Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.906
Filter
1.
Cell Commun Signal ; 22(1): 257, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711089

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cells , Prostatic Hyperplasia , Transforming Growth Factor beta , rho-Associated Kinases , rho-Associated Kinases/metabolism , Male , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Prostate/pathology , Prostate/metabolism , Cell Movement , Mice , Stromal Cells/metabolism , Stromal Cells/pathology
2.
Nat Commun ; 15(1): 4139, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755155

ABSTRACT

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.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells , Multiple Myeloma , Tumor Microenvironment , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Humans , Tumor Microenvironment/genetics , Cell Line, Tumor , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Transcription, Genetic , Bone Marrow Cells/metabolism , Cell Movement/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Female , Male
3.
Am J Pathol ; 194(6): 1137-1153, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749609

ABSTRACT

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.


Subject(s)
Adenocarcinoma , Mesenchymal Stem Cells , Mice, Inbred BALB C , Receptor, ErbB-2 , Receptors, Estrogen , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Female , Humans , Receptor, ErbB-2/metabolism , Mice , Receptors, Estrogen/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Receptors, Progesterone/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/metabolism
4.
Exp Cell Res ; 438(1): 114032, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38583856

ABSTRACT

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.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplastic Stem Cells , Sequestosome-1 Protein , Single-Cell Analysis , Triple Negative Breast Neoplasms , Wnt Signaling Pathway , Humans , Epithelial-Mesenchymal Transition/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Wnt Signaling Pathway/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Female , Single-Cell Analysis/methods , Transcriptome/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , beta Catenin/metabolism , beta Catenin/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Tumor Microenvironment/genetics
5.
Cancer Res Commun ; 4(4): 1150-1164, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38598843

ABSTRACT

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.


Subject(s)
Cell Adhesion , Cell Proliferation , Mesenchymal Stem Cells , Multiple Myeloma , Humans , Multiple Myeloma/pathology , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Apoptosis , Coculture Techniques , Cell Line, Tumor , Cell Aggregation , Cell Survival
6.
J Bone Miner Res ; 39(2): 161-176, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38477740

ABSTRACT

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.


Subject(s)
Adipose Tissue , Mesenchymal Stem Cells , Synovial Membrane , Animals , Synovial Membrane/pathology , Synovial Membrane/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Mice , Osteoarthritis/pathology , Osteoarthritis/metabolism , Patella/pathology , Patella/metabolism
7.
Oncogene ; 43(18): 1353-1368, 2024 May.
Article in English | MEDLINE | ID: mdl-38459120

ABSTRACT

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.


Subject(s)
ErbB Receptors , Sarcoma , Single-Cell Analysis , Humans , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , Mice , Sarcoma/pathology , Sarcoma/genetics , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays , Cetuximab/pharmacology , Cetuximab/therapeutic use , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic
8.
Exp Hematol ; 133: 104212, 2024 May.
Article in English | MEDLINE | ID: mdl-38552942

ABSTRACT

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.


Subject(s)
Leukemia , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Leukemia/pathology , Leukemia/drug therapy , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Animals , Bone Marrow/pathology , Bone Marrow/drug effects , Bone Marrow/metabolism , Stromal Cells/pathology , Stromal Cells/metabolism , Stromal Cells/drug effects , Coculture Techniques , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology
9.
Biomed Pharmacother ; 173: 115790, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38431436

ABSTRACT

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.


Subject(s)
Glioblastoma , Mesenchymal Stem Cells , Animals , Mice , Glioblastoma/pathology , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Immunotherapy/methods , Interleukin-12 , Cell Line, Tumor , Disease Models, Animal , Mesenchymal Stem Cells/pathology , Tumor Microenvironment
10.
Sci Rep ; 14(1): 6649, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38503815

ABSTRACT

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.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Intestinal Pseudo-Obstruction , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice , Animals , Bone Marrow/pathology , Colitis/chemically induced , Mesenchymal Stem Cells/pathology , Inflammation , Anti-Inflammatory Agents/adverse effects , Disease Models, Animal
11.
J Neuroimmunol ; 389: 578313, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38401393

ABSTRACT

The clinical effect of human Mesenchymal stem cells (hMSCs) transplanted into EAE mice/MS patients is short lived due to poor survival of the transplanted cells. Since Granagard, a nanoformulation of pomegranate seed oil, extended the presence of Neuronal Stem cells transplanted into CJD mice brains, we tested whether this safe food supplement can also elongate the survival of hMSCs transplanted into EAE mice. Indeed, pathological studies 60 days post transplantation identified human cells only in brains of Granagard treated mice, concomitant with increased clinical activity. We conclude that Granagard may prolong the activity of stem cell transplantation in neurological diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Multiple Sclerosis , Humans , Animals , Mice , Multiple Sclerosis/therapy , Multiple Sclerosis/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Brain/pathology , Stem Cell Transplantation , Immunologic Factors , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology
12.
Oncol Res ; 32(3): 433-437, 2024.
Article in English | MEDLINE | ID: mdl-38361752

ABSTRACT

The main aim of antineoplastic treatment is to maximize patient benefit by augmenting the drug accumulation within affected organs and tissues, thus incrementing drug effects and, at the same time, reducing the damage of non-involved tissues to cytotoxic agents. Mesenchymal stromal cells (MSC) represent a group of undifferentiated multipotent cells presenting wide self-renewal features and the capacity to differentiate into an assortment of mesenchymal family cells. During the last year, they have been proposed as natural carriers for the selective release of antitumor drugs to malignant cells, thus optimizing cytotoxic action on cancer cells, while significantly reducing adverse side effects on healthy cells. MSC chemotherapeutic drug loading and delivery is an encouraging new area of cell therapy for several tumors, especially for those with unsatisfactory prognosis and limited treatment options available. Although some experimental models have been successfully developed, phase I clinical studies are needed to confirm this potential application of cell therapy, in particular in the case of primary and secondary lung cancers.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Mesenchymal Stem Cells , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Technology
13.
Pak J Biol Sci ; 27(1): 18-26, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38413394

ABSTRACT

<b>Background and Objective:</b> Liver fibrosis (LF) is a most common pathological process characterized by the activation of hepatocytes leading to the accumulation of extracellular matrix (ECM). Hypoxia precondition treated in MSCs (H-MSCs) could enhance their immunomodulatory and regeneration capability, through expressing robust anti-inflammatory cytokines and growth factors, known as H-MSCs secretome (SH-MSCs) that are critical for the improvement of liver fibrosis. However, the study regarding the efficacy and mechanism of action of SH-MSCs in ameliorating liver fibrosis is still inconclusive. In this study, the therapeutic potential and underlying mechanism for SH-MSCs in the treatment of liver fibrosis were investigated. <b>Materials and Methods:</b> A rat model with liver fibrosis induced by CCl<sub>4</sub> was created and maintained for 8 weeks. The rats received intravenous doses of SH-MSCs and secretome derived from normoxia MSCs (SN-MSCs), filtered using a tangential flow filtration (TFF) system with different molecular weight cut-off categories, both at a dosage of 0.5 mL. The ELISA assay was employed to examine the cytokines and growth factors present in both SH-MSCs and SN-MSCs. On the ninth day, the rats were euthanized and liver tissues were collected for subsequent histological examination and analysis of mRNA expression. <b>Results:</b> The ELISA test revealed that SH-MSCs exhibited higher levels of VEGF, PDGF, bFGF, IL-10, TGF-ß and IL-6 compared to SN-MSCs. <i>In vivo</i>, administration of SH-MSCs notably decreased mortality rates. It also demonstrated a reduction in liver fibrosis, collagen fiber areas, α-SMA positive staining and relative mRNA expression of TGF-ß. Conversely, SN-MSCs also contributed to liver fibrosis improvement, although SH-MSCs demonstrated more favorable outcomes. <b>Conclusion:</b> Current findings suggested that SH-MSCs could improve CCl<sub>4</sub>-induced liver fibrosis and decrease α-SMA and TGF-ß expression.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rats , Animals , Liver Regeneration , Secretome , Liver Cirrhosis/metabolism , Fibrosis , Hypoxia/metabolism , Hypoxia/pathology , Transforming Growth Factor beta/adverse effects , Transforming Growth Factor beta/metabolism , Disease Models, Animal , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , RNA, Messenger/metabolism
14.
Br J Pharmacol ; 181(2): 216-237, 2024 01.
Article in English | MEDLINE | ID: mdl-36609915

ABSTRACT

The bone marrow (BM) is the primary site of adult haematopoiesis, where stromal elements (e.g. fibroblasts and mesenchymal stem cells [MSCs]) work in concert to support blood cell development. However, the establishment of an abnormal clone can lead to a blood malignancy, such as acute myeloid leukaemia (AML). Despite our increased understanding of the pathophysiology of the disease, patient survival remains suboptimal, mainly driven by the development of therapy resistance. In this review, we highlight the importance of bone marrow fibroblasts and MSCs in health and acute myeloid leukaemia and their impact on patient prognosis. We discuss how stromal elements reduce the killing effects of therapies via a combination of contact-dependent (e.g. integrins) and contact-independent (i.e. secreted factors) mechanisms, accompanied by the establishment of an immunosuppressive microenvironment. Importantly, we underline the challenges of therapeutically targeting the bone marrow stroma to improve acute myeloid leukaemia patient outcomes, due to the inherent heterogeneity of stromal cell populations. LINKED ARTICLES: This article is part of a themed issue on Cancer Microenvironment and Pharmacological Interventions. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.2/issuetoc.


Subject(s)
Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Adult , Humans , Bone Marrow/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mesenchymal Stem Cells/pathology , Stromal Cells , Fibroblasts/pathology , Tumor Microenvironment , Bone Marrow Cells/pathology
15.
Asia Pac J Clin Oncol ; 20(1): 7-15, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37190944

ABSTRACT

Despite rapid advancement in research of diagnostics and therapeutics, cancer is the most dangerous disease-causing millions of deaths worldwide. Many of the conventional anticancer therapies can even lead to developing resistance to therapy and recurrence of cancer. To find a new, alternative treatment strategy for a variety of ailments scientists and researchers have turned their attention to cell therapies and regenerative medicine. Stem cells are now being researched for their extensive potential application in therapy for several incurable illnesses including cancer. One of the most often employed cell types for regenerative medicine is mesenchymal stem cells. Mesenchymal stem cells (MSCs) are considered a promising source of stem cells in personalized cell-based therapies. The inherent tumor tropic and immune-modulatory properties of MSCs can be used to target cancer cells. This review aims to focus on the anticancer properties of MSCs and their effect on different signaling pathways. Later on, we discuss the advantages of engineered MSCs over non-engineered MSCsin cancer therapy.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Regenerative Medicine
16.
Physiol Rev ; 104(2): 659-725, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37589393

ABSTRACT

Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.


Subject(s)
Heart Failure , Mesenchymal Stem Cells , Myocardial Infarction , Humans , Myocardial Infarction/therapy , Myocardial Infarction/pathology , Myocardium/metabolism , Cardiovascular Physiological Phenomena , Heart Failure/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology
17.
BJOG ; 131(6): 759-767, 2024 May.
Article in English | MEDLINE | ID: mdl-37492999

ABSTRACT

OBJECTIVE: To investigate the effects of an adjuvant allogenic umbilical cord mesenchymal stromal cell (UC-MSC) patch applied during fetal surgery on motor and sphincter function in the ovine MMC model. DESIGN: MMC defects were surgically created at 75 days of gestation and repaired 14 days later. POPULATION: Ovine MMC model: fetal lambs. METHODS: We compared lambs that received a UC-MSC patch with a control group of lambs that received an acellular patch. MAIN OUTCOME MEASURES: Clinical neurological assessment was performed at 2 and 24 hours of life and included determination of the Sheep Locomotor Rating scale (SLR), which has been validated in the ovine MMC model. Electrophysical examinations, spine scans and histological analyses were also performed. RESULTS: Of the 13 operated lambs, nine were born alive: five had of these had received a UC-MSC patch and four an acellular patch. At 24 hours of life, lambs in the UC-MSC group had a significantly higher score (14 versus 5, P = 0.04). Amyotrophy was significantly more common in the control group (75% versus 0%, P = 0.02). All the lambs in the control group and none of those in the UC-MSC group were incontinent. No significant differences were observed between the UC-MSC and control groups in terms of the presence of spontaneous EMG activity, nerve conduction or spinal evoked potentials. In the microscopic examination, lambs in the UC-MSC group had less fibrosis between the spinal cord and the dermis (mean thickness, 453 versus 3921 µm, P = 0.03) and around the spinal cord (mean thickness, 47 versus 158 µm, P < 0.001). Examination of the spinal cord in the area of the MMC defect showed a higher large neuron density in the UC-MSC group (14.5 versus 5.6 neurons/mm2, P < 0.001). No tumours were observed. CONCLUSIONS: Fetal repair of MMC using UC-MSC patches improves motor and sphincter function as well as spinal preservation and reduction of fibrosis.


Subject(s)
Meningomyelocele , Mesenchymal Stem Cells , Pregnancy , Female , Sheep , Animals , Humans , Meningomyelocele/surgery , Umbilical Cord , Spinal Cord/pathology , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Fibrosis
18.
Environ Res ; 244: 117783, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38048862

ABSTRACT

Although bone marrow mesenchymal stem cells (BM-MSCs)-derived exosomes have been reported to be closely associated with acute myeloid leukemia (AML) progression and chemo-resistance, but its detailed functions and molecular mechanisms have not been fully delineated. Besides, serum RNA m6A demethylase fat mass and obesity-associated protein (FTO)-containing exosomes are deemed as important indicators for cancer progression, and this study aimed to investigate the role of BM-MSCs-derived FTO-exosomes in regulating the malignant phenotypes of AML cells. Here, we verified that BM-MSCs-derived exosomes delivered FTO to promote cancer aggressiveness, stem cell properties and Cytosine arabinoside (Ara-C)-chemoresistance in AML cells, and the underlying mechanisms were also uncovered. Our data suggested that BM-MSCs-derived FTO-exo demethylated m6A modifications in the m6A-modified LncRNA GLCC1 to facilitate its combination with the RNA-binding protein Hu antigen R (HuR), which further increased the stability and expression levels of LncRNA GLCC1. In addition, LncRNA GLCC1 was verified as an oncogene to facilitate cell proliferation and enhanced Ara-C-chemoresistance in AML cells. Further experiments confirmed that demethylated LncRNA GLCC1 served as scaffold to facilitate the formation of the IGF2 mRNA binding protein 1 (IGF2BP1)-c-Myc complex, which led to the activation of the downstream tumor-promoting c-Myc-associated signal pathways. Moreover, our rescuing experiments validated that the promoting effects of BM-MSCs-derived FTO-exo on cancer aggressiveness and drug resistance in AML cells were abrogated by silencing LncRNA GLCC1 and c-Myc. Thus, the present firstly investigated the functions and underlying mechanisms by which BM-MSCs-derived FTO-exo enhanced cancer aggressiveness and chemo-resistance in AML by modulating the LncRNA GLCC1-IGF2BP1-c-Myc signal pathway, and our work provided novel biomarkers for the diagnosis, treatment and therapy of AML in clinic.


Subject(s)
Adenine/analogs & derivatives , Exosomes , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , RNA, Long Noncoding , Humans , Exosomes/metabolism , Exosomes/pathology , Drug Resistance, Neoplasm , RNA, Long Noncoding/metabolism , Leukemia, Myeloid, Acute/genetics , Cytarabine/pharmacology , Cytarabine/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Demethylation , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism
19.
Stem Cells Transl Med ; 13(2): 107-115, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38016185

ABSTRACT

Alcohol-associated liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) represent pathological conditions that include many distinct stages, potentially leading to the final stage of cirrhotic liver. To date, liver transplantation is the sole successful treatment with concomitant limitations related to donor organ shortage and the need of life-long immunosuppressive therapy. Recently, cell-based therapies for ALD and NAFLD have been proposed with mesenchymal stem/stromal cells (MSCs) as promising effectors. MSC therapeutic applications offer hepatoprotection, regulation of the inflammatory process and angiogenesis particularly in ALD and NAFLD pre-clinical disease models. Recent studies suggested that hepatospecific MSC-based therapies could benefit liver diseases by restoring liver function and decreasing inflammation and fibrosis. Similarly to solid-organ transplantation, limitations in MSC approaches include donor availability exacerbated by high number of cells and cell trapping into lungs. Herein, based on recent advances, we discuss the use of MSCs as a therapeutic approach for ALD and NAFLD and we provide the available information for the establishment of a framework toward a potential clinical application.


Subject(s)
Liver Diseases, Alcoholic , Mesenchymal Stem Cells , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Liver Diseases, Alcoholic/complications , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/therapy , Liver/pathology , Liver Cirrhosis/pathology , Mesenchymal Stem Cells/pathology
20.
Biomed Pharmacother ; 170: 116043, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38128186

ABSTRACT

Cancer stem cells are the key link between malignant tumor progression and drug resistance. This cell population has special properties that are different from those of conventional tumor cells, and the role of cancer stem cell-related exosomes in progression of tumor malignancy is becoming increasingly clear. Cancer stem cell-derived exosomes carry a variety of functional molecules involved in regulation of the microenvironment, especially with regard to immune cells, but how these exosomes exert their functions and the specific mechanisms need to be further clarified. Here, we summarize the role of cancer stem cell exosomes in regulating immune cells in detail, aiming to provide new insights for subsequent targeted drug development and clinical strategy formulation.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Mesenchymal Stem Cells/pathology , Tumor Microenvironment , Neoplasms/drug therapy , Immunotherapy , Neoplastic Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...