Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.916
Filtrer
1.
Discov Med ; 36(188): 1772-1779, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39327240

RÉSUMÉ

Aging is frequently associated with a progressive increase in chronic low-grade inflammation, known as "inflammaging". Numerous studies have shown that inflammaging is closely linked to the development of several age-related diseases. However, the underlying mechanism and its causal role are still not fully understood despite this association. In the complex context of aging, mesenchymal stem cells (MSCs) undergo changes in behavior and functionality. This narrative topical review examines the recent advances in aging research, specifically focusing on the role of inflammaging and related mechanisms that contribute to age-related chronic diseases. The authors critically investigated whether and how inflammaging, epigenetic damage, mitochondrial changes, and macrophage alterations may influence stem cell behavior, highlighting the interplay between these factors and their potential therapeutic implications. By elucidating the mechanisms underlying these processes, we can gain valuable insights into the maintenance and regeneration of stem cell populations, providing the basis for novel therapeutic strategies targeting age-related decline and disease progression.


Sujet(s)
Vieillissement , Épigenèse génétique , Inflammation , Macrophages , Mitochondries , Humains , Macrophages/métabolisme , Macrophages/anatomopathologie , Mitochondries/métabolisme , Mitochondries/génétique , Mitochondries/anatomopathologie , Inflammation/anatomopathologie , Inflammation/génétique , Vieillissement/génétique , Vieillissement/anatomopathologie , Vieillissement/physiologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Animaux
2.
Cells ; 13(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39272976

RÉSUMÉ

3D bioprinting has become a valuable tool for studying the biology of solid tumors, including glioblastoma multiforme (GBM). Our analysis of publicly available bulk RNA and single-cell sequencing data has allowed us to define the chemotactic profile of GBM tumors and identify the cell types that secrete particular chemokines in the GBM tumor microenvironment (TME). Our findings indicate that primary GBM tissues express multiple chemokines, whereas spherical monocultures of GBM cells significantly lose this diversity. Subsequently, the comparative analysis of GBM spherical monocultures vs. 3D-bioprinted multicultures of cells showed a restoration of chemokine profile diversity in 3D-bioprinted cultures. Furthermore, single-cell RNA-Seq analysis showed that cells of the perivascular niche (pericytes and endocytes) express multiple chemokines in the GBM TME. Next, we 3D-bioprinted cells from two glioblastoma cell lines, U-251 and DK-MG, alone and as co-cultures with mesenchymal stromal cells (representing cells of the perivascular niche) and assessed the chemokine secretome. The results clearly demonstrated that the interaction of tumors and mesenchymal cells leads to in a significant increase in the repertoire and levels of secreted chemokines under culture in 21% O2 and 1% O2. Our study indicates that cells of the perivascular niche may perform a substantial role in shaping the chemokine microenvironment in GBM tumors.


Sujet(s)
Chimiokines , Techniques de coculture , Glioblastome , Cellules souches mésenchymateuses , Microenvironnement tumoral , Humains , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Chimiokines/métabolisme , Lignée cellulaire tumorale , Impression tridimensionnelle , Bio-impression , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme
3.
Front Endocrinol (Lausanne) ; 15: 1359052, 2024.
Article de Anglais | MEDLINE | ID: mdl-39157681

RÉSUMÉ

Introduction: Changes to bone physiology play a central role in the development of osteoarthritis with the mechanosensing osteocyte releasing factors that drive disease progression. This study developed a humanised in vitro model to detect osteocyte responses to either interleukin-6, a driver of degeneration and bone remodelling in animal and human joint injury, or mechanical loading, to mimic osteoarthritis stimuli in joints. Methods: Human MSC cells (Y201) were differentiated in 3-dimensional type I collagen gels in osteogenic media and osteocyte phenotype assessed by RTqPCR and immunostaining. Gels were subjected to a single pathophysiological load or stimulated with interleukin-6 with unloaded or unstimulated cells as controls. RNA was extracted 1-hour post-load and assessed by RNAseq. Markers of pain, bone remodelling, and inflammation were quantified by RT-qPCR and ELISA. Results: Y201 cells embedded within 3D collagen gels assumed dendritic morphology and expressed mature osteocytes markers. Mechanical loading of the osteocyte model regulated 7564 genes (Padj p<0.05, 3026 down, 4538 up). 93% of the osteocyte transcriptome signature was expressed in the model with 38% of these genes mechanically regulated. Mechanically loaded osteocytes regulated 26% of gene ontology pathways linked to OA pain, 40% reflecting bone remodelling and 27% representing inflammation. Load regulated genes associated with osteopetrosis, osteoporosis and osteoarthritis. 42% of effector genes in a genome-wide association study meta-analysis were mechanically regulated by osteocytes with 10 genes representing potential druggable targets. Interleukin-6 stimulation of osteocytes at concentrations reported in human synovial fluids from patients with OA or following knee injury, regulated similar readouts to mechanical loading including markers of pain, bone remodelling, and inflammation. Discussion: We have developed a reproducible model of human osteocyte like cells that express >90% of the genes in the osteocyte transcriptome signature. Mechanical loading and inflammatory stimulation regulated genes and proteins implicated in osteoarthritis symptoms of pain as well as inflammation and degeneration underlying disease progression. Nearly half of the genes classified as 'effectors' in GWAS were mechanically regulated in this model. This model will be useful in identifying new mechanisms underlying bone and joint pathologies and testing drugs targeting those mechanisms.


Sujet(s)
Inflammation , Cellules souches mésenchymateuses , Arthrose , Ostéocytes , Humains , Ostéocytes/métabolisme , Ostéocytes/anatomopathologie , Arthrose/anatomopathologie , Arthrose/métabolisme , Inflammation/anatomopathologie , Inflammation/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Interleukine-6/métabolisme , Remodelage osseux , Cellules cultivées , Différenciation cellulaire
4.
J Clin Invest ; 134(16)2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39145446

RÉSUMÉ

Tissue fibrosis remains unamenable to meaningful therapeutic interventions and is the primary cause of chronic graft failure after organ transplantation. Eukaryotic translation initiation factor (eIF4E), a key translational regulator, serves as convergent target of multiple upstream profibrotic signaling pathways that contribute to mesenchymal cell (MC) activation. Here, we investigate the role of MAP kinase-interacting serine/threonine kinase-induced (MNK-induced) direct phosphorylation of eIF4E at serine 209 (Ser209) in maintaining fibrotic transformation of MCs and determine the contribution of the MNK/eIF4E pathway to the pathogenesis of chronic lung allograft dysfunction (CLAD). MCs from patients with CLAD demonstrated constitutively higher eIF4E phosphorylation at Ser209, and eIF4E phospho-Ser209 was found to be critical in regulating key fibrogenic protein autotaxin, leading to sustained ß-catenin activation and profibrotic functions of CLAD MCs. MNK1 signaling was upregulated in CLAD MCs, and genetic or pharmacologic targeting of MNK1 activity inhibited eIF4E phospho-Ser209 and profibrotic functions of CLAD MCs in vitro. Treatment with an MNK1/2 inhibitor (eFT-508) abrogated allograft fibrosis in an orthotopic murine lung-transplant model. Together these studies identify what we believe is a previously unrecognized MNK/eIF4E/ATX/ß-catenin signaling pathway of fibrotic transformation of MCs and present the first evidence, to our knowledge, for the utility of MNK inhibitors in fibrosis.


Sujet(s)
Allogreffes , Facteur-4E d'initiation eucaryote , Transplantation pulmonaire , Protein-Serine-Threonine Kinases , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Animaux , Souris , Phosphorylation , Humains , Facteur-4E d'initiation eucaryote/métabolisme , Facteur-4E d'initiation eucaryote/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Mâle , Fibrose , Femelle , Transduction du signal
5.
Tissue Cell ; 90: 102527, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39181089

RÉSUMÉ

Chronic kidney disease (CKD) and end-stage renal disease (ESRD) are prevalent and debilitating conditions with a significant impact on patients' quality of life. In this study, we conducted a comprehensive investigation into the histological characteristics of renal progenitor/stem cells (RPCs), renal mesenchymal stem-like cells, and endothelial progenitor cells (EPCs) in CKD and ESRD patients. Additionally, we performed a molecular docking analysis to explore potential drug-receptor interactions involving common medications prescribed to CKD patients. Our histological examination revealed a noteworthy increase in the number of CD24- and CD133-positive cells in CKD and ESRD patients, representing RPCs. These cells are implicated in kidney repair and regeneration, underscoring their potential role in CKD management. Moreover, we observed an elevation in the number of EPCs within the kidneys of CKD and ESRD patients, suggesting a protective role of EPCs in kidney preservation. The molecular docking analysis unveiled intriguing insights into potential drug interventions. Notably, digoxin exhibited the highest in-silico binding affinity to numerous receptors associated with the functions of RPCs, renal mesenchymal stem-like cells, and EPCs, emphasizing the potential multifaceted effects of this cardiac glycoside in CKD patients. Other drugs, including apixaban, glimepiride, and glibenclamide, also displayed strong in-silico affinities to specific receptors, indicating their potential influence on various renal cell functions. In conclusion, this study provides valuable insights into the histological alterations in renal cell populations in CKD and ESRD patients and underscores the potential roles of RPCs and EPCs in kidney repair and preservation. The molecular docking analysis reveals the complex interactions between common drugs and renal cells, suggesting the need for further in-vitro and in-vivo research to fully understand these relationships. These findings contribute to our understanding of CKD and offer new avenues for research into potential therapeutic interventions.


Sujet(s)
Progéniteurs endothéliaux , Défaillance rénale chronique , Cellules souches mésenchymateuses , Simulation de docking moléculaire , Insuffisance rénale chronique , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Défaillance rénale chronique/anatomopathologie , Défaillance rénale chronique/métabolisme , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/anatomopathologie , Rein/anatomopathologie , Rein/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte
6.
Exp Hematol ; 138: 104282, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39032857

RÉSUMÉ

We and others have previously shown that TAZ plays a tumor suppressive role in multiple myeloma. However, recent reports suggest that molecular crosstalk between the myeloma cells and bone marrow stromal components contributes to the myeloma cell survival and drug resistance. These reports further point to reciprocal interaction via adhesion molecules as the most prominent mechanism of intercellular crosstalk between myeloma cells and bone marrow mesenchymal stromal cells (BM-MSCs). YAP/TAZ silencing/expression has been shown to correlate across all cancers with a set of adhesion/extracellular matrix proteins. Therefore, we hypothesized that TAZ may regulate myeloma cell interaction with BM stromal cells by influencing the expression of distinct cell adhesion signatures. We used previously established TAZ myeloma cell line models, including DELTA47-pLENTI or TAZ knockout DELTA47 cells cocultured with or without BM-MSCs, as our study models. Using RNA sequencing analysis, we performed the first comprehensive screen for cell adhesion-related transcriptional targets of TAZ in multiple myeloma (MM). In doing so, we uncovered an enrichment of cell adhesion-related genes in TAZ knockout DELTA47 cells relatively to pLENTI-DELTA47 cells, including 11 genes with log2 fold change > 2 (p < 0.05), namely, ANXA1, ADGRL2, NCAM1, NCAM2, ADGRL3, CXADR, ALCAM, JAM2, KIRREL1, KIRREL2, and ADGRG7, suggesting possible relationship with TAZ. We validated ANXA1 as a bona fide target of TAZ in MM. We show that TAZ represses myeloma cell migration and interaction with BM-MSCs by transcriptionally downregulating ANXA1 expression via TEAD-dependent mechanism. Our data provide new insights into the understanding of the role of TAZ in the intercellular communication signals between myeloma cells and BM-MSCs. Our findings also suggest that ANXA1 represents a putative cell adhesion target to attenuate BM-MSC driven, tumor-promoting interaction with myeloma cells.


Sujet(s)
Annexine A1 , Communication cellulaire , Régulation négative , Régulation de l'expression des gènes tumoraux , Cellules souches mésenchymateuses , Myélome multiple , Transcriptional coactivator with PDZ-binding motif proteins , Myélome multiple/anatomopathologie , Myélome multiple/métabolisme , Myélome multiple/génétique , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Lignée cellulaire tumorale , Annexine A1/génétique , Annexine A1/métabolisme , Adhérence cellulaire , Techniques de coculture , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Protéines tumorales/biosynthèse
7.
Cells ; 13(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38891074

RÉSUMÉ

Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.


Sujet(s)
Tumeurs du cerveau , Reprogrammation cellulaire , Évolution clonale , Glioblastome , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Évolution clonale/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Reprogrammation cellulaire/génétique , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie
8.
Cancer Lett ; 596: 217022, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38849014

RÉSUMÉ

We previously reported that extracellular matrix protein 1 isoform a (ECM1a) promotes epithelial ovarian cancer (EOC) through autocrine signaling by binding to cell surface receptors αXß2. However, the role of ECM1a as a secretory molecule in the tumor microenvironment is rarely reported. In this study, we constructed murine Ecm1-knockout mice and human ECM1a-knockin mice and further generated orthotopic or peritoneal xenograft tumor models to mimic the different metastatic stages of EOC. We show that ECM1a induces oncogenic metastasis of orthotopic xenograft tumors, but inhibits early-metastasis of peritoneal xenograft tumors. ECM1a remodels extracellular matrices (ECM) and promotes remote metastases by recruiting and transforming bone marrow mesenchymal stem cells (BMSCs) into platelet-derived growth factor receptor beta (PDGFRß+) cancer-associated fibroblasts (CAFs) and facilitating the secretion of angiopoietin-like protein 2 (ANGPTL2). Competing with ECM1a, ANGPTL2 also binds to integrin αX through the P1/P2 peptides, resulting in negative effects on BMSC differentiation. Collectively, this study reveals the dual functions of ECM1a in remodeling of TME during tumor progression, emphasizing the complexity of EOC phenotypic heterogeneity and metastasis.


Sujet(s)
Protéine-2 de type angiopoïétine , Fibroblastes associés au cancer , Protéines de la matrice extracellulaire , Souris knockout , Tumeurs de l'ovaire , Microenvironnement tumoral , Animaux , Femelle , Humains , Souris , Protéines semblables à l'angiopoïétine/métabolisme , Protéines semblables à l'angiopoïétine/génétique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Carcinome épithélial de l'ovaire/anatomopathologie , Carcinome épithélial de l'ovaire/métabolisme , Carcinome épithélial de l'ovaire/génétique , Lignée cellulaire tumorale , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Métastase tumorale , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme
9.
Clin Cancer Res ; 30(15): 3259-3272, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38775859

RÉSUMÉ

PURPOSE: The genetic intratumoral heterogeneity observed in human osteosarcomas poses challenges for drug development and the study of cell fate, plasticity, and differentiation, which are processes linked to tumor grade, cell metastasis, and survival. EXPERIMENTAL DESIGN: To pinpoint errors in osteosarcoma differentiation, we transcriptionally profiled 31,527 cells from a tissue-engineered model that directs mesenchymal stem cells toward adipogenic and osteoblastic fates. Incorporating preexisting chondrocyte data, we applied trajectory analysis and non-negative matrix factorization to generate the first human mesenchymal differentiation atlas. RESULTS: This "roadmap" served as a reference to delineate the cellular composition of morphologically complex osteosarcoma tumors and quantify each cell's lineage commitment. Projecting a bulk RNA-sequencing osteosarcoma dataset onto this roadmap unveiled a correlation between a stem-like transcriptomic phenotype and poorer survival outcomes. CONCLUSIONS: Our study quantifies osteosarcoma differentiation and lineage, a prerequisite to better understanding lineage-specific differentiation bottlenecks that might someday be targeted therapeutically.


Sujet(s)
Tumeurs osseuses , Différenciation cellulaire , Cellules souches mésenchymateuses , Ostéosarcome , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/mortalité , Humains , Cellules souches mésenchymateuses/anatomopathologie , Cellules souches mésenchymateuses/métabolisme , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Tumeurs osseuses/mortalité , Analyse sur cellule unique/méthodes , Transcriptome , Lignage cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes
10.
Am J Pathol ; 194(6): 1137-1153, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38749609

RÉSUMÉ

Preclinical models that display spontaneous metastasis are necessary to improve the therapeutic options for hormone receptor-positive breast cancers. Within this study, detailed cellular and molecular characterization was conducted on MCa-P1362, a newly established mouse model of metastatic breast cancer that is syngeneic in BALB/c mice. MCa-P1362 cancer cells express estrogen receptor, progesterone receptor, and the human epidermal growth factor receptor 2. MCa-P1362 cancer cells proliferate in vitro and in vivo in response to estrogen, yet do not depend on steroid hormones for growth and tumor progression. Analysis of MCa-P1362 tumor explants revealed the tumors contained a mixture of cancer cells and mesenchymal stromal cells. Through transcriptomic and functional analyses of both cancer and stromal cells, stem cells were detected within both populations. Functional studies demonstrated that MCa-P1362 cancer stem cells drove tumor initiation, whereas stromal cells from these tumors contributed to drug resistance. MCa-P1362 may serve as a useful preclinical model to investigate the cellular and molecular basis of breast tumor progression and therapeutic resistance.


Sujet(s)
Adénocarcinome , Cellules souches mésenchymateuses , Souris de lignée BALB C , Récepteur ErbB-2 , Récepteurs des oestrogènes , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Femelle , Humains , Récepteur ErbB-2/métabolisme , Souris , Récepteurs des oestrogènes/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome/métabolisme , Récepteurs à la progestérone/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Lignée cellulaire tumorale , Tumeurs expérimentales de la mamelle/anatomopathologie , Tumeurs expérimentales de la mamelle/métabolisme
11.
Nat Commun ; 15(1): 4139, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755155

RÉSUMÉ

The natural history of multiple myeloma is characterized by its localization to the bone marrow and its interaction with bone marrow stromal cells. The bone marrow stromal cells provide growth and survival signals, thereby promoting the development of drug resistance. Here, we show that the interaction between bone marrow stromal cells and myeloma cells (using human cell lines) induces chromatin remodeling of cis-regulatory elements and is associated with changes in the expression of genes involved in the cell migration and cytokine signaling. The expression of genes involved in these stromal interactions are observed in extramedullary disease in patients with myeloma and provides the rationale for survival of myeloma cells outside of the bone marrow microenvironment. Expression of these stromal interaction genes is also observed in a subset of patients with newly diagnosed myeloma and are akin to the transcriptional program of extramedullary disease. The presence of such adverse stromal interactions in newly diagnosed myeloma is associated with accelerated disease dissemination, predicts the early development of therapeutic resistance, and is of independent prognostic significance. These stromal cell induced transcriptomic and epigenomic changes both predict long-term outcomes and identify therapeutic targets in the tumor microenvironment for the development of novel therapeutic approaches.


Sujet(s)
Assemblage et désassemblage de la chromatine , Régulation de l'expression des gènes tumoraux , Cellules souches mésenchymateuses , Myélome multiple , Microenvironnement tumoral , Myélome multiple/génétique , Myélome multiple/anatomopathologie , Myélome multiple/métabolisme , Humains , Microenvironnement tumoral/génétique , Lignée cellulaire tumorale , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Transcription génétique , Cellules de la moelle osseuse/métabolisme , Mouvement cellulaire/génétique , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Femelle , Mâle
12.
Cell Commun Signal ; 22(1): 257, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38711089

RÉSUMÉ

Benign prostatic hyperplasia (BPH) is a multifactorial disease in which abnormal growth factor activation and embryonic reawakening are considered important factors. Here we demonstrated that the aberrant activation of transforming growth factor ß (TGF-ß)/Rho kinase 1 (ROCK1) increased the stemness of BPH tissue by recruiting mesenchymal stem cells (MSCs), indicating the important role of embryonic reawakening in BPH. When TGF-ß/ROCK1 is abnormally activated, MSCs are recruited and differentiate into fibroblasts/myofibroblasts, leading to prostate stromal hyperplasia. Further research showed that inhibition of ROCK1 activation suppressed MSC migration and their potential for stromal differentiation. Collectively, our findings suggest that abnormal activation of TGF-ß/ROCK1 regulates stem cell lineage specificity, and the small molecule inhibitor GSK269962A could target ROCK1 and may be a potential treatment for BPH.


Sujet(s)
Cellules souches mésenchymateuses , Hyperplasie de la prostate , Facteur de croissance transformant bêta , rho-Associated Kinases , rho-Associated Kinases/métabolisme , Mâle , Hyperplasie de la prostate/anatomopathologie , Hyperplasie de la prostate/métabolisme , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Animaux , Différenciation cellulaire , Prostate/anatomopathologie , Prostate/métabolisme , Mouvement cellulaire , Souris , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie
13.
Cancer Res Commun ; 4(4): 1150-1164, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38598843

RÉSUMÉ

Multiple myeloma involves early dissemination of malignant plasma cells across the bone marrow; however, the initial steps of dissemination remain unclear. Human bone marrow-derived mesenchymal stromal cells (hMSC) stimulate myeloma cell expansion (e.g., IL6) and simultaneously retain myeloma cells via chemokines (e.g., CXCL12) and adhesion factors. Hence, we hypothesized that the imbalance between cell division and retention drives dissemination. We present an in vitro model using primary hMSCs cocultured with INA-6 myeloma cells. Time-lapse microscopy revealed proliferation and attachment/detachment dynamics. Separation techniques (V-well adhesion assay and well plate sandwich centrifugation) were established to isolate MSC-interacting myeloma subpopulations that were characterized by RNA sequencing, cell viability, and apoptosis. Results were correlated with gene expression data (n = 837) and survival of patients with myeloma (n = 536). On dispersed hMSCs, INA-6 saturate hMSC surface before proliferating into large homotypic aggregates, from which single cells detached completely. On confluent hMSCs, aggregates were replaced by strong heterotypic hMSC-INA-6 interactions, which modulated apoptosis time dependently. Only INA-6 daughter cells (nMA-INA6) detached from hMSCs by cell division but sustained adherence to hMSC-adhering mother cells (MA-INA6). Isolated nMA-INA6 indicated hMSC autonomy through superior viability after IL6 withdrawal and upregulation of proliferation-related genes. MA-INA6 upregulated adhesion and retention factors (CXCL12), that, intriguingly, were highly expressed in myeloma samples from patients with longer overall and progression-free survival, but their expression decreased in relapsed myeloma samples. Altogether, in vitro dissemination of INA-6 is driven by detaching daughter cells after a cycle of hMSC-(re)attachment and proliferation, involving adhesion factors that represent a bone marrow-retentive phenotype with potential clinical relevance. SIGNIFICANCE: Novel methods describe in vitro dissemination of myeloma cells as detachment of daughter cells after cell division. Myeloma adhesion genes were identified that counteract in vitro detachment with potential clinical relevance.


Sujet(s)
Adhérence cellulaire , Prolifération cellulaire , Cellules souches mésenchymateuses , Myélome multiple , Humains , Myélome multiple/anatomopathologie , Myélome multiple/génétique , Myélome multiple/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Apoptose , Techniques de coculture , Lignée cellulaire tumorale , Agrégation cellulaire , Survie cellulaire
14.
Exp Cell Res ; 438(1): 114032, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38583856

RÉSUMÉ

Triple-negative breast cancer (TNBC) is characterized by the complex tumor microenvironment (TME) consisting of an abundance of mesenchymal stem cells (MSCs), which is known to facilitate epithelial-to-mesenchymal transition (EMT). The development of single-cell genomics is a powerful method for defining the intricate genetic landscapes of malignancies. In this study, we have employed single-cell RNA sequencing (scRNA-seq) to dissect the intra-tumoral heterogeneity and analyze the single-cell transcriptomic landscape to detect rare consequential cell subpopulations of significance. The scRNA-seq analysis of TNBC and Normal patient derived samples revealed that EMT markers and transcription factors were most upregulated in MSC population. Further, exploration of gene expression analysis among TNBC and Normal patient-derived MSCs ascertained the role of SQSTM1/P62 and Wnt/ß-catenin in TNBC progression. Wnt/ß-catenin and Wnt/PCP signaling pathways are prominent contributors of EMT, stemness, and cancer stem cell (CSC) properties of TNBC. SQSTM1/P62 cooperates with the components of the Wnt/PCP signaling pathway and is critically involved at the interface of autophagy and EMT. Moreover, siRNA targeting SQSTM1/P62 and inhibitor of Wnt/ß-catenin (FH535) in conjunction was used to explore molecular modification of EMT and stemness markers. Although SQSTM1/P62 is not crucial for cell survival, cytotoxicity assay revealed synergistic interaction between the siRNA/inhibitor. Modulation of these important pathways helped in reduction of expression of genes and proteins contributing to CSC properties. Gene and protein expression analysis revealed the induction of EMT to MET. Moreover, co-treatment resulted in inactivation of non-canonical Wnt VANGL2-JNK signaling axis. The synergistic impact of inhibition of SQSTM1/P62 and Wnt/ß-catenin signaling facilitates the development of a potential therapeutic regimen for TNBC.


Sujet(s)
Transition épithélio-mésenchymateuse , Cellules souches tumorales , Séquestosome-1 , Analyse sur cellule unique , Tumeurs du sein triple-négatives , Voie de signalisation Wnt , Femelle , Humains , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Séquestosome-1/métabolisme , Séquestosome-1/génétique , Analyse sur cellule unique/méthodes , Transcriptome/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Microenvironnement tumoral/génétique , Voie de signalisation Wnt/génétique
15.
Sci Rep ; 14(1): 6649, 2024 03 20.
Article de Anglais | MEDLINE | ID: mdl-38503815

RÉSUMÉ

Current treatments for inflammatory bowel disease (IBD) are often inadequate due to limited efficacy and toxicity, leading to surgical resection in refractory cases. IBD's broad and complex pathogenesis involving the immune system, enteric nervous system, microbiome, and oxidative stress requires more effective therapeutic strategies. In this study, we investigated the therapeutic potential of bone marrow-derived mesenchymal stem cell (BM-MSC) treatments in spontaneous chronic colitis using the Winnie mouse model which closely replicates the presentation and inflammatory profile of ulcerative colitis. The 14-day BM-MSC treatment regimen reduced the severity of colitis, leading to the attenuation of diarrheal symptoms and recovery in body mass. Morphological and histological abnormalities in the colon were also alleviated. Transcriptomic analysis demonstrated that BM-MSC treatment led to alterations in gene expression profiles primarily downregulating genes related to inflammation, including pro-inflammatory cytokines, chemokines and other biomarkers of inflammation. Further evaluation of immune cell populations using immunohistochemistry revealed a reduction in leukocyte infiltration upon BM-MSC treatment. Notably, enteric neuronal gene signatures were the most impacted by BM-MSC treatment, which correlated with the restoration of neuronal density in the myenteric ganglia. Moreover, BM-MSCs exhibited neuroprotective effects against oxidative stress-induced neuronal loss through antioxidant mechanisms, including the reduction of mitochondrial-derived superoxide and attenuation of oxidative stress-induced HMGB1 translocation, potentially relying on MSC-derived SOD1. These findings suggest that BM-MSCs hold promise as a therapeutic intervention to mitigate chronic colitis by exerting anti-inflammatory effects and protecting the enteric nervous system from oxidative stress-induced damage.


Sujet(s)
Colite , Maladies inflammatoires intestinales , Pseudo-obstruction intestinale , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Souris , Animaux , Moelle osseuse/anatomopathologie , Colite/induit chimiquement , Cellules souches mésenchymateuses/anatomopathologie , Inflammation , Anti-inflammatoires/effets indésirables , Modèles animaux de maladie humaine
16.
Haematologica ; 109(7): 2073-2084, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38426282

RÉSUMÉ

B-cell precursor acute lymphoblastic leukemia (BCP-ALL) can hijack the normal bone marrow microenvironment to create a leukemic niche which facilitates blast cell survival and promotes drug resistance. Bone marrow-derived mesenchymal stromal cells (MSC) mimic this protective environment in ex vivo co-cultures with leukemic cells obtained from children with newly diagnosed BCP-ALL. We examined the potential mechanisms of this protection by RNA sequencing of flow-sorted MSC after co-culture with BCP-ALL cells. Leukemic cells induced an interferon (IFN)-related gene signature in MSC, which was partially dependent on direct cell-cell signaling. The signature was selectively induced by BCP-ALL cells, most profoundly by ETV6-RUNX1-positive ALL cells, as co-culture of MSC with healthy immune cells did not provoke a similar IFN signature. Leukemic cells and MSC both secreted IFNα and IFNß, but not IFNγ. In line, the IFN gene signature was sensitive to blockade of IFNα/ß signaling, but less to that of IFNγ. The viability of leukemic cells and level of resistance to three chemotherapeutic agents was not affected by interference with IFN signaling using selective IFNα/ß inhibitors or silencing of IFN-related genes. Taken together, our data suggest that the leukemia-induced expression of IFNα/ß-related genes by MSC does not support survival of BCP-ALL cells but may serve a different role in the pathobiology of BCP-ALL.


Sujet(s)
Techniques de coculture , Interféron alpha , Interféron bêta , Cellules souches mésenchymateuses , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Interféron alpha/pharmacologie , Interféron bêta/métabolisme , Microenvironnement tumoral , Transduction du signal , Enfant , Lignée cellulaire tumorale , Transcriptome , Résistance aux médicaments antinéoplasiques , Analyse de profil d'expression de gènes , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Régulation de l'expression des gènes dans la leucémie , ETS Translocation Variant 6 Protein , Sous-unité alpha 2 du facteur CBF
17.
J Bone Miner Res ; 39(2): 161-176, 2024 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-38477740

RÉSUMÉ

Osteoarthritis (OA) affects multiple tissues in the knee joint, including the synovium and intra-articular adipose tissue (IAAT) that are attached to each other. However, whether these two tissues share the same progenitor cells and hence function as a single unit in joint homeostasis and diseases is largely unknown. Single-cell transcriptomic profiling of synovium and infrapatellar fat pad (IFP), the largest IAAT, from control and OA mice revealed five mesenchymal clusters and predicted mesenchymal progenitor cells (MPCs) as the common progenitors for other cells: synovial lining fibroblasts (SLFs), myofibroblasts (MFs), and preadipocytes 1 and 2. Histologic examination of joints in reporter mice having Dpp4-CreER and Prg4-CreER that label MPCs and SLFs, respectively, demonstrated that Dpp4+ MPCs reside in the synovial sublining layer and give rise to Prg4+ SLFs and Perilipin+ adipocytes during growth and OA progression. After OA injury, both MPCs and SLFs gave rise to MFs, which remained in the thickened synovium at later stages of OA. In culture, Dpp4+ MPCs possessed mesenchymal progenitor properties, such as proliferation and multilineage differentiation. In contrast, Prg4+ SLFs did not contribute to adipocytes in IFP and Prg4+ cells barely grew in vitro. Taken together, we demonstrate that the synovium and joint fat pad are one integrated functional tissue sharing common mesenchymal progenitors and undergoing coordinated changes during OA progression.


Both synovium and intra-articular adipose tissue (IAAT) in knee joint play a critical role in joint health and osteoarthritis (OA) progression. Recent single-cell RNA-sequencing studies have been performed on the mouse and human synovium. However, IAATs residing in close proximity to the synovium have not been studied yet. Our study reveals mesenchymal cell heterogeneity of synovium/infrapatellar fat pad (Syn/IFP) tissue and their OA responses. We identify Dpp4+ multipotent progenitors as a source that give rise to Prg4+ lining layer fibroblasts in the synovium, adipocytes in the IFP, and myofibroblasts in the OA Syn/IFP tissue. Our work demonstrates that Syn/IFP is a functionally connected tissue that shares common mesenchymal progenitors and undergoes coordinated OA changes. This novel insight advances our knowledge of previously understudied joint tissues and provides new directions for drug discovery to treat joint disorders.


Sujet(s)
Tissu adipeux , Cellules souches mésenchymateuses , Membrane synoviale , Animaux , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Souris , Arthrose/anatomopathologie , Arthrose/métabolisme , Patella/anatomopathologie , Patella/métabolisme
18.
Biomed Pharmacother ; 173: 115790, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38431436

RÉSUMÉ

BACKGROUND: Although PD-1 blockade is effective for treating several types of cancer, the efficacy of this agent in glioblastoma is largely limited. To overcome non-responders and the immunosuppressive tumor microenvironment, combinational immunotherapeutic strategies with anti-PD-1 need to be considered. Here, we developed IL-12-secreting mesenchymal stem cells (MSC_IL-12) with glioblastoma tropism and evaluated the therapeutic effects of anti-PD-1, MSC_IL-12, and their combination against glioblastoma. METHODS: Therapeutic responses were evaluated using an immunocompetent mouse orthotopic model. Tumor-infiltrating lymphocytes (TILs) were analyzed using immunofluorescent imaging. Single-cell transcriptome was obtained from mouse brains after treatments. RESULTS: Anti-PD-1 and MSC_IL-12 showed complete tumor remission in 25.0% (4/16) and 23.1% (3/13) of glioblastoma-implanted mice, respectively, and their combination yielded synergistic antitumor efficacy indicated by 50.0% (6/12) of complete tumor remission. Analyses of TILs revealed that anti-PD-1 increased CD8+ T cells, while MSC_IL-12 led to infiltration of CD4+ T cells and NK cells. Both therapies reduced frequencies of Tregs. All these aspects observed in each monotherapy group were superimposed in the combination group. Notably, no tumor growth was observed upon rechallenge in cured mice, indicating long-term immunity against glioblastoma provoked by the therapies. Single-cell RNA-seq data confirmed these results and revealed that the combined treatment led to immune-favorable tumor microenvironment-CD4+, CD8+ T cells, effector memory T cells, and activated microglia were increased, whereas exhausted T cells, Tregs, and M2 polarized microglia were reduced. CONCLUSION: Anti-PD-1 and MSC_IL-12 monotherapies show long-term therapeutic responses, and their combination further enhances antitumor efficacy against glioblastoma via inducing immune-favorable tumor microenvironment.


Sujet(s)
Glioblastome , Cellules souches mésenchymateuses , Animaux , Souris , Glioblastome/anatomopathologie , Lymphocytes T CD8+ , Récepteur-1 de mort cellulaire programmée , Immunothérapie/méthodes , Interleukine-12 , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Cellules souches mésenchymateuses/anatomopathologie , Microenvironnement tumoral
19.
Oncogene ; 43(18): 1353-1368, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38459120

RÉSUMÉ

Undifferentiated pleomorphic sarcoma (UPS) is a highly aggressive malignant soft tissue tumor with a poor prognosis; however, the identity and heterogeneity of tumor populations remain elusive. Here, eight major cell clusters were identified through the RNA sequencing of 79,569 individual cells of UPS. UPS originates from mesenchymal stem cells (MSCs) and features undifferentiated subclusters. UPS subclusters were predicted to exist in two bulk RNA datasets, and had a prognostic value in The Cancer Genome Atlas (TCGA) dataset. The functional heterogeneity of malignant UPS cells and the immune microenvironment were characterized. Additionally, the fused cells were innovatively detected by expressing both monocyte/macrophage markers and other subcluster-associated genes. Based on the ligand-receptor interaction analysis, cellular interactions with epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) were abundant. Furthermore, 73% of patients with UPS (48/66) showed positive EGFR expression, which was associated with a poor prognosis. EGFR blockade with cetuximab inhibited tumor growth in a patient-derived xenograft model. Our transcriptomic studies delineate the landscape of UPS intratumor heterogeneity and serve as a foundational resource for further discovery and therapeutic exploration.


Sujet(s)
Récepteurs ErbB , Sarcomes , Analyse sur cellule unique , Humains , Animaux , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Souris , Sarcomes/anatomopathologie , Sarcomes/génétique , Pronostic , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Cétuximab/pharmacologie , Cétuximab/usage thérapeutique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Lignée cellulaire tumorale , Femelle , Régulation de l'expression des gènes tumoraux
20.
Exp Hematol ; 133: 104212, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38552942

RÉSUMÉ

Extensive research over the past 50 years has resulted in significant improvements in survival for patients diagnosed with leukemia. Despite this, a subgroup of patients harboring high-risk genetic alterations still suffer from poor outcomes. There is a desperate need for new treatments to improve survival, yet consistent failure exists in the translation of in vitro drug development to clinical application. Preclinical screening conventionally utilizes tumor cell monocultures to assess drug activity; however, emerging research has acknowledged the vital role of the tumor microenvironment in treatment resistance and disease relapse. Current co-culture drug screening methods frequently employ fibroblasts as the designated stromal cell component. Alternative stromal cell types that are known to contribute to chemoresistance are often absent in preclinical evaluations of drug efficacy. This review highlights mechanisms of chemoresistance by a range of different stromal constituents present in the bone marrow microenvironment. Utilizing an array of stromal cell types at the early stages of drug screening may enhance the translation of in vitro drug development to clinical use. Ultimately, we highlight the need to consider the bone marrow microenvironment in drug screening platforms for leukemia to develop superior therapies for the treatment of high-risk patients with poor prognostic outcomes.


Sujet(s)
Leucémies , Microenvironnement tumoral , Humains , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Leucémies/anatomopathologie , Leucémies/traitement médicamenteux , Tests de criblage d'agents antitumoraux/méthodes , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Résistance aux médicaments antinéoplasiques , Animaux , Moelle osseuse/anatomopathologie , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/métabolisme , Cellules stromales/anatomopathologie , Cellules stromales/métabolisme , Cellules stromales/effets des médicaments et des substances chimiques , Techniques de coculture , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE