Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 314
Filtrar
1.
J Cell Mol Med ; 27(23): 3692-3705, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37830980

RESUMEN

Perineural invasion (PNI) has emerged as a key pathological feature and be considered as a poor prognostic factor in cervical cancer. However, the underlying molecular mechanisms are largely unknown. Here, PNI status of 269 cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) samples were quantified by using whole-slide diagnostic images obtained from The Cancer Genome Atlas. Integrated analyses revealed that PNI was an indicative marker of poorer disease-free survival for CESC patients. Among the differentially expressed genes, ADCYAP1 were identified. Clinical specimens supported that high expression of PACAP (encoded by ADCYAP1) contributed to PNI in CESC. Mechanistically, PACAP, secreted from cervical cancer cells, reversed myelin differentiation of Schwann cells (SCs). Then, dedifferentiated SCs promoted PNI by producing chemokine FGF17 and by degrading extracellular matrix through secretion of Cathepsin S and MMP-12. In conclusion, this study identified PACAP was associated with PNI in cervical cancer and suggested that tumour-derived PACAP reversed myelin differentiation of SCs to aid PNI.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias del Cuello Uterino , Femenino , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Invasividad Neoplásica/patología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Células de Schwann/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Comunicación Paracrina/genética
2.
Stem Cell Rev Rep ; 19(5): 1554-1575, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060532

RESUMEN

Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Cicatrización de Heridas , Heridas y Lesiones , Humanos , Becaplermina/genética , Becaplermina/metabolismo , Supervivencia Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucosa/farmacología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/genética , Comunicación Paracrina/genética , Cordón Umbilical/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
3.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36835007

RESUMEN

Hypoxia is an important risk for renal disease. The mitochondrial enzyme arginase-II (Arg-II) is expressed and/or induced by hypoxia in proximal tubular epithelial cells (PTECs) and in podocytes, leading to cellular damage. Because PTECs are vulnerable to hypoxia and located in proximity to podocytes, we examined the role of Arg-II in the crosstalk of PTECs under hypoxic conditions with podocytes. A human PTEC cell line (HK2) and a human podocyte cell line (AB8/13) were cultured. Arg-ii gene was ablated by CRISPR/Case9 in both cell types. HK2 cells were exposed to normoxia (21% O2) or hypoxia (1% O2) for 48 h. Conditioned medium (CM) was collected and transferred to the podocytes. Podocyte injuries were then analyzed. Hypoxic (not normoxic) HK2-CM caused cytoskeletal derangement, cell apoptosis, and increased Arg-II levels in differentiated podocytes. These effects were absent when arg-ii in HK2 was ablated. The detrimental effects of the hypoxic HK2-CM were prevented by TGF-ß1 type-I receptor blocker SB431542. Indeed, TGF-ß1 levels in hypoxic HK2-CM (but not arg-ii-/--HK2-CM) were increased. Furthermore, the detrimental effects of TGF-ß1 on podocytes were prevented in arg-ii-/--podocytes. This study demonstrates crosstalk between PTECs and podocytes through the Arg-II-TGF-ß1 cascade, which may contribute to hypoxia-induced podocyte damage.


Asunto(s)
Túbulos Renales Proximales , Comunicación Paracrina , Podocitos , Humanos , Apoptosis , Arginasa/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Comunicación Paracrina/genética , Podocitos/metabolismo , Podocitos/patología , Factor de Crecimiento Transformador beta1/metabolismo
4.
Hepatology ; 78(5): 1478-1491, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35950514

RESUMEN

BACKGROUND AND AIMS: The mammalian liver harbors heterogeneous cell types that communicate via local paracrine signaling. Recent studies have delineated the transcriptomic landscape of the liver in NASH that provides insights into liver cell heterogeneity, intercellular crosstalk, and disease-associated reprogramming. However, the nature of intrahepatic signaling and its role in NASH progression remain obscure. APPROACH AND RESULTS: Here, we performed transcriptomic analyses and identified cardiotrophin-like cytokine factor 1 (CLCF1), a member of the IL-6 family cytokines, as a cholangiocyte-derived paracrine factor that was elevated in the liver from diet-induced NASH mice and patients with NASH. Adenovirus-associated virus-mediated overexpression of CLCF1 in the liver ameliorated NASH pathologies in two diet-induced NASH models in mice, illustrating that CLCF1 induction may serve an adaptive and protective role during NASH pathogenesis. Unexpectedly, messenger RNA and protein levels of leukemia inhibitory factor receptor (LIFR), a subunit of the receptor complex for CLCF1, were markedly downregulated in NASH liver. Hepatocyte-specific inactivation of LIFR accelerated NASH progression in mice, supporting an important role of intrahepatic cytokine signaling in maintaining tissue homeostasis under metabolic stress conditions. CONCLUSIONS: Together, this study sheds light on the molecular nature of intrahepatic paracrine signaling during NASH pathogenesis and uncovers potential targets for therapeutic intervention.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Comunicación Paracrina , Animales , Humanos , Ratones , Citocinas/genética , Citocinas/metabolismo , Dieta/efectos adversos , Modelos Animales de Enfermedad , Interleucinas/metabolismo , Hígado/metabolismo , Mamíferos , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Comunicación Paracrina/genética , Comunicación Paracrina/fisiología
5.
Cells ; 11(2)2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35053392

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is a highly invasive brain tumour, characterized by its ability to secrete factors promoting its virulence. Brain endothelial cells (BECs) in the GBM environment are physiologically modulated. The present study investigated the modulatory effects of normoxically and hypoxically induced glioblastoma U-87 cell secretions on BECs. METHODS: Conditioned media (CM) were derived by cultivating U-87 cells under hypoxic incubation (5% O2) and normoxic incubation (21% O2). Treated bEnd.3 cells were evaluated for mitochondrial dehydrogenase activity, mitochondrial membrane potential (ΔΨm), ATP production, transendothelial electrical resistance (TEER), and endothelial tight-junction (ETJ) gene expression over 96 h. RESULTS: The coculture of bEnd.3 cells with U-87 cells, or exposure to either hypoxic or normoxic U-87CM, was associated with low cellular viability. The ΔΨm in bEnd.3 cells was hyperpolarized after hypoxic U-87CM treatment (p < 0.0001). However, normoxic U-87CM did not affect the state of ΔΨm. BEC ATP levels were reduced after being cocultured with U-87 cells, or with hypoxic and normoxic CM (p < 0.05). Suppressed mitochondrial activity in bEnd.3 cells was associated with increased transendothelial permeability, while bEnd.3 cells significantly increased the gene expression levels of ETJs (p < 0.05) when treated with U-87CM. CONCLUSIONS: Hypoxic and normoxic glioblastoma paracrine factors differentially suppressed mitochondrial activity in BECs, increasing the BECs' barrier permeability.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Células Endoteliales/patología , Glioblastoma/patología , Comunicación Paracrina , Hipoxia Tumoral , Adenosina Trifosfato/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Claudina-5/genética , Claudina-5/metabolismo , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ocludina/genética , Ocludina/metabolismo , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/genética , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética
6.
PLoS One ; 17(1): e0261498, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35025920

RESUMEN

BACKGROUND: A paracrine mechanism is thought to mediate the proangiogenic capacity of adipose-derived stromal/stem cells (ASCs). However, the precise mechanism by which ASCs promote the formation of blood vessels by endothelial progenitor cells (EPCs) is unclear. METHODS: The EPCs-ASCs cocultures prepared in different ratios were subjected to tube formations assay to verify whether ASCs could directly participate in the tube genesis. The supernatant from cultured ASCs was used to stimulate EPCs to evaluate the effects on the angiogenic property of EPCs, as well as capacity for migration and invasion. A coculture model with transwell chamber were used to explore the regulation of angiogenesis markers expression in EPCs by ASCs. We then mixed ASCs with EPCs and transplanted them with adipose tissue into nude mice to evaluate the effects on angiogenesis in adipose tissue grafts. RESULTS: In the EPCs-ASCs cocultures, the tube formation was significantly decreased as the relative abundance of ASCs increased, while the ASCs was found to migrate and integrated into the agglomerates formed by EPCs. The supernatant from ASCs cultures promoted the migration and invasion of EPCs and the ability to form capillary-like structures. The expression of multiple angiogenesis markers in EPCs were significantly increased when cocultured with ASCs. In vivo, ASCs combined with EPC promoted vascularization in the fat transplant. Immunofluorescence straining of Edu and CD31 indicated that the Edu labeled EPC did not directly participate in the vascularization inside the fat tissue. CONCLUSIONS: ADSC can participate in the tube formation of EPC although it cannot form canonical capillary structures. Meanwhile, Soluble factors secreted by ASCs promotes the angiogenic potential of EPCs. ASCs paracrine signaling appears to promote angiogenesis by increasing the migration and invasion of EPCs and simultaneously upregulating the expression of angiogenesis markers in EPCs. The results of in vivo experiments showed that ASCs combined with EPCs significantly promote the formation of blood vessels in the fat implant. Remarkably, EPCs may promote angiogenesis by paracrine regulation of endogenous endothelial cells (ECs) rather than direct participation in the formation of blood vessels.


Asunto(s)
Células Progenitoras Endoteliales/trasplante , Supervivencia de Injerto/fisiología , Neovascularización Fisiológica/fisiología , Células del Estroma/trasplante , Tejido Adiposo/citología , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Técnicas de Cultivo de Célula , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Ratones , Ratones Desnudos , Comunicación Paracrina/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Células del Estroma/citología , Células del Estroma/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
FEBS J ; 289(2): 417-435, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34355516

RESUMEN

Sepsis is a life-threatening process related to a dysregulated host response to an underlying infection, which results in organ dysfunction and poor outcomes. Therapeutic strategies using mesenchymal stromal cells (MSCs) are under investigation for sepsis, with efforts to improve cellular utility. Syndecan (SDC) proteins are transmembrane proteoglycans involved with cellular signaling events including tissue repair and modulating inflammation. Bone marrow-derived human MSCs express syndecan-2 (SDC2) at a level higher than other SDC family members; thus, we explored SDC2 in MSC function. Administration of human MSCs silenced for SDC2 in experimental sepsis resulted in decreased bacterial clearance, and increased tissue injury and mortality compared with wild-type MSCs. These findings were associated with a loss of resolution of inflammation in the peritoneal cavity, and higher levels of proinflammatory mediators in organs. MSCs silenced for SDC2 had a decreased ability to promote phagocytosis of apoptotic neutrophils by macrophages in the peritoneum, and also a diminished capability to convert macrophages from a proinflammatory to a proresolution phenotype via cellular or paracrine actions. Extracellular vesicles are a paracrine effector of MSCs that may contribute to resolution of inflammation, and their production was dramatically reduced in SDC2-silenced human MSCs. Collectively, these data demonstrate the importance of SDC2 for cellular and paracrine function of human MSCs during sepsis.


Asunto(s)
Vesículas Extracelulares/genética , Inflamación/genética , Sepsis/genética , Sindecano-2/genética , Animales , Polaridad Celular/genética , Polaridad Celular/inmunología , Modelos Animales de Enfermedad , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/microbiología , Regulación del Desarrollo de la Expresión Génica/genética , Silenciador del Gen , Humanos , Inmunidad/genética , Inflamación/microbiología , Inflamación/patología , Inflamación/terapia , Macrófagos/inmunología , Macrófagos/microbiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Neutrófilos/inmunología , Neutrófilos/microbiología , Comunicación Paracrina/genética , Fagocitosis/genética , Sepsis/microbiología , Sepsis/patología , Sepsis/terapia
8.
Hum Cell ; 35(1): 111-124, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34792755

RESUMEN

Fanconi anemia (FA) is a rare genetic disorder characterized by genomic instability, developmental defects, and bone marrow (BM) failure. Hematopoietic stem cells (HSCs) in BM interact with the mesenchymal stem/stromal cells (MSCs); and this partly sustains the tissue homeostasis. MicroRNAs (miRNAs) can play a critical role during these interactions possibly via paracrine mechanisms. This is the first study addressing the miRNA profile of FA BM-MSCs obtained before and after BM transplantation (preBMT and postBMT, respectively). Non-coding RNA expression profiling and quality control analyses were performed in Donors (n = 13), FA preBMT (n = 11), and FA postBMT (n = 6) BM-MSCs using GeneChip miRNA 2.0 Array. Six Donor-FA preBMT pairs were used to identify a differentially expressed miRNA expression signature containing 50 miRNAs, which exhibited a strong correlation with the signature obtained from unpaired samples. Five miRNAs (hsa-miR-146a-5p, hsa-miR-148b-3p, hsa-miR-187-3p, hsa-miR-196b-5p, and hsa-miR-25-3p) significantly downregulated in both the paired and unpaired analyses were used to generate the BM-MSCs' miRNA-BM mononuclear mRNA networks upon integration of a public dataset (GSE16334; studying Donor versus FA samples). Functionally enriched KEGG pathways included cellular senescence, miRNAs, and pathways in cancer. Here, we showed that hsa-miR-146a-5p and hsa-miR-874-3p were rescued upon BMT (n = 3 triplets). The decrease in miR-146a-5p was also validated using RT-qPCR and emerged as a strong candidate as a modulator of BM mRNAs in FA patients.


Asunto(s)
Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Inestabilidad Genómica/genética , Células Madre Hematopoyéticas/fisiología , Humanos , MicroARNs/fisiología , Comunicación Paracrina/genética , Comunicación Paracrina/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575956

RESUMEN

Umbilical cord blood (UCB) has long been seen as a rich source of naïve cells with strong regenerative potential, likely mediated by paracrine signals. More recently, small extracellular vesicles (sEV), such as exosomes, have been shown to play essential roles in cell-to-cell communication, via the transport of numerous molecules, including small RNAs. Often explored for their potential as biomarkers, sEV are now known to have regenerative and immunomodulating characteristics, particularly if isolated from stem cell-rich tissues. In this study, we aim to characterize the immunomodulating properties of umbilical cord blood mononuclear cell-derived sEV (UCB-MNC-sEV) and explore their therapeutic potential for inflammatory skin diseases. UCB-MNC-sEV were shown to shift macrophages toward an anti-inflammatory phenotype, which in turn exert paracrine effects on fibroblasts, despite previous inflammatory stimuli. Additionally, the incubation of PBMC with UCB-MNC-sEV resulted in a reduction of total CD4+ and CD8+ T-cell proliferation and cytokine release, while specifically supporting the development of regulatory T-cells (Treg), by influencing FOXP3 expression. In a 3D model of psoriatic skin, UCB-MNC-sEV reduced the expression of inflammatory and psoriatic markers IL6, IL8, CXCL10, COX2, S100A7, and DEFB4. In vivo, UCB-MNC-sEV significantly prevented or reversed acanthosis in imiquimod-induced psoriasis, and tendentially increased the number of Treg in skin, without having an overall impact on disease burden. This work provides evidence for the anti-inflammatory and tolerogenic effect of UCB-MNC-sEV, which may be harnessed for the treatment of Th17-driven inflammatory skin diseases, such as psoriasis.


Asunto(s)
Exosomas/inmunología , Factores de Transcripción Forkhead/genética , Inmunomodulación/inmunología , Inflamación/terapia , Psoriasis/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/genética , Comunicación Celular/inmunología , Proliferación Celular/genética , Citocinas/genética , Exosomas/genética , Exosomas/trasplante , Vesículas Extracelulares/trasplante , Femenino , Sangre Fetal/inmunología , Sangre Fetal/trasplante , Humanos , Inmunomodulación/genética , Inflamación/sangre , Inflamación/patología , Macrófagos/inmunología , Masculino , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Psoriasis/sangre , Psoriasis/patología , Linfocitos T Reguladores/inmunología
10.
Nat Commun ; 12(1): 3403, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099707

RESUMEN

Developmental outcomes are shaped by the interplay between intrinsic and external factors. The production of stomata-essential pores for gas exchange in plants-is extremely plastic and offers an excellent system to study this interplay at the cell lineage level. For plants, light is a key external cue, and it promotes stomatal development and the accumulation of the master stomatal regulator SPEECHLESS (SPCH). However, how light signals are relayed to influence SPCH remains unknown. Here, we show that the light-regulated transcription factor ELONGATED HYPOCOTYL 5 (HY5), a critical regulator for photomorphogenic growth, is present in inner mesophyll cells and directly binds and activates STOMAGEN. STOMAGEN, the mesophyll-derived secreted peptide, in turn stabilizes SPCH in the epidermis, leading to enhanced stomatal production. Our work identifies a molecular link between light signaling and stomatal development that spans two tissue layers and highlights how an environmental signaling factor may coordinate growth across tissue types.


Asunto(s)
Arabidopsis/crecimiento & desarrollo , Regulación de la Expresión Génica de las Plantas/efectos de la radiación , Luz , Desarrollo de la Planta/genética , Estomas de Plantas/crecimiento & desarrollo , Arabidopsis/genética , Arabidopsis/efectos de la radiación , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Hipocótilo/metabolismo , Células del Mesófilo/metabolismo , Comunicación Paracrina/genética , Comunicación Paracrina/efectos de la radiación , Desarrollo de la Planta/efectos de la radiación , Epidermis de la Planta/metabolismo , Estomas de Plantas/efectos de la radiación , Plantas Modificadas Genéticamente , Estabilidad Proteica/efectos de la radiación
11.
Am J Physiol Cell Physiol ; 321(1): C82-C93, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34038245

RESUMEN

The resolution of inflammation is closely linked with tissue repair. Recent studies have revealed that macrophages suppress inflammatory reactions by producing lipid mediators, called specialized proresolving mediators (SPMs); however, the biological significance of SPMs in tissue repair remains to be fully elucidated in the heart. In this study, we focused on maresin-1 (MaR1) and examined the reparative effects of MaR1 in cardiomyocytes. The treatment with MaR1 increased cell size in cultured neonatal rat cardiomyocytes. Since the expression of fetal cardiac genes was unchanged by MaR1, physiological hypertrophy was induced by MaR1. SR3335, an inhibitor of retinoic acid-related orphan receptor α (RORα), mitigated MaR1-induced cardiomyocyte hypertrophy, consistent with the recent report that RORα is one of MaR1 receptors. Importantly, in response to MaR1, cardiomyocytes produced IGF-1 via RORα. Moreover, MaR1 activated phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and wortmannin, a PI3K inhibitor, or triciribine, an Akt inhibitor, abrogated MaR1-induced cardiomyocyte hypertrophy. Finally, the blockade of IGF-1 receptor by NVP-AEW541 inhibited MaR-1-induced cardiomyocyte hypertrophy as well as the activation of PI3K/Akt pathway. These data indicate that MaR1 induces cardiomyocyte hypertrophy through RORα/IGF-1/PI3K/Akt pathway. Considering that MaR1 is a potent resolving factor, MaR1 could be a key mediator that orchestrates the resolution of inflammation with myocardial repair.


Asunto(s)
Cardiomegalia/genética , Cardiotónicos/farmacología , Ácidos Docosahexaenoicos/efectos adversos , Factor I del Crecimiento Similar a la Insulina/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/efectos de los fármacos , Comunicación Paracrina/genética , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/antagonistas & inhibidores , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Comunicación Paracrina/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ribonucleósidos/farmacología , Transducción de Señal , Sulfonamidas/farmacología , Tiofenos/farmacología , Wortmanina/farmacología
12.
PLoS Biol ; 19(4): e3001204, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33891586

RESUMEN

Many cell types display the remarkable ability to alter their cellular phenotype in response to specific external or internal signals. Such phenotypic plasticity is apparent in the nematode Caenorhabditis elegans when adverse environmental conditions trigger entry into the dauer diapause stage. This entry is accompanied by structural, molecular, and functional remodeling of a number of distinct tissue types of the animal, including its nervous system. The transcription factor (TF) effectors of 3 different hormonal signaling systems, the insulin-responsive DAF-16/FoxO TF, the TGFß-responsive DAF-3/SMAD TF, and the steroid nuclear hormone receptor, DAF-12/VDR, a homolog of the vitamin D receptor (VDR), were previously shown to be required for entering the dauer arrest stage, but their cellular and temporal focus of action for the underlying cellular remodeling processes remained incompletely understood. Through the generation of conditional alleles that allowed us to spatially and temporally control gene activity, we show here that all 3 TFs are not only required to initiate tissue remodeling upon entry into the dauer stage, as shown before, but are also continuously required to maintain the remodeled state. We show that DAF-3/SMAD is required in sensory neurons to promote and then maintain animal-wide tissue remodeling events. In contrast, DAF-16/FoxO or DAF-12/VDR act cell-autonomously to control anatomical, molecular, and behavioral remodeling events in specific cell types. Intriguingly, we also uncover non-cell autonomous function of DAF-16/FoxO and DAF-12/VDR in nervous system remodeling, indicating the presence of several insulin-dependent interorgan signaling axes. Our findings provide novel perspectives into how hormonal systems control tissue remodeling.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Comunicación Celular/genética , Plasticidad de la Célula/genética , Factores de Transcripción Forkhead/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , Especificidad de Órganos/genética , Organogénesis/genética , Comunicación Paracrina/genética , Receptores de Calcitriol/genética , Receptores de Calcitriol/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/genética
13.
FEBS J ; 288(18): 5406-5429, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33705609

RESUMEN

Transcription factor SOX9 was a biomarker for prostate cancer (Pca) with poor prognosis. Nevertheless, the regulatory mechanism underlying SOX9 upregulation still remains unclear. Several cytokines have been reported to be involved in the regulation of SOX9, suggesting that cancer-associated fibroblasts (CAFs), one of the main sources of secreted factors in the tumor microenvironment (TME), may play a role in regulating SOX9 expression. Herein, an in vitro model of paracrine interaction between primary CAFs and Pca cells was applied to investigate the molecular mechanism of SOX9 upregulation during Pca progression. The regulatory axis was validated by in vivo experiments and The Cancer Genome Atlas data. Conditional medium of CAFs (CAF-CM) upregulated the expression of SOX9, which was mutually proved to be essential for CAF-induced tumor progression. Further analysis showed that hepatocyte growth factor (HGF) secreted by CAFs was responsible for SOX9 elevation in Pca cells, via the activation of c-Met signaling. Mechanistically, HGF/c-Met signaling specifically activated MEK1/2-ERK1/2 pathway, which induced phosphorylation and upregulation of FRA1, which then transcriptionally upregulated SOX9 by binding to the promoter of SOX9 gene. Moreover, we identified that HGF/c-Met-ERK1/2-FRA1-SOX9 axis was relatively conserved between human and mouse species by validating in mouse Pca cells. Our results reveal a novel insight into the molecular mechanism that SOX9 in Pca cells is promoted by CAFs through HGF/c-Met-ERK1/2-FRA1 axis. Furthermore, SOX9 may serve as an alternative marker for the activated HGF/c-Met signaling to enroll the optimal Pca patients for HGF/c-Met inhibition treatment, since it is much more stable and easier to detect.


Asunto(s)
Factor de Crecimiento de Hepatocito/genética , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-met/genética , Factor de Transcripción SOX9/genética , Anciano , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Persona de Mediana Edad , Comunicación Paracrina/genética , Activación Transcripcional/genética , Microambiente Tumoral/genética
14.
Mol Cancer Res ; 19(6): 1051-1062, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33619227

RESUMEN

Placental growth factor (PlGF) is a pro-angiogenic, N-glycosylated growth factor, which is secreted under pathologic situations. Here, we investigated the regulation of PlGF in response to ionizing radiation (IR) and its role for tumor angiogenesis and radiosensitivity. Secretion and expression of PlGF was induced in multiple tumor cell lines (medulloblastoma, colon and lung adenocarcinoma) in response to irradiation in a dose- and time-dependent manner. Early upregulation of PlGF expression and secretion in response to irradiation was primarily observed in p53 wild-type tumor cells, whereas tumor cells with mutated p53 only showed a minimal or delayed response. Mechanistic investigations with genetic and pharmacologic targeting of p53 corroborated regulation of PlGF by the tumor suppressor p53 in response to irradiation under normoxic and hypoxic conditions, but with so far unresolved mechanisms relevant for its minimal and delayed expression in tumor cells with a p53-mutated genetic background. Probing a paracrine role of IR-induced PlGF secretion in vitro, migration of endothelial cells was specifically increased towards irradiated PlGF wild type but not towards irradiated PlGF-knockout (PIGF-ko) medulloblastoma cells. Tumors derived from these PlGF-ko cells displayed a reduced growth rate, but similar tumor vasculature formation as in their wild-type counterparts. Interestingly though, high-dose irradiation strongly reduced microvessel density with a concomitant high rate of complete tumor regression only in the PlGF-ko tumors. IMPLICATIONS: Our study shows a strong paracrine vasculature-protective role of PlGF as part of a p53-regulated IR-induced resistance mechanism and suggest PlGF as a promising target for a combined treatment modality with RT.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Comunicación Paracrina/efectos de la radiación , Factor de Crecimiento Placentario/genética , Tolerancia a Radiación/genética , Radiación Ionizante , Proteína p53 Supresora de Tumor/genética , Células A549 , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/efectos de la radiación , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Comunicación Paracrina/genética , Factor de Crecimiento Placentario/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Carcinogenesis ; 42(5): 714-723, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33421057

RESUMEN

Melanoma is one of the most aggressive forms of human cancer and its incidence has significantly increased worldwide over the last decades. This neoplasia has been characterized by the release of a wide variety of soluble factors, which could stimulate tumor cell proliferation and survival in an autocrine and paracrine manner. Consequently, we sought to evaluate the pattern of soluble factors produced by pre-metastatic and metastatic melanoma established cultures, and to determine whether these factors can be detected in the autologous serum of malignant melanoma patients. Our results showed that both melanoma cultures had a common profile of 27 soluble factors mainly characterized by the high expression of VEGF-A, IL-6, MCP-1, IL-8, and SDF-1. In addition, when we compared supernatants, we observed significant differences in VEGF-A, BDNF, FGF-2, and NGF-ß concentrations. As we found in melanoma cultures, serum samples also had their specific production pattern composed by 21 soluble factors. Surprisingly, PDGF-BB and EGF were only found in serum, whereas IL-2, IL-4, IL-8, IL31, FGF2, and GRO-α were only expressed in the supernatant. Significant differences in PDGF-BB, MIP-1ß, HGF, PIGF-1, BDNF, EGF, Eotaxin, and IP-10 were also found after comparing autologous serum with healthy controls. According to this, no correlation was found between culture supernatants and autologous serum samples, which suggests that some factors may act locally, and others systemically. Nonetheless, after validation of our results in an independent cohort of patients, we concluded that PDGF-BB, VEGF-A, and IP-10 serum levels could be used to monitor different melanoma stages.


Asunto(s)
Becaplermina/sangre , Quimiocina CXCL10/sangre , Melanoma/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Comunicación Autocrina/genética , Becaplermina/genética , Proliferación Celular/genética , Quimiocina CCL2/sangre , Quimiocina CXCL10/genética , Quimiocina CXCL12/sangre , Citocinas/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Interleucina-6/sangre , Interleucina-8/sangre , Masculino , Melanoma/genética , Melanoma/patología , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/genética , Comunicación Paracrina/genética , Factor A de Crecimiento Endotelial Vascular/genética
16.
Mol Oncol ; 15(5): 1528-1542, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33508878

RESUMEN

Chemotherapeutic resistance is a major obstacle in the control of advanced breast cancer (BCa). We have previously shown that small extracellular vesicles (sEVs) can transmit adriamycin resistance between BCa cells. Here, we describe that sEV-mediated TGF-ß1 intercellular transfer is involved in the drug-resistant transmission. sEVs were isolated and characterized from both sensitive and resistant cells. sEVs derived from the resistant cells were incubated with the sensitive cells and resulted in transmitting the resistant phenotype to the recipient cells. Cytokine antibody microarray revealed that most metastasis-associated cytokines present at the high levels in sEVs from the resistant cells compared with their levels in sEVs from the sensitive cells, particularly TGF-ß1 is enriched in sEVs from the resistant cells. The sEV-mediated TGF-ß1 intercellular transfer led to increasing Smad2 phosphorylation and improving cell survival by suppressing apoptosis and enhancing cell mobility. Furthermore, sEV-mediated drug-resistant transmission by delivering TGF-ß1 was validated using a zebrafish xenograft tumor model. These results elaborated that sEV-mediated TGF-ß1 intercellular transfer contributes to adriamycin resistance in BCa.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Vesículas Extracelulares/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Embrión no Mamífero , Vesículas Extracelulares/fisiología , Femenino , Humanos , Células MCF-7 , Comunicación Paracrina/genética , Factor de Crecimiento Transformador beta1/genética , Microambiente Tumoral/genética , Pez Cebra/embriología
17.
Curr Opin Genet Dev ; 66: 83-92, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33477017

RESUMEN

Exosomes are secreted small extracellular vesicles (EVs) packaged with diverse biological cargo. They mediate complex intercellular communications among cells in maintenance of normal physiology or to trigger profound disease progression. Increasing numbers of studies have identified exosome-mediated functions contributing to cancer progression, including roles in paracrine cell-to-cell communication, stromal reprogramming, angiogenesis, and immune responses. Despite the growing body of knowledge, the specific role of exosomes in mediating pre-cancerous conditions is not fully understood and their ability to transform a healthy cell is still controversial. Here we review recent studies describing functions attributed to exosomes in different stages of carcinogenesis. We also explore how exosomes ultimately contribute to the progression of a primary tumor to metastatic disease.


Asunto(s)
Carcinogénesis/genética , Exosomas/genética , Neoplasias/genética , Microambiente Tumoral/genética , Comunicación Celular/genética , Vesículas Extracelulares/genética , Humanos , Neoplasias/patología , Comunicación Paracrina/genética
18.
J Surg Res ; 257: 294-305, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32871430

RESUMEN

BACKGROUND: Drug-eluting stents impair post-angioplasty re-endothelialization thus compromising restenosis prevention while heightening thrombotic risks. We recently found that inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) effectively mitigated both restenosis and thrombosis in rodent models. This motivated us to determine how PERK inhibition impacts re-endothelialization. METHODS: Re-endothelialization was evaluated in endothelial-denuded rat carotid arteries after balloon angioplasty and periadventitial administration of PERK inhibitor in a hydrogel. To study whether PERK in smooth muscle cells (SMCs) regulates re-endothelialization by paracrinally influencing endothelial cells (ECs), denuded arteries exposing SMCs were lentiviral-infected to silence PERK; in vitro, the extracellular vesicles isolated from the medium of PDGF-activated, PERK-upregulating human primary SMCs were transferred to human primary ECs. RESULTS: Treatment with PERK inhibitor versus vehicle control accelerated re-endothelialization in denuded arteries. PERK-specific silencing in the denuded arterial wall (mainly SMCs) also enhanced re-endothelialization compared to scrambled shRNA control. In vitro, while medium transfer from PDGF-activated SMCs impaired EC viability and increased the mRNA levels of dysfunctional EC markers, either PERK inhibition or silencing in donor SMCs mitigated these EC changes. Furthermore, CXCL10, a paracrine cytokine detrimental to ECs, was increased by PDGF activation and decreased after PERK inhibition or silencing in SMCs. CONCLUSIONS: Attenuating PERK activity pharmacologically or genetically provides an approach to accelerating post-angioplasty re-endothelialization in rats. The mechanism may involve paracrine factors regulated by PERK in SMCs that impact neighboring ECs. This study rationalizes future development of PERK-targeted endothelium-friendly vascular interventions.


Asunto(s)
Angioplastia de Balón/efectos adversos , Reestenosis Coronaria/prevención & control , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Repitelización/efectos de los fármacos , eIF-2 Quinasa/antagonistas & inhibidores , Angioplastia de Balón/instrumentación , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Reestenosis Coronaria/etiología , Modelos Animales de Enfermedad , Stents Liberadores de Fármacos/efectos adversos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/genética , ARN Interferente Pequeño/metabolismo , Ratas , Repitelización/genética , eIF-2 Quinasa/genética
19.
Antioxid Redox Signal ; 34(9): 736-749, 2021 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32390459

RESUMEN

Significance: Perivascular adipose tissue (PVAT), which is present surrounding most blood vessels, from the aorta to the microvasculature of the dermis, is mainly composed of fat cells, fibroblasts, stem cells, mast cells, and nerve cells. Although the PVAT is objectively present, its physiological and pathological significance has long been ignored. Recent Advances: PVAT was considered as a supporting component of blood vessels and a protective cushion to the vessel wall from the neighboring tissues during relaxation and contraction. Nonetheless, further extensive research found that PVAT actively regulates blood vessel tone through PVAT-derived vasoactive factors, including both relaxing and contracting factors. In addition, PVAT contributes to atherosclerosis through paracrine secretion of a large number of bioactive factors such as adipokines and cytokines. Thereby, PVAT regulates the functions of blood vessels through various mechanisms operating directly on PVAT or on the underlying vessel layers, including vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). Critical Issues: PVAT is a unique adipose tissue that plays an essential role in maintaining the vascular structure and regulating vascular function and homeostasis. This review focuses on recent updates on the various PVAT roles in hypertension and atherosclerosis. Future Directions: Future studies should further investigate the actual contribution of alterations in PVAT metabolism to the overall systemic outcomes of cardiovascular disease, which remains largely unknown. In addition, the messengers and underlying mechanisms responsible for the crosstalk between PVAT and ECs and VSMCs in the vascular wall should be systematically addressed, as well as the contributions of PVAT aging to vascular dysfunction.


Asunto(s)
Tejido Adiposo/metabolismo , Aterosclerosis/metabolismo , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Aterosclerosis/genética , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Hipertensión/genética , Hipertensión/patología , Células Musculares/metabolismo , Células Musculares/patología , Músculo Liso Vascular/patología , Comunicación Paracrina/genética
20.
Int J Biochem Cell Biol ; 131: 105898, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33285290

RESUMEN

Abnormal intraglandular stromal-epithelial interactions have been known as a main key contributing factor for development of Benign Prostatic Hyperplasia (BPH). However, the underlying mechanism for the dysregulated intercellular communication remains unclear. In this study we compared the proteomic profiles of hyperplastic tissue with adjacent normal tissue of BPH and identified Rab27B small GTPase, a key regulator of exocytosis, as a protein that was overexpressed in the epithelium of BPH tissue. Overexpression of Rab27B in prostatic epithelial cells strongly increased the signaling activities of the PI3K/AKT and ERK1/2 pathways, whereas, downregulation of Rab27B expression in the epithelial cells of BPH reduced the signaling activities and decreased cell proliferation. The elevated Rab27B expression caused an overall increase in cell surface presentation of growth factor receptors without affecting their expression. However, the small GTPase also possesses an inhibitory activity against mTORC1 independent of its role in cell surface presentation of growth factor receptors. Our findings demonstrate a pivotal role of the small GTPase in autocrine and paracrine signaling and suggest that its abnormal expression underlies the dysregulated stromal-epithelial interactions in BPH.


Asunto(s)
Comunicación Autocrina/genética , Células Epiteliales/metabolismo , Comunicación Paracrina/genética , Hiperplasia Prostática/genética , Células del Estroma/metabolismo , Proteínas de Unión al GTP rab/genética , Línea Celular , Conjuntos de Datos como Asunto , Células Epiteliales/patología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Microtomía , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Próstata/metabolismo , Próstata/patología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Células del Estroma/patología , Proteínas de Unión al GTP rab/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...