Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.154
Filtrar
1.
Gen Comp Endocrinol ; 356: 114580, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38964421

RESUMEN

Thyroid stimulating hormone (TSH), a glycoprotein synthesized and secreted from thyrotrophs of the pituitary gland, is composed of a glycoprotein hormone common alpha subunit (CGA) and a specific beta subunit (TSHB). The major biological function of TSH is to stimulate thyroidal follicles to synthesize and secrete thyroid hormones through activating its cognate receptor, the thyroid stimulating hormone receptor (TSHR). In the present study, polyclonal antisera against ricefield eel Tshb and Tshr were generated respectively, and the expression of Tshb and Tshr was examined at mRNA and protein levels. RT-PCR analysis showed that tshb mRNA was expressed mainly in the pituitary as well as in some extrapituitary tissues including the ovary and testis. Tshr mRNA was also expressed in a tissue-specific manner, with transcripts detected in tissues including the kidney, ovary, and testis. The immunoreactive Tshb signals in the pituitary were shown to be localized to the inner areas of adenohypophysis which are close to the neurohypophysis of adult ricefield eels. Tshb-immunoreatvie cells in the pituitary of ricefield eel larvae were firstly observed at hatching. The expression of immunoreactive Tshb and Cga was also detected in ricefield eel ovary and testis together with Tshr. In the ovary, immunoreactive Tshb, Cga, and Tshr were observed in oocytes and granulosa cells. In the testis, immunoreactive Tshb was mainly observed in Sertoli cells while immunoreactive Cga and Tshr were detected in germ cells as well as somatic cells. Results of the present study suggest that Tsh may be synthesized both in the ovary and testis locally, which may play paracrine and/or autocrine roles in gonadal development in ricefield eels.


Asunto(s)
Anguilas , Receptores de Tirotropina , Animales , Receptores de Tirotropina/metabolismo , Receptores de Tirotropina/genética , Femenino , Masculino , Anguilas/metabolismo , Anguilas/genética , Testículo/metabolismo , Gónadas/metabolismo , Comunicación Paracrina/fisiología , Ovario/metabolismo , Hipófisis/metabolismo , Tirotropina de Subunidad beta/metabolismo , Tirotropina de Subunidad beta/genética , Comunicación Autocrina/fisiología
2.
Biomed Pharmacother ; 163: 114799, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37121147

RESUMEN

Cardiac hypertrophy is characterized by cardiac structural remodeling, fibrosis, microvascular rarefaction, and chronic inflammation. The heart is structurally organized by different cell types, including cardiomyocytes, fibroblasts, endothelial cells, and immune cells. These cells highly interact with each other by a number of paracrine or autocrine factors. Cell-cell communication is indispensable for cardiac development, but also plays a vital role in regulating cardiac response to damage. Although cardiomyocytes and fibroblasts are deemed as key regulators of hypertrophic stimulation, other cells, including endothelial cells, also exert important effects on cardiac hypertrophy. More particularly, endothelial cells are the most abundant cells in the heart, which make up the basic structure of blood vessels and are widespread around other cells in the heart, implicating the great and inbuilt advantage of intercellular crosstalk. Cardiac microvascular plexuses are essential for transport of liquids, nutrients, molecules and cells within the heart. Meanwhile, endothelial cell-mediated paracrine signals have multiple positive or negative influences on cardiac hypertrophy. However, a comprehensive discussion of these influences and consequences is required. This review aims to summarize the basic function of endothelial cells in angiogenesis, with an emphasis on angiogenic molecules under hypertrophic conditions. The secondary objective of the research is to fully discuss the key molecules involved in the intercellular crosstalk and the endothelial cell-mediated protective or detrimental effects on other cardiac cells. This review provides a more comprehensive understanding of the overall role of endothelial cells in cardiac hypertrophy and guides the therapeutic approaches and drug development of cardiac hypertrophy.


Asunto(s)
Cardiomegalia , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Comunicación Paracrina/fisiología , Fibroblastos/metabolismo
3.
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
4.
Nat Commun ; 13(1): 4130, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840551

RESUMEN

Fate determination and maintenance of fetal testes in most mammals occur cell autonomously as a result of the action of key transcription factors in Sertoli cells. However, the cases of freemartin, where an XX twin develops testis structures under the influence of an XY twin, imply that hormonal factor(s) from the XY embryo contribute to sex reversal of the XX twin. Here we show that in mouse XY embryos, Sertoli cell-derived anti-Mullerian hormone (AMH) and activin B together maintain Sertoli cell identity. Sertoli cells in the gonadal poles of XY embryos lacking both AMH and activin B transdifferentiate into their female counterpart granulosa cells, leading to ovotestis formation. The ovotestes remain to adulthood and produce both sperm and oocytes, although there are few of the former and the latter fail to mature. Finally, the ability of XY mice to masculinize ovaries is lost in the absence of these two factors. These results provide insight into fate maintenance of fetal testes through the action of putative freemartin factors.


Asunto(s)
Activinas , Hormona Antimülleriana , Diferenciación Celular , Testículo , Activinas/metabolismo , Activinas/farmacología , Animales , Hormona Antimülleriana/metabolismo , Hormona Antimülleriana/farmacología , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/fisiología , Diferenciación Celular/fisiología , Femenino , Masculino , Mamíferos , Ratones , Comunicación Paracrina/fisiología , Semen , Células de Sertoli , Testículo/metabolismo
5.
J Exp Clin Cancer Res ; 41(1): 48, 2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35109895

RESUMEN

BACKGROUND: Perineural invasion (PNI) and autophagy are two common features in the tumor microenvironment of pancreatic cancer (PanCa) and have a negative effect on prognosis. Potential mediator cells and the molecular mechanism underlying their relationships need to be fully elucidated. METHODS: To investigate the autophagy of Schwann cells (SCs) in PNI, we reproduced the microenvironment of PNI by collecting clinical PNI tissue, performing sciatic nerve injection of nude mice with cancer cells and establishing a Dorsal root ganglion (DRG) coculture system with cancer cell lines. Autophagy was detected by IHC, IF, transmission electron microscopy (TEM) and western blotting assays. Apoptosis was detected by IF, TEM and western blotting. NGF targeting molecular RO 08-2750(RO) and the autophagy inhibitor Chloroquine (CQ) were utilized to evaluate the effect on autophagy and apoptosis in SCs and PanCa cells in PNI samples. RESULTS: SC autophagy is activated in PNI by paracrine NGF from PanCa cells. Autophagy-activated Schwann cells promote PNI through a) enhanced migration and axon guidance toward PanCa cells and b) increased chemoattraction to PanCa cells. The NGF-targeting reagent RO and autophagy inhibitor CQ inhibited Schwann cell autophagic flux and induced Schwann cell apoptosis. Moreover, RO and CQ could induce PanCa cell apoptosis and showed good therapeutic effects in the PNI model. CONCLUSIONS: PanCa cells can induce autophagy in SCs through paracrine pathways such as the NGF/ATG7 pathway. Autophagic SCs exert a "nerve-repair like effect", induce a high level of autophagy of cancer cells, provide a "beacon" for the invasion of cancer cells to nerve fibers, and induce directional growth of cancer cells. Targeting NGF and autophagy for PNI treatment can block nerve infiltration and is expected to provide new directions and an experimental basis for the research and treatment of nerve infiltration in pancreatic cancer.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Factor de Crecimiento Nervioso/metabolismo , Comunicación Paracrina/fisiología , Células de Schwann/metabolismo , Animales , Autofagia , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Invasividad Neoplásica/patología , Ratas , Transfección , Microambiente Tumoral
6.
Aging (Albany NY) ; 14(1): 195-224, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35020601

RESUMEN

Endothelial defects significantly contribute to cardiovascular pathology in the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS). Using an endothelium-specific progeria mouse model, we identify a novel, endothelium-specific microRNA (miR) signature linked to the p53-senescence pathway and a senescence-associated secretory phenotype (SASP). Progerin-expressing endothelial cells exert profound cell-non-autonomous effects initiating senescence in non-endothelial cell populations and causing immune cell infiltrates around blood vessels. Comparative miR expression analyses revealed unique upregulation of senescence-associated miR34a-5p in endothelial cells with strong accumulation at atheroprone aortic arch regions but also, in whole cardiac- and lung tissues as well as in the circulation of progeria mice. Mechanistically, miR34a-5p knockdown reduced not only p53 levels but also late-stage senescence regulator p16 with no effect on p21 levels, while p53 knockdown reduced miR34a-5p and partially rescued p21-mediated cell cycle inhibition with a moderate effect on SASP. These data demonstrate that miR34a-5p reinforces two separate senescence regulating branches in progerin-expressing endothelial cells, the p53- and p16-associated pathways, which synergistically maintain a senescence phenotype that contributes to cardiovascular pathology. Thus, the key function of circulatory miR34a-5p in endothelial dysfunction-linked cardiovascular pathology offers novel routes for diagnosis, prognosis and treatment for cardiovascular aging in HGPS and potentially geriatric patients.


Asunto(s)
Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/fisiología , Lamina Tipo A/metabolismo , MicroARNs/metabolismo , Progeria/metabolismo , Regulación hacia Arriba/fisiología , Envejecimiento , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aterosclerosis/metabolismo , Senescencia Celular , Regulación hacia Abajo , Lamina Tipo A/genética , Ratones , MicroARNs/genética , Comunicación Paracrina/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
J Neuroimmunol ; 364: 577792, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35030439

RESUMEN

Intense mental stimulation and stress often directly induce or exacerbate psoriasis. On the contrary, patients with nerve injury and nervous system dysfunction have psoriasis remission. The nervous system plays an important role in the inflammatory process of psoriasis, and neuropeptides are considered as local mediators of disease maintenance. To examine the molecular mechanism involved in this, first we analyzed calcitonin gene-related peptide (CGRP)-treated langerhans Cells and γδ-T cells separately. CGRP induced IL-23 mRNA and protein expression via PDK1-Rsk signaling pathway. However, CGRP had no effect on secretion of IL-17A and IL-22 in γδ-T cells. Then we treated LCs/γδ-T cells Co-culture Model with CGRP. CGRP upregulated IL-17A and IL-22 expression in co-culture model through the paracrine effect of LCs. IL-17A and IL-22 are key cytokines of psoriasis. These findings provide a potential mechanism by which nerve factors affect the development of psoriasis.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Linfocitos Intraepiteliales/metabolismo , Células de Langerhans/metabolismo , Comunicación Paracrina/fisiología , Psoriasis/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Humanos , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Neuroinmunomodulación/fisiología , Regulación hacia Arriba , Interleucina-22
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.
Mech Ageing Dev ; 200: 111593, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34756925

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease. Exosomes are extracellular vesicles secreted by a variety of cells, and they participate in intercellular communication by transferring microRNA (miRNA) and other substances. Among the various internal and external factors involved in the occurrence and development of AD, exosome-derived miRNAs have become essential in the pathogenesis and treatment of AD. As nanocarriers of miRNA, exosomes are expected to become an important tool in the pathogenesis, diagnosis, and treatment of AD. This article reviews the roles of exosomal miRNAs in the pathophysiological process, diagnostic biomarkers and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer , Exosomas/genética , MicroARNs/metabolismo , Comunicación Paracrina/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Biomarcadores/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Humanos , Transducción de Señal/fisiología
10.
Development ; 148(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34651174

RESUMEN

During embryonic development and tissue homeostasis, reproducible proportions of differentiated cell types are specified from populations of multipotent precursor cells. Molecular mechanisms that enable both robust cell-type proportioning despite variable initial conditions in the precursor cells, and the re-establishment of these proportions upon perturbations in a developing tissue remain to be characterized. Here, we report that the differentiation of robust proportions of epiblast-like and primitive endoderm-like cells in mouse embryonic stem cell cultures emerges at the population level through cell-cell communication via a short-range fibroblast growth factor 4 (FGF4) signal. We characterize the molecular and dynamical properties of the communication mechanism and show how it controls both robust cell-type proportioning from a wide range of experimentally controlled initial conditions, as well as the autonomous re-establishment of these proportions following the isolation of one cell type. The generation and maintenance of reproducible proportions of discrete cell types is a new function for FGF signaling that might operate in a range of developing tissues.


Asunto(s)
Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Células Madre Embrionarias de Ratones/citología , Animales , Tipificación del Cuerpo , Desarrollo Embrionario , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Factor 4 de Crecimiento de Fibroblastos/genética , Estratos Germinativos/citología , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Comunicación Paracrina/fisiología , Transducción de Señal
11.
Toxicology ; 460: 152883, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34352351

RESUMEN

3,5,6-Trichloro-2-pyridinol (TCP) is an important biomarker and one of the final metabolites of chlorpyrifos (CPF). TCP inhibits secretion of sex hormones. Similar to CPF, TCP can bind to sex steroid hormone receptors and decrease the secretion of sex hormones. However, little attention has been paid to the ability of TCP and CPF to interfere with androgen receptor (AR) in Sertoli cells. This study aimed to explain how TCP promotes the inhibitory effect of CPF on the paracrine function of Sertoli cells. Western blotting indicated that after 20 weeks of exposure, expression of AR in testes was significantly reduced by CPF. An in vitro assay measured the cytotoxicity of CPF, TCP and diethylphosphate (DEP) on viability of Sertoli cells by Cell Counting Kit-8. CPF cytotoxicity was greater than that of TCP, and TCP cytotoxicity was greater than that of DEP at concentrations of 1000 µmol/L. Western blotting indicated that TCP and CPF both decreased expression of AR and cAMP-response element binding protein phosphorylation, while DEP had no effect in Sertoli cells, which are important in regulating paracrine function of Sertoli cells. The fluorescence measurements and docking studies revealed that testosterone, CPF and TCP showed four types of intermolecular interactions with AR, highlighting alkyl bonds with some of the same amino acids. Compared with testosterone, CPF and TCP also showed significant synergistic interaction with AR. CPF interacted with more amino acids and interaction energy than TCP did. This research elucidates TCP in the antiandrogenic effect of CPF on the paracrine function and suggests that TCP or chemicals with a trichloropyridine structure must be considered during reproductive toxicity assessment of potential environmental pollutants.


Asunto(s)
Antagonistas de Receptores Androgénicos/toxicidad , Cloropirifos/toxicidad , Comunicación Paracrina/efectos de los fármacos , Piridonas/toxicidad , Receptores Androgénicos/metabolismo , Células de Sertoli/metabolismo , Antagonistas de Receptores Androgénicos/administración & dosificación , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cloropirifos/administración & dosificación , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Herbicidas/administración & dosificación , Herbicidas/toxicidad , Humanos , Insecticidas/administración & dosificación , Insecticidas/toxicidad , Masculino , Comunicación Paracrina/fisiología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Piridonas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Células de Sertoli/efectos de los fármacos
12.
Prostate ; 81(13): 926-937, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34254335

RESUMEN

Advances in prostatic stroma studies over the past few decades have demonstrated that the stroma not only supports and nourishes the gland's secretory epithelium but also participates in key aspects of morphogenesis, in the prostate's hormonal metabolism, and in the functionality of the secretory epithelium. Furthermore, the stroma is implicated in the onset and progression of prostate cancer through the formation of the so-called reactive stroma, which corresponds to a tumorigenesis-permissive microenvironment. Prostatic stromal cells are interconnected and exchange paracrine signals among themselves in a gland that is highly sensitive to endocrine hormones. There is a growing body of evidence that telocytes, recently detected interstitial cells that are also present in the prostate, are involved in stromal organization, so that their processes form a network of interconnections with both the epithelium and the other stromal cells. The present review provides an update on the different types of prostate stromal cells, their interrelationships and implications for prostate development, physiology and pathological conditions.


Asunto(s)
Próstata/patología , Células del Estroma/patología , Animales , Humanos , Masculino , Comunicación Paracrina/fisiología , Neoplasias de la Próstata/patología
13.
Am J Physiol Heart Circ Physiol ; 321(1): H112-H127, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34085844

RESUMEN

Mesenchymal stromal cells (MSCs) have been studied for nearly two decades as a therapy for myocardial restoration. An emerging direction to repair myocardium is through their paracrine function, which includes the utilization of MSC-derived conditioned medium or extracellular vesicles. In this review, we go over the unique characteristics of MSCs that make it suitable for "off the shelf," cell-free regenerative therapy, current MSC-derived cell-free approaches including their advantages and disadvantages, and the known mechanisms of action of the paracrine effect of MSCs. With a summary of the clinical trials and preclinical studies of MSC-derived cell-free therapy, we classify the aforementioned mechanisms into angiogenesis, immunomodulation, extracellular matrix remodeling, antiapoptosis, and antioxidation. Particularly, we discuss on ways researchers have worked toward enhancing these desired properties to improve the therapeutic outcomes and the investigation of mechanobiology involved in MSC paracrine function. Lastly, we bring up the remaining challenges in this arising field and suggestions for future directions to improve our understanding and control over the potential of MSC paracrine function for myocardial restoration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Miocardio/citología , Comunicación Paracrina/fisiología , Animales , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Medicina Regenerativa
14.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166187, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34102256

RESUMEN

Deficiency of angiogenic and neurotrophic factors under long term diabetes is known to lead to Schwann cell degeneration, clinically manifested as Diabetic Neuropathy (DN). While the transplantation of exogenous allogenic Mesenchymal Stromal Cells (MSCs) has shown amelioration of DN through paracrine action, it is not known what functional changes occur in endogenous bone-marrow MSCs under chronic diabetes in terms of homing, migration and/or paracrine signalling with reference to the end-point clinical manifestation of Diabetic Neuropathy. We thus aimed at determining the changes in BM-MSCs under Type 1 Diabetes with respect to survival, self-renewal, oxidative status, paracrine activity, intracellular Ca2+ response and migration in response to pathological cytokine/chemokine, in reference to the time-point of decline in Nerve Conduction Velocity (NCV) in a rat model. Within one week of diabetes induction, BM-MSCs underwent apoptosis, and compromised their self-renewal capacity, antioxidant defence mechanism and migration toward cytokine/chemokine; whereas epineurial blood vessel thickening and demyelination resulting in NCV decline were observed only after three weeks. By two- and three-weeks post diabetes induction, BM-MSC apoptosis reduced and proliferative ability was restored; however, their self-renewal, migration and intracellular Ca2+ response toward pathological cytokine/chemokine remained impaired. These results indicate that T1D induced intrinsic functional impairments in endogenous BM-MSCs occur before neuropathy onset. This timeline of functional alterations in BM-MSCs also suggest that treatment strategies that target the bone marrow niche early on may help to modulate BM-MSC functional impairments and thus slow down the progression of neuropathy.


Asunto(s)
Médula Ósea/patología , Diabetes Mellitus Tipo 1/patología , Neuropatías Diabéticas/patología , Células Madre Mesenquimatosas/patología , Animales , Apoptosis/fisiología , Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Neuropatías Diabéticas/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina/fisiología , Ratas , Ratas Wistar
15.
Purinergic Signal ; 17(3): 345-370, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33982134

RESUMEN

Cancer comprises a collection of diseases that occur in almost any tissue and it is characterized by an abnormal and uncontrolled cell growth that results in tumor formation and propagation to other tissues, causing tissue and organ malfunction and death. Despite the undeniable improvement in cancer diagnostics and therapy, there is an urgent need for new therapeutic and preventive strategies with improved efficacy and fewer side effects. In this context, purinergic signaling emerges as an interesting candidate as a cancer biomarker or therapeutic target. There is abundant evidence that tumor cells have significant changes in the expression of purinergic receptors, which comprise the G-protein coupled P2Y and AdoR families of receptors and the ligand-gated ion channel P2X receptors. Tumor cells also exhibit changes in the expression of nucleotidases and other enzymes involved in nucleotide metabolism, and the concentrations of extracellular nucleotides are significantly higher than those observed in normal cells. In this review, we will focus on the potential role of purinergic signaling in the ten most lethal cancers (lung, breast, colorectal, liver, stomach, prostate, cervical, esophagus, pancreas, and ovary), which together are responsible for more than 5 million annual deaths.


Asunto(s)
Adenosina Trifosfato/metabolismo , Comunicación Autocrina/fisiología , Neoplasias/metabolismo , Comunicación Paracrina/fisiología , Receptores Purinérgicos/metabolismo , Adenosina Trifosfato/genética , Animales , Humanos , Neoplasias/genética , Neoplasias/mortalidad , Receptores Purinérgicos/genética , Transducción de Señal/fisiología
16.
Cell Mol Life Sci ; 78(10): 4521-4544, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34019103

RESUMEN

Cellular senescence is a process that can prevent tumour development in a cell autonomous manner by imposing a stable cell cycle arrest after oncogene activation. Paradoxically, senescence can also promote tumour growth cell non-autonomously by creating a permissive tumour microenvironment that fuels tumour initiation, progression to malignancy and metastasis. In a pituitary tumour known as adamantinomatous craniopharyngioma (ACP), cells that carry oncogenic ß-catenin mutations and overactivate the WNT signalling pathway form cell clusters that become senescent and activate a senescence-associated secretory phenotype (SASP). Research in mouse models of ACP has provided insights into the function of the senescent cell clusters and revealed a critical role for SASP-mediated activities in paracrine tumour initiation. In this review, we first discuss this research on ACP and subsequently explore the theme of paracrine tumourigenesis in other tumour models available in the literature. Evidence is accumulating supporting the notion that paracrine signalling brought about by senescent cells may underlie tumourigenesis across different tumours and cancer models.


Asunto(s)
Carcinogénesis/patología , Senescencia Celular/fisiología , Craneofaringioma/patología , Comunicación Paracrina/fisiología , Animales , Humanos , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
17.
Prostate ; 81(7): 407-417, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33734457

RESUMEN

Growing evidence supports the pivotal role played by periprostatic adipose tissue (PPAT) in prostate cancer (PCa) microenvironment. We investigated whether PPAT can affect response to Docetaxel (DCTX) and the mechanisms associated. Conditioned medium was collected from the in vitro differentiated adipocytes isolated from PPAT which was isolated from PCa patients, during radical prostatectomy. Drug efficacy was studied by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide citotoxicity assay. Culture with CM of human PPAT (AdipoCM) promotes DCTX resistance in two different human prostate cancer cell lines (DU145 and PC3) and upregulated the expression of BCL-xL, BCL-2, and TUBB2B. AG1024, a well-known IGF-1 receptor inhibitor, counteracts the decreased response to DCTX observed in presence of AdipoCM and decreased TUBB2B expression, suggesting that a paracrine secretion of IGF-1 by PPAT affect DCTX response of PCa cell. Collectively, our study showed that factors secreted by PPAT elicits DCTX resistance through antiapoptotic proteins and TUBB2B upregulation in androgen independent PCa cell lines. These findings reveal the potential of novel therapeutic strategies targeting adipocyte-released factors and IGF-1 axis to overcome DCTX resistance in patients with PCa.


Asunto(s)
Tejido Adiposo/metabolismo , Antineoplásicos/uso terapéutico , Docetaxel/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Tubulina (Proteína)/metabolismo , Tejido Adiposo/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Comunicación Paracrina/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Tubulina (Proteína)/genética , Regulación hacia Arriba
18.
Drug Discov Today ; 26(8): 2036-2044, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33775925

RESUMEN

Aging and pre-existing conditions in older patients increase severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) severity and its complications, although the causes remain unclear. Apart from acute pulmonary syndrome, Coronavirus 2019 (COVID-19) can increasingly induce chronic conditions. Importantly, SARS-CoV-2 triggers de novo type 2 diabetes mellitus (T2DM) linked to age-associated cardiovascular disease (CVD), cancers, and neurodegeneration. Mechanistically, SARS-CoV-2 induces inflammation, possibly through damage-associated molecular pattern (DAMP) signaling and 'cytokine storm,' causing insulin resistance and the adiponectin (APN) paradox, a phenomenon linking metabolic dysfunction to chronic disease. Accordingly, preventing the APN paradox by suppressing APN-related inflammatory signaling might prove beneficial. A better understanding could uncover novel therapies for SARS-CoV-2 and its chronic disorders.


Asunto(s)
Adiponectina/metabolismo , Envejecimiento/fisiología , COVID-19 , Diabetes Mellitus Tipo 2/inmunología , Inflamación/metabolismo , SARS-CoV-2 , COVID-19/inmunología , COVID-19/metabolismo , Enfermedad Crónica , Humanos , Comunicación Paracrina/fisiología , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología
19.
Reprod Sci ; 28(6): 1718-1732, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33751459

RESUMEN

Human umbilical cord mesenchymal stem cell (UC-MSC) application is a promising arising therapy for the treatment of premature ovarian failure (POF). However, little is known about the inflammation regulatory effects of human umbilical cord MSCs (UC-MSCs) on chemotherapy-induced ovarian damage, regardless of in vivo or in vitro. This study was designed to investigate the therapeutic effects of UC-MSC transplantation and underlying mechanisms regarding both apoptosis and inflammation in POF mice. The chemotherapy-induced POF models were induced by intraperitoneal injection of cyclophosphamide. Ovarian function parameters, granulosa cell (GC) apoptosis, and inflammation were examined. Morphological staining showed that UC-MSC treatment increased the ovary size, and the numbers of primary and secondary follicles, but decreased the number of atretic follicles. Estradiol levels in the UC-MSC-treated group were increased while follicle-stimulating hormone levels were reduced compared to those in the POF group. UC-MSCs inhibited cyclophosphamide-induced GC apoptosis and inflammation. Meanwhile, phosphorylation of AKT and P38 was elevated after UC-MSC treatment. Tracking of UC-MSCs in vivo indicated that transplanted UC-MSCs were only located in the interstitium of ovaries rather than in follicles. Importantly, UC-MSC-derived extracellular vesicles protected GCs from alkylating agent-induced apoptosis and inflammation in vitro. Our results suggest that UC-MSC transplantation can reduce ovary injury and improve ovarian function in chemotherapy-induced POF mice through anti-apoptotic and anti-inflammatory effects via a paracrine mechanism.


Asunto(s)
Apoptosis/fisiología , Inflamación/prevención & control , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Insuficiencia Ovárica Primaria/terapia , Cordón Umbilical/citología , Animales , Antineoplásicos/efectos adversos , Femenino , Células de la Granulosa/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ovario/fisiopatología , Comunicación Paracrina/fisiología , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/fisiopatología
20.
J Neurochem ; 158(2): 153-168, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33704788

RESUMEN

γ-Aminobutyric acid (GABA) is thought to play a paracrine role in adrenal medullary chromaffin (AMC) cells. Comparative physiological and immunocytochemical approaches were used to address the issue of how the paracrine function of GABA in AMC cells is established. GABAA receptor Cl- channel activities in AMC cells of rats and mice, where corticosterone is the major glucocorticoid, were much smaller than those in AMC cells of guinea-pigs and cattle, where cortisol is the major. The extent of enhancement of GABAA receptor α3 subunit expression in rat pheochromocytoma (PC12) cells by cortisol was larger than that by corticosterone in parallel with their glucocorticoid activities. Thus, the species difference in GABAA receptor expression may be ascribed to a difference in glucocorticoid activity between corticosterone and cortisol. GABAA receptor Cl- channel activity in mouse AMC cells was enhanced by allopregnanolone, as noted with that in guinea-pig AMC cells, and the enzymes involved in allopregnanolone production were immunohistochemically detected in the zona fasciculata in both mice and guinea pigs. The expression of glutamic acid decarboxylase 67 (GAD67), one of the GABA synthesizing enzymes, increased after birth, whereas GABAA receptors already developed at birth. Stimulation of pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, but not nicotinic or muscarinic receptors, in PC12 cells, resulted in an increase in GAD67 expression in a protein-kinase A-dependent manner. The results indicate that glucocorticoid and PACAP are mainly responsible for the expressions of GABAA receptors and GAD67 involved in GABA signaling in AMC cells, respectively.


Asunto(s)
Médula Suprarrenal/fisiología , Células Cromafines/fisiología , Comunicación Paracrina/fisiología , Ácido gamma-Aminobutírico/fisiología , Médula Suprarrenal/citología , Animales , Bovinos , Canales de Cloruro/metabolismo , Cricetinae , Glutamato Descarboxilasa/metabolismo , Cobayas , Hidrocortisona/metabolismo , Inmunohistoquímica , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Células PC12 , Pregnanolona/farmacología , Ratas , Receptores de GABA-A/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA