the vascular endothelium. Here, we aimed to determine the effect of Del-1-secreting S.t△ppGpp on anti-tumor activity and tumor-infiltrating neutrophil recruitment in a mouse model of colon cancer. METHODS: We investigated the anti-cancer activity of Del-1-secreting engineered Salmonella (△ppGpp S. Typhimurium) in the mice colon cancer models. RESULTS: In the present study, we identified that Del-1-secreting engineered Salmonella had more potent anti-cancer activity compared with normal S.t△ppGpp without Del-1 secretion. We postulated that Del-1 expression increased M1 macrophage recruitment to tumors by decreasing tumor-infiltrating neutrophils. This approach could enhance the anti-cancer effects of S.t△ppGpp. CONCLUSIONS: Collectively, the approach of using engineered bacteria that deliver Del-1 to block tumor-infiltrating neutrophil recruitment is a potential therapeutic approach.


Asunto(s)
Terapia Biológica/métodos , Proteínas de Unión al Calcio/fisiología , Moléculas de Adhesión Celular/fisiología , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Infiltración Neutrófila , Salmonella typhimurium , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C , Resultado del Tratamiento
4.
Front Immunol ; 12: 753929, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950135

RESUMEN

Background: Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS) mediated by autoimmunity. No objective clinical indicators are available for the diagnosis and prognosis of MS. Extracellular proteins are most glycosylated and likely to enter into the body fluid to serve as potential biomarkers. Our work will contribute to the in-depth study of the functions of extracellular proteins and the discovery of disease biomarkers. Methods: MS expression profiling data of the human brain was downloaded from the Gene Expression Omnibus (GEO). Extracellular protein-differentially expressed genes (EP-DEGs) were screened by protein annotation databases. GO and KEGG were used to analyze the function and pathway of EP-DEGs. STRING, Cytoscape, MCODE and Cytohubba were used to construct a protein-protein interaction (PPI) network and screen key EP-DEGs. Key EP-DEGs levels were detected in the CSF of MS patients. ROC curve and survival analysis were used to evaluate the diagnostic and prognostic ability of key EP-DEGs. Results: We screened 133 EP-DEGs from DEGs. EP-DEGs were enriched in the collagen-containing extracellular matrix, signaling receptor activator activity, immune-related pathways, and PI3K-Akt signaling pathway. The PPI network of EP-DEGs had 85 nodes and 185 edges. We identified 4 key extracellular proteins IL17A, IL2, CD44, IGF1, and 16 extracellular proteins that interacted with IL17A. We clinically verified that IL17A levels decreased, but Del-1 and resolvinD1 levels increased. The diagnostic accuracy of Del-1 (AUC: 0.947) was superior to that of IgG (AUC: 0.740) with a sensitivity of 82.4% and a specificity of 100%. High Del-1 levels were significantly associated with better relapse-free and progression-free survival. Conclusion: IL17A, IL2, CD44, and IGF1 may be key extracellular proteins in the pathogenesis of MS. IL17A, Del-1, and resolvinD1 may co-regulate the development of MS and Del-1 is a potential biomarker of MS. We used bioinformatics methods to explore the biomarkers of MS and validated the results in clinical samples. The study provides a theoretical and experimental basis for revealing the pathogenesis of MS and improving the diagnosis and prognosis of MS.


Asunto(s)
Líquido Extracelular/química , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Proteínas/análisis , Adulto , Biomarcadores , Química Encefálica , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/fisiología , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/fisiología , Proteínas del Líquido Cefalorraquídeo/análisis , Proteínas del Líquido Cefalorraquídeo/genética , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Cefalea/genética , Cefalea/metabolismo , Humanos , Interleucina-17/análisis , Interleucina-17/fisiología , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Supervivencia sin Progresión , Análisis por Matrices de Proteínas , Mapas de Interacción de Proteínas , Proteínas/genética , Sensibilidad y Especificidad
5.
Theranostics ; 11(20): 9738-9751, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34815781

RESUMEN

Rationale: Angiogenesis and osteogenesis are highly coupled processes which are indispensable to bone repair. However, the underlying mechanism(s) remain elusive. To bridge the gap in understanding the coupling process is crucial to develop corresponding solutions to abnormal bone healing. Epidermal growth factor-like protein 6 (EGFL6) is an angiogenic factor specifically and distinctively up-regulated during osteoblast differentiation. In contrast with most currently known osteoblast-derived coupling factors, EGFL6 is highlighted with little or low expression in other cells and tissues. Methods: In this study, primary bone marrow mesenchymal stem cells (MSCs) and osteoblastic cell line (MC3T3-E1) were transduced with lentiviral silencing or overexpression constructs targeting EGFL6. Cells were induced by osteogenic medium, followed by the evaluation of mineralization as well as related gene and protein expression. Global and conditional knockout mice were established to examine the bone phenotype under physiological condition. Furthermore, bone defect models were created to investigate the outcome of bone repair in mice lacking EGFL6 expression. Results: We show that overexpression of EGFL6 markedly enhances osteogenic capacity in vitro by augmenting bone morphogenic protein (BMP)-Smad and MAPK signaling, whereas downregulation of EGFL6 diminishes osteoblastic mineralization. Interestingly, osteoblast differentiation was not affected by the exogenous addition of EGFL6 protein, thereby indicating that EGFL6 may regulate osteoblastic function in an intracrine manner. Mice with osteoblast-specific and global knockout of EGFL6 surprisingly exhibit a normal bone phenotype under physiological conditions. However, EGFL6-deficiency leads to compromised bone repair in a bone defect model which is characterized by decreased formation of type H vessels as well as osteoblast lineage cells. Conclusions: Together, these data demonstrate that EGFL6 serves as an essential regulator to couple osteogenesis to angiogenesis during bone repair.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neovascularización Fisiológica/fisiología , Osteogénesis/fisiología , Animales , Células de la Médula Ósea/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Regeneración Ósea/fisiología , Huesos/metabolismo , Proteínas de Unión al Calcio/fisiología , Moléculas de Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Femenino , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Cultivo Primario de Células , Transducción de Señal , Proteínas Smad/metabolismo
6.
Int J Oncol ; 59(5)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34664682

RESUMEN

The Nectin cell adhesion molecule (Nectin) family members are Ca2+­independent immunoglobulin­like cellular adhesion molecules (including Nectins 1­4), involved in cell adhesion via homophilic/heterophilic interplay. In addition, the Nectin family plays a significant role in enhancing cellular viability and movement ability. In contrast to enrichment of Nectins 1­3 in normal tissues, Nectin­4 is particularly overexpressed in a number of tumor types, including breast, lung, urothelial, colorectal, pancreatic and ovarian cancer. Moreover, the upregulation of Nectin­4 is an independent biomarker for overall survival in numerous cancer types. A large number of studies have revealed that high expression of Nectin­4 is closely related to tumor occurrence and development in various cancer types, but the manner in which Nectin­4 protein contributes to the onset and development of these malignancies is yet unknown. The present review summarizes the molecular mechanisms and functions of Nectin­4 protein in the biological processes and current advances with regard to its expression and regulation in various cancer types.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Neoplasias/etiología , Anticuerpos Monoclonales/farmacología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/química , Ensayos Clínicos como Asunto , Transición Epitelial-Mesenquimal , Humanos , Neoplasias/terapia , Neovascularización Patológica/etiología , Viroterapia Oncolítica , Transducción de Señal/fisiología
7.
PLoS Genet ; 17(9): e1009778, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491997

RESUMEN

Meiosis initiation and progression are regulated by both germ cells and gonadal somatic cells. However, little is known about what genes or proteins connecting somatic and germ cells are required for this regulation. Our results show that deficiency for adhesion molecule IGSF11, which is expressed in both Sertoli cells and germ cells, leads to male infertility in mice. Combining a new meiotic fluorescent reporter system with testicular cell transplantation, we demonstrated that IGSF11 is required in both somatic cells and spermatogenic cells for primary spermatocyte development. In the absence of IGSF11, spermatocytes proceed through pachytene, but the pericentric heterochromatin of nonhomologous chromosomes remains inappropriately clustered from late pachytene onward, resulting in undissolved interchromosomal interactions. Hi-C analysis reveals elevated levels of interchromosomal interactions occurring mostly at the chromosome ends. Collectively, our data elucidates that IGSF11 in somatic cells and germ cells is required for pericentric heterochromatin dissociation during diplotene in mouse primary spermatocytes.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Heterocromatina/metabolismo , Inmunoglobulinas/fisiología , Profase Meiótica I , Animales , Moléculas de Adhesión Celular/genética , Cromosomas , Femenino , Genes Reporteros , Humanos , Inmunoglobulinas/genética , Masculino , Ratones , Células de Sertoli/metabolismo , Espermatocitos/metabolismo
8.
Mol Nutr Food Res ; 65(20): e2001010, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390195

RESUMEN

SCOPE: Konjac glucomannan oligosaccharides (KMOS) are prebiotics and may improve intestinal immunity through modulation of macrophage function. However, the underlying molecular mechanisms were unclear. METHODS AND RESULTS: Using a mouse model of dextran sulfated sodium (DSS)-induced acute colitis, the study demonstrates here that KMOS (400 mg-1 kg-1 d-1 ) can ameliorate intestinal inflammation in a macrophage dependent manner. Oral exposure to KMOS prevents DSS-induced intestinal pathology, improves epithelial integrity, and decreases accumulation of colonic inflammatory leukocytes and cytokines. The therapeutic effects of KMOS are dependent on the function of macrophages, as depletion of macrophages abolished the effects. In colonic lamina propria of DSS-treated mice, as well as in vitro culture of bone marrow derived macrophages (BMDMs), KMOS skews reprogramming of classically activated macrophages (CAM/M1) into alternatively activated macrophages (AAM/M2). The study further determines that the activation of SIGNR1/phospho-c-Raf (S338)/phospho-p65 (S276)/acetyl-p65 (K310) pathway is responsible for KMOS-induced AAM/M2 polarization. Blockage of SIGNR1 abolishes KMOS-induced AAM/M2 polarization of activated macrophages, expression of phospho-p65 (S276) in colonic macrophages, and alleviation of DSS-induced colitis in mice, suggesting that SIGNR1 is critical for macrophage responses to KMOS. CONCLUSIONS: This study reveals a SIGNR1-mediated macrophage-dependent pathway that supports regulatory function of KMOS in host immunity and intestinal homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Colitis/prevención & control , Lectinas Tipo C/fisiología , Activación de Macrófagos/fisiología , Mananos/farmacología , Oligosacáridos/farmacología , Prebióticos , Receptores de Superficie Celular/fisiología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
9.
Drug Dev Res ; 82(8): 1096-1110, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34462935

RESUMEN

Trophoblast cell-surface antigen 2 (Trop 2) is a transmembrane glycoprotein that is highly expressed in various cancer types with relatively low or no baseline expression in most normal tissues. Its overexpression is associated with tumor growth and poor prognosis; Trop 2 is, therefore, an ideal therapeutic target for epithelial cancers. Several Trop 2 targeted therapeutics have recently been developed for the treatment of cancers, such as anti-Trop 2 antibodies and antibody-drug conjugates (ADCs), as well as Trop 2-specific cell therapy. In particular, the safety and clinical benefit of Trop 2-based ADCs have been demonstrated in clinical trials across multiple tumor types, including those with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and heavily pretreated non-small cell lung cancer. In this review, we elaborate on recent advances in Trop 2 targeted modalities and provide an overview of novel insights for future developments in this field.


Asunto(s)
Moléculas de Adhesión Celular/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva
10.
J Clin Invest ; 131(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34403362

RESUMEN

The secreted protein developmental endothelial locus 1 (DEL-1) regulates inflammatory cell recruitment and protects against inflammatory pathologies in animal models. Here, we investigated DEL-1 in inflammatory arthritis using collagen-induced arthritis (CIA) and collagen Ab-induced arthritis (CAIA) models. In both models, mice with endothelium-specific overexpression of DEL-1 were protected from arthritis relative to WT controls, whereas arthritis was exacerbated in DEL-1-deficient mice. Compared with WT controls, mice with collagen VI promoter-driven overexpression of DEL-1 in mesenchymal cells were protected against CIA but not CAIA, suggesting a role for DEL-1 in the induction of the arthritogenic Ab response. Indeed, DEL-1 was expressed in perivascular stromal cells of the lymph nodes and inhibited Tfh and germinal center B cell responses. Mechanistically, DEL-1 inhibited DC-dependent induction of Tfh cells by targeting the LFA-1 integrin on T cells. Overall, DEL-1 restrained arthritis through a dual mechanism, one acting locally in the joints and associated with the anti-recruitment function of endothelial cell-derived DEL-1; the other mechanism acting systemically in the lymph nodes and associated with the ability of stromal cell-derived DEL-1 to restrain Tfh responses. DEL-1 may therefore be a promising therapeutic for the treatment of inflammatory arthritis.


