1
|
Wang R, Wang Y, Niu Y, He D, Jin S, Li Z, Zhu L, Chen L, Wu X, Ding C, Wu T, Shi X, Zhang H, Li C, Wang X, Xie Z, Li W, Liu Y. Deep Learning-Predicted Dihydroartemisinin Rescues Osteoporosis by Maintaining Mesenchymal Stem Cell Stemness through Activating Histone 3 Lys 9 Acetylation. ACS CENTRAL SCIENCE 2023; 9:1927-1943. [PMID: 37901168 PMCID: PMC10604014 DOI: 10.1021/acscentsci.3c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Indexed: 10/31/2023]
Abstract
Maintaining the stemness of bone marrow mesenchymal stem cells (BMMSCs) is crucial for bone homeostasis and regeneration. However, in vitro expansion and bone diseases impair BMMSC stemness, limiting its functionality in bone tissue engineering. Using a deep learning-based efficacy prediction system and bone tissue sequencing, we identify a natural small-molecule compound, dihydroartemisinin (DHA), that maintains BMMSC stemness and enhances bone regeneration. During long-term in vitro expansion, DHA preserves BMMSC stemness characteristics, including its self-renewal ability and unbiased differentiation. In an osteoporosis mouse model, oral administration of DHA restores the femur trabecular structure, bone density, and BMMSC stemness in situ. Mechanistically, DHA maintains BMMSC stemness by promoting histone 3 lysine 9 acetylation via GCN5 activation both in vivo and in vitro. Furthermore, the bone-targeted delivery of DHA by mesoporous silica nanoparticles improves its therapeutic efficacy in osteoporosis. Collectively, DHA could be a promising therapeutic agent for treating osteoporosis by maintaining BMMSC stemness.
Collapse
Affiliation(s)
- Ruoxi Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yu Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yuting Niu
- Central
Laboratory, National Center for Stomatology & National Clinical
Research Center for Oral Diseases & National Engineering Laboratory
for Digital and Material Technology of Stomatology & Beijing Key
Laboratory of Digital Stomatology & Research Center of Engineering
and Technology for Computerized Dentistry Ministry of Health &
NMPA Key Laboratory for Dental Materials & Translational Research
Center for Orocraniofacial Stem Cells and Systemic Health, Central
Laboratory, Peking University School and
Hospital for Stomatology, Beijing 100081, China
| | - Danqing He
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Shanshan Jin
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Zixin Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Lisha Zhu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Liyuan Chen
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xiaolan Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chengye Ding
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Tianhao Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xinmeng Shi
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - He Zhang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chang Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xin Wang
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Zhengwei Xie
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Weiran Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yan Liu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| |
Collapse
|
2
|
Giuliani A, Sabbatinelli J, Amatori S, Graciotti L, Silvestrini A, Matacchione G, Ramini D, Mensà E, Prattichizzo F, Babini L, Mattiucci D, Busilacchi EM, Bacalini MG, Espinosa E, Lattanzio F, Procopio AD, Olivieri F, Poloni A, Fanelli M, Rippo MR. MiR-422a promotes adipogenesis via MeCP2 downregulation in human bone marrow mesenchymal stem cells. Cell Mol Life Sci 2023; 80:75. [PMID: 36847916 PMCID: PMC9971129 DOI: 10.1007/s00018-023-04719-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 12/16/2022] [Accepted: 01/22/2023] [Indexed: 03/01/2023]
Abstract
Methyl-CpG binding protein 2 (MeCP2) is a ubiquitous transcriptional regulator. The study of this protein has been mainly focused on the central nervous system because alterations of its expression are associated with neurological disorders such as Rett syndrome. However, young patients with Rett syndrome also suffer from osteoporosis, suggesting a role of MeCP2 in the differentiation of human bone marrow mesenchymal stromal cells (hBMSCs), the precursors of osteoblasts and adipocytes. Here, we report an in vitro downregulation of MeCP2 in hBMSCs undergoing adipogenic differentiation (AD) and in adipocytes of human and rat bone marrow tissue samples. This modulation does not depend on MeCP2 DNA methylation nor on mRNA levels but on differentially expressed miRNAs during AD. MiRNA profiling revealed that miR-422a and miR-483-5p are upregulated in hBMSC-derived adipocytes compared to their precursors. MiR-483-5p, but not miR-422a, is also up-regulated in hBMSC-derived osteoblasts, suggesting a specific role of the latter in the adipogenic process. Experimental modulation of intracellular levels of miR-422a and miR-483-5p affected MeCP2 expression through direct interaction with its 3' UTR elements, and the adipogenic process. Accordingly, the knockdown of MeCP2 in hBMSCs through MeCP2-targeting shRNA lentiviral vectors increased the levels of adipogenesis-related genes. Finally, since adipocytes released a higher amount of miR-422a in culture medium compared to hBMSCs we analyzed the levels of circulating miR-422a in patients with osteoporosis-a condition characterized by increased marrow adiposity-demonstrating that its levels are negatively correlated with T- and Z-scores. Overall, our findings suggest that miR-422a has a role in hBMSC adipogenesis by downregulating MeCP2 and its circulating levels are associated with bone mass loss in primary osteoporosis.
Collapse
Affiliation(s)
- Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.,SOD Medicina di Laboratorio, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
| | - Stefano Amatori
- Department of Biomolecular Sciences, Molecular Pathology Laboratory "PaoLa", University of Urbino Carlo Bo, Fano, PU, Italy
| | - Laura Graciotti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.,Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Silvestrini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
| | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
| | - Deborah Ramini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Emanuela Mensà
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
| | | | - Lucia Babini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
| | - Domenico Mattiucci
- Section of Hematology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Elena Marinelli Busilacchi
- Section of Hematology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Maria Giulia Bacalini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Laboratorio Brain Aging, Bologna, Italy
| | - Emma Espinosa
- Geriatrics, Santa Croce Hospital, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Fano, Italy
| | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.,Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.,Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Antonella Poloni
- Section of Hematology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Mirco Fanelli
- Department of Biomolecular Sciences, Molecular Pathology Laboratory "PaoLa", University of Urbino Carlo Bo, Fano, PU, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.
| |
Collapse
|
3
|
Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells. Cells 2023; 12:cells12020285. [PMID: 36672222 PMCID: PMC9857206 DOI: 10.3390/cells12020285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Bone marrow mesenchymal stromal cells (BMSCs) are multipotent cells able to self-renew and differentiate, depending on the microenvironment, into adipocytes and osteoblasts. These cells have a limited number of replications and enter replicative senescence during in vitro expansion. The role of DNA methylation (DNAm) assumes importance in cell function and commitment; however, its exact contribution to BMSC differentiation and replicative senescence is still unclear. We performed a genome-wide DNAm analysis on BMSCs cultured in vitro at early passages and induced to differentiate into adipocytes and osteoblasts, and on replicative senescent BMSCs and HUVECs, to identify DNAm patterns of senescence and differentiation. We also compared BMSCs and HUVECs in replicative senescence and found that, in both cellular systems, genome-wide hypomethylation was accompanied by a higher-than-expected overlap of differentially methylated positions (DMPs) and concordance in terms of direction of the change. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis on lineage-independent senescence-associated DMPs revealed 16 common pathways, including Insulin resistance, Molecule adhesion, and Wnt/β-catenin signaling. In both adipogenesis and osteogenesis, we observed a general demethylation of CpG sites compared with undifferentiated BMSCs with a higher number of DMPs in osteogenesis. KEGG analysis resulted in 30 pathways enriched in osteoblasts and only 2 in adipocytes when compared to undifferentiated cells. When comparing differentiated BMSCs with senescent ones, osteogenesis exhibited a greater overlap with senescence in terms of number of DMPs and direction of methylation change compared to adipogenesis. In conclusion, this study may be useful for future research on general mechanisms that occur in replicative senescence and furthermore to identify trajectories of BMSC differentiation and common aspects of differentiated and senescent cells.
Collapse
|
4
|
Kholodenko IV, Gisina AM, Manukyan GV, Majouga AG, Svirshchevskaya EV, Kholodenko RV, Yarygin KN. Resistance of Human Liver Mesenchymal Stem Cells to FAS-Induced Cell Death. Curr Issues Mol Biol 2022; 44:3428-3443. [PMID: 36005132 PMCID: PMC9406952 DOI: 10.3390/cimb44080236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/05/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a pronounced therapeutic potential in various pathological conditions. Though therapeutic effects of MSC transplantation have been studied for a long time, the underlying mechanisms are still not clear. It has been shown that transplanted MSCs are rapidly eliminated, presumably by apoptosis. As the mechanisms of MSC apoptosis are not fully understood, in the present work we analyzed MSC sensitivity to Fas-induced apoptosis using MSCs isolated from the biopsies of liver fibrosis patients (L-MSCs). The level of cell death was analyzed by flow cytometry in the propidium iodide test. The luminescent ATP assay was used to measure cellular ATP levels; and the mitochondrial membrane potential was assessed using the potential-dependent dye JC-1. We found that human L-MSCs were resistant to Fas-induced cell death over a wide range of FasL and anti-Fas mAb concentrations. At the same time, intrinsic death signal inducers CoCl2 and staurosporine caused apoptosis of L-MSCs in a dose-dependent manner. Despite the absence of Fas-induced cell death treatment of L-MSCs with low concentrations of FasL or anti-Fas mAb resulted in a cellular ATP level decrease, while high concentrations of the inducers caused a decline of the mitochondrial membrane potential. Pre-incubation of L-MSCs with the pro-inflammatory cytokine TNF-α did not promote L-MSC cell death. Our data indicate that human L-MSCs have increased resistance to receptor-mediated cell death even under inflammatory conditions.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia; (A.M.G.); (K.N.Y.)
- Correspondence: ; Tel.: +7-(905)7765062; Fax: +7-(499)2450857
| | - Alisa M. Gisina
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia; (A.M.G.); (K.N.Y.)
| | - Garik V. Manukyan
- Petrovsky Russian Research Center of Surgery, 119991 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Elena V. Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (E.V.S.); (R.V.K.)
| | - Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (E.V.S.); (R.V.K.)
| | - Konstantin N. Yarygin
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia; (A.M.G.); (K.N.Y.)
| |
Collapse
|
5
|
Treatment with Mesenchymal Stromal Cells Overexpressing Fas-Ligand Ameliorates Acute Graft-versus-Host Disease in Mice. Int J Mol Sci 2022; 23:ijms23010534. [PMID: 35008964 PMCID: PMC8745472 DOI: 10.3390/ijms23010534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/22/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) has the potential to cure malignant and non-malignant hematological disorders, but because of the serious side effects of this intervention its applications are limited to a restricted number of diseases. Graft-versus-host disease (GvHD) is the most frequent complication and the leading cause of mortality and morbidity following allo-HCT. It results from the attack of the transplanted T cells from the graft against the cells of the recipient. There is no clear treatment for this severe complication. Due to their immunomodulatory properties, mesenchymal stromal cells (MSC) have been proposed to treat GvHD, but the results did not meet expectations. We have previously showed that the immunomodulatory effect of the MSC was significantly enhanced through adenoviral-mediated overexpression of FasL. In this study, we have tested the properties of FasL-overexpressing MSC in vivo, in a mouse model for acute GvHD. We found that treatment with FasL-overexpressing MSC delayed the onset of the disease and increased survival of the mice.
Collapse
|
6
|
Mohr A, Chu T, Clarkson CT, Brooke GN, Teif VB, Zwacka RM. Fas-threshold signalling in MSCs promotes pancreatic cancer progression and metastasis. Cancer Lett 2021; 519:63-77. [PMID: 34171406 DOI: 10.1016/j.canlet.2021.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) belong to the tumour microenvironment and have been implicated in tumour progression. We found that the number of MSCs significantly increased in tumour-burdened mice driven by Fas-threshold signalling. Consequently, MSCs lacking Fas lost their ability to induce metastasis development in a pancreatic cancer model. Mixing of MSCs with pancreatic cancer cells led to sustained production of the pro-metastatic cytokines CCL2 and IL6 by the stem cells. The levels of these cytokines were dependent on the number of MSCs, linking Fas-mediated MSC-proliferation to their capacity to promote tumour progression. Furthermore, we discovered that CCL2 and IL6 were induced by pancreatic cancer cell-derived IL1. Importantly, analysis of patient transcriptomic data revealed that high FasL expression correlates with high levels of MSC markers as well as increased IL6 and CCL2 levels in pancreatic tumours. Moreover, both FasL and CCL2 are linked to elevated levels of markers specific for monocytes known to possess further pro-metastatic activities. These results confirm our experimental findings of a FasL-MSC-IL1-CCL2/IL6 axis in pancreatic cancer and highlights the role of MSCs in tumour progression.
Collapse
Affiliation(s)
- Andrea Mohr
- School of Life Sciences, Protein Structure and Mechanism of Disease Group, Cancer and Stem Cell Biology Laboratory, University of Essex, Colchester, CO4 3SQ, UK.
| | - Tianyuan Chu
- School of Life Sciences, Protein Structure and Mechanism of Disease Group, Cancer and Stem Cell Biology Laboratory, University of Essex, Colchester, CO4 3SQ, UK
| | - Christopher T Clarkson
- School of Life Sciences, Genomics and Computational Biology Group, Gene Regulation Laboratory, University of Essex, Colchester, CO4 3SQ, UK
| | - Greg N Brooke
- School of Life Sciences, Protein Structure and Mechanism of Disease Group, Molecular Oncology Laboratory, University of Essex, Colchester, CO4 3SQ, UK
| | - Vladimir B Teif
- School of Life Sciences, Genomics and Computational Biology Group, Gene Regulation Laboratory, University of Essex, Colchester, CO4 3SQ, UK
| | - Ralf M Zwacka
- School of Life Sciences, Protein Structure and Mechanism of Disease Group, Cancer and Stem Cell Biology Laboratory, University of Essex, Colchester, CO4 3SQ, UK.
| |
Collapse
|
7
|
Apaza Alccayhuaman KA, Heimel P, Lee JS, Tangl S, Strauss FJ, Stähli A, Matalová E, Gruber R. FasL Is Required for Osseous Healing in Extraction Sockets in Mice. Front Immunol 2021; 12:678873. [PMID: 34135904 PMCID: PMC8200669 DOI: 10.3389/fimmu.2021.678873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/10/2021] [Indexed: 01/15/2023] Open
Abstract
Fas ligand (FasL) is a member of the tumor necrosis factor (TNF) superfamily involved in the activation of apoptosis. Assuming that apoptosis is initiated after tooth extraction it is reasonable to suggest that FasL may play a pivotal role in the healing of extraction sockets. Herein, we tested the hypothesis of whether the lack of FasL impairs the healing of extraction sockets. To this end, we extracted upper right incisors of FasL knockout (KO) mice and their wildtype (WT) littermates. After a healing period of two weeks, bone volume over total volume (BV/TV) via µCT and descriptive histological analyses were performed. µCT revealed that BV/TV in the coronal region of the socket amounted to 39.4% in WT and 21.8% in KO, with a significant difference between the groups (p=0.002). Likewise, in the middle region of the socket, BV/TV amounted to 50.3% in WT and 40.8% in KO (p<0.001). In the apical part, however, no difference was noticed. Consistently, WT mice displayed a significantly higher median trabecular thickness and a lower trabecular separation when compared to the KO group at the coronal and central region of the socket. There was the overall tendency that in both, female and male mice, FasL affects bone regeneration. Taken together, these findings suggest that FasL deficiency may reduce bone regeneration during the healing process of extraction sockets.
Collapse
Affiliation(s)
- Karol Alí Apaza Alccayhuaman
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Karl Donath Laboratory for Hard Tissue and Biomaterial Research, School of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Patrick Heimel
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research, School of Dentistry, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Vienna, Austria
| | - Jung-Seok Lee
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Stefan Tangl
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research, School of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Vienna, Austria
| | - Franz J Strauss
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Clinic of Reconstructive Dentistry, University of Zurich, Zurich, Switzerland.,Department of Conservative Dentistry, School of Dentistry, University of Chile, Santiago, Chile
| | - Alexandra Stähli
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Eva Matalová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Vienna, Austria.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Alvarado-Velez M, Enam SF, Mehta N, Lyon JG, LaPlaca MC, Bellamkonda RV. Immuno-suppressive hydrogels enhance allogeneic MSC survival after transplantation in the injured brain. Biomaterials 2020; 266:120419. [PMID: 33038594 DOI: 10.1016/j.biomaterials.2020.120419] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/15/2020] [Accepted: 09/20/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) triggers multiple biochemical and cellular processes that exacerbate brain tissue damage through a secondary injury. Therapies that prevent or limit the evolution of secondary injury could significantly reduce the neurological deficits associated with TBI. Mesenchymal stem cell (MSC) transplantation after TBI can ameliorate neurological deficits by modulating inflammation and enhancing the expression of neurotrophic factors. However, transplanted MSCs can be actively rejected by host immune responses, such as those mediated by cytotoxic CD8+ T cells, thereby limiting their therapeutic efficacy. Here, we designed an agarose hydrogel that releases Fas ligand (FasL), a protein that can induce apoptosis of cytotoxic CD8+ T cells. We studied the immunosuppressive effect of this hydrogel near the allogeneic MSC transplantation site and its impact on the survival of transplanted MSCs in the injured brain. Agarose-FasL hydrogels locally reduced the host cytotoxic CD8+ T cell population and enhanced the survival of allogeneic MSCs transplanted near the injury site. Furthermore, the expression of crucial neurotrophic factors was elevated in the injury penumbra, suggesting an enhanced therapeutic effect of MSCs. These results suggest that the development of immunosuppressive hydrogels for stem cell delivery can enhance the benefits of stem cell therapy for TBI.
Collapse
Affiliation(s)
- Melissa Alvarado-Velez
- Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Syed Faaiz Enam
- Dept. of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nalini Mehta
- Dept. of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Johnathan G Lyon
- Dept. of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Michelle C LaPlaca
- Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ravi V Bellamkonda
- Dept. of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
9
|
The Experimental Pathology at Ancona: 50 Years of Exciting and Pioneering Research on Human Pathology. THE FIRST OUTSTANDING 50 YEARS OF “UNIVERSITÀ POLITECNICA DELLE MARCHE” 2020. [PMCID: PMC7120276 DOI: 10.1007/978-3-030-33832-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Half century ago, a few academic pioneers founded the laboratories of experimental and ultrastructural pathology in Ancona. From this origin, a new phase of experimental studies developed aimed at translational and clinical research up to the present, when our group is internationally recognized for its fundamental contributions in gerontological research and molecular diagnostic pathology. Since the desire of immortality and of eternal youth seems to be as old as mankind, in the future we plan to focus our scientific research on Regenerative Medicine and Rejuvenation strategies. This is the most ambitious aim in the framework of the world aging population. We do not know whether we would achieve these results by ourselves. We are confident that, as in the past, new generations of scientist of the school of experimental pathology at Ancona will get the baton by the older one and lead the future with the same enthusiasm, love and commitment.
Collapse
|
10
|
Zhang WL, Chi CT, Meng XH, Liang SD. miRNA‑15a‑5p facilitates the bone marrow stem cell apoptosis of femoral head necrosis through the Wnt/β‑catenin/PPARγ signaling pathway. Mol Med Rep 2019; 19:4779-4787. [PMID: 30957181 PMCID: PMC6522831 DOI: 10.3892/mmr.2019.10130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
Bone marrow stem cells (BMSCs) are a group cells that function as an underlying cell source for bone tissue regeneration. However, the molecular mechanisms of how BMSCs are induced into apoptosis remains unclear. In the present study, it was demonstrated that the molecular mechanisms of BMSCs were exerted via microRNA-15a-5p (miR-15a-5p) in femoral head necrosis (FHN). Briefly, miRNA-15a-5p expression was elevated in a rat model of FHN. Overexpression of miR-15a-5p promoted the apoptosis of BMSCs and reduced cell growth through the Wnt/β-catenin/peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway. Downregulation of miR-15a-5p reduced the apoptosis of BMSCs and promoted cell growth through the Wnt/β-catenin/PPARγ signaling pathway. The activation of Wnt attenuated the effects of miR-15a-5p on the apoptosis of BMSCs via the β-catenin/PPARγ signaling pathway. In conclusion, the present results indicated that miRNA-15a-5p was involved in the regulation of the apoptosis of BMSCs through regulating the Wnt/β-catenin/PPARγ signaling pathway, which may serve an important role in the regulation of FHN.
Collapse
Affiliation(s)
- Wan-Li Zhang
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Cheng-Tao Chi
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiang-Hui Meng
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Shao-Dong Liang
- Department of Neurosurgery, Hongqi Hospital Affiliated with Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
11
|
Svandova E, Vesela B, Lesot H, Sadoine J, Poliard A, Matalova E. FasL Modulates Expression of Mmp2 in Osteoblasts. Front Physiol 2018; 9:1314. [PMID: 30283358 PMCID: PMC6157335 DOI: 10.3389/fphys.2018.01314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022] Open
Abstract
FasL is a well-known actor in the apoptotic pathways but recent reports have pointed to its important novel roles beyond cell death, as observed also for bone cells. This is supported by non-apoptotic appearance of FasL during osteogenesis and by significant bone alterations unrelated to apoptosis in FasL deficient (gld) mice. The molecular mechanism behind this novel role has not yet been revealed. In this report, intramembranous bone, where osteoblasts differentiate directly from mesenchymal precursors without intermediary chondrogenic step, was investigated. Mouse mandibular bone surrounding the first lower molar was used as a model. The stage where a complex set of bone cells (osteoblasts, osteocytes, osteoclasts) is first present during development was selected for an initial examination. Immunohistochemical staining detected FasL in non-apoptotic cells at this stage. Further, FasL deficient vs. wild type samples subjected to osteogenic PCR Array analysis displayed a significantly decreased expression of Mmp2 in gld bone. To examine the possibility of this novel FasL–Mmp2 relationship, intramembranous bone-derived osteoblastic cells (MC3T3-E1) were treated with anti-FasL antibody or rmFasL. Indeed, the FasL neutralization caused a decreased expression of Mmp2 and rmFasL added to the cells resulted in the opposite effect. Since Mmp2-/- mice display age-dependent alterations in the intramembranous bone, early stages of gld mandibular bone were examined and age-dependent phenotype was confirmed also in gld mice. Taken together, the present in vivo and in vitro findings point to a new non-apoptotic function of FasL in bone development associated with Mmp2 expression.
Collapse
Affiliation(s)
- Eva Svandova
- Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia.,Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, The Czech Academy of Sciences, Brno, Czechia
| | - Barbora Vesela
- Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia.,Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, The Czech Academy of Sciences, Brno, Czechia
| | - Hervé Lesot
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, The Czech Academy of Sciences, Brno, Czechia
| | - Jeremy Sadoine
- Faculté de Chirurgie Dentaire, Université Paris Descartes, Paris, France
| | - Anne Poliard
- Faculté de Chirurgie Dentaire, Université Paris Descartes, Paris, France
| | - Eva Matalova
- Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia.,Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, The Czech Academy of Sciences, Brno, Czechia
| |
Collapse
|
12
|
Fas-L promotes the stem cell potency of adipose-derived mesenchymal cells. Cell Death Dis 2018; 9:695. [PMID: 29891848 PMCID: PMC5995957 DOI: 10.1038/s41419-018-0702-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/26/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Fas-L is a TNF family member known to trigger cell death. It has recently become evident that Fas-L can transduce also non-apoptotic signals. Mesenchymal stem cells (MSCs) are multipotent cells that are derived from various adult tissues. Although MSCs from different tissues display common properties they also display tissue-specific characteristics. Previous works have demonstrated massive apoptosis following Fas-L treatment of bone marrow-derived MSCs both in vitro and following their administration in vivo. We therefore set to examine Fas-L-induced responses in adipose-derived stem cells (ASCs). Human ASCs were isolated from lipoaspirates and their reactivity to Fas-L treatment was examined. ASCs responded to Fas-L by simultaneous apoptosis and proliferation, which yielded a net doubling of cell quantities and a phenotypic shift, including reduced expression of CD105 and increased expression of CD73, in association with increased bone differentiation potential. Treatment of freshly isolated ASCs led to an increase in large colony forming unit fibroblasts, likely produced by early stem cell progenitor cells. Fas-L-induced apoptosis and proliferation signaling were found to be independent as caspase inhibition attenuated Fas-L-induced apoptosis without impacting proliferation, whereas inhibition of PI3K and MEK, but not of JNK, attenuated Fas-L-dependent proliferation, but not apoptosis. Thus, Fas-L signaling in ASCs leads to their expansion and phenotypic shift toward a more potent stem cell state. We speculate that these reactions ensure the survival of ASC progenitor cells encountering Fas-L-enriched environments during tissue damage and inflammation and may also enhance ASC survival following their administration in vivo.
Collapse
|
13
|
Zhao T, Du H, Blum JS, Yan C. Critical role of PPARγ in myeloid-derived suppressor cell-stimulated cancer cell proliferation and metastasis. Oncotarget 2016; 7:1529-43. [PMID: 26625314 PMCID: PMC4811478 DOI: 10.18632/oncotarget.6414] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/17/2015] [Indexed: 12/04/2022] Open
Abstract
Lysosomal acid lipase (LAL) is a key enzyme controlling neutral lipid metabolic signaling in myeloid-derived suppressor cells (MDSCs). MDSCs from LAL-deficient (lal−/−) mice directly stimulate cancer cell proliferation. PPARγ ligand treatment inhibited lal−/− MDSCs stimulation of tumor cell growth and metastasis in vivo, and tumor cell proliferation and migration in vitro. In addition, PPARγ ligand treatment impaired lal−/− MDSCs transendothelial migration, and differentiation from lineage-negative cells. The corrective effects of PPARγ ligand on lal−/− MDSCs functions were mediated by regulating the mammalian target of rapamycin (mTOR) pathway, and subsequently blocking MDSCs ROS overproduction. Furthermore, in the myeloid-specific dominant-negative PPARγ (dnPPARγ) overexpression bitransgenic mouse model, tumor growth and metastasis were enhanced, and MDSCs from these mice stimulated tumor cell proliferation and migration. MDSCs with dnPPARγ overexpression showed increased transendothelial migration, overactivation of the mTOR pathway, and ROS overproduction. These results indicate that PPARγ plays a critical role in neutral lipid metabolic signaling controlled by LAL, which provides a mechanistic basis for clinically targeting MDSCs to reduce the risk of cancer proliferation, growth and metastasis.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hong Du
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Janice S Blum
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cong Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Zoller V, Funcke JB, Keuper M, Abd El Hay M, Debatin KM, Wabitsch M, Fischer-Posovszky P. TRAIL (TNF-related apoptosis-inducing ligand) inhibits human adipocyte differentiation via caspase-mediated downregulation of adipogenic transcription factors. Cell Death Dis 2016; 7:e2412. [PMID: 27735943 PMCID: PMC5133965 DOI: 10.1038/cddis.2016.286] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/13/2016] [Accepted: 08/10/2016] [Indexed: 01/20/2023]
Abstract
Tumor necrosis factor-α (TNFα) and other ligands of the TNF superfamily are potent regulators of adipose tissue metabolism and play a crucial role in the obesity-induced inflammation of adipose tissue. Adipose tissue expression levels of TRAIL (TNF-related apoptosis-inducing ligand) and its receptor were shown to be upregulated by overfeeding and decreased by fasting in mice. In the present study we aimed to elucidate the impact of TRAIL on adipogenesis. To this end, human Simpson-Golabi-Behmel syndrome (SGBS) preadipocytes as well as stromal-vascular cells isolated from human white adipose tissue were used as model systems. Human recombinant TRAIL inhibited adipogenic differentiation in a dose-dependent manner. It activated the cleavage of caspase-8 and -3, which in turn resulted in a downregulation of the key adipogenic transcription factors C/EBPα, C/EBPδ, and PPARγ. The effect was completely blocked by pharmacological or genetic inhibition of caspases. Taken together we discovered a so far unrecognized function of TRAIL in the regulation of adipogenesis. Targeting the TRAIL/TRAIL receptor system might provide a novel strategy to interfere with adipose tissue homeostasis.
Collapse
Affiliation(s)
- Verena Zoller
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jan-Bernd Funcke
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Michaela Keuper
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Muad Abd El Hay
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
15
|
Xuan W, Zhao H, Hankin J, Chen L, Yao S, Ma D. Local anesthetic bupivacaine induced ovarian and prostate cancer apoptotic cell death and underlying mechanisms in vitro. Sci Rep 2016; 6:26277. [PMID: 27195613 PMCID: PMC4872542 DOI: 10.1038/srep26277] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 04/25/2016] [Indexed: 12/12/2022] Open
Abstract
Retrospective studies indicate that the use of regional anesthesia can reduce cancer recurrence after surgery which could be due to ranging from immune function preservation to direct molecular mechanisms. This study was to investigate the effects of bupivacaine on ovarian and prostate cancer cell biology and the underlying molecular mechanisms. Cell viability, proliferation and migration of ovarian carcinoma (SKOV-3) and prostate carcinoma (PC-3) were examined following treatment with bupivacaine. Cleaved caspase 3, 8 and 9, and GSK-3β, pGSK-3βtyr216 and pGSK-3βser9 expression were assessed by immunofluorescence. FAS ligand neutralization, caspase and GSK-3 inhibitors and GSK-3β siRNA were applied to further explore underlying mechanisms. Clinically relevant concentrations of bupivacaine reduced cell viability and inhibited cellular proliferation and migration in both cell lines. Caspase 8 and 9 inhibition generated partial cell death reversal in SKOV-3, whilst only caspase 9 was effective in PC-3. Bupivacaine increased the phosphorylation of GSK-3βTyr216 in SKOV-3 but without measurable effect in PC3. GSK-3β inhibition and siRNA gene knockdown decreased bupivacaine induced cell death in SKOV-3 but not in PC3. Our data suggests that bupivacaine has direct ‘anti-cancer’ properties through the activation of intrinsic and extrinsic apoptotic pathways in ovarian cancer but only the intrinsic pathway in prostate cancer.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhoung University of Science and Technology, Wuhan, Hubei, China.,Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea &Westminster Hospital, London, UK
| | - Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea &Westminster Hospital, London, UK
| | - James Hankin
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea &Westminster Hospital, London, UK
| | - Lin Chen
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhoung University of Science and Technology, Wuhan, Hubei, China.,Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea &Westminster Hospital, London, UK
| | - Shanglong Yao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhoung University of Science and Technology, Wuhan, Hubei, China
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea &Westminster Hospital, London, UK
| |
Collapse
|
16
|
Ottoboni L, De Feo D, Merlini A, Martino G. Commonalities in immune modulation between mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs). Immunol Lett 2015; 168:228-39. [DOI: 10.1016/j.imlet.2015.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
|
17
|
Funcke JB, Zoller V, El Hay MA, Debatin KM, Wabitsch M, Fischer-Posovszky P. TNF-related apoptosis-inducing ligand promotes human preadipocyte proliferation via ERK1/2 activation. FASEB J 2015; 29:3065-75. [PMID: 25857555 DOI: 10.1096/fj.14-267278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/16/2015] [Indexed: 12/31/2022]
Abstract
Upon obesity, adipose tissue is excessively expanded and characterized by pathologic processes like hypoxia, fibrosis, and inflammation. Death ligands belonging to the TNF superfamily such as TNF-α are important contributors to these derangements and exert a pronounced influence on the metabolic and cellular homeostasis of adipose tissue. Here, we sought to identify the effect of the death ligand TNF-related apoptosis-inducing ligand (TRAIL) on the adipose tissue precursor cell pool and therefore investigated its influence on preadipocyte proliferation. Treatment of human preadipocytes with TRAIL resulted in a time- and dose-dependent increase in proliferation (EC50 3.4 ng/ml) comparable to IGF-1. Although no apoptosis was observed, TRAIL triggered a rapid cleavage of caspase-8 and -3. Neither inhibition of caspase activity by zVAD.fmk (20 µM) nor ablation of caspase-8 expression by lentivirus-delivered small hairpin RNA (shRNA) abolished the proliferative response. TRAIL triggered a delayed and sustained activation of ERK1/2, leaving Akt, p38, JNK, and NF-κB unaffected. Importantly, inhibition of ERK1/2 activation by PD0325901 (300 nM) or AZD6244 (5 or 10 µM) completely abolished the proliferative response. We thus reveal a hitherto unknown function of TRAIL in regulating adipose tissue homeostasis by promoting the proliferation of tissue-resident precursor cells.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| | - Verena Zoller
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| | - Muad Abd El Hay
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| | - Klaus-Michael Debatin
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| | - Martin Wabitsch
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| | - Pamela Fischer-Posovszky
- *Division of Pediatric Endocrinology and Diabetes and Department of Pediatrics and Adolescent Medicine, Ulm Medical Center, Ulm, Germany
| |
Collapse
|
18
|
Immunologic Network and Response to Intramyocardial CD34+ Stem Cell Therapy in Patients With Dilated Cardiomyopathy. J Card Fail 2015; 21:572-82. [PMID: 25863169 DOI: 10.1016/j.cardfail.2015.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/28/2015] [Accepted: 03/30/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Although stem cell therapy (SCT) is emerging as a potential treatment for patients with dilated cardiomyopathy (DCM), clinical response remains variable. Our objective was to determine whether baseline differences in circulating immunologic and nonimmunologic biomarkers may help to identify patients more likely to respond to intramyocardial injection of CD34(+)-based SCT. METHODS AND RESULTS We enrolled from January 3, 2011 to March 5, 2012 37 patients with longstanding DCM (left ventricular ejection fraction [LVEF] <40%, New York Heart Association functional class III) who underwent peripheral CD34(+) stem cell mobilization with granulocyte colony-stimulating factor (G-CSF) and collection by means of apheresis. CD34(+) cells were labeled with (99m)Tc-hexamethylpropyleneamine oxime to allow assessment of stem cell retention at 18 hours. Response to SCT was predefined as an increase in LVEF of ≥5% at 3 months. The majority (84%) of patients were male with an overall mean LVEF of 27 ± 7% and a median N-terminal pro-B-type natriuretic peptide (NT-proBNP) level of 2,774 pg/mL. Nineteen patients (51%) were responders to SCT. There was no significant difference between responders and nonresponders regarding to age, sex, baseline LVEF, NT-proBNP levels, or 6-minute walking distance. With the use of a partial least squares (PLS) predictive model, we identified 9 baseline factors that were associated with both stem cell response and stem cell retention (mechanistic validation). Among the baseline factors positively associated with both clinical response and stem cell retention were G-CSF, SDF-1, LIF, MCP-1, and MCP-3. Among baseline factors negatively associated with both clinical response and retention were IL-12p70, FASL, ICAM-1, and GGT. A decrease in G-CSF at 3-month follow-up was also observed in responders compared with nonresponders (P = .02). CONCLUSIONS If further validated, baseline immunologic and nonimmunologic biomarkers may help to identify patients with DCM who are more likely to respond to CD34(+)-based SCT.
Collapse
|
19
|
Ming L, Jin F, Huang P, Luo H, Liu W, Zhang L, Yuan W, Zhang Y, Jin Y. Licochalcone A up-regulates of FasL in mesenchymal stem cells to strengthen bone formation and increase bone mass. Sci Rep 2014; 4:7209. [PMID: 25428397 PMCID: PMC4245531 DOI: 10.1038/srep07209] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/14/2014] [Indexed: 12/29/2022] Open
Abstract
The role of bone marrow-derived mesenchymal stem cells(BMSCs)in the pathogenesis and therapy of osteoporosis has drawn increasing attention in recent years. In the development of osteoporosis, it has been demonstrated that many changes occurred in the behavior of BMSCs. For example, the biological system of FasL pathways mediated differentiation of ERK and GSK-3β-catenin pathway was damaged. Here we found that 0.35 mg/L Licochalcone A (L-A) had a strong effect in increasing the osteogenic differentiation and mineralization of BMSCs both in vivo and in vitro by up-regulating FasL and further playing a role in regulating the ERK and GSK-3β-catenin systems. It has also demonstrated that the administration of L-A could restore the biological function of the damaged BMSCs differentiation by recovering or protecting bone mass in a disease state through activating the endosteal bone formation and partially inhibiting bone resorption in acute estrogen deficiency model. Results of our study suggested that careful titration of MSC was response to L-A and up-regulated FasL pathways mediating differentiation of ERK and GSK-3β-catenin biological systems under disease state in vivo, restore the impaired function, is one of the ways of L-A relieve or treatment osteoporosis.
Collapse
Affiliation(s)
- Leiguo Ming
- 1] State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [3] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| | - Fang Jin
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ping Huang
- Department of Clinical Laboratory, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi. 710032, China
| | - Hailang Luo
- 1] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| | - Wenjia Liu
- 1] Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [3] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| | - Leilei Zhang
- 1] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| | - Wei Yuan
- 1] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| | - Yongjie Zhang
- 1] State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [3] State Key Laboratory of Military Stomatology, Department of Oral Histology and Pathology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yan Jin
- 1] State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China [2] Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, China [3] Institute forTissue Engineering and Regenerative Medicine Research of Xi'an, Xi'an, China
| |
Collapse
|
20
|
Tanabe S. Role of mesenchymal stem cells in cell life and their signaling. World J Stem Cells 2014; 6:24-32. [PMID: 24567785 PMCID: PMC3927011 DOI: 10.4252/wjsc.v6.i1.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/18/2013] [Accepted: 12/13/2013] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have various roles in the body and cellular environment, and the cellular phenotypes of MSCs changes in different conditions. MSCs support the maintenance of other cells, and the capacity of MSCs to differentiate into several cell types makes the cells unique and full of possibilities. The involvement of MSCs in the epithelial-mesenchymal transition is an important property of these cells. In this review, the role of MSCs in cell life, including their application in therapy, is first described, and the signaling mechanism of MSCs is investigated for a further understanding of these cells.
Collapse
|