Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
FASEB J ; 38(9): e23637, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38720403

RESUMEN

Vascular smooth muscle cell (VSMC) plasticity is fundamental in uterine spiral artery remodeling during placentation in Eutherian mammals. Our previous work showed that the invasion of trophoblast cells into uterine myometrium coincides with a phenotypic change of VSMCs. Here, we elucidate the mechanism by which trophoblast cells confer VSMC plasticity. Analysis of genetic markers on E13.5, E16.5, and E19.5 in the rat metrial gland, the entry point of uterine arteries, revealed that trophoblast invasion is associated with downregulation of MYOCARDIN, α-smooth muscle actin, and calponin1, and concomitant upregulation of Smemb in VSMCs. Myocardin overexpression or knockdown in VSMCs led to upregulation or downregulation of contractile markers, respectively. Co-culture of trophoblast cells with VSMCs decreased MYOCARDIN expression along with compromised expression of contractile markers in VSMCs. However, co-culture of trophoblast cells with VSMCs overexpressing MYOCARDIN inhibited their change in phenotype, whereas, overexpression of transactivation domain deleted MYOCARDIN failed to elicit this response. Furthermore, the co-culture of trophoblast cells with VSMCs led to the activation of NFκß signaling. Interestingly, despite producing IL-1ß, trophoblast cells possess only the decoy receptor, whereas, VSMCs possess the IL-1ß signaling receptor. Treatment of VSMCs with exogenous IL-1ß led to a decrease in MYOCARDIN and an increase in phosphorylation of NFκß. The effect of trophoblast cells in the downregulation of MYOCARDIN in VSMCs was reversed by blocking NFκß translocation to the nucleus. Together, these data highlight that trophoblast cells direct VSMC plasticity, and trophoblast-derived IL-1ß is a key player in downregulating MYOCARDIN via the NFκß signaling pathway.


Asunto(s)
Interleucina-1beta , Músculo Liso Vascular , Miocitos del Músculo Liso , FN-kappa B , Proteínas Nucleares , Transducción de Señal , Transactivadores , Trofoblastos , Animales , Trofoblastos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Transactivadores/metabolismo , Transactivadores/genética , Ratas , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Transducción de Señal/fisiología , FN-kappa B/metabolismo , Femenino , Miocitos del Músculo Liso/metabolismo , Interleucina-1beta/metabolismo , Embarazo , Técnicas de Cocultivo , Ratas Sprague-Dawley , Células Cultivadas , Plasticidad de la Célula/fisiología , Calponinas
2.
Cancer Metastasis Rev ; 43(1): 321-362, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38517618

RESUMEN

Recent advances have brought forth the complex interplay between tumor cell plasticity and its consequential impact on drug resistance and tumor recurrence, both of which are critical determinants of neoplastic progression and therapeutic efficacy. Various forms of tumor cell plasticity, instrumental in facilitating neoplastic cells to develop drug resistance, include epithelial-mesenchymal transition (EMT) alternatively termed epithelial-mesenchymal plasticity, the acquisition of cancer stem cell (CSC) attributes, and transdifferentiation into diverse cell lineages. Nuclear receptors (NRs) are a superfamily of transcription factors (TFs) that play an essential role in regulating a multitude of cellular processes, including cell proliferation, differentiation, and apoptosis. NRs have been implicated to play a critical role in modulating gene expression associated with tumor cell plasticity and drug resistance. This review aims to provide a comprehensive overview of the current understanding of how NRs regulate these key aspects of cancer biology. We discuss the diverse mechanisms through which NRs influence tumor cell plasticity, including EMT, stemness, and metastasis. Further, we explore the intricate relationship between NRs and drug resistance, highlighting the impact of NR signaling on chemotherapy, radiotherapy and targeted therapies. We also discuss the emerging therapeutic strategies targeting NRs to overcome tumor cell plasticity and drug resistance. This review also provides valuable insights into the current clinical trials that involve agonists or antagonists of NRs modulating various aspects of tumor cell plasticity, thereby delineating the potential of NRs as therapeutic targets for improved cancer treatment outcomes.


Asunto(s)
Plasticidad de la Célula , Neoplasias , Humanos , Plasticidad de la Célula/fisiología , Neoplasias/patología , Transducción de Señal , Transición Epitelial-Mesenquimal/fisiología , Resistencia a Antineoplásicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Células Madre Neoplásicas/patología
3.
Science ; 383(6687): eadi7342, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38452090

RESUMEN

Lineage plasticity-a state of dual fate expression-is required to release stem cells from their niche constraints and redirect them to tissue compartments where they are most needed. In this work, we found that without resolving lineage plasticity, skin stem cells cannot effectively generate each lineage in vitro nor regrow hair and repair wounded epidermis in vivo. A small-molecule screen unearthed retinoic acid as a critical regulator. Combining high-throughput approaches, cell culture, and in vivo mouse genetics, we dissected its roles in tissue regeneration. We found that retinoic acid is made locally in hair follicle stem cell niches, where its levels determine identity and usage. Our findings have therapeutic implications for hair growth as well as chronic wounds and cancers, where lineage plasticity is unresolved.


Asunto(s)
Células Madre Adultas , Plasticidad de la Célula , Epidermis , Folículo Piloso , Tretinoina , Cicatrización de Heridas , Animales , Ratones , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/fisiología , Epidermis/efectos de los fármacos , Epidermis/fisiología , Folículo Piloso/citología , Folículo Piloso/efectos de los fármacos , Folículo Piloso/fisiología , Tretinoina/metabolismo , Tretinoina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Rejuvenecimiento/fisiología , Técnicas de Cultivo de Célula , Neoplasias/patología , Ratones Endogámicos C57BL
4.
Nat Cancer ; 4(8): 1063-1082, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37537300

RESUMEN

Cell plasticity represents the ability of cells to be reprogrammed and to change their fate and identity, enabling homeostasis restoration and tissue regeneration following damage. Cell plasticity also contributes to pathological conditions, such as cancer, enabling cells to acquire new phenotypic and functional features by transiting across distinct cell states that contribute to tumor initiation, progression, metastasis and resistance to therapy. Here, we review the intrinsic and extrinsic mechanisms driving cell plasticity that promote tumor growth and proliferation as well as metastasis and drug tolerance. Finally, we discuss how cell plasticity could be exploited for anti-cancer therapy.


Asunto(s)
Plasticidad de la Célula , Neoplasias , Humanos , Plasticidad de la Célula/fisiología , Neoplasias/tratamiento farmacológico , Transformación Celular Neoplásica , Homeostasis
5.
Cell Mol Gastroenterol Hepatol ; 14(5): 1025-1051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35835391

RESUMEN

BACKGROUND & AIMS: Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS: Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS: GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS: GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.


Asunto(s)
Plasticidad de la Célula , Neuroglía , Animales , Ratones , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Plasticidad de la Célula/genética , Plasticidad de la Célula/fisiología , Gastrinas , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Hedgehog , Hiperplasia/patología , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Células Neuroendocrinas/fisiología , Neuroglía/metabolismo , Proteínas Proto-Oncogénicas , ARN Interferente Pequeño
6.
Sci China Life Sci ; 65(8): 1481-1497, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35696016

RESUMEN

The CREB1 gene encodes an exceptionally pleiotropic transcription factor that frequently dysregulated in human cancers. CREB1 can regulate tumor cell status of proliferation and/or migration; however, the molecular basis for this switch involvement in cell plasticity has not fully been understood yet. Here, we first show that knocking out CREB1 triggers a remarkable effect of epithelial-mesenchymal transition (EMT) and leads to the occurrence of inhibited proliferation and enhanced motility in HCT116 colorectal cancer cells. By monitoring 45 cellular signaling pathway activities, we find that multiple growth-related pathways decline significantly while inflammatory pathways including NF-κB are largely upregulated in comparing between the CREB1 wild-type and knocked out cells. Mechanistically, cells with CREB1 knocked out show downregulation of MYC as a result of impaired CREB1-dependent transcription of the oncogenic lncRNA CCAT1. Interestingly, the unbalanced competition between the coactivator CBP/p300 for CREB1 and p65 leads to the activation of the NF-κB pathway in cells with CREB1 disrupted, which induces an obvious EMT phenotype of the cancer cells. Taken together, these studies identify previously unknown mechanisms of CREB1 in CRC cell plasticity via regulating lncRNA CCAT1 and NF-κB pathways, providing a critical insight into a combined strategy for CREB1-targeted tumor therapies.


Asunto(s)
Plasticidad de la Célula , Neoplasias Colorrectales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , FN-kappa B , ARN Largo no Codificante , Línea Celular Tumoral , Movimiento Celular/genética , Plasticidad de la Célula/genética , Plasticidad de la Célula/fisiología , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/fisiopatología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
7.
Cell Rep ; 39(1): 110595, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385726

RESUMEN

Bioinformatic analysis of 94 patient-derived xenografts (PDXs), cell lines, and organoids (PCOs) identifies three intrinsic transcriptional subtypes of metastatic castration-resistant prostate cancer: androgen receptor (AR) pathway + prostate cancer (PC) (ARPC), mesenchymal and stem-like PC (MSPC), and neuroendocrine PC (NEPC). A sizable proportion of castration-resistant and metastatic stage PC (M-CRPC) cases are admixtures of ARPC and MSPC. Analysis of clinical datasets and mechanistic studies indicates that MSPC arises from ARPC as a consequence of therapy-induced lineage plasticity. AR blockade with enzalutamide induces (1) transcriptional silencing of TP53 and hence dedifferentiation to a hybrid epithelial and mesenchymal and stem-like state and (2) inhibition of BMP signaling, which promotes resistance to AR inhibition. Enzalutamide-tolerant LNCaP cells re-enter the cell cycle in response to neuregulin and generate metastasis in mice. Combined inhibition of HER2/3 and AR or mTORC1 exhibits efficacy in models of ARPC and MSPC or MSPC, respectively. These results define MSPC, trace its origin to therapy-induced lineage plasticity, and reveal its sensitivity to HER2/3 inhibition.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata Resistentes a la Castración , Transducción de Señal , Animales , Antineoplásicos/farmacología , Benzamidas , Carcinoma Neuroendocrino , Línea Celular Tumoral , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/fisiología , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Nitrilos , Feniltiohidantoína , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología
8.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35193955

RESUMEN

In search of redox mechanisms in breast cancer, we uncovered a striking role for glutathione peroxidase 2 (GPx2) in oncogenic signaling and patient survival. GPx2 loss stimulates malignant progression due to reactive oxygen species/hypoxia inducible factor-α (HIF1α)/VEGFA (vascular endothelial growth factor A) signaling, causing poor perfusion and hypoxia, which were reversed by GPx2 reexpression or HIF1α inhibition. Ingenuity Pathway Analysis revealed a link between GPx2 loss, tumor angiogenesis, metabolic modulation, and HIF1α signaling. Single-cell RNA analysis and bioenergetic profiling revealed that GPx2 loss stimulated the Warburg effect in most tumor cell subpopulations, except for one cluster, which was capable of oxidative phosphorylation and glycolysis, as confirmed by coexpression of phosphorylated-AMPK and GLUT1. These findings underscore a unique role for redox signaling by GPx2 dysregulation in breast cancer, underlying tumor heterogeneity, leading to metabolic plasticity and malignant progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Plasticidad de la Célula/fisiología , Glutatión Peroxidasa/metabolismo , Animales , Línea Celular Tumoral , Femenino , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/fisiología , Glucólisis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metabolismo/fisiología , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Oxidación-Reducción , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042776

RESUMEN

Sympathetic innervation regulates energy balance, and the nerve density in the adipose tissues changes under various metabolic states, resulting in altered neuronal control and conferring resilience to metabolic challenges. However, the impact of the immune milieu on neuronal innervation is not known. Here, we examined the regulatory role on nerve plasticity by eosinophils and found they increased cell abundance in response to cold and produced nerve growth factor (NGF) in the white adipose tissues (WAT). Deletion of Ngf from eosinophils or depletion of eosinophils impairs cold-induced axonal outgrowth and beiging process. The spatial proximity between sympathetic nerves, IL-33-expressing stromal cells, and eosinophils was visualized in both human and mouse adipose tissues. At the cellular level, the sympathetic adrenergic signal induced calcium flux in the stromal cells and subsequent release of IL-33, which drove the up-regulation of IL-5 from group 2 innate lymphoid cells (ILC2s), leading to eosinophil accretion. We propose a feed-forward loop between sympathetic activity and type 2 immunity that coordinately enhances sympathetic innervation and promotes energy expenditure.


Asunto(s)
Tejido Adiposo/metabolismo , Axones/metabolismo , Plasticidad de la Célula/fisiología , Eosinófilos/inmunología , Tejido Adiposo Blanco/metabolismo , Adulto , Animales , Calcio , Femenino , Humanos , Inmunidad Innata , Interleucina-33/metabolismo , Linfocitos/inmunología , Ratones , Persona de Mediana Edad , Factor de Crecimiento Nervioso/metabolismo , Células del Estroma/metabolismo , Sistema Nervioso Simpático/fisiología
10.
Gastroenterology ; 162(2): 415-430, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34728185

RESUMEN

The mucosa of the body of the stomach (ie, the gastric corpus) uses 2 overlapping, depth-dependent mechanisms to respond to injury. Superficial injury heals via surface cells with histopathologic changes like foveolar hyperplasia. Deeper, usually chronic, injury/inflammation, most frequently induced by the carcinogenic bacteria Helicobacter pylori, elicits glandular histopathologic alterations, initially manifesting as pyloric (also known as pseudopyloric) metaplasia. In this pyloric metaplasia, corpus glands become antrum (pylorus)-like with loss of acid-secreting parietal cells (atrophic gastritis), expansion of foveolar cells, and reprogramming of digestive enzyme-secreting chief cells into deep antral gland-like mucous cells. After acute parietal cell loss, chief cells can reprogram through an orderly stepwise progression (paligenosis) initiated by interleukin-13-secreting innate lymphoid cells (ILC2s). First, massive lysosomal activation helps mitigate reactive oxygen species and remove damaged organelles. Second, mucus and wound-healing proteins (eg, TFF2) and other transcriptional alterations are induced, at which point the reprogrammed chief cells are recognized as mucus-secreting spasmolytic polypeptide-expressing metaplasia cells. In chronic severe injury, glands with pyloric metaplasia can harbor both actively proliferating spasmolytic polypeptide-expressing metaplasia cells and eventually intestine-like cells. Gastric glands with such lineage confusion (mixed incomplete intestinal metaplasia and proliferative spasmolytic polypeptide-expressing metaplasia) may be at particular risk for progression to dysplasia and cancer. A pyloric-like pattern of metaplasia after injury also occurs in other gastrointestinal organs including esophagus, pancreas, and intestines, and the paligenosis program itself seems broadly conserved across tissues and species. Here we discuss aspects of metaplasia in stomach, incorporating data derived from animal models and work on human cells and tissues in correlation with diagnostic and clinical implications.


Asunto(s)
Plasticidad de la Célula/fisiología , Reprogramación Celular/fisiología , Mucosa Gástrica/fisiología , Regeneración/fisiología , Estómago/fisiología , Animales , Mucosa Gástrica/citología , Mucosa Gástrica/patología , Infecciones por Helicobacter/fisiopatología , Humanos , Hiperplasia , Metaplasia , Células Parietales Gástricas/fisiología , Estómago/citología , Estómago/patología
11.
Physiol Rep ; 9(19): e15066, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34605201

RESUMEN

The gut wall houses mast cells that are anatomically situated near enteric neuronal fibers. Roles of specific neuropeptides in modulating function of immune components like mast cells in response to challenge with bacterial components are relatively unknown. Investigating such interactions requires models that include diverse cellular elements in native anatomic arrangements. Using an organotypic slice model that maintains gut wall cellular diversity ex vivo, the present study compared responses between tissues derived from male and female mice to examine neural-immune signaling in the gut wall after selected treatments. Ileum slices were treated with pharmacological reagents that block neuronal function (e.g., tetrodotoxin) or vasoactive intestinal peptide (VIP) receptors prior to challenge with lipopolysaccharide (LPS) to assess their influence on anatomic plasticity of VIP fibers and activation of mast cells. Sex differences were observed in the number of mucosal mast cells (c-kit/ACK2 immunoreactive) at baseline, regardless of treatment, with female ileum tissue having 46% more ACK2-IR mast cells than males. After challenge with LPS, male mast cell counts rose to female levels. Furthermore, sex differences were observed in the percentage of ACK2-IR cells within 1 µm of a VIP+ neuronal fiber, and mast cell size, a metric previously tied to activation, with females having larger cells at baseline. Male mast cell sizes reached female levels after LPS challenge. This study suggests sex differences in neural-immune plasticity and in mast cell activation both basally and in response to challenge with LPS. These sex differences could potentially impact functional neuroimmune response to pathogens.


Asunto(s)
Plasticidad de la Célula/fisiología , Íleon/citología , Mastocitos/citología , Neuronas/citología , Caracteres Sexuales , Animales , Femenino , Masculino , Ratones
12.
J Clin Invest ; 131(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34596051

RESUMEN

The cardiac conduction system (CCS) ensures regular contractile function, and injury to any of its components can cause cardiac dysrhythmia. Although all cardiomyocytes (CMs) originate from common progenitors, the CCS is composed of biologically distinct cell types with unique functional and developmental characteristics. In contrast to ventricular cardiomyocytes, which continue to proliferate after birth, most CCS cells terminally exit the cell cycle during fetal development. Although the CCS should thus provide a poor substrate for postnatal injury repair, its regenerative capacity remains untested. Here, we describe a genetic system for ablating CMs that reside within the atrioventricular conduction system (AVCS). Adult mouse AVCS ablation resulted in regenerative failure characterized by persistent atrioventricular conduction defects and contractile dysfunction. In contrast, AVCS injury in neonatal mice led to recovery in a subset of these mice, thus providing evidence for CCS plasticity. Furthermore, CM proliferation did not appear to completely account for the observed functional recovery, suggesting that mechanisms regulating recovery from dysrhythmia are likely to be distinct from cardiac regeneration associated with ventricular injury. Taken together, we anticipate that our results will motivate further mechanistic studies of CCS plasticity and enable the exploration of rhythm restoration as an alternative therapeutic strategy.


Asunto(s)
Nodo Atrioventricular/lesiones , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Animales , Nodo Atrioventricular/fisiología , Plasticidad de la Célula/fisiología , Ratones , Ratones Endogámicos C57BL
15.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34299340

RESUMEN

Unlike some lower vertebrates which can completely regenerate their heart, the human heart is a terminally differentiated organ. Cardiomyocytes lost during cardiac injury and heart failure cannot be replaced due to their limited proliferative capacity. Therefore, cardiac injury generally leads to progressive failure. Here, we summarize the latest progress in research on methods to induce cardiomyocyte cell cycle entry and heart repair through the alteration of cardiomyocyte plasticity, which is emerging as an effective strategy to compensate for the loss of functional cardiomyocytes and improve the impaired heart functions.


Asunto(s)
Proliferación Celular/fisiología , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/metabolismo , Animales , Ciclo Celular , Plasticidad de la Célula/genética , Plasticidad de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Corazón/fisiología , Insuficiencia Cardíaca/fisiopatología , Lesiones Cardíacas/terapia , Humanos , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Transducción de Señal
17.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067929

RESUMEN

Cutaneous melanoma (CM) tissue represents a network constituted by cancer cells and tumor microenvironment (TME). A key feature of CM is the high structural and cellular plasticity of TME, allowing its evolution with disease and adaptation to cancer cell and environmental alterations. In particular, during melanoma development and progression each component of TME by interacting with each other and with cancer cells is subjected to dramatic structural and cellular modifications. These alterations affect extracellular matrix (ECM) remodelling, phenotypic profile of stromal cells, cancer growth and therapeutic response. The stromal fibroblast populations of the TME include normal fibroblasts and melanoma-associated fibroblasts (MAFs) that are highly abundant and flexible cell types interacting with melanoma and stromal cells and differently influencing CM outcomes. The shift from the normal microenvironment to TME and from normal fibroblasts to MAFs deeply sustains CM growth. Hence, in this article we review the features of the normal microenvironment and TME and describe the phenotypic plasticity of normal dermal fibroblasts and MAFs, highlighting their roles in normal skin homeostasis and TME regulation. Moreover, we discuss the influence of MAFs and their secretory profiles on TME remodelling, melanoma progression, targeted therapy resistance and immunosurveillance, highlighting the cellular interactions, the signalling pathways and molecules involved in these processes.


Asunto(s)
Fibroblastos/fisiología , Melanoma/metabolismo , Microambiente Tumoral/fisiología , Fibroblastos Asociados al Cáncer/metabolismo , Comunicación Celular , Plasticidad de la Célula/fisiología , Matriz Extracelular/metabolismo , Humanos , Melanoma/patología , Melanoma/fisiopatología , Transducción de Señal , Neoplasias Cutáneas/patología , Células del Estroma/metabolismo , Melanoma Cutáneo Maligno
18.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071193

RESUMEN

Cutaneous melanoma represents one of the deadliest types of skin cancer. The prognosis strongly depends on the disease stage, thus early detection is crucial. New therapies, including BRAF and MEK inhibitors and immunotherapies, have significantly improved the survival of patients in the last decade. However, intrinsic and acquired resistance is still a challenge. In this review, we discuss two major aspects that contribute to the aggressiveness of melanoma, namely, the embryonic origin of melanocytes and melanoma cells and cellular plasticity. First, we summarize the physiological function of epidermal melanocytes and their development from precursor cells that originate from the neural crest (NC). Next, we discuss the concepts of intratumoral heterogeneity, cellular plasticity, and phenotype switching that enable melanoma to adapt to changes in the tumor microenvironment and promote disease progression and drug resistance. Finally, we further dissect the connection of these two aspects by focusing on the transcriptional regulators MSX1, MITF, SOX10, PAX3, and FOXD3. These factors play a key role in NC initiation, NC cell migration, and melanocyte formation, and we discuss how they contribute to cellular plasticity and drug resistance in melanoma.


Asunto(s)
Plasticidad de la Célula/fisiología , Resistencia a Antineoplásicos/fisiología , Melanoma/metabolismo , Cresta Neural/metabolismo , Neoplasias Cutáneas/metabolismo , Acrilonitrilo/análogos & derivados , Acrilonitrilo/farmacología , Compuestos de Anilina/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Diferenciación Celular , Movimiento Celular , Resistencia a Antineoplásicos/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Transcripción MSX1/genética , Melanocitos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción PAX3/genética , Fenotipo , Pirimidinonas/farmacología , Factores de Transcripción SOXE/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología
19.
Commun Biol ; 4(1): 747, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135460

RESUMEN

Tumour recurrence is a serious impediment to cancer treatment, but the mechanisms involved are poorly understood. The most frequently used anti-tumour therapies-chemotherapy and radiotherapy-target highly proliferative cancer cells. However non- or slow-proliferative dormant cancer cells can persist after treatment, eventually causing tumour relapse. Whereas the reversible growth arrest mechanism allows quiescent cells to re-enter the cell cycle, senescent cells are largely thought to be irreversibly arrested, and may instead contribute to tumour growth and relapse through paracrine signalling mechanisms. Thus, due to the differences in their growth arrest mechanism, metabolic features, plasticity and adaptation to their respective tumour microenvironment, dormant-senescent and -quiescent cancer cells could have different but complementary roles in fuelling tumour growth. In this review article, we discuss the implication of dormant cancer cells in tumour relapse and the need to understand how quiescent and senescent cells, respectively, may play a part in this process.


Asunto(s)
Plasticidad de la Célula/fisiología , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Recurrencia Local de Neoplasia/patología , Neoplasias/patología , Ciclo Celular/fisiología , Humanos , Neoplasias/terapia , Transducción de Señal , Microambiente Tumoral/fisiología
20.
Oncogene ; 40(23): 4033-4049, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017076

RESUMEN

Intrahepatic cholangiocarcinoma (iCCA) is a rare malignancy of the intrahepatic biliary tract with a very poor prognosis. Although some clinicopathological parameters can be prognostic factors for iCCA, the molecular prognostic markers and potential mechanisms of iCCA have not been well investigated. Here, we report that the Fragile X mental retardation protein (FMRP), a RNA binding protein functionally absent in patients with the Fragile X syndrome (FXS) and also involved in several types of cancers, is overexpressed in human iCCA and its expression is significantly increased in iCCA metastatic tissues. The silencing of FMRP in metastatic iCCA cell lines affects cell migration and invasion, suggesting a role of FMRP in iCCA progression. Moreover, we show evidence that FMRP is localized at the invasive front of human iCCA neoplastic nests and in pseudopodia and invadopodia protrusions of migrating and invading iCCA cancer cells. Here FMRP binds several mRNAs encoding key proteins involved in the formation and/or function of these protrusions. In particular, we find that FMRP binds to and regulates the expression of Cortactin, a critical regulator of invadopodia formation. Altogether, our findings suggest that FMRP could promote cell invasiveness modulating membrane plasticity and invadopodia formation at the leading edges of invading iCCA cells.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Plasticidad de la Célula/fisiología , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Cortactina/metabolismo , Humanos , Masculino , Ratones Desnudos , Metástasis de la Neoplasia , Podosomas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...