Asunto(s)
Artritis Experimental/prevención & control , Proteínas de Unión al Calcio/fisiología , Moléculas de Adhesión Celular/fisiología , Activación de Linfocitos , Células T Auxiliares Foliculares/inmunología , Animales , Diferenciación Celular , Femenino , Centro Germinal/inmunología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Células del Estroma/química , Células T Auxiliares Foliculares/citología
11.
J Biol Chem ; 297(3): 101080, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403696

RESUMEN

TIN2 is a core component of the shelterin complex linking double-stranded telomeric DNA-binding proteins (TRF1 and TRF2) and single-strand overhang-binding proteins (TPP1-POT1). In vivo, the large majority of TRF1 and TRF2 exist in complexes containing TIN2 but lacking TPP1/POT1; however, the role of TRF1-TIN2 interactions in mediating interactions with telomeric DNA is unclear. Here, we investigated DNA molecular structures promoted by TRF1-TIN2 interaction using atomic force microscopy (AFM), total internal reflection fluorescence microscopy (TIRFM), and the DNA tightrope assay. We demonstrate that the short (TIN2S) and long (TIN2L) isoforms of TIN2 facilitate TRF1-mediated DNA compaction (cis-interactions) and DNA-DNA bridging (trans-interactions) in a telomeric sequence- and length-dependent manner. On the short telomeric DNA substrate (six TTAGGG repeats), the majority of TRF1-mediated telomeric DNA-DNA bridging events are transient with a lifetime of ~1.95 s. On longer DNA substrates (270 TTAGGG repeats), TIN2 forms multiprotein complexes with TRF1 and stabilizes TRF1-mediated DNA-DNA bridging events that last on the order of minutes. Preincubation of TRF1 with its regulator protein Tankyrase 1 and the cofactor NAD+ significantly reduced TRF1-TIN2 mediated DNA-DNA bridging, whereas TIN2 protected the disassembly of TRF1-TIN2 mediated DNA-DNA bridging upon Tankyrase 1 addition. Furthermore, we showed that TPP1 inhibits TRF1-TIN2L-mediated DNA-DNA bridging. Our study, together with previous findings, supports a molecular model in which protein assemblies at telomeres are heterogeneous with distinct subcomplexes and full shelterin complexes playing distinct roles in telomere protection and elongation.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Moléculas de Adhesión Celular/fisiología , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Microscopía de Fuerza Atómica/métodos , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Complejo Shelterina/metabolismo , Complejo Shelterina/fisiología , Telómero/metabolismo , Proteínas de Unión a Telómeros/fisiología , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/fisiología , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología
12.
Int J Mol Sci ; 22(12)2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-34205668

RESUMEN

Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Proteínas de la Matriz Extracelular/fisiología , Boca/crecimiento & desarrollo , Osteonectina/fisiología , Trombospondinas/fisiología , Animales , Proteínas CCN de Señalización Intercelular/fisiología , Neoplasias de Cabeza y Cuello/etiología , Humanos , Boca/embriología , Tenascina/fisiología , Cicatrización de Heridas
13.
Biochem Pharmacol ; 192: 114672, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34237338

RESUMEN

Pnn, a multiple functional protein, plays roles in embryonic development, cellular differentiation, tumorigenesis, and metastasis. In the past two decades, the functions of Pnn in regulating RNA alternative splicing, gene regulation, and cell-cell connection have been revealed. Although Pnn is originally identified as a desmosome-associated protein for linking desmosome and intermediated filament, emerging evidence implies that Pnn not only is a desmosome protein but also plays critical roles in the nucleus. To date, through cell biology investigation and the generation of animal models with genetic manipulation, the physiological role of Pnn has been characterized in the research fields of developmental biology, tumor biology, and neuroscience. Through proteomic and molecular biology studies, transcription regulators, splicing regulators, and cytoskeletal proteins were found to interact with Pnn. In addition, histopathological and biochemical evidence has pointed to an association of Pnn expression level with tumorigenesis and metastasis. A previous clinical study also demonstrated a correlation between a reduced expression of Pnn and human dementia. Besides, experimental studies showed a protective role of Pnn against ischemic stress in astrocytes. All indicated a variety of roles of Pnn in different cell types. In this review article, we introduced the role of Pnn in embryogenesis and pathogenesis as well as discussed its potential clinical application.


Asunto(s)
Carcinogénesis/metabolismo , Moléculas de Adhesión Celular/fisiología , Proliferación Celular/fisiología , Desarrollo Embrionario/fisiología , Proteínas Nucleares/fisiología , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Humanos
14.
Front Immunol ; 12: 705206, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34290715

RESUMEN

Different body systems (epidermis, respiratory tract, cornea, oral cavity, and gastrointestinal tract) are in continuous direct contact with innocuous and/or potentially harmful external agents, exhibiting dynamic and highly selective interaction throughout the epithelia, which function as both a physical and chemical protective barrier. Resident immune cells in the epithelia are constantly challenged and must distinguish among antigens that must be either tolerated or those to which a response must be mounted for. When such a decision begins to take place in lymphoid foci and/or mucosa-associated lymphoid tissues, the epithelia network of immune surveillance actively dominates both oral and gastrointestinal compartments, which are thought to operate in the same immune continuum. However, anatomical variations clearly differentiate immune processes in both the mouth and gastrointestinal tract that demonstrate a wide array of independent immune responses. From single vs. multiple epithelia cell layers, widespread cell-to-cell junction types, microbial-associated recognition receptors, dendritic cell function as well as related signaling, the objective of this review is to specifically contrast the current knowledge of oral versus gut immune niches in the context of epithelia/lymphoid foci/MALT local immunity and systemic output. Related differences in 1) anatomy 2) cell-to-cell communication 3) antigen capture/processing/presentation 4) signaling in regulatory vs. proinflammatory responses and 5) systemic output consequences and its relations to disease pathogenesis are discussed.


Asunto(s)
Alostasis , Homeostasis , Inmunidad Mucosa/inmunología , Vigilancia Inmunológica/inmunología , Mucosa Intestinal/inmunología , Mucosa Bucal/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Traslocación Bacteriana/inmunología , Moléculas de Adhesión Celular/fisiología , Comunicación Celular , Células Dendríticas/inmunología , Disbiosis/inmunología , Células Epiteliales/inmunología , Humanos , Inflamación , Uniones Intercelulares/fisiología , Mucosa Intestinal/citología , Microbiota , Mucosa Bucal/citología , Moco/fisiología , Especificidad de Órganos , Saliva/inmunología , Transducción de Señal
15.
Commun Biol ; 4(1): 763, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34155338

RESUMEN

Mechanical forces control cell behavior, including cancer progression. Cells sense forces through actomyosin to activate YAP. However, the regulators of F-actin dynamics playing relevant roles during mechanostransduction in vitro and in vivo remain poorly characterized. Here we identify the Fascin1 F-actin bundling protein as a factor that sustains YAP activation in response to ECM mechanical cues. This is conserved in the mouse liver, where Fascin1 regulates YAP-dependent phenotypes, and in human cholangiocarcinoma cell lines. Moreover, this is relevant for liver tumorigenesis, because Fascin1 is required in the AKT/NICD cholangiocarcinogenesis model and it is sufficient, together with AKT, to induce cholangiocellular lesions in mice, recapitulating genetic YAP requirements. In support of these findings, Fascin1 expression in human intrahepatic cholangiocarcinomas strongly correlates with poor patient prognosis. We propose that Fascin1 represents a pro-oncogenic mechanism that can be exploited during intrahepatic cholangiocarcinoma development to overcome a mechanical tumor-suppressive environment.


Asunto(s)
Neoplasias de los Conductos Biliares/etiología , Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , Colangiocarcinoma/etiología , Mecanotransducción Celular/fisiología , Proteínas de Microfilamentos/fisiología , Factores de Transcripción/fisiología , Complejo 2-3 Proteico Relacionado con la Actina/fisiología , Animales , Proteína CapZ/fisiología , Moléculas de Adhesión Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Fosfoproteínas/fisiología
16.
J Invest Dermatol ; 141(11): 2602-2610.e3, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33965403

RESUMEN

The skin epidermis is attached to the underlying dermis by a laminin 332 (Lm332)-rich basement membrane. Consequently, loss of Lm332 leads to the severe blistering disorder epidermolysis bullosa junctionalis in humans and animals. Owing to the indispensable role of Lm332 in keratinocyte adhesion in vivo, the severity of the disease has limited research into other functions of the protein. We have conditionally disrupted Lm332 expression in basal keratinocytes of adult mice. Although blisters develop along the interfollicular epidermis, hair follicle basal cells provide sufficient anchorage of the epidermis to the dermis, making inducible deletion of the Lama3 gene compatible with life. Loss of Lm332 promoted the thickening of the epidermis and exaggerated desquamation. Global RNA expression analysis revealed major changes in the expression of keratins, cornified envelope proteins, and cellular stress markers. These modifications of the keratinocyte genetic program are accompanied by changes in cell shape and disorganization of the actin cytoskeleton. These data indicate that loss of Lm332-mediated progenitor cell adhesion alters cell fate and disturbs epidermal homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Homeostasis/fisiología , Queratinocitos/citología , Citoesqueleto de Actina/fisiología , Alarminas/fisiología , Animales , Vesícula/etiología , Diferenciación Celular , Epidermis/patología , Queratinas/análisis , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Kalinina
17.
J Invest Dermatol ; 141(10): 2338-2343, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34023128

RESUMEN

Periostin, an extracellular matrix and matricellular protein, binds to several types of integrins that transduce its signals. Its function in allergic inflammation is the establishment of sustained chronic inflammation through an amplification of T helper type 2‒immune responses. In addition, recent studies have shown a significant role of periostin in itch sensation through direct integrin-mediated stimulation of nerve fibers and interaction with immune and nonimmune cells (e.g., macrophages, eosinophils, basophils, and keratinocytes). The objective of this review is to describe the role of periostin in itch induction in human and animal models and its expression in human pruritic conditions.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Prurito/etiología , Animales , Humanos , Integrinas/fisiología , Péptido Natriurético Encefálico/fisiología , Sensación , Canal Catiónico TRPA1/fisiología
18.
Curr Eye Res ; 46(11): 1751-1761, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33988065

RESUMEN

Purpose: Circular RNAs (circRNAs) are essential regulators in tumorigenesis and development. In this study, we focused on the functions of circRNA muskelin 1 (circMKLN1) in retinoblastoma (RB) progression.Materials and Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) assay was conducted to determine the levels of circMKLN1, microRNA-425-5p (miR-425-5p) and programmed cell death 4 (PDCD4). The characteristic of circMKLN1 was analyzed using RNase R assay. Cell Counting Kit-8 (CCK-8) assay and colony formation assay were employed to explore cell proliferation ability. The transwell assay was utilized for cell migration and invasion. A Western blot assay was performed for protein levels. The dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to demonstrate the relationships among circMKLN1, miR-425-5p and PDCD4. Murine xenograft model assay was adopted to investigate the role of circMKLN1 in vivo.Results: CircMKLN1 was downregulated in RB tissues and cells. High levels of circMKLN1 were related to a favorable outcome of RB patients. CircMKLN1 was resistant to RNase R digestion and circMKLN1 overexpression repressed RB cell proliferation, migration and invasion in vitro. MiR-425-5p was identified as the target of circMKLN1 and miR-425-5p elevation reversed the effects of circMKLN1 overexpression on RB cell malignant behaviors. Furthermore, as the target gene of miR-425-5p, PDCD4 silencing could ameliorate the suppressive roles of circMKLN1 in RB cell growth and metastasis. Additionally, circMKLN1 overexpression hampered tumor growth in vivo.Conclusions: CircMKLN1 overexpression decelerated the progression of RB through sponging miR-425-5p and elevating PDCD4.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Moléculas de Adhesión Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , MicroARNs/genética , ARN Circular/fisiología , Proteínas de Unión al ARN/genética , Neoplasias de la Retina/fisiopatología , Retinoblastoma/fisiopatología , Animales , Western Blotting , Recuento de Células , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Progresión de la Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Retina/genética , Retinoblastoma/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807264

RESUMEN

Skeletal muscle regeneration is a well-organized process that requires remodeling of the extracellular matrix (ECM). In this study, we revealed the protective role of periostin, a matricellular protein that binds to several ECM proteins during muscle regeneration. In intact muscle, periostin was localized at the neuromuscular junction, muscle spindle, and myotendinous junction, which are connection sites between muscle fibers and nerves or tendons. During muscle regeneration, periostin exhibited robustly increased expression and localization at the interstitial space. Periostin-null mice showed decreased muscle weight due to the loss of muscle fibers during repeated muscle regeneration. Cultured muscle progenitor cells from periostin-null mice showed no deficiencies in their proliferation, differentiation, and the expression of Pax7, MyoD, and myogenin, suggesting that the loss of muscle fibers in periostin-null mice was not due to the impaired function of muscle stem/progenitor cells. Periostin-null mice displayed a decreased number of CD31-positive blood vessels during muscle regeneration, suggesting that the decreased nutritional supply from blood vessels was the cause of muscle fiber loss in periostin-null mice. These results highlight the novel role of periostin in maintaining muscle mass during muscle regeneration.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Regeneración/fisiología , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Diferenciación Celular , Uniones Célula-Matriz/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Tendones/metabolismo , Cicatrización de Heridas/fisiología
20.
J Virol ; 95(14): e0052821, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33910952

RESUMEN

Measles virus (MeV), an enveloped RNA virus in the family Paramyxoviridae, is still an important cause of childhood morbidity and mortality worldwide. MeV usually causes acute febrile illness with skin rash, but in rare cases persists in the brain, causing a progressive neurological disorder, subacute sclerosing panencephalitis (SSPE). The disease is fatal, and no effective therapy is currently available. Although transsynaptic cell-to-cell transmission is thought to account for MeV propagation in the brain, neurons do not express the known receptors for MeV. Recent studies have shown that hyperfusogenic changes in the MeV fusion (F) protein play a key role in MeV propagation in the brain. However, how such mutant viruses spread in neurons remains unexplained. Here, we show that cell adhesion molecule 1 (CADM1; also known as IGSF4A, Necl-2, and SynCAM1) and CADM2 (also known as IGSF4D, Necl-3, SynCAM2) are host factors that enable MeV to cause membrane fusion in cells lacking the known receptors and to spread between neurons. During enveloped virus entry, a cellular receptor generally interacts in trans with the attachment protein on the envelope. However, CADM1 and CADM2 interact in cis with the MeV attachment protein on the same cell membrane, causing the fusion protein triggering and membrane fusion. Knockdown of CADM1 and CADM2 inhibits syncytium formation and virus transmission between neurons that are both mediated by hyperfusogenic F proteins. Thus, our results unravel the molecular mechanism (receptor-mimicking cis-acting fusion triggering) by which MeV spreads transsynaptically between neurons, thereby causing SSPE. IMPORTANCE Measles virus (MeV), an enveloped RNA virus, is the causative agent of measles, which is still an important cause of childhood morbidity and mortality worldwide. Persistent MeV infection in the brain causes a fatal progressive neurological disorder, subacute sclerosing panencephalitis (SSPE), several years after acute infection. However, how MeV spreads in neurons, which are mainly affected in SSPE, remains largely unknown. In this study, we demonstrate that cell adhesion molecule 1 (CADM1) and CADM2 are host factors enabling MeV spread between neurons. During enveloped virus entry, a cellular receptor generally interacts in trans with the attachment protein on the viral membrane (envelope). Remarkably, CADM1 and CADM2 interact in cis with the MeV attachment protein on the same membrane, triggering the fusion protein and causing membrane fusion, as viral receptors usually do in trans. Careful screening may lead to more examples of such "receptor-mimicking cis-acting fusion triggering" in other viruses.


Asunto(s)
Molécula 1 de Adhesión Celular/fisiología , Moléculas de Adhesión Celular/fisiología , Virus del Sarampión/patogenicidad , Panencefalitis Esclerosante Subaguda/virología , Internalización del Virus , Animales , Línea Celular , Chlorocebus aethiops , Células Gigantes/virología , Humanos , Ratones , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...