Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 547
Filtrar
2.
J Hepatol ; 81(1): 135-148, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38460791

RESUMEN

BACKGROUND & AIMS: NOTCH signaling in liver sinusoidal endothelial cells (LSECs) regulates liver fibrosis, a pathological feature of chronic liver diseases. POFUT1 is an essential regulator of NOTCH signaling. Here, we investigated the role of LSEC-expressed POFUT1 in liver fibrosis. METHODS: Endothelial-specific Pofut1 knockout mice were generated and experimental liver fibrosis was induced by chronic carbon tetrachloride exposure or common bile duct ligation. Liver samples were assessed by ELISA, histology, electron microscopy, immunostaining and RNA in situ hybridization. LSECs and hepatic stellate cells (HSCs) were isolated for gene expression analysis by RNA sequencing, qPCR, and western blotting. Signaling crosstalk between LSECs and HSCs was investigated by treating HSCs with supernatant from LSEC cultures. Liver single-cell RNA sequencing datasets from patients with cirrhosis and healthy individuals were analyzed to evaluate the clinical relevance of gene expression changes observed in mouse studies. RESULTS: POFUT1 loss promoted injury-induced LSEC capillarization and HSC activation, leading to aggravated liver fibrosis. RNA sequencing analysis revealed that POFUT1 deficiency upregulated fibrinogen expression in LSECs. Consistently, fibrinogen was elevated in LSECs of patients with cirrhosis. HSCs treated with supernatant from LSECs of Pofut1 null mice showed exacerbated activation compared to those treated with supernatant from control LSECs, and this effect was attenuated by knockdown of fibrinogen or by pharmacological inhibition of fibrinogen receptor signaling, altogether suggesting that LSEC-derived fibrinogen induced the activation of HSCs. Mechanistically, POFUT1 loss augmented fibrinogen expression by enhancing NOTCH/HES1/STAT3 signaling. CONCLUSIONS: Endothelial POFUT1 prevents injury-induced liver fibrosis by repressing the expression of fibrinogen, which functions as a profibrotic paracrine signal to activate HSCs. Therapies targeting the POFUT1/fibrinogen axis offer a promising strategy for the prevention and treatment of fibrotic liver diseases. IMPACT AND IMPLICATIONS: Paracrine signals produced by liver vasculature play a major role in the development of liver fibrosis, which is a pathological hallmark of most liver diseases. Identifying those paracrine signals is clinically relevant in that they may serve as therapeutic targets. In this study, we discovered that genetic deletion of Pofut1 aggravated experimental liver fibrosis in mouse models. Moreover, fibrinogen was identified as a downstream target repressed by Pofut1 in liver endothelial cells and functioned as a novel paracrine signal that drove liver fibrosis. In addition, fibrinogen was found to be relevant to cirrhosis and may serve as a potential therapeutic target for this devastating human disease.


Asunto(s)
Células Endoteliales , Fibrinógeno , Células Estrelladas Hepáticas , Cirrosis Hepática , Ratones Noqueados , Animales , Humanos , Masculino , Ratones , Tetracloruro de Carbono/toxicidad , Tetracloruro de Carbono/efectos adversos , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Fibrinógeno/metabolismo , Fibrinógeno/biosíntesis , Fibrinógeno/genética , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Receptores Notch/metabolismo , Receptores Notch/fisiología , Transducción de Señal
3.
Med Oncol ; 38(12): 139, 2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34633549

RESUMEN

Notch signaling involves cell to cell contact. It is an ancient signaling mechanism that is conserved throughout the animal kingdom. The basic function of Notch signaling is to decide cell fate and execute asymmetrical division. Notch signaling is indispensable for embryo growth. Aberrant Notch signaling involves in cancer progression by altering cell proliferation rate, tumor micro-environment, stem cell activities. The role of Notch signaling in cancer progression is context-dependent. In breast cancer and T cell lymphoma Notch signaling is highly active, whereas in squamous cell carcinoma (SCC) as oral and skin cancer, the signaling is suppressed. It is believed that in SCC, Notch-mediated tumor growth is due to the cell non-autonomous function. Oral cancer is the 6th most risky cancer worldwide. In many patients, oral cancer is preceded by pre-cancer conditions. In this review, we have summarized the research knowledge related to the role of Notch signaling in oral cancer and pre-cancer conditions and the therapeutic options available targeting different components of Notch pathways.


Asunto(s)
Neoplasias de la Boca/etiología , Lesiones Precancerosas/etiología , Receptores Notch/fisiología , Humanos , Células Madre Neoplásicas/fisiología , Receptores Notch/antagonistas & inhibidores , Transducción de Señal/fisiología
4.
Future Oncol ; 17(31): 4185-4206, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34342489

RESUMEN

Triple-negative breast cancer (TNBC) is the most complex, aggressive and fatal subtype of breast cancer. Owing to the lack of targeted therapy and heterogenic nature of TNBC, chemotherapy remains the sole treatment option for TNBC, with taxanes and anthracyclines representing the general chemotherapeutic regimen in TNBC therapy. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. Breast cancer stem cells (BCSCs) are one of the major causes for the development of chemoresistance in TNBC patients. After surviving the chemotherapy damage, the presence of BCSCs results in relapse and recurrence of TNBC. Several pathways are known to regulate BCSCs' survival, such as the Wnt/ß-catenin, Hedgehog, JAK/STAT and HIPPO pathways. Therefore it is imperative to target these pathways in the context of eliminating chemoresistance. In this review we will discuss the novel strategies and various preclinical and clinical studies to give an insight into overcoming TNBC chemoresistance. We present a detailed account of recent studies carried out that open an exciting perspective in relation to the mechanisms of chemoresistance.


Asunto(s)
Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP/fisiología , Supervivencia Celular , Resistencia a Antineoplásicos , Femenino , Proteínas Hedgehog/fisiología , Vía de Señalización Hippo , Humanos , FN-kappa B/fisiología , Receptores Notch/fisiología , Neoplasias de la Mama Triple Negativas/patología , Vía de Señalización Wnt
5.
Biomolecules ; 11(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34200313

RESUMEN

Notch signaling forms an evolutionarily conserved juxtacrine pathway crucial for cellular development. Initially identified in Drosophila wing morphogenesis, Notch signaling has since been demonstrated to play pivotal roles in governing mammalian cellular development in a large variety of cell types. Indeed, abolishing Notch constituents in mouse models result in embryonic lethality, demonstrating that Notch signaling is critical for development and differentiation. In this review, we focus on the crucial role of Notch signaling in governing embryogenesis and differentiation of multiple progenitor cell types. Using hematopoiesis as a diverse cellular model, we highlight the role of Notch in regulating the cell fate of common lymphoid progenitors. Additionally, the influence of Notch through microenvironment interplay with lymphoid cells and how dysregulation influences disease processes is explored. Furthermore, bi-directional and lateral Notch signaling between ligand expressing source cells and target cells are investigated, indicating potentially novel therapeutic options for treatment of Notch-mediated diseases. Finally, we discuss the role of cis-inhibition in regulating Notch signaling in mammalian development.


Asunto(s)
Linaje de la Célula/fisiología , Desarrollo Embrionario/fisiología , Linfopoyesis/fisiología , Receptores Notch/fisiología , Animales , Humanos , Linfocitos/fisiología , Transducción de Señal/fisiología
6.
Neural Plast ; 2021: 5522717, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194486

RESUMEN

The loss of auditory sensory hair cells (HCs) is the most common cause of sensorineural hearing loss (SNHL). As the main sound transmission structure in the cochlea, it is necessary to maintain the normal shape and survival of HCs. In this review, we described and summarized the signaling pathways that regulate the development and survival of auditory HCs in SNHL. The role of the mitogen-activated protein kinase (MAPK), phosphoinositide-3 kinase/protein kinase B (PI3K/Akt), Notch/Wnt/Atoh1, calcium channels, and oxidative stress/reactive oxygen species (ROS) signaling pathways are the most relevant. The molecular interactions of these signaling pathways play an important role in the survival of HCs, which may provide a theoretical basis and possible therapeutic interventions for the treatment of hearing loss.


Asunto(s)
Células Ciliadas Auditivas/fisiología , Transducción de Señal/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Canales de Calcio/fisiología , Supervivencia Celular , Pérdida Auditiva Sensorineural/patología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores Notch/fisiología , Vía de Señalización Wnt/fisiología
7.
Theranostics ; 11(13): 6526-6541, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995674

RESUMEN

The treatment for metastatic castration-resistant prostate cancer patients remains a great challenge in the clinic and continuously demands discoveries of new targets and therapies. Here, we assess the function and therapeutic value of SIRT6 in metastatic castration-resistant prostate cancer. Methods: The expression of SIRT6 was examined in prostate cancer tissue microarray by immunohistochemistry staining. The functions of SIRT6 and underlying mechanisms were elucidated by in vitro and in vivo experiments. We also developed an efficient method to silence SIRT6 by aptamer-modified exosomes carrying small interfering RNA and tested the therapeutic effect in the xenograft mice models. Results: SIRT6 expression is positively correlated with prostate cancer progression. Loss of SIRT6 significantly suppressed proliferation and metastasis of prostate cancer cell lines both in vitro and in vivo. SIRT6-driven prostate cancer displays activation of multiple cancer-related signaling pathways, especially the Notch pathway. Silencing SIRT6 by siRNA delivered through engineered exosomes inhibited tumor growth and metastasis. Conclusions: SIRT6 is identified as a driver and therapeutic target for metastatic prostate cancer in our findings, and inhibition of SIRT6 by engineered exosomes can serve as a promising therapeutic tool for clinical application.


Asunto(s)
Adenocarcinoma/terapia , Exosomas , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Próstata/terapia , Sirtuinas/antagonistas & inhibidores , Adenocarcinoma/patología , Animales , Aptámeros de Nucleótidos , Carcinogénesis , Línea Celular Tumoral , ADN Complementario/genética , Progresión de la Enfermedad , Electroporación , Vectores Genéticos/farmacología , Vectores Genéticos/uso terapéutico , Células HEK293 , Humanos , Lentivirus/genética , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Receptores Notch/fisiología , Transducción de Señal , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Commun Biol ; 4(1): 290, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674719

RESUMEN

SARS-CoV-2 virus has infected more than 92 million people worldwide resulting in the Coronavirus disease 2019 (COVID-19). Using a rhesus macaque model of SARS-CoV-2 infection, we have characterized the transcriptional signatures induced in the lungs of juvenile and old macaques following infection. Genes associated with Interferon (IFN) signaling, neutrophil degranulation and innate immune pathways are significantly induced in macaque infected lungs, while pathways associated with collagen formation are downregulated, as also seen in lungs of macaques with tuberculosis. In COVID-19, increasing age is a significant risk factor for poor prognosis and increased mortality. Type I IFN and Notch signaling pathways are significantly upregulated in lungs of juvenile infected macaques when compared with old infected macaques. These results are corroborated with increased peripheral neutrophil counts and neutrophil lymphocyte ratio in older individuals with COVID-19 disease. Together, our transcriptomic studies have delineated disease pathways that improve our understanding of the immunopathogenesis of COVID-19.


Asunto(s)
COVID-19/inmunología , Degranulación de la Célula , Interferones/fisiología , Neutrófilos/fisiología , SARS-CoV-2 , Anciano , Animales , Antígenos CD36/fisiología , COVID-19/etiología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Pulmón/metabolismo , Macaca mulatta , Masculino , Persona de Mediana Edad , Receptores Notch/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/fisiología
10.
Cells ; 10(1)2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33374160

RESUMEN

Notch receptors are single-pass transmembrane proteins that play a critical role in cell fate decisions and have been implicated in the regulation of many developmental processes. The human Notch family comprises of four receptors (Notch 1 to 4) and five ligands. Their signaling can regulate extremely basic cellular processes such as differentiation, proliferation and death. Notch is also involved in hematopoiesis and angiogenesis, and increasing evidence suggests that these genes are involved and frequently deregulated in several human malignancies, contributing to cell autonomous activities that may be either oncogenic or tumor suppressive. It was recently proposed that Notch signaling could play an active role in promoting and sustaining a broad spectrum of lymphoid malignancies as well as mutations in Notch family members that are present in several disorders of T- and B-cells, which could be responsible for altering the related signaling. Therefore, different Notch pathway molecules could be considered as potential therapeutic targets for hematological cancers. In this review, we will summarize and discuss compelling evidence pointing to Notch receptors as pleiotropic regulators of hematologic malignancies biology, first describing the physiological role of their signaling in T- and B-cell development and homeostasis, in order to fully understand the pathological alterations reported.


Asunto(s)
Linfocitos B , Neoplasias Hematológicas/metabolismo , Receptores Notch/fisiología , Linfocitos T , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Linfocitos B/patología , Humanos , Ratones , Transducción de Señal , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/patología
11.
Cells ; 9(10)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33076453

RESUMEN

Although the role of NOTCH signaling has been extensively studied in health and disease, many questions still remain unresolved. Being crucial for tissue homeostasis, NOTCH signaling is also implicated in multiple cancers by either promoting or suppressing tumor development. In this review we illustrate the context-dependent role of NOTCH signaling during tumorigenesis with a particular focus on gliomas, the most frequent and aggressive brain tumors in adults. For a long time, NOTCH has been considered an oncogene in glioma mainly by virtue of its neural stem cell-promoting activity. However, the recent identification of NOTCH-inactivating mutations in some glioma patients has challenged this notion, prompting a re-examination of the function of NOTCH in brain tumor subtypes. We discuss recent findings that might help to reconcile the controversial role of NOTCH signaling in this disease, and pose outstanding questions that still remain to be addressed.


Asunto(s)
Neoplasias Encefálicas/genética , Genes Supresores de Tumor/fisiología , Glioma/genética , Oncogenes/fisiología , Receptores Notch/fisiología , Transducción de Señal , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Carcinogénesis/patología , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Homeostasis , Humanos , Células Madre Neoplásicas/patología , Células-Madre Neurales
12.
Front Immunol ; 11: 368, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265900

RESUMEN

Introduction: Leprosy is an infectious disease caused by Mycobacterium leprae, a debilitating disease that affects the skin and peripheral nerves. It is possible that tissue changes during infection with leprosy are related to alterations in the activity of the Notch signaling pathway, an innate signaling pathway in the physiology of the skin and peripheral nerves. Methods: This is a descriptive observational study. Thirty skin biopsies from leprosy patients and 15 from individuals with no history of this disease were evaluated. In these samples, gene expressions of cellular components associated with the Notch signaling pathway, Hes-1, Hey-1, Runx-1 Jagged-1, Notch-1, and Numb, were evaluated using q-PCR, and protein expression was evaluated using immunohistochemistry of Runx-1 and Hes-1. Results: Changes were observed in the transcription of Notch signaling pathway components; Hes-1 was downregulated and Runx-1 upregulated in the skin of infected patients. These results were confirmed by immunohistochemistry, where reduction of Hes-1 expression was found in the epidermis, eccrine glands, and hair follicles. Increased expression of Runx-1 was found in inflammatory cells in the dermis of infected patients; however, it is not related to tissue changes. With these results, a multivariate analysis was performed to determine the causes of transcription factor Hes-1 reduction. It was concluded that tissue inflammation was the main cause. Conclusions: The tissue changes found in the skin of infected patients could be associated with a reduction in the expression of Hes-1, a situation that would promote the survival and proliferation of M. leprae in this tissue.


Asunto(s)
Lepra/metabolismo , Fibras Nerviosas/patología , Receptores Notch/fisiología , Piel/metabolismo , Adulto , Anciano , Subunidad alfa 2 del Factor de Unión al Sitio Principal/análisis , Ciclina D1/análisis , Femenino , Humanos , Inmunohistoquímica , Lepra/patología , Masculino , Persona de Mediana Edad , Fibras Nerviosas/química , Transducción de Señal/fisiología , Piel/patología , Factor de Transcripción HES-1/análisis
13.
Front Immunol ; 11: 499, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265938

RESUMEN

Generating inhibitors for A Disintegrin And Metalloproteinase 10 (ADAM10), a zinc-dependent protease, was heavily invested in by the pharmaceutical industry starting over 20 years ago. There has been much enthusiasm in basic research for these inhibitors, with a multitude of studies generating significant data, yet the clinical trials have not replicated the same results. ADAM10 is ubiquitously expressed and cleaves many important substrates such as Notch, PD-L1, EGFR/HER ligands, ICOS-L, TACI, and the "stress related molecules" MIC-A, MIC-B and ULBPs. This review goes through the most recent pre-clinical data with inhibitors as well as clinical data supporting the use of ADAM10 inhibitor use in cancer and autoimmunity. It additionally addresses how ADAM10 inhibitor therapy can be improved and if inhibitor therapy can be paired with other drug treatments to maximize effectiveness in various disease states. Finally, it examines the ADAM10 substrates that are important to each disease state and if any of these substrates or ADAM10 itself is a potential biomarker for disease.


Asunto(s)
Proteína ADAM10/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Enfermedades Autoinmunes/tratamiento farmacológico , Proteínas de la Membrana/antagonistas & inhibidores , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Ensayos Clínicos como Asunto , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Estudios Multicéntricos como Asunto , Neoplasias/enzimología , Neoplasias/inmunología , Inhibidores de Proteasas/farmacología , Receptores Notch/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Especificidad por Sustrato
14.
Biomed Pharmacother ; 127: 110141, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32334375

RESUMEN

Osteosarcoma is a bone tumor prevalent in children and young adults. LncRNAs are a family of non-protein-coding transcripts longer than 200 nucleotides. The tumor-related pathological functions of lncRNAs include proliferation, migration, and chemotherapy resistance, all of which have been widely acknowledged in research on osteosarcoma. In addition, compelling evidence suggests that lncRNAs could serve as diagnostic indicators, prognostic biomarkers, and targets for disease treatment. In this review, we systematically summarize how lncRNAs regulate tumorigenesis, invasion and therapeutic resistance. By deepening our knowledge of the relationship between lncRNAs and osteosarcoma, we hope to translate research findings into clinical applications as soon as possible.


Asunto(s)
Neoplasias Óseas/etiología , Osteosarcoma/etiología , ARN Largo no Codificante/fisiología , Neoplasias Óseas/patología , Carcinogénesis , Progresión de la Enfermedad , Humanos , Osteosarcoma/patología , Fosfatidilinositol 3-Quinasas/fisiología , Pronóstico , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores Notch/fisiología , Vía de Señalización Wnt/fisiología
15.
Dev Biol ; 462(1): 101-115, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32243888

RESUMEN

Drosophila Larval hematopoiesis takes place at the lymph gland, where myeloid-like progenitors differentiate into Plasmatocytes and Crystal Cells, under regulation of conserved signaling pathways. It has been established that the Notch pathway plays a specific role in Crystal Cell differentiation and maintenance. In mammalian hematopoiesis, the Notch pathway has been proposed to fulfill broader functions, including Hematopoietic Stem Cell maintenance and cell fate decision in progenitors. In this work we describe different roles that Notch plays in the lymph gland. We show that Notch, activated by its ligand Serrate, expressed at the Posterior Signaling Center, is required to restrain Core Progenitor differentiation. We define a novel population of blood cell progenitors that we name Distal Progenitors, where Notch, activated by Serrate expressed in Lineage Specifying Cells at the Medullary Zone/Cortical Zone boundary, regulates a binary decision between Plasmatocyte and Crystal Cell fates. Thus, Notch plays context-specific functions in different blood cell progenitor populations of the Drosophila lymph gland.


Asunto(s)
Células Madre Hematopoyéticas/citología , Ganglios Linfáticos/metabolismo , Receptores Notch/metabolismo , Animales , Células Sanguíneas/citología , Diferenciación Celular/fisiología , Linaje de la Célula , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Hematopoyesis/fisiología , Proteína Jagged-1/metabolismo , Larva/metabolismo , Receptores Notch/fisiología , Transducción de Señal/fisiología
16.
Stem Cells Transl Med ; 9(7): 799-812, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32297712

RESUMEN

Hyperactivity of the NOTCH pathway is associated with tumor growth and radiotherapy resistance in lung cancer, and NOTCH/γ-secretase inhibitors (GSIs) are a potential therapeutic target. The therapeutic outcome, however, is often restricted by the dose-limiting toxicity of combined treatments on the surrounding healthy tissue. The NOTCH signaling pathway is also crucial for homeostasis and repair of the normal airway epithelium. The effects of NOTCH/γ-secretase inhibition on the irradiation of normal lung epithelium are unknown and may counteract antitumor activity. Here we, therefore, investigated whether normal tissue toxicity to radiation is altered upon NOTCH pathway inhibition. We established air-liquid interface pseudostratified and polarized cultures from primary human bronchial epithelial cells and blocked NOTCH signaling alone or after irradiation with small-molecule NOTCH inhibitor/GSI. We found that the reduction in proliferation and viability of bronchial stem cells (TP63+) in response to irradiation is rescued with concomitant NOTCH inhibition. This correlated with reduced activation of the DNA damage response and accelerated repair by 24 hours and 3 days postirradiation. The increase in basal cell proliferation and viability in GSI-treated and irradiated cultures resulted in an improved epithelial barrier function. Comparable results were obtained after in vivo irradiation, where the combination of NOTCH inhibition and irradiation increased the percentage of stem cells and ciliated cells ex vivo. These encourage further use of normal patient tissue for toxicity screening of combination treatments and disclose novel interactions between NOTCH inhibition and radiotherapy and opportunities for tissue repair after radiotherapy.


Asunto(s)
Lesión Pulmonar/fisiopatología , Receptores Notch/fisiología , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Epiteliales , Humanos , Transducción de Señal
17.
Dev Genes Evol ; 230(3): 213-225, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31960122

RESUMEN

Size and shape constitute fundamental aspects in the description of morphology. Yet while the developmental-genetic underpinnings of trait size, in particular with regard to scaling relationships, are increasingly well understood, those of shape remain largely elusive. Here we investigate the potential function of the Notch signaling pathway in instructing the shape of beetle horns, a highly diversified and evolutionarily novel morphological structure. We focused on the bull-headed dung beetle Onthophagus taurus due to the wide range of horn sizes and shapes present among males in this species, in order to assess the potential function of Notch signaling in the specification of horn shape alongside the regulation of shape changes with allometry. Using RNA interference-mediated transcript depletion of Notch and its ligands, we document a highly conserved role of Notch signaling in general appendage formation. By integrating our functional genetic approach with a geometric morphometric analysis, we find that Notch signaling moderately but consistently affects horn shape, and does so differently for the horns of minor, intermediate-sized, and major males. Our results suggest that the function of Notch signaling during head horn formation may vary in a complex manner across male morphs, and highlights the power of integrating functional genetic and geometric morphometric approaches in analyzing subtle but nevertheless biologically important phenotypes in the face of significant allometric variation.


Asunto(s)
Tipificación del Cuerpo , Escarabajos/crecimiento & desarrollo , Escarabajos/genética , Receptores Notch/fisiología , Proteínas Serrate-Jagged/metabolismo , Transducción de Señal , Animales , Evolución Biológica , Escarabajos/anatomía & histología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes de Insecto , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Morfogénesis , Fenotipo , Interferencia de ARN , Proteínas Serrate-Jagged/genética , Caracteres Sexuales
18.
Cell Tissue Res ; 379(1): 169-179, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31781870

RESUMEN

Osteogenic differentiation is a tightly regulated process realized by progenitor cell osteoblasts. Notch signaling pathway plays a critical role in skeletal development and bone remodeling. Controversial data exist regarding the role of Notch activation in promoting or preventing osteogenic differentiation. This study aims to investigate the effect of several Notch components and their dosage on osteogenic differentiation of mesenchymal stem cells of adipose tissue. Osteogenic differentiation was induced in the presence of either of Notch components (NICD, Jag1, Dll1, Dll4) dosed by lentiviral transduction. We show that osteogenic differentiation was increased by NICD and Jag1 transduction in a dose-dependent manner; however, a high dosage of both NICD and Jag1 decreased the efficiency of osteogenic differentiation. NICD dose-dependently increased activity of the CSL luciferase reporter but a high dosage of NICD caused a decrease in the activity of the reporter. A high dosage of both Notch components NICD and Jag1 induced apoptosis. In co-culture experiments where only half of the cells were transduced with either NICD or Jag1, only NICD increased osteogenic differentiation according to the dosage, while Jag1-transduced cells differentiated almost equally independently on dosage. In conclusion, activation of Notch promotes osteogenic differentiation in a tissue-specific dose-dependent manner; both NICD and Jag1 are able to increase osteogenic potential but at moderate doses only and a high dosage of Notch activation is detrimental to osteogenic differentiation. This result might be especially important when considering possibilities of using Notch activation to promote osteogenesis in clinical applications to bone repair.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Osteogénesis , Receptores Notch/fisiología , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/fisiología , Lentivirus/genética , Osteogénesis/genética , Receptores Notch/genética , Transducción Genética
19.
Hepatology ; 71(1): 196-213, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211856

RESUMEN

Cholangiocarcinoma (CCA) mortality rates are increasing as a result of rising incidence and limited curative treatment(s) for patients with advanced disease. NOTCH pathway reactivation has been reported in biliary malignancies to conflicting degrees, hindering prioritization of key therapeutic targets within the network and identification of candidate responder patients for NOTCH-directed therapies. We analyzed genomic data from 341 patients with CCA and identified NOTCH1 significantly increased in a subgroup characterized by distinct stromal infiltration. Network-wide imbalance of the NOTCH pathway was seen in CCA, including correlation of NOTCH1 with NOTCH3 and NOTCH ligands. Given the diversity of observed NOTCH receptor engagement, γ-secretase modulation was rationalized as a therapeutic option. Indeed, subcutaneous transplantation of sensitive and resistant CCA cell lines pretreated with a γ-secretase inhibitor (GSi) cocktail demonstrated the antineoplastic effects of GSi in a subset of CCA and led to the development of a 225-gene responder signature. This signature was validated in an independent cohort of 119 patients. Further, this signature was enriched in liver tumors initiated by hydrodynamic injections of activated-NOTCH as compared with the AKT-RAS-driven tumors. Candidate GSi-responder patients were characterized by distinct transcriptomes overlapping with previous hepatobiliary metastasis and stemness, unique stromal properties, and dysfunctional intratumoral immune infiltration. Pan-cancer analysis identified 41.9% of cancer types to harbor prospective GSi-responder patients, which was adapted into a 20-gene GSi-sensitivity score metric capable of discriminating nanomolar versus micromolar sensitivity to a cell-permeable GSi (Z-LLNle-CHO) across 60 diverse tumor lines (area under the curve = 1). Conclusion: We have established a GSi-responder signature with evidence across several patient cohorts, as well as in vitro and in vivo models, to enable precision medicine application of NOTCH-directed therapy in CCA as well as prospectively across diverse malignancies.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Benzazepinas/farmacología , Benzazepinas/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/etiología , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/etiología , Dibenzazepinas/farmacología , Dibenzazepinas/uso terapéutico , Fluorenos/farmacología , Fluorenos/uso terapéutico , Cetonas/farmacología , Cetonas/uso terapéutico , Receptores Notch/efectos de los fármacos , Receptores Notch/fisiología , Línea Celular Tumoral , Humanos , Resultado del Tratamiento
20.
Proc Natl Acad Sci U S A ; 116(47): 23551-23561, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31685607

RESUMEN

Angiogenesis frequently occurs in the context of acute or persistent inflammation. The complex interplay of proinflammatory and proangiogenic cues is only partially understood. Using an experimental model, permitting exposure of developing blood vessel sprouts to multiple combinations of diverse biochemical stimuli and juxtacrine cell interactions, we present evidence that a proinflammatory cytokine, tumor necrosis factor (TNF), can have both proangiogenic and antiangiogenic effects, depending on the dose and the presence of pericytes. In particular, we find that pericytes can rescue and enhance angiogenesis in the presence of otherwise-inhibitory high TNF doses. This sharp switch from proangiogenic to antiangiogenic effect of TNF observed with an escalating dose of this cytokine, as well as the effect of pericytes, are explained by a mathematical model trained on the biochemical data. Furthermore, this model was predictive of the effects of diverse combinations of proinflammatory and antiinflammatory cues, and variable pericyte coverage. The mechanism supports the effect of TNF and pericytes as modulating signaling networks impinging on Notch signaling and specification of the Tip and Stalk phenotypes. This integrative analysis elucidates the plasticity of the angiogenic morphogenesis in the presence of diverse and potentially conflicting cues, with immediate implications for many physiological and pathological settings.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Pericitos/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Comunicación Celular , Técnicas de Cultivo de Célula , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Lisofosfolípidos/farmacología , Modelos Biológicos , Neovascularización Patológica/patología , Pericitos/efectos de los fármacos , Receptores Notch/fisiología , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/farmacología , Acetato de Tetradecanoilforbol/farmacología , Ingeniería de Tejidos , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA