Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Diagnostics (Basel) ; 14(15)2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39125555

RESUMEN

Dedifferentiated liposarcoma (DDLPS) is a non-lipogenic sarcoma, generally arising from well-differentiated liposarcoma (WDLPS), although it can develop de novo. DDLPS tumors rarely trans-differentiate into non-adipose mesenchymal tissues; however, the latter lack notable variety and mostly show striated muscle or osteogenic/chondrogenic differentiation. Here, we report a case of DDLPS that contained numerous atypical vessels. A man in his sixties presented with a large tumor in his right thigh, and the tumor was surgically resected. Microscopically, most of the tumor was WDLPS, but a minor portion showed DDLPS, consisting of high-grade spindle cells. Remarkably, the DDLPS contained vessels of various sizes with atypical cytoarchitecture, including vessels with seemingly muscular layers. Immunohistochemically, the atypical cells within the vascular wall expressed aSMA, consistent with smooth muscle cells or pericytes, whereas surrounding high-grade spindle cells only focally expressed it, and these aSMA-positive cells within the vessels exhibited MDM2 amplification by immuno-fluorescence in situ hybridization. Our results demonstrate that DDLPS can trans-differentiate into smooth muscle cells of various-sized accompanying vessels, which may support their survival and proliferation.

2.
ACS Appl Mater Interfaces ; 16(29): 37418-37434, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38980153

RESUMEN

The re-epithelialization process gets severely dysregulated in chronic nonhealing diabetic foot ulcers/wounds. Keratinocyte growth factor (KGF or FGF-7) is the major modulator of the re-epithelialization process, which regulates the physiological phenotypes of cutaneous keratinocytes. The existing therapeutic strategies of growth factor administration have several limitations. To overcome these, we have designed a KGF-mimetic peptide (KGFp, 13mer) based on the receptor interaction sites in murine KGF. KGFp enhanced migration and transdifferentiation of mouse bone marrow-derived MSCs toward keratinocyte-like cells (KLCs). A significant increase in the expression of skin-specific markers Bnc1 (28.5-fold), Ck5 (14.6-fold), Ck14 (26.1-fold), Ck10 (187.7-fold), and epithelial markers EpCam (23.3-fold) and Cdh1 (64.2-fold) was associated with the activation of ERK1/2 and STAT3 molecular signaling in the KLCs. Further, to enhance the stability of KGFp in the wound microenvironment, it was conjugated to biocompatible 3D porous polymer scaffolds without compromising its active binding sites followed by chemical characterization using Fourier transform infrared spectroscopy, field-emission scanning electron microscopy, dynamic mechanical analysis, and thermogravimetry. In vitro evaluation of the KGFp-conjugated 3D polymer scaffolds revealed its potential for transdifferentiation of MSCs into KLCs. Transplantation of allogeneic MSCGFP using KGFp-conjugated 3D polymer scaffolds in chronic nonhealing type 2 diabetic wounds (db/db transgenic, 50-52 weeks old male mice) significantly enhanced re-epithelialization-mediated wound closure rate (79.3%) as compared to the control groups (Untransplanted -22.4%, MSCGFP-3D polymer scaffold -38.5%). Thus, KGFp-conjugated 3D porous polymer scaffolds drive the fate of the MSCs toward keratinocytes that may serve as potential stem cell delivery platform technology for tissue engineering and transplantation.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos , Queratinocitos , Andamios del Tejido , Animales , Ratones , Andamios del Tejido/química , Queratinocitos/efectos de los fármacos , Factor 7 de Crecimiento de Fibroblastos/química , Factor 7 de Crecimiento de Fibroblastos/farmacología , Porosidad , Péptidos/química , Péptidos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Piel/efectos de los fármacos , Piel/patología , Polímeros/química , Polímeros/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Masculino , Regeneración/efectos de los fármacos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Pie Diabético/tratamiento farmacológico , Pie Diabético/patología , Pie Diabético/terapia , Humanos
3.
Head Neck Pathol ; 18(1): 40, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727794

RESUMEN

BACKGROUND: Odontogenic lesions constitute a heterogeneous group of lesions. CLIC4 protein regulates different cellular processes, including epithelial-mesenchymal transition and fibroblast-myofibroblast transdifferentiation. This study analyzed CLIC4, E-cadherin, Vimentin, and α-SMA immunoexpression in epithelial odontogenic lesions that exhibit different biological behavior. METHODS: It analyzed the immunoexpression of CLIC4, E-cadherin, and Vimentin in the epithelial cells, as well as CLIC4 and α-SMA in the mesenchymal cells, of ameloblastoma (AM) (n = 16), odontogenic keratocyst (OKC) (n = 20), and adenomatoid odontogenic tumor (AOT) (n = 8). Immunoexpressions were categorized as score 0 (0% positive cells), 1 (< 25%), 2 (≥ 25% - < 50%), 3 (≥ 50% - < 75%), or 4 (≥ 75%). RESULTS: Cytoplasmic CLIC4 immunoexpression was higher in AM and AOT (p < 0.001) epithelial cells. Nuclear-cytoplasmic CLIC4 was higher in OKC's epithelial lining (p < 0.001). Membrane (p = 0.012) and membrane-cytoplasmic (p < 0.001) E-cadherin immunoexpression were higher in OKC, while cytoplasmic E-cadherin expression was higher in AM and AOT (p < 0.001). Vimentin immunoexpression was higher in AM and AOT (p < 0.001). Stromal CLIC4 was higher in AM and OKC (p = 0.008). Similarly, α-SMA immunoexpression was higher in AM and OKC (p = 0.037). Correlations in these proteins' immunoexpression were observed in AM and OKC (p < 0.05). CONCLUSIONS: CLIC4 seems to regulate the epithelial-mesenchymal transition, modifying E-cadherin and Vimentin expression. In mesenchymal cells, CLIC4 may play a role in fibroblast-myofibroblast transdifferentiation. CLIC4 may be associated with epithelial odontogenic lesions with aggressive biological behavior.


Asunto(s)
Ameloblastoma , Cadherinas , Canales de Cloruro , Transición Epitelial-Mesenquimal , Tumores Odontogénicos , Vimentina , Humanos , Transición Epitelial-Mesenquimal/fisiología , Canales de Cloruro/metabolismo , Canales de Cloruro/análisis , Cadherinas/metabolismo , Tumores Odontogénicos/patología , Tumores Odontogénicos/metabolismo , Ameloblastoma/patología , Ameloblastoma/metabolismo , Vimentina/metabolismo , Adulto , Femenino , Quistes Odontogénicos/patología , Quistes Odontogénicos/metabolismo , Masculino , Actinas/metabolismo , Adulto Joven , Persona de Mediana Edad , Antígenos CD/metabolismo , Adolescente
4.
J Appl Toxicol ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38644760

RESUMEN

Prolonged exposure to environments with high concentrations of crystalline silica (CS) can lead to silicosis. Macrophages play a crucial role in the pathogenesis of silicosis. In the process of silicosis, silica (SiO2) invades alveolar macrophages (AMs) and induces mitophagy which usually exists in three states: normal, excessive, and/or deficiency. Different mitophagy states lead to corresponding toxic responses, including successful macrophage repair, injury, necrosis, apoptosis, and even pulmonary fibrosis. This is a complex process accompanied by various cytokines. Unfortunately, the details have not been fully systematically summarized. Therefore, it is necessary to elucidate the role of macrophage mitophagy in SiO2-induced pulmonary fibrosis by systematic analysis on the literature reports. In this review, we first summarized the current data on the macrophage mitophagy in the development of SiO2-induced pulmonary fibrosis. Then, we introduce the molecular mechanism on how SiO2-induced mitophagy causes pulmonary fibrosis. Finally, we focus on introducing new therapies based on newly developed mitophagy-inducing strategies. We conclude that macrophage mitophagy plays a multifaceted role in the progression of SiO2-induced pulmonary fibrosis, and reprogramming the macrophage mitophagy state accordingly may be a potential means of preventing and treating pulmonary fibrosis.

5.
Life Sci ; 346: 122645, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614297

RESUMEN

The increasing global prevalence and associated comorbidities need innovative approaches for type 2 diabetes mellitus (T2DM) prevention and treatment. Genetics contributes significantly to T2DM susceptibility, and genetic counseling is significant in detecting and informing people about the diabetic risk. T2DM is also intricately linked to overnutrition and obesity, and nutritional advising is beneficial to mitigate diabetic evolution. However, manipulating pancreatic cell plasticity and transdifferentiation could help beta cell regeneration and glucose homeostasis, effectively contributing to the antidiabetic fight. Targeted modulation of transcription factors is highlighted for their roles in various aspects of pancreatic cell differentiation and function, inducing non-beta cells' conversion into functional beta cells (responsive to glucose). In addition, pharmacological interventions targeting specific receptors and pathways might facilitate cell transdifferentiation aiming to maintain or increase beta cell mass and function. However, the mechanisms underlying cellular reprogramming are not yet well understood. The present review highlights the primary transcriptional factors in the endocrine pancreas, focusing on transdifferentiation as a primary mechanism. Therefore, islet cell reprogramming, converting one cell type to another and transforming non-beta cells into insulin-producing cells, depends, among others, on transcription factors. It is a promising fact that new transcription factors are discovered every day, and their actions on pancreatic islet cells are revealed. Exploring these pathways associated with pancreatic development and islet endocrine cell differentiation could unravel the molecular intricacies underlying transdifferentiation processes, exploring novel therapeutic strategies to treat diabetes. The medical use of this biotechnology is expected to be achievable within a short time.


Asunto(s)
Transdiferenciación Celular , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Diabetes Mellitus Tipo 2/terapia , Diabetes Mellitus Tipo 2/metabolismo , Animales , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular , Páncreas/metabolismo , Páncreas/patología
6.
Cells ; 13(6)2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38534325

RESUMEN

Calcific Aortic Valve Disease (CAVD) is a significant concern for cardiovascular health and is closely associated with chronic kidney disease (CKD). Aortic valve endothelial cells (VECs) play a significant role in the onset and progression of CAVD. Previous research has suggested that uremic toxins, particularly indoxyl sulfate (IS), induce vascular calcification and endothelial dysfunction, but the effect of IS on valve endothelial cells (VECs) and its contribution to CAVD is unclear. Our results show that IS reduced human VEC viability and increased pro-calcific markers RUNX2 and alkaline phosphatase (ALP) expression. Additionally, IS-exposed VECs cultured in pro-osteogenic media showed increased calcification. Mechanistically, IS induced endothelial-to-mesenchymal transition (EndMT), evidenced by the loss of endothelial markers and increased expression of mesenchymal markers. IS triggered VEC inflammation, as revealed by NF-kB activation, and decreased integrin-linked kinase (ILK) expression. ILK overexpression reversed the loss of endothelial phenotype and RUNX2, emphasizing its relevance in the pathogenesis of CAVD in CKD. Conversely, a lower dose of IS intensified some of the effects in EndMT caused by silencing ILK. These findings imply that IS affects valve endothelium directly, contributing to CAVD by inducing EndMT and calcification, with ILK acting as a crucial modulator.


Asunto(s)
Estenosis de la Válvula Aórtica , Válvula Aórtica/patología , Calcinosis , Proteínas Serina-Treonina Quinasas , Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , Indicán , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Endoteliales/metabolismo , Células Cultivadas , Calcificación Vascular/metabolismo , Insuficiencia Renal Crónica/patología
7.
Theranostics ; 14(4): 1701-1719, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38389831

RESUMEN

Human somatic cells can be reprogrammed into neuron cell fate through regulation of a single transcription factor or application of small molecule cocktails. Methods: Here, we report that forskolin efficiently induces the conversion of human somatic cells into induced neurons (FiNs). Results: A large population of neuron-like phenotype cells was observed as early as 24-36 h post-induction. There were >90% TUJ1-, >80% MAP2-, and >80% NEUN-positive neurons at 5 days post-induction. Multiple subtypes of neurons were present among TUJ1-positive cells, including >60% cholinergic, >20% glutamatergic, >10% GABAergic, and >5% dopaminergic neurons. FiNs exhibited typical neural electrophysiological activity in vitro and the ability to survive in vitro and in vivo more than 2 months. Mechanistically, forskolin functions in FiN reprogramming by regulating the cAMP-CREB1-JNK signals, which upregulates cAMP-CREB1 expression and downregulates JNK expression. Conclusion: Overall, our studies identify a safer and efficient single-small-molecule-driven reprogramming approach for induced neuron generation and reveal a novel regulatory mechanism of neuronal cell fate acquisition.


Asunto(s)
Reprogramación Celular , Factores de Transcripción , Humanos , Colforsina/farmacología , Diferenciación Celular/fisiología , Factores de Transcripción/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico
8.
Artículo en Inglés | MEDLINE | ID: mdl-38325871

RESUMEN

Background: Tubulointerstitial renal fibrosis is an essential feature of diabetic nephropathy (DN). Pericytes play a critical role in microvascular diseases and renal fibrogenesis. However, the role of pericytes in DN remains unclear. Herein, we aimed to explore the properties and possible mechanisms of pericytes in renal fibrosis in DN. Methods: We used multiplex immunofluorescence staining to evaluate the location and expression of activated pericytes and to assess capillary dilation and interstitial fibrosis in the kidneys of db/db mice. Pericytes were co-stained for alpha-smooth muscle actin (α-SMA) to determine which ones differentiate into myofibroblasts in db/db mice. Expression of CD34 and platelet-derived growth factor receptor beta (PDGFR-ß) was assessed in kidney tissue from patients with DN by immunohistochemical staining. Results: We found that cell staining for nerve/glial antigen 2 (NG2)+ and PDGFR-ß+ was greater in the kidneys of db/db mice than in those of db/m mice. There was impaired pericyte coverage of blood vessels and capillary dilation in the renal interstitium. These changes were accompanied by increased collagen I staining and an increase in the number of pericytes with profibrotic phenotypes, as identified by increased NG2+/PDGFR-ß+/α-SMA+ and decreased NG2+/PDGFR-ß+/α-SMA- staining. In DN patients, expression of PDGFR-ß was stronger and there was loss of CD34 compared with the findings in control patients with minor glomerular lesions. Conclusion: In this study, we demonstrated that pericyte activation accompanied by peritubular capillary dysfunction and pericytemyofibroblast transition is associated with renal fibrosis in DN.

9.
Theriogenology ; 215: 259-271, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38103403

RESUMEN

Mesenchymal stem cells (MSC) display self-renewal and mesodermal differentiation potentials. These characteristics make them potentially useful for in vitro derivation of gametes, which may constitute experimental therapies for human and animal reproduction. Organoids provide a spatial support and may simulate a cellular niche for in vitro studies. In this study, we aimed at evaluating the potential integration of fetal bovine MSCs derived from adipose tissue (AT-MSCs) in testicular organoids (TOs), their spatial distribution with testicular cells during TO formation and their potential for germ cell differentiation. TOs were developed using Leydig, Sertoli, and peritubular myoid cells that were previously isolated from bovine testes (n = 6). Thereafter, TOs were characterized using immunofluorescence and Q-PCR to detect testicular cell-specific markers. AT-MSCs were labeled with PKH26 and then cultured with testicular cells at a concentration of 1 × 106 cells per well in Ultra Low Attachment U-shape bottom (ULA) plates. TOs formed by testicular cells and AT-MSCs (TOs + AT-MSCs) maintained a rounded structure throughout the 28-day culture period and did not show significant differences in their diameters. Conversely, control TOs exhibited a compact structure until day 7 of culture, while on day 28 they displayed cellular extensions around their structure. Control TOs had greater (P < 0.05) diameters compared to TOs + AT-MSCs. AT-MSCs induced an increase in proportion of Leydig and peritubular myoid cells in TOs + AT-MSCs; however, did not induce changes in the overall gene expression of testicular cell-specific markers. STAR immunolabelling detected Leydig cells that migrated from the central area to the periphery and formed brunches in control TOs. However, in TOs + AT-MSCs, Leydig cells formed a compact peripheral layer. Sertoli cells immunodetected using WT1 marker were observed within the central area forming clusters of cells in TOs + AT-MSCs. The expression of COL1A associated to peritubular myoids cells was restricted to the central region in TOs + AT-MSCs. Thus, during a 28-day culture period, fetal bovine AT-MSCs integrated and modified the structure of the TOs, by restricting formation of branches, limiting the overall increase in diameters and increasing the proportions of Leydig and peritubular myoid cells. AT-MSCs also induced a reorganization of testicular cells, changing their distribution and particularly the location of Leydig cells.


Asunto(s)
Células Madre Mesenquimatosas , Testículo , Masculino , Animales , Bovinos , Humanos , Testículo/metabolismo , Células de Sertoli/metabolismo , Células Intersticiales del Testículo/metabolismo , Organoides
10.
Kidney Int ; 105(3): 562-581, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38142040

RESUMEN

The parathyroid gland is one of the main organs that regulate calcium and phosphorus metabolism. It is mainly composed of chief cells and oxyphil cells. Oxyphil cell counts are low in the parathyroid glands of healthy adults but are dramatically increased in patients with uremia and secondary hyperparathyroidism (SHPT). Increased oxyphil cell counts are related to drug treatment resistance, but the origin of oxyphil cells and the mechanism of proliferation remain unknown. Herein, three types of parathyroid nodules (chief cell nodules, oxyphil cell nodules and mixed nodules, respectively) excised from parathyroid glands of uremic SHPT patients were used for single-cell RNA sequencing (scRNA-seq), other molecular biology studies, and transplantation into nude mice. Through scRNA-seq of parathyroid mixed nodules from three patients with uremic SHPT, we established the first transcriptomic map of the human parathyroid and found a chief-to-oxyphil cell transdifferentiation characterized by gradual mitochondrial enrichment associated with the uremic milieu. Notably, the mitochondrial enrichment and cellular proliferation of chief cell and oxyphil cell nodules decreased significantly after leaving the uremic milieu via transplantation into nude mice. Remarkably, the phenotype of oxyphil cell nodules improved significantly in the nude mice as characterized by decreased mitochondrial content and the proportion of oxyphil cells to chief cells. Thus, our study provides a comprehensive single-cell transcriptome atlas of the human parathyroid and elucidates the origin of parathyroid oxyphil cells and their underlying transdifferentiating mechanism. These findings enhance our understanding of parathyroid disease and may open new treatment perspectives for patients with chronic kidney disease.


Asunto(s)
Hiperparatiroidismo Secundario , Glándulas Paratiroides , Adulto , Animales , Ratones , Humanos , Glándulas Paratiroides/metabolismo , Células Oxífilas , Ratones Desnudos , Transdiferenciación Celular , Hiperparatiroidismo Secundario/genética , Hiperparatiroidismo Secundario/terapia , Análisis de Secuencia de ARN
11.
Cell Reprogram ; 25(5): 183-186, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37847897

RESUMEN

Reprogramming is traditionally defined as the fate conversion of a cell to a stage of increased developmental potential. In its broader meaning, the reprogramming term is also applied to all forms of cell fate conversion that do not follow a developmental trajectory. Reprogramming is now a well-established field of research that gained rapid progress upon the advent of induced pluripotency. In this perspective, I reflect on the reprogramming lessons of the past, in the contributions to other fields of research and on the potential transformative future use of reprogrammed cells and of its cell derivatives.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Diferenciación Celular
12.
Islets ; 15(1): 2219477, 2023 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258189

RESUMEN

Previous studies indicated that ductal cells can contribute to endocrine neogenesis in adult rodents after alpha cells convert into beta cells. This can occur through Pax4 mis-expression in alpha cells or through long-term administration of gamma-aminobutyric acid (GABA) to healthy mice. GABA has also been reported to increase the number of beta cells through direct effects on their proliferation, but only in specific genetic mouse backgrounds. To test whether GABA induces neogenesis of beta cells from ductal cells or affects pancreatic cell proliferation, we administered GABA or saline over 2 or 6 months to Sox9CreER;R26RYFP mice in which 60-80% of large or small ducts were efficiently lineage labeled. We did not observe any increases in islet neogenesis from ductal cells between 1 and 2 months of age in saline treated mice, nor between 2 and 6 months of saline treatment, supporting previous studies indicating that adult ductal cells do not give rise to new endocrine cells during homeostasis. Unlike previous reports, we did not observe an increase in beta cell neogenesis after 2 or 6 months of GABA administration. Nor did we observe a significant increase in the pancreatic islet area, the number of insulin and glucagon double positive cells, or cell proliferation in the pancreas. This indicates that the effect of long term GABA administration on the pancreas is minimal or highly context dependent.


Asunto(s)
Células Endocrinas , Células Secretoras de Glucagón , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Conductos Pancreáticos , Ácido gamma-Aminobutírico/farmacología
13.
Development ; 150(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36897579

RESUMEN

Pancreatic ε-cells producing ghrelin are one type of endocrine cell found in islets, which have been shown to influence other intra-islet cells, especially in regulating the function of ß cells. However, the role of such cells during ß-cell regeneration is currently unknown. Here, using a zebrafish nitroreductase (NTR)-mediated ß-cell ablation model, we reveal that ghrelin-positive ε-cells in the pancreas act as contributors to neogenic ß-cells after extreme ß-cell loss. Further studies show that the overexpression of ghrelin or the expansion of ε-cells potentiates ß-cell regeneration. Lineage tracing confirms that a proportion of embryonic ε-cells can transdifferentiate to ß-cells, and that the deletion of Pax4 enhances this transdifferentiation of ε-cells to ß-cells. Mechanistically, Pax4 binds to the ghrelin regulatory region and represses its transcription. Thus, deletion of Pax4 derepresses ghrelin expression and causes producing more ghrelin-positive cells, enhancing the transdifferentiation of ε-cells to ß-cells and consequently potentiating ß-cell regeneration. Our findings reveal a previously unreported role for ε-cells during zebrafish ß-cell regeneration, indicating that Pax4 regulates ghrelin transcription and mediates the conversion of embryonic ε-cells to ß-cells after extreme ß-cell loss.


Asunto(s)
Factores de Transcripción , Pez Cebra , Animales , Ghrelina/metabolismo , Proteínas de Homeodominio/metabolismo , Páncreas , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
14.
Biofactors ; 49(3): 646-662, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36714992

RESUMEN

The semi-essential ubiquitous amino acid taurine has been shown to alleviate obesity and hyperglycemia in humans; however, the pathways underlying the antidiabetic actions have not been characterized. We explored the effect of chronic taurine exposure on cell biology of pancreatic islets, in degenerative type 1-like diabetes. The latter was modeled by small dose of streptozotocin (STZ) injection for 5 days in mice, followed by a 10-day administration of taurine (2% w/v, orally) in the drinking water. Taurine treatment opposed the detrimental changes in islet morphology and ß-/α-cell ratio, induced by STZ diabetes, coincidentally with a significant 3.9 ± 0.7-fold enhancement of proliferation and 40 ± 5% reduction of apoptosis in ß-cells. In line with these findings, the treatment counteracted an upregulation of antioxidant (Sod1, Sod2, Cat, Gpx1) and downregulation of islet expansion (Ngn3, Itgb1) genes induced by STZ, in a pancreatic ß-cell line. At the same time, taurine enhanced the transdifferentiation of α-cells into ß-cells by 2.3 ± 0.8-fold, echoed in strong non-metabolic elevation of cytosolic Ca2+ levels in pancreatic α-cells. Our data suggest a bimodal effect of dietary taurine on islet ß-cell biology, which combines the augmentation of α-/ß-cell transdifferentiation with downregulation of apoptosis. The dualism of action, stemming presumably from the intra- and extracellular modality of the signal, is likely to explain the antidiabetic potential of taurine supplementation.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Ratones , Animales , Taurina/farmacología , Transdiferenciación Celular , Glucemia/metabolismo , Islotes Pancreáticos/metabolismo , Hipoglucemiantes/farmacología , Estreptozocina , Insulina/metabolismo
15.
Front Physiol ; 13: 1032774, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36467676

RESUMEN

Islet ß-cell biomarkers can reflect changes in the number and function of islet ß-cells in the prediabetes or early diabetes stage. CD34 is a commonly used stem cell biomarker; however, its expression and function in pancreatic islets remain unclear. In the present study, double immunofluorescence staining, proteomic bioinformatics analysis, and correlation analysis were used to explore the potential of CD34 as an islet ß-cell biomarker. Bioinformatics analysis revealed that the amino acid sequence of CD34 was conserved among multiple species and abundantly expressed on mouse and human pancreatic tissues. Immunofluorescence demonstrated that in the control rat pancreas, CD34 was expressed on glucagon-labeled islet α-cells but not on insulin-labeled islet ß-cells. Furthermore, the proportion of CD34-positive cells, which were also positive for glucagon, was significantly increased in alloxan-induced diabetes models. Statistical analysis revealed that the expression of CD34 was negatively correlated with the number of insulin-labeled islet ß-cells during diabetes progression in dose-dependent fashion in alloxan-induced diabetes models. Furthermore, the results suggested that the transdifferentiation of islet ß-cells into islet α-cells may occur in the process of diabetes. Thus, the present study demonstrated that CD34 is expressed on islet α-cells, and its number is linearly and negatively correlated with the number of islet ß-cells, suggesting that CD34 can be used as a prospective biomarker for islet ß-cells in the early diagnosis of diabetes. The study also suggests the transformation of ß-cells to α-cells in diabetes which provide a potential to be applied towards diabetes mechanism research.

16.
Stem Cell Res Ther ; 13(1): 306, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35841112

RESUMEN

BACKGROUND: Current treatments for salivary gland (SG) hypofunction are palliative and do not address the underlying cause or progression of the disease. SG-derived stem cells have the potential to treat SG hypofunction, but their isolation is challenging, especially when the tissue has been damaged by disease or irradiation for head and neck cancer. In the current study, we test the hypothesis that multipotent bone marrow-derived mesenchymal stem cells (BM-MSCs) in a rat model are capable of trans-differentiating to the SG epithelial cell lineage when induced by a native SG-specific extracellular matrix (SG-ECM) and thus may be a viable substitute for repairing damaged SGs. METHODS: Rat BM-MSCs were treated with homogenates of decellularized rat SG-ECM for one hour in cell suspension and then cultured in tissue culture plates for 7 days in growth media. By day 7, the cultures contained cell aggregates and a cell monolayer. The cell aggregates were hand-selected under a dissecting microscope, transferred to a new tissue culture dish, and cultured for an additional 7 days in epithelial cell differentiation media. Cell aggregates and cells isolated from the monolayer were evaluated for expression of SG progenitor and epithelial cell specific markers, cell morphology and ultrastructure, and ability to form SG-like organoids in vivo. RESULTS: The results showed that this approach was very effective and guided the trans-differentiation of a subpopulation of CD133-positive BM-MSCs to the SG epithelial cell lineage. These cells expressed amylase, tight junction proteins (Cldn 3 and 10), and markers for SG acinar (Aqp5 and Mist 1) and ductal (Krt 14) cells at both the transcript and protein levels, produced intracellular secretory granules which were morphologically identical to those found in submandibular gland, and formed SG-like organoids when implanted in the renal capsule in vivo. CONCLUSIONS: The results of this study suggest the feasibility of using autologous BM-MSCs as an abundant source of stem cells for treating SG hypofunction and restoring the production of saliva in these patients.


Asunto(s)
Células Madre Mesenquimatosas , Organoides , Animales , Diferenciación Celular , Transdiferenciación Celular , Matriz Extracelular/metabolismo , Ratas , Glándulas Salivales
17.
Cell Rep ; 39(9): 110872, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649369

RESUMEN

Type 1 diabetes mellitus (T1D) is a chronic disease with potentially severe complications, and ß-cell deficiency underlies this disease. Despite active research, no therapy to date has been able to induce ß-cell regeneration in humans. Here, we discover the ß-cell regenerative effects of glucagon receptor antibody (anti-GcgR). Treatment with anti-GcgR in mouse models of ß-cell deficiency leads to reversal of hyperglycemia, increase in plasma insulin levels, and restoration of ß-cell mass. We demonstrate that both ß-cell proliferation and α- to ß-cell transdifferentiation contribute to anti-GcgR-induced ß-cell regeneration. Interestingly, anti-GcgR-induced α-cell hyperplasia can be uncoupled from ß-cell regeneration after antibody clearance from the body. Importantly, we are able to show that anti-GcgR-induced ß-cell regeneration is also observed in non-human primates. Furthermore, anti-GcgR and anti-CD3 combination therapy reverses diabetes and increases ß-cell mass in a mouse model of autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Glucagón , Hiperglucemia , Células Secretoras de Insulina , Animales , Modelos Animales de Enfermedad , Glucagón , Hiperglucemia/tratamiento farmacológico , Ratones , Receptores de Glucagón
18.
Int J Mol Sci ; 23(11)2022 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-35682655

RESUMEN

The vertebrate musculoskeletal system is known to be formed by mesenchymal stem cells condensing into tissue elements, which then differentiate into cartilage, bone, tendon/ligament, and muscle cells. These lineage-committed cells mature into end-stage differentiated cells, like hypertrophic chondrocytes and osteocytes, which are expected to expire and to be replaced by newly differentiated cells arising from the same lineage pathway. However, there is emerging evidence of the role of cell transdifferentiation in bone development and disease. Although the concept of cell transdifferentiation is not new, a breakthrough in cell lineage tracing allowed scientists to trace cell fates in vivo. Using this powerful tool, new theories have been established: (1) hypertrophic chondrocytes can transdifferentiate into bone cells during endochondral bone formation, fracture repair, and some bone diseases, and (2) tendon cells, beyond their conventional role in joint movement, directly participate in normal bone and cartilage formation, and ectopic ossification. The goal of this review is to obtain a better understanding of the key roles of cell transdifferentiation in skeletal development and diseases. We will first review the transdifferentiation of chondrocytes to bone cells during endochondral bone formation. Specifically, we will include the history of the debate on the fate of chondrocytes during bone formation, the key findings obtained in recent years on the critical factors and molecules that regulate this cell fate change, and the role of chondrocyte transdifferentiation in skeletal trauma and diseases. In addition, we will also summarize the latest discoveries on the novel roles of tendon cells and adipocytes on skeletal formation and diseases.


Asunto(s)
Transdiferenciación Celular , Osteogénesis , Cartílago/metabolismo , Diferenciación Celular/fisiología , Condrocitos/metabolismo , Condrogénesis/fisiología , Osteogénesis/fisiología
20.
Int Wound J ; 19(7): 1860-1869, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35315582

RESUMEN

Keloid is commonly regarded as a benign skin tumour. Some keloids clinically exhibit hard tissue texture similar to that of cartilage or bone. We hypothesized that the keloid pathological niche environment is likely to induce keloid MSCs towards chondrogenic or osteogenic differentiation and leads to cartilage or bone-like tissue formation. The differences in tissue ossification, histology, mechanical properties, abnormal extracellular matrices and chondrogenic/osteogenic gene expression among sclerous keloids (SKs), regular keloids (RKs) and normal skins (NKs) were carefully examined. The sporadic ossified islets existed in SK group whereas no ossified/chondrified islet was found in other groups by micro-CT reconstruction. H&E, Masson trichrome and safranin O staining revealed lacuna-like structures in SKs, which were featured as bone/cartilage histology. Immunohistochemical staining showed overproduction of osteoprotegerin, type I and III collagen in SK group but similar production level of aggrecan among three groups. The biomechanical analysis demonstrated the weakest compliance of SK tissues. In addition, SK fibroblasts exhibited a relatively slower proliferation rate but higher expression levels of osteogenic and chondrogenic genes among all three groups. These cell populations also showed the strongest potential for lineage transformation. In conclusion, we first reported the presence of ossified and chondrified matrices in some extremely hard keloids in the present study.


Asunto(s)
Queloide , Humanos , Queloide/cirugía , Osteogénesis , Condrogénesis , Diferenciación Celular , Colágeno/metabolismo , Fibroblastos/metabolismo , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA