Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.946
Filtrar
1.
Sci Rep ; 14(1): 8321, 2024 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594471

RESUMO

Endometrial fibrosis is the histologic appearance of intrauterine adhesion (IUA). Emerging evidences demonstrated umbilical cord mesenchymal stem cell-derived exosomes (UCMSC-exo) could alleviate endometrial fibrosis. But the specific mechanism is not clear. In this study, we explored the effect of UCMSC-exo on endometrial fibrosis, and investigated the possible role of miR-140-3p/FOXP1/Smad axis in anti-fibrotic properties of UCMSC-exo. UCMSC-exo were isolated and identified. Transforming growth factor-ß (TGF-ß) was used to induce human endometrial stromal cell (HESC) fibrosis. Dual luciferase assay was performed to verify the relationship between miR-140-3p and FOXP1. The expressions of fibrotic markers, SIP1, and p-Smad2/p-Smad3 in HESCs stimulated with UCMSC-exo were detected by western blot. In addition, the effects of miR-140-3p mimic, miR-140-3p inhibitor and FOXP1 over-expression on endometrial fibrosis were assessed. The isolated UCMSC-exo had a typical cup-shaped morphology and could be internalized into HESCs. The expressions of fibrotic markers were significantly increased by TGF-ß, which was reversed by UCMSC-exo. MiR-140-3p in UCMSC-exo ameliorated TGf-ß-induced HESCs fibrosis. FOXP1 was identified as the direct target of miR-140-3p, which could inversely regulate miR-140-3p's function on HESCs fibrosis. Furthermore, we demonstrated that miR-140-3p in UCMSC-exo regulated Smad signal pathway to exert the anti-fibrotic effect in HESCs. The anti-fibrotic effect of UCMSC-derived exosomes against HESC fibrosis was at least partially achieved by miR-140-3p/FOXP1/Smad axis.


Assuntos
Exossomos , Células-Tronco Mesenquimais , MicroRNAs , Doenças Uterinas , Humanos , Feminino , Exossomos/genética , Células Estromais , Fator de Crescimento Transformador beta , Cordão Umbilical , MicroRNAs/genética , Fibrose , Proteínas Repressoras , Fatores de Transcrição Forkhead/genética
2.
Sci Rep ; 14(1): 7726, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565619

RESUMO

Decidualization can be induced by culturing human endometrial stromal cells (ESCs) with several decidualization stimuli, such as cAMP, medroxyprogesterone acetate (MPA) or Estradiol (E2). However, it has been unclear how decidualized cells induced by different stimuli are different. We compared transcriptomes and cellular functions of decidualized ESCs induced by different stimuli (MPA, E2 + MPA, cAMP, and cAMP + MPA). We also investigated which decidualization stimulus induces a closer in vivo decidualization. Differentially expressed genes (DEGs) and altered cellular functions by each decidualization stimuli were identified by RNA-sequence and gene-ontology analysis. DEGs was about two times higher for stimuli that use cAMP (cAMP and cAMP + MPA) than for stimuli that did not use cAMP (MPA and E2 + MPA). cAMP-using stimuli altered the cellular functions including angiogenesis, inflammation, immune system, and embryo implantation whereas MPA-using stimuli (MPA, E2 + MPA, and cAMP + MPA) altered the cellular functions associated with insulin signaling. A public single-cell RNA-sequence data of the human endometrium was utilized to analyze in vivo decidualization. The altered cellular functions by in vivo decidualization were close to those observed by cAMP + MPA-induced decidualization. In conclusion, decidualized cells induced by different stimuli have different transcriptome and cellular functions. cAMP + MPA may induce a decidualization most closely to in vivo decidualization.


Assuntos
Endométrio , Acetato de Medroxiprogesterona , Feminino , Humanos , Células Cultivadas , Endométrio/metabolismo , Acetato de Medroxiprogesterona/farmacologia , Células Estromais/metabolismo , Expressão Gênica , RNA/metabolismo , Decídua/metabolismo
3.
Sci Rep ; 14(1): 8404, 2024 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-38600158

RESUMO

The survival of leukemic cells is significantly influenced by the bone marrow microenvironment, where stromal cells play a crucial role. While there has been substantial progress in understanding the mechanisms and pathways involved in this crosstalk, limited data exist regarding the impact of leukemic cells on bone marrow stromal cells and their potential role in drug resistance. In this study, we identify that leukemic cells prime bone marrow stromal cells towards osteoblast lineage and promote drug resistance. This biased differentiation of stroma is accompanied by dysregulation of the canonical Wnt signaling pathway. Inhibition of Wnt signaling in stroma reversed the drug resistance in leukemic cells, which was further validated in leukemic mice models. This study evaluates the critical role of leukemic cells in establishing a drug-resistant niche by influencing the bone marrow stromal cells. Additionally, it highlights the potential of targeting Wnt signaling in the stroma by repurposing an anthelmintic drug to overcome the microenvironment-mediated drug resistance.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Mesenquimais , Animais , Camundongos , Via de Sinalização Wnt , Leucemia Mieloide Aguda/metabolismo , Medula Óssea/metabolismo , Células Estromais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Resistência a Medicamentos , Células da Medula Óssea , Microambiente Tumoral/fisiologia
5.
FASEB J ; 38(7): e23604, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38591106

RESUMO

With no lysine/K kinases (WNKs) promote vasocontraction and vascular smooth muscle cell proliferation. In the prostate, smooth muscle contraction and growth may be critical for the development and medical treatment of voiding symptoms in benign prostatic hyperplasia. Here, we examined the effects of isoform-specific WNK silencing and of the WNK inhibitor WNK463 on growth-related functions and contraction in prostate stromal cells, and in human prostate tissues. Impacts of WNK silencing by transfection of cultured stromal cells with isoform-specific siRNAs were qualitatively and quantitatively similar for each WNK isoform. Effects of silencing were largest on cell death (3-5 fold increase in annexin V-positive/7-AAD-positive cells), on proliferation rate, Ki-67 mRNA expression and actin organization (reduced around two-thirds). Contraction in matrix contraction assays and viability were reduced to a lower degree (approximately half), but again to a similar extent for each WNK isoform. Effects of silencing were quantitatively and qualitatively reproduced by 10 µM WNK463, while 1 µM still induced cell death and breakdown in actin organization, without affecting proliferation or viability. Using 500 nM and 10 µM, WNK463 partly inhibited neurogenic and U46619-induced contractions of human prostate tissues (around half), while inhibition of α1-adrenergic contractions (around half) was limited to 10 µM. All four WNK isoforms suppress cell death and promote proliferation in prostate stromal cells. WNK-driven contraction of stromal cells appears possible, even though to a limited extent. Outcomes of isoform-specific WNK silencing can be fully reproduced by WNK463, including inhibition of smooth muscle contraction in human prostate tissues, but require high concentrations.


Assuntos
Actinas , Próstata , Masculino , Humanos , Actinas/metabolismo , Contração Muscular/fisiologia , Células Estromais/metabolismo , Proliferação de Células , Isoformas de Proteínas/metabolismo
6.
Front Immunol ; 15: 1363517, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38562923

RESUMO

Background: Treatment of heart failure post myocardial infarction (post-MI HF) with mesenchymal stem/stromal cells (MSCs) holds great promise. Nevertheless, 2-dimensional (2D) GMP-grade MSCs from different labs and donor sources have different therapeutic efficacy and still in a low yield. Therefore, it is crucial to increase the production and find novel ways to assess the therapeutic efficacy of MSCs. Materials and methods: hUC-MSCs were cultured in 3-dimensional (3D) expansion system for obtaining enough cells for clinical use, named as 3D MSCs. A post-MI HF mouse model was employed to conduct in vivo and in vitro experiments. Single-cell and bulk RNA-seq analyses were performed on 3D MSCs. A total of 125 combination algorithms were leveraged to screen for core ligand genes. Shinyapp and shinycell workflows were used for deploying web-server. Result: 3D GMP-grade MSCs can significantly and stably reduce the extent of post-MI HF. To understand the stable potential cardioprotective mechanism, scRNA-seq revealed the heterogeneity and division-of-labor mode of 3D MSCs at the cellular level. Specifically, scissor phenotypic analysis identified a reported wound-healing CD142+ MSCs subpopulation that is also associated with cardiac protection ability and CD142- MSCs that is in proliferative state, contributing to the cardioprotective function and self-renewal, respectively. Differential expression analysis was conducted on CD142+ MSCs and CD142- MSCs and the differentially expressed ligand-related model was achieved by employing 125 combination algorithms. The present study developed a machine learning predictive model based on 13 ligands. Further analysis using CellChat demonstrated that CD142+ MSCs have a stronger secretion capacity compared to CD142- MSCs and Flow cytometry sorting of the CD142+ MSCs and qRT-PCR validation confirmed the significant upregulation of these 13 ligand factors in CD142+ MSCs. Conclusion: Clinical GMP-grade 3D MSCs could serve as a stable cardioprotective cell product. Using scissor analysis on scRNA-seq data, we have clarified the potential functional and proliferative subpopulation, which cooperatively contributed to self-renewal and functional maintenance for 3D MSCs, named as "division of labor" mode of MSCs. Moreover, a ligand model was robustly developed for predicting the secretory efficacy of MSCs. A user-friendly web-server and a predictive model were constructed and available (https://wangxc.shinyapps.io/3D_MSCs/).


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Camundongos , Animais , Ligantes , Infarto do Miocárdio/genética , Coração , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/terapia , Células Estromais
7.
Cells ; 13(7)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38607021

RESUMO

Human adipose-derived stromal cells (ADSCs) are an important resource for cell-based therapies. However, the dynamics of ADSCs after transplantation and their mechanisms of action in recipients remain unclear. Herein, we generated genetically engineered mouse ADSCs to clarify their biodistribution and post-transplantation status and to analyze their role in recipient mesenchymal tissue modeling. Immortalized ADSCs (iADSCs) retained ADSC characteristics such as stromal marker gene expression and differentiation potential. iADSCs expressing a fluorescent reporter gene were seeded into biocompatible nonwoven fabric sheets and transplanted into the dorsal subcutaneous region of neonatal mice. Transplanted donor ADSCs were distributed as CD90-positive stromal cells on the sheets and survived 1 month after transplantation. Although accumulation of T lymphocytes or macrophages inside the sheet was not observed with or without donor cells, earlier migration and accumulation of recipient blood vascular endothelial cells (ECs) inside the sheet was observed in the presence of donor cells. Thus, our mouse model can help in studying the interplay between donor ADSCs and recipient cells over a 1-month period. This system may be of value for assessing and screening bioengineered ADSCs in vivo for optimal cell-based therapies.


Assuntos
Tecido Adiposo , Células Endoteliais , Humanos , Camundongos , Animais , Distribuição Tecidual , Adipócitos , Células Estromais
8.
Reprod Biol Endocrinol ; 22(1): 37, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38576003

RESUMO

Inadequate endometrial receptivity often results in embryo implantation failure and miscarriage. Human chorionic gonadotropin (hCG) is a key signaling molecule secreted during early embryonic development, which regulates embryonic maternal interface signaling and promotes embryo implantation. This study aimed to examine the impact of hCG on endometrial receptivity and its underlying mechanisms. An exploratory study was designed, and endometrial samples were obtained from women diagnosed with simple tubal infertility or male factor infertile (n = 12) and recurrent implantation failure (RIF, n = 10). Using reverse transcription-quantitative PCR and western blotting, luteinizing hormone (LH)/hCG receptor (LHCGR) levels and autophagy were detected in the endometrial tissues. Subsequently, primary endometrial stromal cells (ESCs) were isolated from these control groups and treated with hCG to examine the presence of LHCGR and markers of endometrial receptivity (HOXA10, ITGB3, FOXO1, LIF, and L-selectin ligand) and autophagy-related factors (Beclin1, LC3, and P62). The findings revealed that the expressions of receptivity factors, LHCGR, and LC3 were reduced in the endometrial tissues of women with RIF compared with the control group, whereas the expression of P62 was elevated. The administration of hCG to ESCs specifically activated LHCGR, stimulating an increase in the endometrial production of HOXA10, ITGB3, FOXO1, LIF and L-selectin ligands. Furthermore, when ESCs were exposed to 0.1 IU/mL hCG for 72 h, the autophagy factors Beclin1 and LC3 increased within the cells and P62 decreased. Moreover, the apoptotic factor Bax increased and Bcl-2 declined. However, when small interfering RNA was used to knock down LHCGR, hCG was less capable of controlling endometrial receptivity and autophagy molecules in ESCs. In addition, hCG stimulation enhanced the phosphorylation of ERK1/2 and mTOR proteins. These results suggest that women with RIF exhibit lower levels of LHCGR and compromised autophagy function in their endometrial tissues. Thus, hCG/LHCGR could potentially improve endometrial receptivity by modulating autophagy and apoptosis.


Assuntos
Endométrio , Selectina L , Gravidez , Humanos , Masculino , Feminino , Proteína Beclina-1 , Selectina L/metabolismo , Endométrio/metabolismo , Gonadotropina Coriônica/farmacologia , Gonadotropina Coriônica/metabolismo , Implantação do Embrião/fisiologia , Autofagia , Células Estromais/metabolismo , Apoptose
9.
Front Immunol ; 15: 1352819, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38455063

RESUMO

Bone fracture repair is a complex, multi-step process that involves communication between immune and stromal cells to coordinate the repair and regeneration of damaged tissue. In the US, 10% of all bone fractures do not heal properly without intervention, resulting in non-union. Complications from non-union fractures are physically and financially debilitating. We now appreciate the important role that immune cells play in tissue repair, and the necessity of the inflammatory response in initiating healing after skeletal trauma. The temporal dynamics of immune and stromal cell populations have been well characterized across the stages of fracture healing. Recent studies have begun to untangle the intricate mechanisms driving the immune response during normal or atypical, delayed healing. Various in vivo models of fracture healing, including genetic knockouts, as well as in vitro models of the fracture callus, have been implemented to enable experimental manipulation of the heterogeneous cellular environment. The goals of this review are to (1): summarize our current understanding of immune cell involvement in fracture healing (2); describe state-of-the art approaches to study inflammatory cells in fracture healing, including computational and in vitro models; and (3) identify gaps in our knowledge concerning immune-stromal crosstalk during bone healing.


Assuntos
Consolidação da Fratura , Fraturas Ósseas , Humanos , Calo Ósseo , Células Estromais , Comunicação Celular
10.
FASEB J ; 38(6): e23548, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38491832

RESUMO

Colorectal cancer (CRC) is a multifaceted disease characterized by a complex interaction between tumor cells and the surrounding microenvironment. Within this intricate landscape, exosomes have emerged as pivotal players in the tumor-stroma crosstalk, influencing the immune microenvironment of CRC. These nano-sized vesicles, secreted by both tumoral and stromal cells, serve as molecular transporters, delivering a heterogeneous mix of biomolecules such as RNAs, proteins, and lipids. In the CRC context, exosomes exert dual roles: they promote tumor growth, metastasis, and immune escape by altering immune cell functions and activating oncogenic signaling pathways and offer potential as biomarkers for early CRC detection and treatment targets. This review delves into the multifunctional roles of exosomes in the CRC immune microenvironment, highlighting their potential implications for future therapeutic strategies and clinical outcomes.


Assuntos
Neoplasias Colorretais , Exossomos , Humanos , Exossomos/metabolismo , RNA/metabolismo , Células Estromais/metabolismo , Neoplasias Colorretais/patologia , Microambiente Tumoral
11.
Exp Mol Med ; 56(3): 527-548, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38443595

RESUMO

Conventional tumor models have critical shortcomings in that they lack the complexity of the human stroma. The heterogeneous stroma is a central compartment of the tumor microenvironment (TME) that must be addressed in cancer research and precision medicine. To fully model the human tumor stroma, the deconstruction and reconstruction of tumor tissues have been suggested as new approaches for in vitro tumor modeling. In this review, we summarize the heterogeneity of tumor-associated stromal cells and general deconstruction approaches used to isolate patient-specific stromal cells from tumor tissue; we also address the effect of the deconstruction procedure on the characteristics of primary cells. Finally, perspectives on the future of reconstructed tumor models are discussed, with an emphasis on the essential prerequisites for developing authentic humanized tumor models.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/patologia , Células Estromais/patologia , Microambiente Tumoral
12.
Stem Cell Res Ther ; 15(1): 68, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38443990

RESUMO

BACKGROUND: Mesenchymal stem/stromal cells (MSCs) are of great therapeutic value due to their role in maintaining the function of hematopoietic stem/progenitor cells (HSPCs). MSCs derived from human pluripotent stem cells represent an ideal alternative because of their unlimited supply. However, the role of MSCs with neural crest origin derived from HPSCs on the maintenance of HSPCs has not been reported. METHODS: Flow cytometric analysis, RNA sequencing and differentiation ability were applied to detect the characteristics of stromal cells from 3D human brain organoids. Human umbilical cord blood CD34+ (UCB-CD34+) cells were cultured in different coculture conditions composed of stromal cells and umbilical cord MSCs (UC-MSCs) with or without a cytokine cocktail. The hematopoietic stroma capacity of stromal cells was tested in vitro with the LTC-IC assay and in vivo by cotransplantation of cord blood nucleated cells and stroma cells into immunodeficient mice. RNA and proteomic sequencing were used to detect the role of MSCs on HSPCs. RESULTS: The stromal cells, derived from both H1-hESCs and human induced pluripotent stem cells forebrain organoids, were capable of differentiating into the classical mesenchymal-derived cells (osteoblasts, chondrocytes, and adipocytes). These cells expressed MSC markers, thus named pluripotent stem cell-derived MSCs (pMSCs). The pMSCs showed neural crest origin with CD271 expression in the early stage. When human UCB-CD34+ HSPCs were cocultured on UC-MSCs or pMSCs, the latter resulted in robust expansion of UCB-CD34+ HSPCs in long-term culture and efficient maintenance of their transplantability. Comparison by RNA sequencing indicated that coculture of human UCB-CD34+ HSPCs with pMSCs provided an improved microenvironment for HSC maintenance. The pMSCs highly expressed the Wnt signaling inhibitors SFRP1 and SFRP2, indicating that they may help to modulate the cell cycle to promote the maintenance of UCB-CD34+ HSPCs by antagonizing Wnt activation. CONCLUSIONS: A novel method for harvesting MSCs with neural crest origin from 3D human brain organoids under serum-free culture conditions was reported. We demonstrate that the pMSCs support human UCB-HSPC expansion in vitro in a long-term culture and the maintenance of their transplantable ability. RNA and proteomic sequencing indicated that pMSCs provided an improved microenvironment for HSC maintenance via mechanisms involving cell-cell contact and secreted factors and suppression of Wnt signaling. This represents a novel method for large-scale production of MSCs of neural crest origin and provides a potential approach for development of human hematopoietic stromal cell therapy for treatment of dyshematopoiesis.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Proteômica , Células Estromais , Antígenos CD34 , Organoides , Prosencéfalo , RNA
13.
Methods Mol Biol ; 2783: 13-24, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478223

RESUMO

Adipose tissue is an abundant and accessible source of stem cells with multipotent properties suitable for tissue engineering and regenerative medical applications. Adipose-derived stromal/stem cells (ASCs) have been widely used in tissue engineering and cell therapy. In addition, the clinical application of ASCs in the treatment of inflammation and injury has been proven a success. Here, we describe methods from our own laboratory and the literature for the isolation and expansion of Adipose-derived stromal/stem cells (ASCs). We present a large-scale procedure suitable for processing >100 mL volumes of lipoaspirate tissue specimens by collagenase digestion, a related procedure suitable for processing adipose tissue aspirates without digestion, and a procedure suitable for intact human adipose tissue, such as buccal fat pads in the maxillofacial region.


Assuntos
Adipócitos , Tecido Adiposo , Humanos , Células Estromais , Células-Tronco , Engenharia Tecidual/métodos , Diferenciação Celular , Células Cultivadas
14.
Methods Mol Biol ; 2783: 3-11, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478222

RESUMO

Human adipose-derived stromal/stem cells (hASCs) are a promising source of adult stem cells used in numerous applications in regenerative medicine. We present the protocols from our laboratory for isolating and expanding hASCs. The isolation of hASCs involves the enzymatic digestion of adipose tissue and subsequent culturing of the isolated cells.


Assuntos
Células-Tronco Mesenquimais , Adulto , Humanos , Adipócitos , Tecido Adiposo , Células Estromais , Medicina Regenerativa , Diferenciação Celular
15.
Methods Mol Biol ; 2783: 109-114, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478228

RESUMO

Adipose tissue provides a valuable cell source for tissue engineering, regenerative medicine, and adipose tissue biology studies. The most widely used adipose-derived stromal/stem cells (ASCs) isolation protocol involves enzymatic digestion with collagenase. However, the yield of the method often proves to be poor if not impossible for collection of sufficient stromal vascular fraction (SVF) for expansion when the sample size is small, for instance when only newborn mice are available for cell culture. Here, we describe an efficient protocol for the isolation and expansion of ASCs using explant culture as an alternative. Briefly, adipose tissue was minced after removing excess liquid. Then, the minced tissue was placed in culture dishes or flasks. The cells will migrate out of tissue and adhere to the culture surface after one or more days.


Assuntos
Adipócitos , Tecido Adiposo , Camundongos , Animais , Células Estromais , Engenharia Tecidual/métodos , Obesidade , Células-Tronco , Diferenciação Celular
16.
Methods Mol Biol ; 2783: 235-262, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478237

RESUMO

Advances in technology and automation over the past several decades have made it feasible to perform high-throughput compound screening with cell spheroids, a valuable approach for drug discovery. It is entirely feasible to generate multiple 384-well plates containing adipose spheroids from cryopreserved, single-donor, adipose stem cells, thus incorporating genetic diversity into the discovery stages of research. In this protocol, we describe our method for isolating primary human adipose stem cells and synthesizing cell spheroids comprised of mature adipocytes and stromal cells. Also included are representative outcome measurements useful for characterizing adipocyte metabolism and health. Wherever possible, we describe technologies that can be used to automate characterization and increase throughput.


Assuntos
Adipócitos , Tecido Adiposo , Humanos , Tecido Adiposo/metabolismo , Adipócitos/metabolismo , Esferoides Celulares , Células Estromais , Obesidade/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular
17.
Methods Mol Biol ; 2783: 177-193, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478233

RESUMO

With the increase in decellularization of different tissue sources, an understanding of the viscoelastic properties of these soft materials is important for determining practical applications. The purpose of this chapter is to better define a series of experiments to profile important rheological properties for adipose-based hydrogels. While there are numerous mechanical characterizations that are done experimentally, the protocol outlined in this chapter provides a step-wise approach to determine the gelation characteristics and native hydrogel network properties. A more complete understanding of adipose-derived hydrogel mechanical properties would provide vital information for downstream applicability in fields such as disease modeling or soft tissue regeneration.


Assuntos
Adipócitos , Hidrogéis , Células Estromais , Cicatrização , Células-Tronco
18.
Methods Mol Biol ; 2783: 303-308, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478242

RESUMO

Breast cancer is an ongoing issue due to its high mortality rates. Obesity enhances the problems associated with breast cancer, meaning there must be a biological connection between them. This crosstalk may be the adipose-derived stem cell. If we can interrupt the communication between adipose-derived stromal/stem cells (ASCs) and breast cancer, we may be able to prevent cancer propagation. Specific kinase inhibition may allow us to downregulate signals, preventing ASC-mediated cancer growth. This chapter provides a critical method for screening a kinase inhibitor drug library for hits on ASCs.


Assuntos
Tecido Adiposo , Neoplasias da Mama , Humanos , Feminino , Adipócitos , Neoplasias da Mama/tratamento farmacológico , Células Estromais/fisiologia , Obesidade , Proliferação de Células
19.
Methods Mol Biol ; 2783: 309-322, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478243

RESUMO

We have developed a hollow fiber bioreactor-based production system for manufacturing large quantities of extracellular vesicles (EVs) containing exosomes from adult human adipose-derived stromal/stem cells (ASCs). By manipulating the cellular bioreactor environment, we have found that we can alter ASC EV production, secretion, and surface protein composition. The aims of this chapter are to describe the methodology for culturing and tuning of adipose ASCs in a bioreactor, along with the collection and isolation of the EVs containing exosomes demonstrating increased HSP70 content.


Assuntos
Exossomos , Vesículas Extracelulares , Adulto , Humanos , Exossomos/metabolismo , Células Estromais , Vesículas Extracelulares/metabolismo , Adipócitos , Obesidade/metabolismo , Células-Tronco , Tecido Adiposo
20.
Cancer Res ; 84(5): 648-649, 2024 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-38437636

RESUMO

Cancer aggressiveness has been linked with obesity, and studies have shown that adipose tissue can enhance cancer progression. In this issue of Cancer Research, Hosni and colleagues discover a paracrine mechanism mediated by adipocyte precursor cells through which urothelial carcinomas become resistant to erdafitinib, a recently approved therapy inhibiting fibroblast growth factor receptors (FGFR). They identified neuregulin 1 (NRG1) secreted by adipocyte precursor cells as an activator of HER3 signaling that enables resistance. The NRG1-mediated FGFR inhibitor resistance was amenable to intervention with pertuzumab, an antibody blocking the NRG1/HER3 axis. To investigate the nature of the resistance-associated NRG1-expressing cells in human patients, the authors analyzed published single-cell RNA sequencing data and observed that such cells appear in a cluster assigned as inflammatory cancer-associated fibroblasts (iCAF). Notably, the gene signature corresponding to these CAFs is highly similar to that shared by adipose stromal cells (ASC) in fat tissue and fibro-adipogenic progenitors (FAP) in skeletal muscle of cancer-free individuals. Because fibroblasts with the ASC/FAP signature are enriched in various carcinomas, it is possible that the paracrine signaling conferred by NRG1 is a pan-cancer mechanism of FGFR inhibitor resistance and tumor aggressiveness. See related article by Hosni et al., p. 725.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma de Células de Transição , Humanos , Adipócitos , Tecido Adiposo , Células Estromais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...