Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 199
Filtrer
1.
J Bone Oncol ; 47: 100620, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39072049

RÉSUMÉ

Therapy-related myeloid neoplasms (t-MN) are a growing concern due to the continued use of cytotoxic therapies to treat malignancies. Cytotoxic therapies have been shown to drive therapy-induced senescence in normal tissues, including in the bone marrow microenvironment (BMME), which plays a crucial role in supporting normal hematopoiesis. This review examines recent work that focuses on the contribution of BMME senescence to t-MN pathogenesis, as well as offers a perspective on potential opportunities for therapeutic intervention.

2.
Stem Cell Res Ther ; 15(1): 178, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38886845

RÉSUMÉ

BACKGROUND: Bone marrow stromal cells (BMSCs) are highly heterogeneous, which may reflect their diverse biological functions, including tissue maintenance, haematopoietic support and immune control. The current understanding of the mechanisms that drive the onset and resolution of heterogeneity, and how BMSCs influence other cells in their environment is limited. Here, we determined how the secretome and importantly the extracellular matrix of BMSCs can influence cellular phenotype. METHODS: We used two immortalised clonal BMSC lines isolated from the same heterogeneous culture as model stromal subtypes with distinct phenotypic traits; a multipotent stem-cell-like stromal line (Y201) and a nullipotent non-stem cell stromal line (Y202), isolated from the same donor BMSC pool. Label-free quantitative phase imaging was used to track cell morphology and migration of the BMSC lines over 96 h in colony-forming assays. We quantified the secreted factors of each cell line by mass spectrometry and confirmed presence of proteins in human bone marrow by immunofluorescence. RESULTS: Transfer of secreted signals from a stem cell to a non-stem cell resulted in a change in morphology and enhanced migration to more closely match stem cell-like features. Mass spectrometry analysis revealed a significant enrichment of extracellular matrix (ECM) proteins in the Y201 stem cell secretome compared to Y202 stromal cells. We confirmed that Y201 produced a more robust ECM in culture compared to Y202. Growth of Y202 on ECM produced by Y201 or Y202 restored migration and fibroblastic morphology, suggesting that it is the deficiency of ECM production that contributes to its phenotype. The proteins periostin and aggrecan, were detected at 71- and 104-fold higher levels in the Y201 versus Y202 secretome and were subsequently identified by immunofluorescence at rare sites on the endosteal surfaces of mouse and human bone, underlying CD271-positive stromal cells. These proteins may represent key non-cellular components of the microenvironment for bona-fide stem cells important for cell maintenance and phenotype in vivo. CONCLUSIONS: We identified plasticity in BMSC morphology and migratory characteristics that can be modified through secreted proteins, particularly from multipotent stem cells. Overall, we demonstrate the importance of specific ECM proteins in co-ordination of cellular phenotype and highlight how non-cellular components of the BMSC microenvironment may provide insights into cell population heterogeneity and the role of BMSCs in health and disease.


Sujet(s)
Matrice extracellulaire , Cellules souches mésenchymateuses , Phénotype , Humains , Matrice extracellulaire/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Mouvement cellulaire , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Cellules stromales/métabolisme , Cellules stromales/cytologie , Lignée cellulaire
3.
Cells ; 13(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38727310

RÉSUMÉ

Fibrous dysplasia (FD) is a mosaic skeletal disorder caused by somatic activating variants of GNAS encoding for Gαs and leading to excessive cyclic adenosine monophosphate signaling in bone-marrow stromal cells (BMSCs). The effect of Gαs activation in the BMSC transcriptome and how it influences FD lesion microenvironment are unclear. We analyzed changes induced by Gαs activation in the BMSC transcriptome and secretome. RNAseq analysis of differential gene expression of cultured BMSCs from patients with FD and healthy volunteers, and from an inducible mouse model of FD, was performed, and the transcriptomic profiles of both models were combined to build a robust FD BMSC genetic signature. Pathways related to Gαs activation, cytokine signaling, and extracellular matrix deposition were identified. To assess the modulation of several key secreted factors in FD pathogenesis, cytokines and other factors were measured in culture media. Cytokines were also screened in a collection of plasma samples from patients with FD, and positive correlations of several cytokines to their disease burden score, as well as to one another and bone turnover markers, were found. These data support the pro-inflammatory, pro-osteoclastic behavior of FD BMSCs and point to several cytokines and other secreted factors as possible therapeutic targets and/or circulating biomarkers for FD.


Sujet(s)
Dysplasie fibreuse des os , Cellules souches mésenchymateuses , Transcriptome , Humains , Animaux , Cellules souches mésenchymateuses/métabolisme , Transcriptome/génétique , Souris , Dysplasie fibreuse des os/génétique , Dysplasie fibreuse des os/métabolisme , Dysplasie fibreuse des os/anatomopathologie , Mâle , Femelle , Cytokines/métabolisme , Sous-unités alpha Gs des protéines G/métabolisme , Sous-unités alpha Gs des protéines G/génétique , Adulte , Adulte d'âge moyen
4.
J Med Virol ; 96(5): e29659, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38747016

RÉSUMÉ

Hepatitis B virus (HBV) infection is a major global health burden with 820 000 deaths per year. In our previous study, we found that the knockdown of autophagy-related protein 5 (ATG5) significantly upregulated the interferon-stimulated genes (ISGs) expression to exert the anti-HCV effect. However, the regulation of ATG5 on HBV replication and its underlying mechanism remains unclear. In this study, we screened the altered expression of type I interferon (IFN-I) pathway genes using RT² Profiler™ PCR array following ATG5 knock-down and we found the bone marrow stromal cell antigen 2 (BST2) expression was significantly increased. We then verified the upregulation of BST2 by ATG5 knockdown using RT-qPCR and found that the knockdown of ATG5 activated the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway. ATG5 knockdown or BST2 overexpression decreased Hepatitis B core Antigen (HBcAg) protein, HBV DNA levels in cells and supernatants of HepAD38 and HBV-infected NTCP-HepG2. Knockdown of BST2 abrogated the anti-HBV effect of ATG5 knockdown. Furthermore, we found that ATG5 interacted with BST2, and further formed a ternary complex together with HBV-X (HBx). In conclusion, our finding indicates that ATG5 promotes HBV replication through decreasing BST2 expression and interacting with it directly to antagonize its antiviral function.


Sujet(s)
Antigènes CD , Protéine-5 associée à l'autophagie , Antigène stromal-2 de la moëlle osseuse , Protéines liées au GPI , Virus de l'hépatite B , Réplication virale , Humains , Antigènes CD/génétique , Antigènes CD/métabolisme , Protéine-5 associée à l'autophagie/génétique , Protéine-5 associée à l'autophagie/métabolisme , Techniques de knock-down de gènes , Protéines liées au GPI/métabolisme , Protéines liées au GPI/génétique , Cellules HepG2 , Hépatite B/virologie , Hépatite B/génétique , Virus de l'hépatite B/physiologie , Virus de l'hépatite B/génétique , Interactions hôte-pathogène , Transduction du signal , Antigène stromal-2 de la moëlle osseuse/métabolisme
5.
Med ; 5(5): 432-444.e4, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38547868

RÉSUMÉ

BACKGROUND: Ischemic stroke is one of the leading causes of death and neurological disability worldwide, and stem cell therapy is highly expected to reverse the sequelae. This phase 1/2, first-in-human study evaluated the safety, feasibility, and monitoring of an intracerebral-transplanted magnetic resonance imaging (MRI)-trackable autologous bone marrow stromal cell (HUNS001-01) for patients with subacute ischemic stroke. METHODS: The study included adults with severe disability due to ischemic stroke. HUNS001-01 cultured with human platelet lysates and labeled with superparamagnetic iron oxide was stereotactically transplanted into the peri-infarct area 47-64 days after ischemic stroke onset (dose: 2 or 5 × 107 cells). Neurological and radiographic evaluations were performed throughout 1 year after cell transplantation. The trial was registered at UMIN Clinical Trial Registry (number UMIN000026130). FINDINGS: All seven patients who met the inclusion criteria successfully achieved cell expansion, underwent intracerebral transplantation, and completed 1 year of follow-up. No product-related adverse events were observed. The median National Institutes of Health Stroke Scale and modified Rankin scale scores before transplantation were 13 and 4, which showed improvements of 1-8 and 0-2, respectively. Cell tracking proved that the engrafted cells migrated toward the infarction border area 1-6 months after transplantation, and the quantitative susceptibility mapping revealed that cell signals at the migrated area constantly increased throughout the follow-up period up to 34% of that of the initial transplanted site. CONCLUSIONS: Intracerebral transplantation of HUNS001-01 was safe and well tolerated. Cell tracking shed light on the therapeutic mechanisms of intracerebral transplantation. FUNDING: This work was supported by the Japan Agency for Medical Research and Development (AMED; JP17bk0104045 and JP20bk0104011).


Sujet(s)
Accident vasculaire cérébral ischémique , Imagerie par résonance magnétique , Humains , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Accident vasculaire cérébral ischémique/thérapie , Accident vasculaire cérébral ischémique/imagerie diagnostique , Accident vasculaire cérébral ischémique/chirurgie , Transplantation autologue/méthodes , Transplantation de cellules souches mésenchymateuses/méthodes , Résultat thérapeutique , Adulte , Études de faisabilité
6.
Heliyon ; 10(3): e25050, 2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38322864

RÉSUMÉ

Background: Bone-marrow-derived mesenchymal stromal (stem) cells [also called MSC(M)] and their extracellular vesicles (EVs) are considered a potentially innovative form of therapy for traumatic brain injury (TBI). Nevertheless, their application to TBI particularly remains preclinical, and the effects of these cells remain unclear and controversial. Therefore, an updated meta-analysis of preclinical studies is necessary to assess the effectiveness of MSC(M) and MSC(M) derived EVs in clinical trials. Methods: The following databases were searched (to December 2022): PubMed, Web of Science, and Embase. In this study, we measured functional outcomes based on the modified neurological severity score (mNSS), cognitive outcomes based on the Morris water maze (MWM), and histopathological outcomes based on lesion volume. A random effects meta-analysis was conducted to evaluate the effect of mNSS, MWM, and lesion volume. Results: A total of 2163 unique records were identified from our search, with Fifty-five full-text articles satisfying inclusion criteria. A mean score of 5.75 was assigned to the studies' quality scores, ranging from 4 to 7. MSC(M) and MSC(M) derived EVs had an overall positive effect on the mNSS score and MWM with SMDs -2.57 (95 % CI -3.26; -1.88; p < 0.01) and - 2.98 (95 % CI -4.21; -1.70; p < 0.01), respectively. As well, MSC(M) derived EVs were effective in reducing lesion volume by an SMD of - 0.80 (95 % CI -1.20; -0.40; p < 0.01). It was observed that there was significant variation among the studies, but further analyses could not determine the cause of this heterogeneity. Conclusions: MSC(M) and MSC(M) derived EVs are promising treatments for TBI in pre-clinical studies, and translation to the clinical domain appears warranted. Besides, large-scale trials in animals and humans are required to support further research due to the limited sample size of MSC(M) derived EVs.

7.
Biomaterials ; 306: 122471, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38377846

RÉSUMÉ

Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Cellules souches mésenchymateuses , Humains , Souris , Animaux , Enfant , Ostéogenèse , Moelle osseuse , Cellules stromales , Différenciation cellulaire , Cellules de la moelle osseuse , Cellules cultivées
8.
Am J Physiol Renal Physiol ; 326(2): F167-F177, 2024 02 01.
Article de Anglais | MEDLINE | ID: mdl-37969103

RÉSUMÉ

This study aimed to investigate the role of bone marrow stromal cell antigen-1 (Bst1; also known as CD157) in acute kidney injury (AKI). Bst1 is a cell surface molecule with various enzymatic activities and downstream intracellular signaling pathways that modulate the immune response. Previous research has linked Bst1 to diseases such as ovarian cancer, Parkinson's disease, and rheumatoid arthritis. We used bilateral ischemia-reperfusion injury (IRI) as an AKI model and created bone marrow chimeric mice to evaluate the role of Bst1 in bone marrow-derived cells. We also used flow cytometry to identify Bst1/CD157 expression in hematopoietic cells and evaluate immune cell dynamics in the kidney. The findings showed that Bst1-deficient (Bst1-/-) mice were protected against renal bilateral IRI. Bone marrow chimera experiments revealed that Bst1 expression on hematopoietic cells, but not parenchymal cells, induced renal IRI. Bst1 was mainly found in B cells and neutrophils by flow cytometry of the spleen and bone marrow. In vitro, migration of neutrophils from Bst1-/- mice was suppressed, and adoptive transfer of neutrophils from wild-type Bst1+/+ mice abolished the renal protective effect in Bst1 knockout mice. In conclusion, the study demonstrated that Bst1-/- mice are protected against renal IRI and that Bst1 expression in neutrophils plays a crucial role in inducing renal IRI. These findings suggest that targeting Bst1 in neutrophils could be a potential therapeutic strategy for AKI.NEW & NOTEWORTHY Acute kidney injury (AKI), a serious disease for which there is no effective Federal Drug Administration-approved treatment, is associated with high mortality rates. Bone marrow stromal cell antigen-1 (Bst1) is a cell surface molecule that can cause kidney fibrosis, but its role in AKI is largely unknown. Our study showed that Bst1-/- mice revealed a protective effect against renal bilateral ischemia-reperfusion injury (IRI). Adoptive transfer studies confirmed that Bst1 expression in hematopoietic cells, especially neutrophils, contributed to renal bilateral IRI.


Sujet(s)
Atteinte rénale aigüe , Cellules souches mésenchymateuses , Lésion d'ischémie-reperfusion , Souris , Animaux , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/prévention et contrôle , Rein/métabolisme , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/prévention et contrôle , Granulocytes neutrophiles/métabolisme , Souris knockout , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL
9.
Genome Med ; 15(1): 66, 2023 09 04.
Article de Anglais | MEDLINE | ID: mdl-37667405

RÉSUMÉ

BACKGROUND: Human bone marrow stromal cells (BMSCs) are an easily accessible and expandable progenitor population with the capacity to generate neural cell types in addition to mesoderm. Lineage tracing studies in transgenic animals have indicated Nestin + BMSCs to be descended from the truncal neural crest. Single-cell analysis provides a means to identify the developmental origin and identity of human BMSC-derived neural progenitors when lineage tracing remains infeasible. This is a prerequisite towards translational application. METHODS: We attained transcriptomic profiles of embryonic long bone, adult human bone marrow, cultured BMSCs and BMSC-derived neurospheres. Integrated scRNAseq analysis was supplemented by characterization of cells during culture expansion and following provision of growth factors and signalling agonists to bias lineage. RESULTS: Reconstructed pseudotime upon the integrated dataset indicated distinct neural and osteogenic differentiation trajectories. The starting state towards the neural differentiation trajectory consisted of Nestin + /MKI67 + BMSCs, which could also be diverted towards the osteogenic trajectory via a branch point. Nestin + /PDGFRA + BMSCs responded to neurosphere culture conditions to generate a subpopulation of cells with a neuronal phenotype according to marker expression and gene ontogeny analysis that occupied the end state along the neural differentiation trajectory. Reconstructed pseudotime also revealed an upregulation of BMP4 expression during culture of BMSC-neurospheres. This provided the rationale for culture supplementation with the BMP signalling agonist SB4, which directed progenitors to upregulate Pax6 and downregulate Nestin. CONCLUSIONS: This study suggested BMSCs originating from truncal neural crest to be the source of cells within long bone marrow possessing neural differentiation potential. Unravelling the transcriptomic dynamics of BMSC-derived neural progenitors promises to enhance differentiation efficiency and safety towards clinical application in cell therapy and disease modelling.


Sujet(s)
Moelle osseuse , Médecine régénérative , Adulte , Animaux , Humains , Nestine/génétique , Ostéogenèse , Neurones
10.
ACS Biomater Sci Eng ; 9(10): 5772-5781, 2023 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-37734919

RÉSUMÉ

The use of bone marrow stromal cells (BMSCs) for bone defect repair has shown great promise due to their differentiation potential. However, isolating the BMSCs from various cell types within the bone marrow remains challenging. To tackle this issue, we utilized semiconducting polymer dots (Pdots) as markers to select the BMSCs within a specific time frame. The therapeutic efficacy of the obtained Pdot-labeled BMSCs was assessed in a bone defect model. Initially, we evaluated the binding capacity of the Pdots with four different types of cells present in the bone marrow including BMSCs, osteoblasts, macrophages, and vascular endothelial cells, in vitro. Notably, BMSCs showed the most rapid uptake of the Pdots, being labeled within only one h of coculture, while other cells took four h to become labeled. Moreover, by colocalizing the Pdots with Prrx1, Sca-1, OSX, F480, and CD105 in the bone marrow cells of monocortical tibial defect (MTD) mice in vivo, we determined the proportions of BMSCs, macrophages, and vascular endothelial cells among all labeled cells from 1 to 8 h after the Pdots injection. It was found that BMSCs have the highest proportion (92%) among all labeled cells extracted after 1 h of Pdots injection. The therapeutic efficacy of the obtained Pdots-labeled BMSCs (1 h) was assessed in a bone defect model. Results showed that the new bone accrual was significantly increased in the treatment of Pdots-labeled BMSCs compared to the bone marrow cell-treated group. Our study revealed that BMSCs screened by the Pdots could improve bone defect repair, suggesting a promising application of the Pdots in bone healing.

11.
Front Bioeng Biotechnol ; 11: 1198545, 2023.
Article de Anglais | MEDLINE | ID: mdl-37496851

RÉSUMÉ

This study aims to present a sustainably releasing system of exosomes-fibrin combinate loaded on tantalum-coating titanium implants. We hope to investigate potential effects of the system on osseointegration between tantalum coating titanium implants and its surrounding bone tissue. Exosomes derived from rabbit bone marrow stromal cells (rBMSCs) and fibrin were deposited onto the micro-nanostructure tantalum coating surface (Ta + exo + FI) and compared to control groups, including tantalum coating (Ta), tantalum coating loaded exosomes (Ta + exo) and tantalum coating loaded fibrin (Ta + FI). The optimal concentration of loading exosomes, exosomes uptake capacity by BMSCs, and the effect of controlled-release by fibrin were assessed by laser scanning confocal microscope (LCSM) and microplate reader. The optimal concentration of exosomes was 1 µg/µL. Adhesion, proliferation, and osteogenic differentiation ability of BMSCs on different materials were assessed in vitro. Finally, osseointegrative capacity of Ta, Ta + exo, Ta + FI, Ta + exo + FI implants in rabbit tibia were respectively evaluated with histology and bone-implant contact ratio (BIC%). It was demonstrated that exosome sustained-release system with fibrin loading on the tantalum coating was successfully established. Fibrin contribute to exosomes release extension from 2d to 6d. Furthermore, Ta + exo + FI significantly promoted adhesion, proliferation, and osteogenic differentiation of BMSCs. In vivo, the implants in Ta + exo + FI group displayed the highest osseointegrative capability than those in other groups. It is concluded that this exosome delivery system on the implants may be an effective way for tantalum coating titanium implants to promote osseointegration between implant and its surrounding bone tissue.

12.
J Vet Sci ; 24(3): e37, 2023 May.
Article de Anglais | MEDLINE | ID: mdl-37271505

RÉSUMÉ

BACKGROUND: Toll-like receptor (TLR) agonists have been used as adjuvants to modulate immune responses in both animals and humans. OBJECTIVES: The objective of this study was to evaluate the combined effects of the TLR 4 agonist monophosphoryl lipid A (MPL) and the TLR 3 agonist polyinosinic:polycytidylic acid (Poly I:C) on equine peripheral blood mononuclear cells (PBMCs), monocyte-derived dendritic cells (MoDCs), and bone marrow-derived mesenchymal stromal cells (BM-MSCs). METHODS: The PBMCs, MoDCs, and BM-MSCs collected from three mixed breed horses were treated with MPL, Poly I:C, and their combination. The mRNA expression of interferon gamma (IFN-γ), interleukin (IL)-1ß, IL-4, IL-6, IL-8, IL-12p40, tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor (VEGF), and monocyte chemoattractant protein-1 (MCP-1) was determined using real-time polymerase chain reaction. RESULTS: The combination of MPL and Poly I:C significantly upregulated immunomodulatory responses in equine cells/ without cytotoxicity. The combination induced greater mRNA expression of pro-inflammatory cytokines IFN-γ and IL-6 than MPL or Poly I:C stimulation alone in PBMCs. In addition, the combination induced significantly higher mRNA expression of IL-1ß, IL-6, and IL-12p40 in MoDCs, and IL-8, MCP-1, and VEGF in BM-MSCs compared to stimulation with a single TLR agonist. CONCLUSIONS: The combination of MPL and Poly I:C can be used as a potential adjuvant candidate for vaccines to aid in preventing infectious diseases in horses.


Sujet(s)
Agranulocytes , Facteur de croissance endothéliale vasculaire de type A , Humains , Equus caballus/génétique , Animaux , Agranulocytes/métabolisme , Interleukine-6 , Sous-unité p40 de l'interleukine-12 , Interleukine-8 , Cytokines/génétique , Cytokines/métabolisme , Interféron gamma , Adjuvants immunologiques/pharmacologie , ARN messager , Poly I
13.
Genomics ; 115(4): 110636, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37150230

RÉSUMÉ

Colorectal cancer (CRC) is the fourth most frequently diagnosed cancer worldwide. Bone marrow stromal cells (BMSCs) play an essential role in tumor development by secreting exosomes. Scavenger receptor class A member 5 (SCARA5) is a newly identified tumor suppressor. This study aimed to investigate the effects of BMSCs-derived exosomes (BMSCs-Exos) on CRC development and to explore their regulatory mechanisms. BMSCs-Exos showed an oval-shaped, bilayer membrane structure. BMSCs-Exos inhibited growth and motility of CRC cells, while BMSCs-Exos with SCARA5 knockdown significantly promoted cell proliferation and movement. Exosomal SCARA5 also effectively suppressed colorectal tumor growth in mouse xenografts. Further analysis revealed that exosomal SCARA5 inhibited the phosphorylation of protein kinase B and phosphoinositide 3-kinase in both CRC cells and tumors. In conclusion, SCARA5 in BMSCs-Exos inhibited CRC progression by inactivating PI3K/Akt, thus suggesting the potential clinical application of SCARA5-containing BMSCs-Exos for CRC treatment.


Sujet(s)
Tumeurs colorectales , Exosomes , Cellules souches mésenchymateuses , microARN , Humains , Souris , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Exosomes/métabolisme , Tumeurs colorectales/métabolisme , Prolifération cellulaire , Cellules souches mésenchymateuses/métabolisme , microARN/métabolisme , Récepteurs éboueurs de classe A/métabolisme
14.
Front Oncol ; 13: 1155621, 2023.
Article de Anglais | MEDLINE | ID: mdl-37091139

RÉSUMÉ

Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells in the bone marrow (BM). MM remains an incurable disease, with the majority of patients experiencing multiple relapses from different drugs. The MM tumor microenvironment (TME) and in particular bone-marrow stromal cells (BMSCs) play a crucial role in the development of drug resistance. Metabolic reprogramming is emerging as a hallmark of cancer that can potentially be exploited for cancer treatment. Recent studies show that metabolism is further adjusted in MM cells during the development of drug resistance. However, little is known about the role of BMSCs in inducing metabolic changes that are associated with drug resistance. In this Perspective, we summarize current knowledge concerning the metabolic reprogramming of MM, with a focus on those changes associated with drug resistance to the proteasome inhibitor Bortezomib (BTZ). In addition, we present proof-of-concept fluxomics (glucose isotope-tracing) and Seahorse data to show that co-culture of MM cells with BMSCs skews the metabolic phenotype of MM cells towards a drug-resistant phenotype, with increased oxidative phosphorylation (OXPHOS), serine synthesis pathway (SSP), TCA cycle and glutathione (GSH) synthesis. Given the crucial role of BMSCs in conveying drug resistance, insights into the metabolic interaction between MM and BMSCs may ultimately aid in the identification of novel metabolic targets that can be exploited for therapy.

15.
Cell Stem Cell ; 30(4): 378-395.e8, 2023 04 06.
Article de Anglais | MEDLINE | ID: mdl-37028404

RÉSUMÉ

Hematopoietic stem cell (HSC) self-renewal and aging are tightly regulated by paracrine factors from the bone marrow niche. However, whether HSC rejuvenation could be achieved by engineering a bone marrow niche ex vivo remains unknown. Here, we show that matrix stiffness fine-tunes HSC niche factor expression by bone marrow stromal cells (BMSCs). Increased stiffness activates Yap/Taz signaling to promote BMSC expansion upon 2D culture, which is largely reversed by 3D culture in soft gelatin methacrylate hydrogels. Notably, 3D co-culture with BMSCs promotes HSC maintenance and lymphopoiesis, reverses aging hallmarks of HSCs, and restores their long-term multilineage reconstitution capacity. In situ atomic force microscopy analysis reveals that mouse bone marrow stiffens with age, which correlates with a compromised HSC niche. Taken together, this study highlights the biomechanical regulation of the HSC niche by BMSCs, which could be harnessed to engineer a soft bone marrow niche for HSC rejuvenation.


Sujet(s)
Moelle osseuse , Cellules souches mésenchymateuses , Animaux , Souris , Moelle osseuse/métabolisme , Rajeunissement , Cellules souches hématopoïétiques/métabolisme , Techniques de coculture , Cellules souches mésenchymateuses/métabolisme , Niche de cellules souches
16.
J Exerc Rehabil ; 19(1): 11-18, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36910676

RÉSUMÉ

The purpose of this study was to investigate the time-dependent alteration in whether concurrent aerobic exercise and bone marrow stromal cell (BMSC) engraftment could regulate myogenic differentiation-related signaling pathway in the soleus up to 35 days after sciatic nerve injury (SNI). The rats were divided as follows: the normal control (CON, n=5), sedentary group (SED, n=20), treadmill exercise group (TEX, n=20), BMSC transplantation group (BMSC, n=20), TEX+BMSC transplantation group (TEX+BMSC, n=20) 7, 14, 21, and 35 days after SNI. SNI was applied into the thigh and treadmill exercise was comprised of walking at a speed of 4 to 8 m/min for 30 min once a day. Harvested BMSC at a density of 5×106 in 50-µL phosphate-buff-ered saline was injected into the injury site. Phosphorylated (p) extracellular signal-regulated kinase 1/2 expression was dramatically upregulated in BMSC and BMSC+EX groups from 21 days after SNI compared to those in the SED group. P-ribosomal s6 kinase (RSK) was sharply increased 14 days later, and then rapidly downregulated from day 21, whereas TEX, BMSC and TEX+ BMSC groups significantly kept up expression levels of p-RSK until 35 days post injury than SED group. TEX+BMSC group significantly increased activation of protein kinase B-mammalian target of rapamycin in the soleus from day 14 and myoblast determination protein 1-myogen-in pathways was activated in TEX+BMSC group from day 21. Present findings provide information that combined intervention of aerobic exercise and BMSC transplantation might be a reliable therapeutic strategy for overcoming the morphological and functional problems in denervated soleus muscle.

17.
Tissue Eng Part B Rev ; 29(2): 103-122, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36066333

RÉSUMÉ

Bone marrow stromal cells (BMSCs) and periosteum-derived cells (PDCs) represent promising skeletal stem cell sources to treat critical-size bone defects. However, the large number of preclinical tests with a variety of in vivo data complicates the selection of cells for further clinical translation. This systematic review aims to analyze the in vivo bone-forming efficacy of BMSCs- and PDCs-based approaches in all published preclinical experiments until November 2020. For this purpose, four databases (PubMed, Embase, Cochrane Central Register of Controlled Trial, and Web of Science) were searched for eligible literature, which yielded a total of 94 full-text articles for systematic review. This review generated an evidence-based list of BMSC- or PDC-based approaches, which have been evaluated for bone formation in different animal models. Among them, 74 studies were included for pairwise and network meta-analysis. The results revealed that both PDC and BMSC had beneficial bone-forming efficacy compared to bare scaffold. In addition, BMSC- and PDC-based approaches had no significant difference regarding in vivo bone-forming efficacy. However, BMSC-based approach had a higher probability to be ranked better than PDC-based approach. Furthermore, the review discusses (i) the possible risk of bias of the in vivo evaluation of cell-based approaches, (ii) the difficulty in replication of such experiments due to frequent poor reporting of the methods and results, and (iii) the clinical relevance of the currently utilized BMSC- and PDC-based approaches. Systematic review registration: The study was prospectively registered in PROSPERO, Registration No. CRD42021270922.


Sujet(s)
Cellules souches mésenchymateuses , Périoste , Animaux , Cellules de la moelle osseuse , Régénération osseuse , Modèles animaux , Ostéogenèse
18.
J Biomed Mater Res A ; 111(1): 106-117, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36194510

RÉSUMÉ

The properties and structure of the cellular microenvironment can influence cell behavior. Sites of cell adhesion to the extracellular matrix (ECM) initiate intracellular signaling that directs cell functions such as proliferation, differentiation, and apoptosis. Electrospun fibers mimic the fibrous nature of native ECM proteins and cell culture in fibers affects cell shape and dimensionality, which can drive specific functions, such as the osteogenic differentiation of primary human bone marrow stromal cells (hBMSCs), by. In order to probe how scaffolds affect cell shape and behavior, cell-fiber contacts were imaged to assess their shape and dimensionality through a novel approach. Fluorescent polymeric fiber scaffolds were made so that they could be imaged by confocal fluorescence microscopy. Fluorescent polymer films were made as a planar control. hBSMCs were cultured on the fluorescent substrates and the cells and substrates were imaged. Two different image analysis approaches, one having geometrical assumptions and the other having statistical assumptions, were used to analyze the 3D structure of cell-scaffold contacts. The cells cultured in scaffolds contacted the fibers in multiple planes over the surface of the cell, while the cells cultured on films had contacts confined to the bottom surface of the cell. Shape metric analysis indicated that cell-fiber contacts had greater dimensionality and greater 3D character than the cell-film contacts. These results suggest that cell adhesion site-initiated signaling could emanate from multiple planes over the cell surface during culture in fibers, as opposed to emanating only from the cell's basal surface during culture on planar surfaces.


Sujet(s)
Cellules souches mésenchymateuses , Ostéogenèse , Humains , Structures d'échafaudage tissulaires/composition chimique , Différenciation cellulaire , Matrice extracellulaire/métabolisme , Cellules cultivées , Ingénierie tissulaire/méthodes , Cellules de la moelle osseuse
19.
Front Endocrinol (Lausanne) ; 13: 1001210, 2022.
Article de Anglais | MEDLINE | ID: mdl-36506047

RÉSUMÉ

Bone marrow adipocytes (BMAds) constitute the most abundant stromal component of adult human bone marrow. Two subtypes of BMAds have been described, the more labile regulated adipocytes (rBMAds) and the more stable constitutive adipocytes (cBMAds), which develop earlier in life and are more resilient to environmental and metabolic disruptions. In vivo, rBMAds are enriched in saturated fatty acids, contain smaller lipid droplets (LDs) and more readily provide hematopoietic support than their cBMAd counterparts. Mouse models have been used for BMAds research, but isolation of primary BMAds presents many challenges, and thus in vitro models remain the current standard to study nuances of adipocyte differentiation. No in vitro model has yet been described for the study of rBMAds/cBMAds. Here, we present an in vitro model of BM adipogenesis with differential rBMAd and cBMAd-like characteristics. We used OP9 BM stromal cells derived from a (C57BL/6xC3H)F2-op/op mouse, which have been extensively characterized as feeder layer for hematopoiesis research. We observed similar canonical adipogenesis transcriptional signatures for spontaneously-differentiated (sOP9) and induced (iOP9) cultures, while fatty acid composition and desaturase expression of Scd1 and Fads2 differed at the population level. To resolve differences at the single adipocyte level we tested Raman microspectroscopy and show it constitutes a high-resolution method for studying adipogenesis in vitro in a label-free manner, with resolution to individual LDs. We found sOP9 adipocytes have lower unsaturation ratios, smaller LDs and higher hematopoietic support than iOP9 adipocytes, thus functionally resembling rBMAds, while iOP9 more closely resembled cBMAds. Validation in human primary samples confirmed a higher unsaturation ratio for lipids extracted from stable cBMAd-rich sites (femoral head upon hip-replacement surgery) versus labile rBMAds (iliac crest after chemotherapy). As a result, the 16:1/16:0 fatty acid unsaturation ratio, which was already shown to discriminate BMAd subtypes in rabbit and rat marrow, was validated to discriminate cBMAds from rBMAd in both the OP9 model in vitro system and in human samples. We expect our model will be useful for cBMAd and rBMAd studies, particularly where isolation of primary BMAds is a limiting step.


Sujet(s)
Moelle osseuse , Gouttelettes lipidiques , Adulte , Humains , Souris , Rats , Animaux , Lapins , Souris de lignée C57BL , Acides gras , Modèles animaux de maladie humaine
20.
Int J Neurosci ; : 1-10, 2022 Dec 16.
Article de Anglais | MEDLINE | ID: mdl-36458531

RÉSUMÉ

OBJECTIVES: This study aimed to produce an acellular spinal cord scaffold-bone marrow stromal cell (ASCS-BMSC) complex in which the growth of BMSCs transplanted into the spinal cord of rats could be simulated in vitro, facilitating the observation and evaluation of the growth of BMSCs on the ASCS for the first time. METHODS: Freeze-thaw, chemical extraction and mechanical shaking approaches were used to remove the cellular components and prepare a rat ASCS containing only the extracellular matrix (ECM) structure from the rat spinal cord. BMSCs were embedded into ASCSs and freeze-dried agarose scaffolds (FASs), and cell migration and proliferation were observed via fluorescence microscopy and the MTT assay. RESULTS: Compared with the normal rat spinal cord, the ASCS had no cell structure and retained ECM components such as type IV collagen, fibronectin and laminin, showing a three-dimensional network structure with good voids. The growth and proliferation of BMSCs on the ASCS was good, as shown by the MTT assay. Scanning electron microscopy showed that BMSCs covered 65% of the ASCS surface, and the mitochondria of BMSCs were developed and adhered to collagen fibres, as demonstrated by transmission electron microscopy. HE staining showed that BMSCs could grow inside the ASCS, and immunohistochemical staining showed that BMSCs still expressed CD44 and CD90 on the ASCS and had stem cell characteristics. CONCLUSIONS: The results of the experiment indicate that the ASCS has the ability to improve cell adhesion and proliferation. Thus, the ASCS-BMSC combination may be used to treat spinal cord injury.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE