Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 785
Filtrar
1.
Cell Rep ; 38(7): 110363, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35172147

RESUMEN

Thymic atrophy reduces naive T cell production and contributes to increased susceptibility to viral infection with age. Expression of tissue-restricted antigen (TRA) genes also declines with age and has been thought to increase autoimmune disease susceptibility. We find that diminished expression of a model TRA gene in aged thymic stromal cells correlates with impaired clonal deletion of cognate T cells recognizing an autoantigen involved in atherosclerosis. Clonal deletion in the polyclonal thymocyte population is also perturbed. Distinct age-associated defects in the generation of antigen-specific T cells include a conspicuous decline in generation of T cells recognizing an immunodominant influenza epitope. Increased catalase activity delays thymic atrophy, and here, we show that it mitigates declining production of influenza-specific T cells and their frequency in lung after infection, but does not reverse declines in TRA expression or efficient negative selection. These results reveal important considerations for strategies to restore thymic function.


Asunto(s)
Envejecimiento/inmunología , Antígenos/inmunología , Inmunidad , Autotolerancia/inmunología , Linfocitos T/inmunología , Animales , Antioxidantes/farmacología , Apolipoproteínas B/metabolismo , Atrofia , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Catalasa/metabolismo , Suplementos Dietéticos , Inmunidad/efectos de los fármacos , Epítopos Inmunodominantes/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/inmunología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Autotolerancia/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Linfocitos T/efectos de los fármacos , Timo/patología
2.
Am J Physiol Renal Physiol ; 322(3): F295-F307, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35037469

RESUMEN

Cyclooxygenase (Cox) inhibitors are known to have severe side effects during renal development. These consist of reduced renal function, underdeveloped subcapsular glomeruli, interstitial fibrosis, and thinner cortical tissue. Global genetic deletion of Cox-2 mimics the phenotype observed after application of Cox inhibitors. This study aimed to investigate which cell types express Cox-2 and prostaglandin E2 receptors and what functions are mediated through this pathway during renal development. Expression of EP2 and EP4 mRNA was detected by RNAscope mainly in descendants of FoxD1+ stromal progenitors; EP1 and EP3, on the other hand, were expressed in tubules. Cox-2 mRNA was detected in medullary interstitial cells and macula densa cells. Functional investigations were performed with a cell-specific approach to delete Cox-2, EP2, and EP4 in FoxD1+ stromal progenitor cells. Our data show that Cox-2 expression in macula densa cells is sufficient to drive renal development. Deletion of EP2 or EP4 in FoxD1+ cells had no functional effect on renal development. Codeletion of EP2 and EP4 in FoxD1+ stromal cells, however, led to severe glomerular defects and a strong decline of glomerular filtration rate (1.316 ± 69.7 µL/min/100 g body wt in controls vs. 644.1 ± 64.58 µL/min/100 g body wt in FoxD1+/Cre EP2-/- EP4ff mice), similar to global deletion of Cox-2. Furthermore, EP2/EP4-deficient mice showed a significant increase in collagen production with a strong downregulation of renal renin expression. This study shows the distinct localization of EP receptors in mice. Functionally, we could identify EP2 and EP4 receptors in stromal FoxD1+ progenitor cells as essential receptor subtypes for normal renal development.NEW & NOTEWORTHY Cyclooxygenase-2 (Cox-2) produces prostaglandins that are essential for normal renal development. It is unclear in which cells Cox-2 and the receptors for prostaglandin E2 (EP receptors) are expressed during late nephrogenesis. This study identified the expression sites for EP subtypes and Cox-2 in neonatal mouse kidneys. Furthermore, it shows that stromal progenitor cells may require intact prostaglandin E2 signaling through EP2 and EP4 receptors for normal renal development.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Corteza Renal/enzimología , Prostaglandinas/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Madre/metabolismo , Células del Estroma/enzimología , Animales , Ciclooxigenasa 2/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corteza Renal/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/genética , Transducción de Señal
3.
Toxicology ; 465: 153050, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34826546

RESUMEN

Emerging evidence suggests that excess iron accumulates in endometriotic and adenomyotic lesions. However, the role iron overload plays in the pathogenesis of endometriosis or adenomyosis remains unknown. Primary human eutopic endometrial stromal cells (EuESCs) from endometriosis or adenomyosis patients were used as the in vitro model of endometriosis or adenomyosis in this study. We found that iron, manifesting as ferric ammonium citrate (FAC; 0.05-4.8 mM), significantly inhibited cell growth, induced oxidative stress through the Fenton reaction, and functionally activated autophagy in EuESCs, as measured by 5-ethynyl-2'-deoxyuridine incorporation assay, MitoSOX™ Red staining, LC3 turnover assay, and tandem mCherry-eGFP-LC3B fluorescence microscopy. Immunohistochemistry analysis of Ki67 expression in proliferative-phase endometrial tissues revealed that cell proliferation in ectopic tissues was dramatically compromised, suggesting that iron overload may play a role in cell growth inhibition in vivo. We observed that autophagy may alleviate the FAC-induced inhibition of endometrial stromal cell proliferation. Furthermore, sequential FAC (0.8 mM, 24 h) and hydrogen peroxide (H2O2; 300 µM, 2 h) treatment successfully induced the Fenton reaction in EuESCs and caused extensive apoptosis, whereas the disruption of autophagy by the knockdown of BECN1 further aggravated cell death. MitoSOX™ Red staining showed that autophagy may promote the survival of EuESCs by decreasing of the Fenton reaction-induced reactive oxygen species generation. In addition, we observed that the Fenton reaction-induced oxidative stress significantly suppressed iron overload-induced autophagy. Moreover, we found that FAC treatment impaired poly(ADP-ribose)-polymerase 1 (PARP1) expression while simultaneously upregulating SIRT1 expression in EuESCs. Our data further showed that PARP1 expression decreased in endometriotic lesions, which may partially result from iron overload. We also found that PARP1 inhibition aggravated iron overload-induced cell growth suppression, and was implicated in iron overload-induced autophagy. In addition, SIRT1 silencing alleviated iron overload-induced PARP1 downregulation and autophagy activation. Overall, our data suggest that iron overload in endometrial stromal cells of endometriotic or adenomyotic lesions may be involved in the inhibition of cell proliferation, simultaneously with the activation of protective autophagy via PARP1/SIRT1 signaling.


Asunto(s)
Adenomiosis/complicaciones , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endometriosis/complicaciones , Endometrio/efectos de los fármacos , Compuestos Férricos/toxicidad , Sobrecarga de Hierro/complicaciones , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Compuestos de Amonio Cuaternario/toxicidad , Sirtuina 1/metabolismo , Células del Estroma/efectos de los fármacos , Adenomiosis/enzimología , Adenomiosis/patología , Adulto , Beclina-1/genética , Beclina-1/metabolismo , Células Cultivadas , Endometriosis/enzimología , Endometriosis/patología , Endometrio/enzimología , Endometrio/patología , Femenino , Humanos , Sobrecarga de Hierro/enzimología , Sobrecarga de Hierro/patología , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1/genética , Transducción de Señal , Sirtuina 1/genética , Células del Estroma/enzimología , Células del Estroma/patología , Adulto Joven
4.
Bioengineered ; 13(1): 800-809, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34964708

RESUMEN

Hypoxia-induced damage in endometrial stromal cells (ESCs) is an important event in the pathological progression of Endometriosis. It is reported that significant inflammation is induced by hypoxia in ESCs, mediated by serval inflammatory progressions, pathways, or factors. Sitagliptin, an important member of the dipeptidyl peptidase-4 (DPP-4) inhibitors family and has been widely used for the management of type 2 diabetes. It has been recently reported to exert significant anti-inflammatory effects. Here, we aim to assess whether Sitagliptin possesses a protective effect against hypoxia-induced damages in ESCs. Our findings indicate that exposure to hypoxia significantly increased oxidative stress in ESCs by increasing the production of reactive oxygen species (ROS) and decreasing the levels of reduced glutathione (GSH), which was ameliorated by Sitagliptin. Additionally, the excessively produced inflammatory mediators, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemoattractant protein-1 (MCP-1), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and high mobility group box (HMGB)-1, in hypoxia-treated HESCs were pronouncedly repressed by Sitagliptin. The activated p38 mitogen-activated protein kinases (MAPK) pathway was observed in hypoxia-stimulated HESCs, then greatly inhibited by the introduction of Sitagliptin. Lastly, hypoxia-induced phosphorylation and degradation of IκBα, as well as the upregulation of nuclear factor kappa-B (NF-κB) p65 and increased transcriptional activity of NF-κB, were dramatically abolished by Sitagliptin. Collectively, Sitagliptin ameliorated hypoxia-induced damages in ESCs by suppressing the inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Endometriosis/metabolismo , Endometrio/citología , Fosfato de Sitagliptina/farmacología , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Progresión de la Enfermedad , Endometriosis/tratamiento farmacológico , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Glutatión/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteolisis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Células del Estroma/citología , Células del Estroma/enzimología , Células del Estroma/metabolismo
6.
Biomed Pharmacother ; 141: 111899, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34346316

RESUMEN

The microenvironment surrounding the tumor affects biological processes, such as cell proliferation, angiogenesis, apoptosis, and invasion. Therefore, the ability to change these environments is an important attribute for tumor cells to obtain specific functions necessary for growth and metastasis. Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic metalloenzymes that facilitate protease-dependent tumor progression by degrading extracellular matrix (ECM) proteins, releasing cytokines, growth factors, and other cell surface molecules. As one of the most widely studied MMPs, MMP-11 is an important protease that is expressed in cancer cells, stromal cells, and the adjacent microenvironment. MMP-11 has a dual effect on tumors. On one hand, MMP-11 promotes tumor development by inhibiting apoptosis and promoting the migration and invasion of cancer cells in the early stage. On the other hand, in animal models, MMP-11 has a protective effect on tumor growth and metastasis at an advanced stage. Based on current findings regarding the importance of MMP-11 in altering the tumor microenvironment, there is a need to further understand how stromal cells and the ECM regulate tumor progression, which may result in the re-examination of MMPs as drug targets for cancer and other diseases. In this review, we summarize the dual role of MMP-11 in cancer and its potential clinical significance.


Asunto(s)
Metaloproteinasa 11 de la Matriz/fisiología , Neoplasias/enzimología , Neoplasias/fisiopatología , Animales , Biomarcadores , Humanos , Metaloproteinasa 11 de la Matriz/metabolismo , Neovascularización Patológica , Células del Estroma/enzimología , Microambiente Tumoral
7.
Am J Reprod Immunol ; 86(5): e13480, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34191381

RESUMEN

PROBLEM: Decidual macrophages (dMφ ) play an important role in the formation of maternal-fetal immune tolerance. However, factors that influence the immune status of dMφ and the related potential mechanisms have not been elucidated to date. METHOD OF STUDY: The gene transcription in dMφ , decidual stromal cells (DSCs), extravillous trophoblasts (EVTs), and peripheral monocytes (pMo) from human samples were measured using real-time polymerase chain reaction (PCR). Monocyte-DSC co-culture was established to explore whether DSCs influenced dMφ polarization via C-C motif ligand 2 (CCL2)-C-C chemokine receptor (CCR2) binding using flow cytometry. In vivo, changes in dMφ percentage and M1 and M2 marker expression after treatment with CCR2 or Janus kinase 2 (JAK2) inhibitor were detected with flow cytometry. Embryo resorption percentages in the above groups were also analyzed. RESULTS: We found that dMφ were an M1/M2 mixed status at the maternal-fetal interface during early pregnancy. CCL2 influenced the immune status of dMφ in an autocrine and paracrine manner. As a downstream regulator of CCR2 and triggers the Stat3 pathway, JAK2 was found to be essential for dMφ homeostasis in vivo. JAK2 inhibitor decreased the dMφ proportion and attenuated Ki67, CD36, CD86, CD206, TNF, and IL-10 expression in dMφ at E8.5 d. Moreover, CCR2-JAK2 pathway inhibition decreased the width of the placental labyrinth layer, further influencing the pregnancy outcome. CONCLUSION: The M1/M2 mixed immune status of dMφ was regulated by DSCs via CCR2, and the CCL2/CCR2/JAK2 pathway was essential for the immune status of dMφ and the outcome of early pregnancy.


Asunto(s)
Quimiocina CCL2/metabolismo , Decidua/enzimología , Histocompatibilidad Materno-Fetal , Tolerancia Inmunológica , Janus Quinasa 2/metabolismo , Macrófagos/enzimología , Receptores CCR2/metabolismo , Células del Estroma/enzimología , Adulto , Animales , Células Cultivadas , Técnicas de Cocultivo , Decidua/efectos de los fármacos , Decidua/inmunología , Pérdida del Embrión/enzimología , Pérdida del Embrión/inmunología , Femenino , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de las Cinasas Janus/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Resultado del Embarazo , Receptores CCR2/antagonistas & inhibidores , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/inmunología , Adulto Joven
8.
J Cell Physiol ; 236(11): 7376-7389, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33959973

RESUMEN

Existing evidence suggests that adverse pregnancy outcomes are closely related to dietary factors. Folate plays an important role in neural tube formation and fetal growth, folate deficiency is a major risk factor of birth defects. Our early studies showed that folate deficiency could impair enddecidualization, however, the mechanism is still unclear. Dysfunctional autophagy is associated with many diseases. Here, we aimed to evaluate the adverse effect of folate deficiency on endometrial decidualization, with a particular focus on endometrial cell autophagy. Mice were fed with no folate diet in vivo and the mouse endometrial stromal cell was cultured in a folate-free medium in vitro. The decrease of the number of endometrial autophagosomes and the protein expressions of autophagy in the folate-deficient group indicated that autophagosome formation, autophagosome-lysosome fusion, and lysosomal degradation were inhibited. Autophagic flux examination using mCherry-GFP-LC3 transfection showed that the fusion of autophagosomes with lysosomes was inhibited by folate deficiency. Autophagy inducer rapamycin could reverse the impairment of folate deficiency on endometrial decidualization. Moreover, folate deficiency could reduce autophagy by disrupting AMPK/mTOR signaling, resulting in aberrant endometrial decidualization and adverse pregnancy outcomes. Further co-immunoprecipitation examination showed that decidual marker protein Hoxa10 could interact with autophagic marker protein Cathepsin L, and the interaction was notably reduced by folate deficiency. In conclusion, AMPK/mTOR downregulated autophagy was essential for aberrant endometrial decidualization in early pregnant mice, which could result in adverse pregnancy outcomes. This provided some new clues for understanding the causal mechanisms of birth defects induced by folate deficiency.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Decidua/enzimología , Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/metabolismo , Células del Estroma/enzimología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagosomas/enzimología , Autofagosomas/ultraestructura , Células Cultivadas , Decidua/ultraestructura , Modelos Animales de Enfermedad , Femenino , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Lisosomas/enzimología , Lisosomas/ultraestructura , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Embarazo , Transducción de Señal , Células del Estroma/ultraestructura
9.
Cardiovasc Res ; 117(6): 1557-1566, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-33705542

RESUMEN

AIMS: Patients with severe respiratory syndrome caused by SARS-CoV-2 undergo cardiac complications due to hyper-inflammatory conditions. Although the presence of the virus has been detected in the myocardium of infected patients, and infection of induced pluripotent cell-derived cardiomyocytes has been demonstrated, the reported expression of Angiotensin-Converting Enzyme-2 (ACE2) in cardiac stromal cells suggests that SARS-CoV-2 may determine cardiac injury by sustaining productive infection and increasing inflammation. METHODS AND RESULTS: We analysed expression of ACE2 receptor in primary human cardiac stromal cells derived from cardiospheres, using proteomics and transcriptomics before exposing them to SARS-CoV-2 in vitro. Using conventional and high sensitivity PCR methods, we measured virus release in the cellular supernatants and monitored the intracellular viral bioprocessing. We performed high-resolution imaging to show the sites of intracellular viral production and demonstrated the presence of viral particles in the cells with electron microscopy. We finally used RT-qPCR assays to detect genes linked to innate immunity and fibrotic pathways coherently regulated in cells after exposure to the virus. CONCLUSIONS: Our findings indicate that cardiac stromal cells are susceptible to SARS-CoV-2 infection and produce variable viral yields depending on the extent of cellular ACE2 receptor expression. Interestingly, these cells also evolved towards hyper-inflammatory/pro-fibrotic phenotypes independently of ACE2 levels. Thus, SARS-CoV-2 infection of myocardial stromal cells could be involved in cardiac injury and explain the high number of complications observed in severe cases of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Cardiopatías/virología , Miocardio/enzimología , Receptores Virales/metabolismo , SARS-CoV-2/patogenicidad , Células del Estroma/virología , Virión/patogenicidad , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/complicaciones , Chlorocebus aethiops , Femenino , Fibrosis , Cardiopatías/enzimología , Cardiopatías/patología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Miocardio/ultraestructura , Fenotipo , Receptores Virales/genética , SARS-CoV-2/ultraestructura , Esferoides Celulares , Células del Estroma/enzimología , Células del Estroma/ultraestructura , Células Vero , Virión/ultraestructura
10.
J Reprod Immunol ; 144: 103252, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33549903

RESUMEN

OBJECTIVES: Uncontrolled TH17 differentiation has been suggested to play a role in the pathogenesis of pregnancy loss. We recently showed that menstrual blood stromal/stem cells (MenSCs) alter functional features of natural killer cells. Here, we hypothesized that MenSCs could modulate differentiation of TH17 cells. METHOD: MenSCs were collected from 18 apparently healthy women and characterized. Bone marrow mesenchymal stem cells (BMSCs) served as a control. TH17 polarization and proliferation of purified T CD4+ cells were assessed by flow cytometry in a well-defined co-culture system containing T CD4+ cells and MenSCs or BMSCs. Indoleamine 2,3-Dioxygenase (IDO) activity was evaluated in MenSC and BMSC culture supernatants by a colorimetric assay. The impact of MenSCs on expression of transcription factors, RORC, T-bet, Gata3, NRP-1 and Helios were studied by qPCR. RESULTS: MenSCs significantly inhibited TH17 differentiation (p = 0.0383) and percentage of the cells co-expressing IL-17 and IFN-γ (p = 0.0023). PGE2 blockade significantly reduced percentage and proliferation of T CD4+IL-17+ (p = 0.003, p = 0.0018), T CD4+ IFN-γ+ (p = 0.002, p = 0.0022) and T CD4+IL-17+ IFN-γ+ (p = 0.004, p = 0.02) cells. MenSCs produced a considerable activity of IDO (p = 0.0002), induced a significant rise in the Treg frequency (p = 0.0091) and a sharp increase in TH17/Tregs ratio (p = 0.0022). MenSCs increased expression of NRP1 (p = 0.001), while downregulated expression of RORC in T cells (p = 0.001). CONCLUSION: Our results suggest a supportive role for MenSCs in establishing a pregnancy-friendly microenvironment in the uterus and put forth the idea that inherent abnormalities of MenSCs may be a basis for dysregulated endometrial immune network leading to pregnancy loss.


Asunto(s)
Células Madre Adultas/inmunología , Menstruación/sangre , Embarazo/inmunología , Células del Estroma/inmunología , Células Th17/inmunología , Adulto , Células Madre Adultas/enzimología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Endometrio/citología , Endometrio/inmunología , Femenino , Voluntarios Sanos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/análisis , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células del Estroma/enzimología
11.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G506-G520, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33470182

RESUMEN

The stem/progenitor cells of the developing intestine are biologically distinct from their adult counterparts. Here, we examine the microenvironmental cues that regulate the embryonic stem/progenitor population, focusing on the role of Notch pathway factor delta-like protein-1 (DLK1). mRNA-seq analyses of intestinal mesenchymal cells (IMCs) collected from embryonic day 14.5 (E14.5) or adult IMCs and a novel coculture system with E14.5 intestinal epithelial organoids were used. Following addition of recombinant DLK1 (rDLK) or Dlk1 siRNA (siDlk1), epithelial characteristics were compared using imaging, replating efficiency assays, qPCR, and immunocytochemistry. The intestinal phenotypes of littermate Dlk1+/+ and Dlk1-/- mice were compared using immunohistochemistry. Using transcriptomic analyses, we identified morphogens derived from the embryonic mesenchyme that potentially regulate the developing epithelial cells, to focus on Notch family candidate DLK1. Immunohistochemistry indicated that DLK1 was expressed exclusively in the intestinal stroma at E14.5 at the top of emerging villi, decreased after birth, and shifted to the intestinal epithelium in adulthood. In coculture experiments, addition of rDLK1 to adult IMCs inhibited organoid differentiation, whereas Dlk1 knockdown in embryonic IMCs increased epithelial differentiation to secretory lineage cells. Dlk1-/- mice had restricted Ki67+ cells in the villi base and increased secretory lineage cells compared with Dlk1+/+ embryos. Mesenchyme-derived DLK1 plays an important role in the promotion of epithelial stem/precursor expansion and prevention of differentiation to secretory lineages in the developing intestine.NEW & NOTEWORTHY Using a novel coculture system, transcriptomics, and transgenic mice, we investigated differential molecular signaling between the intestinal epithelium and mesenchyme during development and in the adult. We show that the Notch pathway factor delta-like protein-1 (DLK1) is stromally produced during development and uncover a new role for DLK1 in the regulation of intestinal epithelial stem/precursor expansion and differentiation to secretory lineages.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/enzimología , Células Epiteliales/enzimología , Mucosa Intestinal/enzimología , Células del Estroma/enzimología , Animales , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/embriología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides , Vías Secretoras , Transducción de Señal , Nicho de Células Madre , Transcriptoma
12.
Reprod Sci ; 28(2): 435-446, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32909189

RESUMEN

Endometriosis is generally characterized as a tumor-like disease because of its potential for distant metastasis and local tissue invasion, while whether osteopontin (OPN) plays a role in the pathogenesis of endometriosis has not been thoroughly investigated. We investigated the expression of OPN, urokinase plasminogen activator (uPA), phosphatidylinositol 3 kinase (PI3K), and phospho-PI3 kinase (p-PI3K) in endometrial stromal cells (ESCs). The serum concentration of OPN was determined by enzyme-linked immunosorbent assays (ELISA). OPN was downregulated to explore the corresponding change of uPA, p-PI3K, F-actin, and α-tubulin. The expression of OPN, uPA, PI3K, and p-PI3K was evaluated by western blot and quantitative real-time PCR (RT-qPCR) and the expression of F-actin and α-tubulin was confirmed by immunofluorescence assay. The proliferation and migration abilities of ESCs were investigated by CCK8, transwell, and wound scratch assays. Endometrial OPN, p-PI3K, and uPA expressions and serum OPN levels were increased in patients with endometriosis compared with the control. The expressions of p-PI3K, uPA, and α-tubulin were decreased by siRNA-OPN interference in ectopic ESCs. Activation and inhibition of the PI3K pathway apparently upregulate and downregulate uPA expression. Knockdown of OPN and inhibition of the PI3K pathway remarkably inhibited cell migration in ectopic ESCs. Meanwhile, activation of the PI3K pathway promoted the migration ability of ectopic ESCs. OPN may regulate the expression of uPA through the PI3K signal pathway to affect the migration ability of ESCs, indicating that OPN, uPA, and the PI3K pathway may be potential targets for interrupting development of endometriosis.


Asunto(s)
Movimiento Celular , Endometriosis/enzimología , Endometrio/enzimología , Osteopontina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Células del Estroma/enzimología , Adulto , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Endometriosis/genética , Endometriosis/patología , Endometrio/patología , Femenino , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Osteopontina/genética , Transducción de Señal , Células del Estroma/patología , Adulto Joven
13.
Exp Mol Pathol ; 118: 104574, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33197426

RESUMEN

BACKGROUND AND AIMS: It is becoming evident that local estrogen exposure is important in postmenopausal breast cancer patients. The microenvironment is established by breast stromal cells based on communication with tumor cells that is essential to cancer development, invasion, and metastasis. Here we investigated aromatase activity levels in both tumor and matched stromal tissues by showing their impact on the manufacturing of local estrogen and tumor progression in cases of invasive ductal carcinoma (IDC). METHODS: Tumor (T) and tumor-associated stroma (TAS) neighboring tissues were acquired from each postmenopausal patient, diagnosed with IDC, and categorized as luminal A (n = 20). The control group was formed from tumor-free breast tissue samples (N, n = 12). A microsomal-based technique was created to compare breast tissue aromatase activities using liquid chromatography - mass spectrometry. FINDINGS: We observed that the TAS tissues have the highest aromatase activities (p < 0.05). High progesterone receptor (PR) intensity levels were found to be decreasing the activity level in these tissues significantly (p < 0.05). Tumor tissue specific aromatase activity levels of postmenopausal patients' were tend to be lower compared to healthy premenopausal subjects' (3 fold, p < 0.001). In addition low activity in tumor tissues were associated with low grade and late stage cancers. CONCLUSIONS: Early detection and personalized therapy is essential for postmenopausal breast cancer patients. Together, our in-house tandem mass spectrometry technique has the potential for further development and standardization for the measurement of aromatase activity and may assist clinicians decide on therapy policies for postmenopausal IDC patients which could be an invaluable asset for precise and specific evaluation.


Asunto(s)
Aromatasa/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Toma de Decisiones , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células del Estroma/patología , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/cirugía , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/cirugía , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Medicina de Precisión , Pronóstico , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Células del Estroma/enzimología , Microambiente Tumoral
14.
Cells ; 9(12)2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33339125

RESUMEN

Transplanted mesenchymal stem/stromal cells (MSCs) are a promising and innovative approach in regenerative medicine. Their regenerative potential is partly based upon their immunomodulatory activities. One of the most investigated immunomediators in MSCs, such as in periodontal ligament-derived MSCs (hPDLSCs), is indoleamine-2,3-dioxygenase-1 (IDO-1) which is upregulated by inflammatory stimuli, like cytokines. However, there are no data concerning continuing IDO-1 expression in hPDLSCs after the removal of inflammatory stimuli, such as cytokines and toll-like receptor (TLR) agonist-2 and TLR-3. Hence, primary hPDLSCs were stimulated with interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, TLR-2 agonist Pam3CSK4 or TLR-3 agonist Poly I/C. IDO-1 gene and protein expression and its enzymatic activity were measured up to five days after removing any stimuli. IL-1ß- and TNF-α-induced IDO-1 expression and enzymatic activity decreased in a time-dependent manner after cessation of stimulation. IFN-γ caused a long-lasting effect on IDO-1 up to five days after removing IFN-γ. Both, TLR-2 and TLR-3 agonists induced a significant increase in IDO-1 gene expression, but only TLR-3 agonist induced significantly higher IDO-1 protein expression and enzymatic activity in conditioned media (CM). IDO-1 activity of Poly I/C- and Pam3CSK4-treated hPDLSCs was higher at one day after removal of stimuli than immediately after stimulation and declined to basal levels after five days. Among all tested stimuli, only IFN-γ was able to induce long-lasting IDO-1 expression and activity in hPDLSCs. The high plasticity of IDO-1 expression and its enzymatic activity in hPDLSCs due to the variable cytokine and virulence factor milieu and the temporal-dependent responsiveness of hPDLSCs may cause a highly dynamic potential of hPDLSCs to modulate immune responses in periodontal tissues.


Asunto(s)
Citocinas/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ligamento Periodontal/citología , Células Madre/enzimología , Receptores Toll-Like/agonistas , Células Cultivadas , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Lipopéptidos/farmacología , Poli I-C/farmacología , Células Madre/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Receptores Toll-Like/metabolismo
15.
Front Immunol ; 11: 601759, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33363543

RESUMEN

Indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO) catalyze the rate-limiting step of tryptophan catabolism along the kynurenine pathway, which has important immuno suppressive properties, particularly in tumor cells and dendritic cells. The prominent expression of IDO1 in the placenta also suggested a role in preventing immune rejection of fetal tissues, and pharmacological inhibition of IDO1 induced abortion of allogeneic fetuses in mice. However, this was later challenged by the lack of rejection of allogeneic fetuses in IDO1-KO mice, suggesting that other mechanisms may compensate for IDO1 deficiency. Here we investigated whether TDO could contribute to feto-maternal tolerance and compensate for IDO1 deficiency in IDO1-KO mice. Expression of TDO mRNA was previously detected in placental tissues. We developed a new chimeric rabbit anti-TDO antibody to confirm TDO expression at the protein level and identify the positive cell type by immunohistochemistry in murine placenta. We observed massive TDO expression in decidual stromal cells, starting at day E3.5, peaking at day E6.5 then declining rapidly while remaining detectable until gestation end. IDO1 was also induced in decidual stromal cells, but only at a later stage of gestation when TDO expression declined. To determine whether TDO contributed to feto-maternal tolerance, we mated TDO-KO and double IDO1-TDO-KO females with allogeneic males. However, we did not observe reduced fertility. These results suggest that, despite its expression in decidual stromal cells, TDO is not a dominant mechanism of feto-maternal tolerance able to compensate for the absence of IDO1. Redundant additional mechanisms of immunosuppression likely take over in these KO mice. The massive expression of TDO during decidualization might suggest a role of TDO in angiogenesis or vessel tonicity, as previously described for IDO1.


Asunto(s)
Decidua/enzimología , Tolerancia Inmunológica , Intercambio Materno-Fetal/inmunología , Células del Estroma/enzimología , Triptófano Oxigenasa/metabolismo , Animales , Decidua/citología , Decidua/inmunología , Femenino , Fertilidad/inmunología , Edad Gestacional , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Células del Estroma/inmunología , Triptófano Oxigenasa/genética
16.
Open Vet J ; 10(2): 157-163, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32821660

RESUMEN

Background: In several mammals, subfertility or infertility associated with endometritis was reported. Although there have been studies about endometritis in bitches, the pathophysiological mechanisms are not completely known. Aim: This study aimed to evaluate the immunohistochemical expression of Cyclooxygenase 2 (COX2) in clinically healthy bitches with normal uterine tissue and bitches with endometritis. Methods: Forty-eight mixed breed bitches in diestrus were used. Uterine biopsies were collected for diagnosis [normal endometrium (n = 15; NE), cystic endometrial hyperplasia (n = 1), atrophy (n= 2), acute endometritis (n = 9; AE), subacute endometritis (n = 7; SE), and chronic endometritis (n = 14; CE)]. Immunostaining and quantification of positively stained cells was performed on full-thickness uterine biopsies. Data were analyzed by the GLIMMIX procedure of SAS. Results: COX2 immunostaining was scattered and restricted to cells in the stroma in bitches with NE. However, in bitches with endometritis, strong staining was observed in the luminal epithelium, glandular epithelium, and stromal cells. Staining was also observed in inflammatory cells localized in the stroma as well as inside of the glands. The percentage of COX2 positive stromal cells in bitches with AE, SE, and CE was significantly higher compared with NE (p < 0.005). In addition, the percentage of COX2 positive stromal cells in bitches with SE, and CE was significantly lower compared with AE (p < 0.003). Conclusion: COX2 could be involved in the pathophysiological mechanisms producing endometritis without the presence of cystic endometrial hyperplasia in bitches. However, further researches on this topic are required.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Enfermedades de los Perros/enzimología , Hiperplasia Endometrial/veterinaria , Endometritis/veterinaria , Animales , Diestro , Enfermedades de los Perros/fisiopatología , Perros , Hiperplasia Endometrial/enzimología , Hiperplasia Endometrial/fisiopatología , Endometritis/enzimología , Endometritis/fisiopatología , Femenino , Inmunohistoquímica/veterinaria , Células del Estroma/enzimología , Útero/enzimología , Útero/fisiopatología
17.
Fertil Steril ; 113(5): 1067-1079.e5, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32386617

RESUMEN

OBJECTIVE: To explore the possible mechanism of protein kinase CK2, which participates in estrogen recruitment of endothelial progenitor cells (EPCs), and its role in the angiogenesis of endometriosis lesions. DESIGN: Laboratory study. SETTING: University. ANIMAL(S): BALB/c mice. INTERVENTION(S): Exposure of human endometrial stromal cells (HESCs) to estrogen and CK2 inhibitor CX-4945 and endometrial stromal cells transfected with the protein kinase CK2 vector (HESC-CK2). Endometriosis models were induced by allogeneic mice transplantation of the endometrium into dorsal skinfold chambers. The mice received an IP injection of 50 mg/kg emodin per day or were treated with 100 µg/kg estrogen by SC injection once a week. MAIN OUTCOME MEASURE(S): The concentration of cytokines in cells was measured with ELISA. The migration of EPCs was examined using the scratch assay method and Transwell, a capillary tube-formation assay to determine EPC tube-forming capacity, and protein and mRNA expression with Western blot and polymerase chain reaction analyses, respectively. RESULT(S): Protein kinase CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in a stromal cell-derived factor-1 (SDF-1)-CXCR4-dependent manner. Conditioned medium from endometrial stromal cells that were stably transfected with the protein kinase CK2 vector (HESC-CK2) or pretreated with estrogen significantly enhanced the migration and recruitment of EPCs. In contrast, conditioned medium from HESCs that were treated with CX-4945, a selective inhibitor of CK2, inhibited the mobility and viability of EPCs. Furthermore, CK2 overexpression significantly upregulated SDF-1 expression and secretion in endometrial stromal cells by activating the AKT/mTOR pathway. Moreover, treatment with the SDF-1 receptor CXCR4-specific inhibitor AMD3100 completely reversed the CK2-enhanced migration of EPCs. CONCLUSION(S): This study demonstrates that CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in an SDF-1-CXCR4-dependent manner and may be a therapeutic target.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Quimiocina CXCL12/metabolismo , Endometriosis/enzimología , Endometrio/enzimología , Células Progenitoras Endoteliales/enzimología , Receptores CXCR4/metabolismo , Células del Estroma/enzimología , Animales , Quinasa de la Caseína II/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Endometriosis/genética , Endometriosis/patología , Endometrio/patología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/patología , Estrógenos/farmacología , Femenino , Humanos , Ratones Endogámicos BALB C , Neovascularización Patológica , Comunicación Paracrina/efectos de los fármacos , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/patología
18.
Pregnancy Hypertens ; 20: 108-110, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32278308

RESUMEN

Endothelin-converting enzyme-1(ECE-1) is a key regulatory enzyme in the processing of endothelin-1 (ET-1). We quantified and localized ECE-1 in normal and preeclamptic placentas. Normal (n=6) and preeclamptic (n=6) placentas were serially sectioned for immunofluorescence (IF). Cell type specific markers identified endothelial, trophoblast, macrophage, smooth muscle, and fibroblast cells. Quantitative analyses were performed by western blot and ELISA. IF identified ECE-1 expression within the stroma and villous space. Cellular localization of ECE-1 was limited to endothelial membranes. There was significantly less ECE-1 in preeclamptic placentas, suggesting ECE-1 is important for proper regulation of ET-1 within the placenta.


Asunto(s)
Enzimas Convertidoras de Endotelina/análisis , Placenta/enzimología , Preeclampsia/enzimología , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Vellosidades Coriónicas/enzimología , Regulación hacia Abajo , Células Endoteliales/enzimología , Femenino , Humanos , Preeclampsia/diagnóstico , Embarazo , Células del Estroma/enzimología
19.
J Biol Chem ; 295(8): 2248-2258, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31937587

RESUMEN

The regulation mechanisms involved in matrix metalloproteinase (MMP) expression and the motility of human endometrial and decidual stromal cells (ESCs and DSCs, respectively) during decidualization remain unclear. DSCs show significant increased cell motility and expression of FOS-like 1 (FOSL1) and MMP1, MMP2, and MMP9 compared with ESCs, whereas lack of decidualization inducers leads to a rapid decrease in FOSL1 and MMP1 and MMP9 expression in DSCs in vitro Therefore, we hypothesized that a link exists between decidualization inducers and FOSL1 in up-regulation of motility during decidualization. Based on the response of ESCs/DSCs to different decidualization systems in vitro, we found that progesterone (P4) alone had no significant effect and that 17ß-estradiol (E2) significantly increased cell motility and FOSL1 and MMP1 and MMP9 expression at the mRNA and protein levels, whereas 8-bromo-cAMP significantly decreased cell motility and FOSL1 and MMP9 expression in the presence of P4. In addition, we showed that E2 triggered phosphorylation of estrogen receptor 1 (ESR1), which could directly bind to the promoter of FOSL1 in ESCs/DSCs. Additionally, we also revealed silencing of ESR1 expression by siRNA abrogated E2-induced FOSL1 expression at the transcript and protein levels. Moreover, silencing of FOSL1 expression by siRNA was able to block E2-induced MMP1 and MMP9 expression and cell motility in ESCs/DSCs. Taken together, our data suggest that, in addition to its enhancement of secretory function, the change in MMP expression and cell motility is another component of the decidualization of ESCs/DSCs, including estrogen-dependent MMP1 and MMP9 expression mediated by E2-ESR1-FOSL1 signaling.


Asunto(s)
Movimiento Celular , Decidua/citología , Endometrio/citología , Estrógenos/farmacología , Metaloproteinasas de la Matriz/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Adulto , Movimiento Celular/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Persona de Mediana Edad , Prolactina/metabolismo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Adulto Joven
20.
Int J Cancer ; 147(1): 266-276, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31904863

RESUMEN

We investigated the value of reactive stroma as a predictor for trastuzumab resistance in patients with early HER2-positive breast cancer receiving adjuvant therapy. The pathological reactive stroma and the mRNA gene signatures that reflect reactive stroma in 209 HER2-positive breast cancer samples from the FinHer adjuvant trial were evaluated. Levels of stromal gene signatures were determined as a continuous parameter, and pathological reactive stromal findings were defined as stromal predominant breast cancer (SPBC; ≥50% stromal) and correlated with distant disease-free survival. Gene signatures associated with reactive stroma in HER2-positive early breast cancer (N = 209) were significantly associated with trastuzumab resistance in estrogen receptor (ER)-negative tumors (hazard ratio [HR] = 1.27 p interaction = 0.014 [DCN], HR = 1.58, p interaction = 0.027 [PLAU], HR = 1.71, p interaction = 0.019 [HER2STROMA, novel HER2 stromal signature]), but not in ER-positive tumors (HR = 0.73 p interaction = 0.47 [DCN], HR = 0.71, p interaction = 0.73 [PLAU], HR = 0.84; p interaction = 0.36 [HER2STROMA]). Pathological evaluation of HER2-positive/ER-negative tumors suggested an association between SPBC and trastuzumab resistance. Reactive stroma did not correlate with tumor-infiltrating lymphocytes (TILs), and the expected benefit from trastuzumab in patients with high levels of TILs was pronounced only in tumors with low stromal reactivity (SPBC <50%). In conclusion, reactive stroma in HER2-positive/ER-negative early breast cancer tumors may predict resistance to adjuvant trastuzumab therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Ensayos Clínicos Fase III como Asunto , Resistencia a Antineoplásicos , Femenino , Expresión Génica , Humanos , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ensayos Clínicos Controlados Aleatorios como Asunto , Células del Estroma/enzimología , Células del Estroma/patología , Transcriptoma , Factor de Crecimiento Transformador beta1/metabolismo , Trastuzumab/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...