1
|
Gurav P, Patade T, Hajare S, Kedar RN. n-3 PUFAs synergistically enhance the efficacy of doxorubicin by inhibiting the proliferation and invasion of breast cancer cells. Med Oncol 2023; 41:2. [PMID: 38017288 DOI: 10.1007/s12032-023-02229-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Breast cancer stands as a prominent contributor to cancer-related fatalities among women globally, characterized by an unfavorable prognosis, low survival rates, and its conventional treatment approach involving chemotherapy. Doxorubicin (DOXO) represents a potent anti-tumor agent widely employed in combating breast cancer. Regrettably, a substantial proportion of patients eventually develop resistance to DOXO treatment, elevating the risk of relapse and adverse clinical outcomes. Omega-3 polyunsaturated fatty acids (n-3 PUFAs), recognized as essential components of the human diet, have exhibited considerable promise in targeting malignant cells, initiating apoptosis, and impeding tumor proliferation and metastatic dissemination. Combining these nutritional supplements, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), with DOXO presents a compelling strategy to augment treatment efficacy. The present study was conducted employing a breast cancer cell line, MCF-7, to assess the synergistic potential of DHA, EPA, and DOXO. Remarkably, the combination treatment yielded a substantial increase in cytotoxicity compared to the administration of DOXO alone. Furthermore, an enhancement in the suppression of metastasis was evident in the combination treatment relative to the exclusive use of DOXO. Cell cycle analysis unveiled that cells subjected to the combination treatment exhibited a more pronounced arrest in the G1 phase, signifying the combination's heightened effectiveness in impeding cell progression into the doubling phase. Collectively, the amalgamation of n-3 polyunsaturated fatty acids (n-3 PUFAs) emerges as a potent strategy for enhancing the therapeutic potential of DOXO, effectively restraining the growth and dissemination of breast cancer cells.
Collapse
Affiliation(s)
- Pradnya Gurav
- School of Bioengineering Sciences and Research, MIT Arts, Design and Technology University, Rajbaugh Campus, Loni Kalbhor, Pune, Maharashtra, 412201, India
| | - Tanvi Patade
- School of Bioengineering Sciences and Research, MIT Arts, Design and Technology University, Rajbaugh Campus, Loni Kalbhor, Pune, Maharashtra, 412201, India
| | - Shubham Hajare
- School of Bioengineering Sciences and Research, MIT Arts, Design and Technology University, Rajbaugh Campus, Loni Kalbhor, Pune, Maharashtra, 412201, India
| | - R N Kedar
- School of Bioengineering Sciences and Research, MIT Arts, Design and Technology University, Rajbaugh Campus, Loni Kalbhor, Pune, Maharashtra, 412201, India.
| |
Collapse
|
2
|
Kaur G, Wang X, Li X, Ong H, He X, Cai C. Overexpression of GREM1 Improves the Survival Capacity of Aged Cardiac Mesenchymal Progenitor Cells via Upregulation of the ERK/NRF2-Associated Antioxidant Signal Pathway. Cells 2023; 12:1203. [PMID: 37190112 PMCID: PMC10136744 DOI: 10.3390/cells12081203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Ischemic heart disease is the leading cause of mortality in the United States. Progenitor cell therapy can restore myocardial structure and function. However, its efficacy is severely limited by cell aging and senescence. Gremlin-1 (GREM1), a member of the bone morphogenetic protein antagonist family, has been implicated in cell proliferation and survival. However, GREM1's role in cell aging and senescence has never been investigated in human cardiac mesenchymal progenitor cells (hMPCs). Therefore, this study assessed the hypothesis that overexpression of GREM1 rejuvenates the cardiac regenerative potential of aging hMPCs to a youthful stage and therefore allows better capacity for myocardial repair. We recently reported that a subpopulation of hMPCs with low mitochondrial membrane potential can be sorted from right atrial appendage-derived cells in patients with cardiomyopathy and exhibit cardiac reparative capacity in a mouse model of myocardial infarction. In this study, lentiviral particles were used to overexpress GREM1 in these hMPCs. Protein and mRNA expression were assessed through Western blot and RT-qPCR. FACS analysis for Annexin V/PI staining and lactate dehydrogenase assay were used to assess cell survival. It was observed that cell aging and cell senescence led to a decrease in GREM1 expression. In addition, overexpression of GREM1 led to a decrease in expression of senescence genes. Overexpression of GREM1 led to no significant change in cell proliferation. However, GREM1 appeared to have an anti-apoptotic effect, with an increase in survival and decrease in cytotoxicity evident in GREM1-overexpressing hMPCs. Overexpressing GREM1 also induced cytoprotective properties by decreasing reactive oxidative species and mitochondrial membrane potential. This result was associated with increased expression of antioxidant proteins, such as SOD1 and catalase, and activation of the ERK/NRF2 survival signal pathway. Inhibition of ERK led to a decrease in GREM1-mediated rejuvenation in terms of cell survival, which suggests that an ERK-dependent pathway may be involved. Taken altogether, these results indicate that overexpression of GREM1 can allow aging hMPCs to adopt a more robust phenotype with improved survival capacity, which is associated with an activated ERK/NRF2 antioxidant signal pathway.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoliang Wang
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiuchun Li
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Hannah Ong
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiangfei He
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Chuanxi Cai
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| |
Collapse
|
3
|
Weber L, Lee BS, Imboden S, Hsieh CJ, Lin NY. Phenotyping senescent mesenchymal stromal cells using AI image translation. CURRENT RESEARCH IN BIOTECHNOLOGY 2023; 5:100120. [PMID: 38045568 PMCID: PMC10691861 DOI: 10.1016/j.crbiot.2023.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) offer promising potential in biomedical research, clinical therapeutics, and immunomodulatory therapies due to their ease of isolation and multipotent, immunoprivileged, and immunosuppersive properties. Extensive efforts have focused on optimizing the cell isolation and culture methods to generate scalable, therapeutically-relevant MSCs for clinical applications. However, MSC-based therapies are often hindered by cell heterogeneity and inconsistency of therapeutic function caused, in part, by MSC senescence. As such, noninvasive and molecular-based MSC characterizations play an essential role in assuring the consistency of MSC functions. Here, we demonstrated that AI image translation algorithms can effectively predict immunofluorescence images of MSC senescence markers from phase contrast images. We showed that the expression level of senescence markers including senescence-associated beta-galactosidase (SABG), p16, p21, and p38 are accurately predicted by deep-learning models for Doxorubicin-induced MSC senescence, irradiation-induced MSC senescence, and replicative MSC senescence. Our AI model distinguished the non-senescent and senescent MSC populations and simultaneously captured the cell-to-cell variability within a population. Our microscopy-based phenotyping platform can be integrated with cell culture routines making it an easily accessible tool for MSC engineering and manufacturing.
Collapse
Affiliation(s)
- Leya Weber
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles 90095, CA, United States
| | - Brandon S. Lee
- Department of Bioengineering, University of California, Los Angeles 90095, CA, United States
| | - Sara Imboden
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles 90095, CA, United States
| | - Cho-Jui Hsieh
- Department of Computer Science, University of California, Los Angeles 90095, CA, United States
| | - Neil Y.C. Lin
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles 90095, CA, United States
- Department of Bioengineering, University of California, Los Angeles 90095, CA, United States
- California NanoSystems Institute, University of California, Los Angeles 90095, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, CA, United States
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles 90095, CA, United States
- Broad Stem Cell Center, University of California, Los Angeles 90095, CA, United States
| |
Collapse
|
4
|
Wahlmueller M, Narzt MS, Missfeldt K, Arminger V, Krasensky A, Lämmermann I, Schaedl B, Mairhofer M, Suessner S, Wolbank S, Priglinger E. Establishment of In Vitro Models by Stress-Induced Premature Senescence for Characterizing the Stromal Vascular Niche in Human Adipose Tissue. Life (Basel) 2022; 12:life12101459. [PMID: 36294893 PMCID: PMC9605485 DOI: 10.3390/life12101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Acting as the largest energy reservoir in the body, adipose tissue is involved in longevity and progression of age-related metabolic dysfunction. Here, cellular senescence plays a central role in the generation of a pro-inflammatory environment and in the evolution of chronic diseases. Within the complexity of a tissue, identification and targeting of senescent cells is hampered by their heterogeneity. In this study, we generated stress-induced premature senescence 2D and 3D in vitro models for the stromal vascular niche of human adipose tissue. We established treatment conditions for senescence induction using Doxorubicin (Dox), starting from adipose-derived stromal/stem cells (ASCs), which we adapted to freshly isolated microtissue-stromal vascular fraction (MT-SVF), where cells are embedded within their native extracellular matrix. Senescence hallmarks for the established in vitro models were verified on different cellular levels, including morphology, cell cycle arrest, senescence-associated β-galactosidase activity (SA-βgal) and gene expression. Two subsequent exposures with 200 nM Dox for six days were suitable to induce senescence in our in vitro models. We demonstrated induction of senescence in the 2D in vitro models through SA-βgal activity, at the mRNA level (LMNB1, CDK1, p21) and additionally by G2/M phase cell cycle arrest in ASCs. Significant differences in Lamin B1 and p21 protein expression confirmed senescence in our MT-SVF 3D model. MT-SVF 3D cultures were composed of multiple cell types, including CD31, CD34 and CD68 positive cells, while cell death remained unaltered upon senescence induction. As heterogeneity and complexity of adipose tissue senescence is given by multiple cell types, our established senescence models that represent the perivascular niche embedded within its native extracellular matrix are highly relevant for future clinical studies.
Collapse
Affiliation(s)
- Marlene Wahlmueller
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Marie-Sophie Narzt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Karin Missfeldt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Verena Arminger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Anna Krasensky
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- Rockfish Bio AG, 1010 Vienna, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Mario Mairhofer
- Department of Hematology and Internal Oncology, Johannes Kepler University, 4020 Linz, Austria
| | - Susanne Suessner
- Austrian Red Cross Blood Transfusion Service for Upper Austria, 4020 Linz, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Eleni Priglinger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
- Correspondence:
| |
Collapse
|
5
|
Bientinesi E, Lulli M, Becatti M, Ristori S, Margheri F, Monti D. Doxorubicin-induced senescence in normal fibroblasts promotes in vitro tumour cell growth and invasiveness: the role of Quercetin in modulating these processes. Mech Ageing Dev 2022; 206:111689. [PMID: 35728630 DOI: 10.1016/j.mad.2022.111689] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/05/2022] [Accepted: 06/16/2022] [Indexed: 01/10/2023]
Abstract
Ageing is a complex biological phenomenon representing the major risk factor for developing age-related diseases, such as cardiovascular pathologies, neurodegenerative diseases, and cancer. Geroscience, the new vision of gerontology, identifies cellular senescence as an interconnected biological process that characterises ageing and age-related diseases. Therefore, many strategies have been employed in the last years to reduce the harmful effects of senescence, and among these, the most intriguing ones use nutraceutical compounds. Here we show that a pre-treatment with Quercetin, a bioactive flavonoid present in many fruits and vegetables, increasing cellular antioxidant defence, can alleviate Doxorubicin (Doxo)-induced cellular senescence in human normal WI-38 fibroblasts. Furthermore, our work demonstrates that Quercetin pre-treatment, reducing the number of senescent cells and the production of the senescence-associated secretory phenotype (SASP) factors, can decrease the pro-tumour effects of conditioned medium from Doxo-induced senescent fibroblasts on osteosarcoma cells. Overall, our findings are consistent with the hypothesis that targeting senescent cells can be an emerging strategy for cancer treatment, especially in elderly patients, in which senescent cells are already abundant in several tissues and organs.
Collapse
Affiliation(s)
- Elisa Bientinesi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Sara Ristori
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy 50134.
| |
Collapse
|
6
|
Enukashvily NI, Semenova N, Chubar AV, Ostromyshenskii DI, Gushcha EA, Gritsaev S, Bessmeltsev SS, Rugal VI, Prikhodko EM, Kostroma I, Zherniakova A, Kotova AV, Belik LA, Shumeev A, Maslennikova II, Ivolgin DI. Pericentromeric Non-Coding DNA Transcription Is Associated with Niche Impairment in Patients with Ineffective or Partially Effective Multiple Myeloma Treatment. Int J Mol Sci 2022; 23:ijms23063359. [PMID: 35328779 PMCID: PMC8951104 DOI: 10.3390/ijms23063359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSC) ‘educated’ by tumor cells are an essential component of the multiple myeloma (MM) tumor microenvironment (TME) involved in tumor progression. Transcription of tandemly repeated (TR) non-coding DNA is often activated in many tumors and is required for tumor progression and cancer cells genome reorganization. The aim of the work was to study functional properties including the TR DNA transcription profile of MSC from the hematopoietic niche of treated MM patients. Healthy donors (HD) and patients after bortezomib-based treatment (with partial or complete response, PoCR, and non-responders, NR) were enrolled in the study. Their trephine biopsies were examined histologically to evaluate the hematopoietic niche. MSC cultures obtained from the biopsies were used for evaluation of the proliferation rate, osteogenic differentiation, presence of tumor MSC markers, resistance to bortezomib, and pericentromeric TR DNA transcription level. The MSC ‘education’ by multiple myeloma cells was mimicked in co-culture experiments with or without bortezomib. The TR DNA transcription profile was accessed. The histological examination revealed the persistence of the tumor microenvironment (especially of the vasculature) in treated patients. In co-culture experiments, MSC of bortezomib-treated patients were more resistant to bortezomib and protected cancer MM cells of the RPMI8226 cell line more effectively than HD-MSC did. The MSC obtained from PoCR and NR samples differed in their functional properties (proliferation capacity, osteogenic potential, and cancer-associated fibroblasts markers). Transcriptome analysis revealed activation of the TR transcription in cells of non-hematopoietic origin from NR patients’ bone marrow. The pericentromeric TR DNA of HS2/HS3 families was among the most upregulated in stromal MSC but not in cancer cells. The highest level of transcription was observed in NR-MSC. Transcription of HS2/HS3 was not detected in healthy donors MSC unless they were co-cultured with MM cancer cells and acquired cancer-associated phenotype. Treatment with TNFα downregulated HS2/HS3 transcription in MSC and upregulated in MM cells. Our results suggest that the hematopoietic niche retains the cancer-associated phenotype after treatment. Pericentromeric non-coding DNA transcription is associated with the MSC cancer-associated phenotype in patients with ineffective or partially effective multiple myeloma treatment.
Collapse
Affiliation(s)
- Natella I. Enukashvily
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
- Cell Technologies Lab, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia; (I.I.M.); (D.I.I.)
- Correspondence: (N.I.E.); (N.S.)
| | - Natalia Semenova
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
- Correspondence: (N.I.E.); (N.S.)
| | - Anna V. Chubar
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
| | - Dmitry I. Ostromyshenskii
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
| | - Ekaterina A. Gushcha
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
| | - Sergei Gritsaev
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
| | - Stanislav S. Bessmeltsev
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
| | - Viktor I. Rugal
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
| | - Egor M. Prikhodko
- Pokrovsky Stem Cell Bank, LLC, 199106 St. Petersburg, Russia; (E.M.P.); (A.S.)
- Faculty of Clinical Propaedeutics, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
| | - Ivan Kostroma
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
| | - Anastasia Zherniakova
- Clinical Department, Russian Research Institute of Hematology and Transfusiology FMBA of Russia, 191024 St. Petersburg, Russia; (S.G.); (S.S.B.); (V.I.R.); (I.K.); (A.Z.)
| | - Anastasia V. Kotova
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
| | - Liubov A. Belik
- Lab of the Non-Coding DNA Studies, Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (A.V.C.); (D.I.O.); (E.A.G.); (A.V.K.); (L.A.B.)
| | - Alexander Shumeev
- Pokrovsky Stem Cell Bank, LLC, 199106 St. Petersburg, Russia; (E.M.P.); (A.S.)
| | - Irina I. Maslennikova
- Cell Technologies Lab, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia; (I.I.M.); (D.I.I.)
- Pokrovsky Stem Cell Bank, LLC, 199106 St. Petersburg, Russia; (E.M.P.); (A.S.)
| | - Dmitry I. Ivolgin
- Cell Technologies Lab, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia; (I.I.M.); (D.I.I.)
- Pokrovsky Stem Cell Bank, LLC, 199106 St. Petersburg, Russia; (E.M.P.); (A.S.)
| |
Collapse
|
7
|
Sanchez-Mata A, Gonzalez-Muñoz E. Understanding menstrual blood-derived stromal/stem cells: Definition and properties. Are we rushing into their therapeutic applications? iScience 2021; 24:103501. [PMID: 34917895 PMCID: PMC8646170 DOI: 10.1016/j.isci.2021.103501] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cells with mesenchymal stem cell properties have been identified in menstrual blood and termed menstrual blood-derived stem/stromal cells (MenSCs). MenSCs have been proposed as ideal candidates for cell-based therapy in regenerative medicine and immune-related diseases. However, MenSCs identity has been loosely defined so far and there is controversy regarding their cell markers and differentiation potential. In this review, we outline the origin of MenSCs in the context of regenerating human endometrium, with attention to endometrial eMSCs as reference cells to understand MenSCs. We summarize the cell identity markers analyzed and the immunomodulatory and reparative properties reported. We also address the recent use of MenSCs in cell reprogramming. The main goal of this review is to contribute to the understanding of the identity and properties of MenSCs as well as to identify potential caveats and new venues that deserve to be explored to strengthen their potential applications.
Collapse
Affiliation(s)
- Alicia Sanchez-Mata
- Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29071 Málaga, Spain
| | - Elena Gonzalez-Muñoz
- Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29071 Málaga, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), 29071 Málaga, Spain
| |
Collapse
|
8
|
Li H, Huang H, Chen X, Chen S, Yu L, Wang C, Liu Y, Zhang K, Wu L, Han ZC, Liu N, Wu J, Li Z. The delivery of hsa-miR-11401 by extracellular vesicles can relieve doxorubicin-induced mesenchymal stem cell apoptosis. Stem Cell Res Ther 2021; 12:77. [PMID: 33482923 PMCID: PMC7821514 DOI: 10.1186/s13287-021-02156-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/11/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Chemotherapy is an effective anti-tumor treatment. Mesenchymal stem cells (MSCs), exerting therapy effect on injured tissues during chemotherapy, may be damaged in the process. The possibility of self-healing through long-range paracrine and the mechanisms are unclear. METHODS Doxorubicin, a commonly used chemotherapy drug, was to treat human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for 6 h as an in vitro cell model of chemotherapy-induced damage. Then we use extracellular vesicles derived from placental mesenchymal stem cells (hP-MSCs) to investigate the therapeutic potential of MSCs-EVs for chemotherapy injury. The mechanism was explored using microRNA sequencing. RESULTS MSC-derived extracellular vesicles significantly alleviated the chemotherapy-induced apoptosis. Using microRNA sequencing, we identified hsa-miR-11401, which was downregulated in the Dox group but upregulated in the EV group. The upregulation of hsa-miR-11401 reduced the expression of SCOTIN, thereby inhibiting p53-dependent cell apoptosis. CONCLUSIONS Hsa-miR-11401 expressed by MSCs can be transported to chemotherapy-damaged cells by EVs, reducing the high expression of SCOTIN in damaged cells, thereby inhibiting SCOTIN-mediated apoptosis.
Collapse
Affiliation(s)
- Huifang Li
- Nankai University School of Medicine, Tianjin, China.,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China
| | - Haoyan Huang
- Nankai University School of Medicine, Tianjin, China
| | - Xiaoniao Chen
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Shang Chen
- Nankai University School of Medicine, Tianjin, China
| | - Lu Yu
- Nankai University School of Medicine, Tianjin, China
| | - Chen Wang
- Nankai University School of Medicine, Tianjin, China
| | - Yue Liu
- Nankai University School of Medicine, Tianjin, China
| | - Kaiyue Zhang
- Nankai University School of Medicine, Tianjin, China
| | - Lingling Wu
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhong-Chao Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China.,Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China.,Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co, Beijing, China
| | - Na Liu
- Nankai University School of Medicine, Tianjin, China.
| | - Jie Wu
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, China. .,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China.
| |
Collapse
|
9
|
Kuhlmann C, Schenck TL, Haas EM, Giunta R, Wiggenhauser PS. [Current review of factors in the stem cell donor that influence the regenerative potential of adipose tissue-derived stem cells]. HANDCHIR MIKROCHIR P 2020; 52:521-532. [PMID: 33291167 DOI: 10.1055/a-1250-7878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Regenerative therapies like cell-assisted lipotransfer or preclinical experimental studies use adipose tissue-derived stem cells (ASCs) as the main therapeutic agent. But there are also factors depending on the clinical donor that influence the cell yield and regenerative potential of human ASCs and stromal vascular fraction (SVF). Therefore, the aim of this review was to identify and evaluate these factors according to current literature. METHODS For this purpose, a systematic literature review was performed with focus on factors affecting the regenerative potential of ASCs and SVF using the National Library of Medicine. RESULTS Currently, there is an abundance of studies regarding clinical donor factors influencing ASCs properties. But there is some contradiction and need for further investigation. Nevertheless, we identified several recurrent factors: age, sex, weight, diabetes, lipoedema, use of antidepressants, anti-hormonal therapy and chemotherapy. CONCLUSION We recommend characterisation of the ASC donor cohort in all publications, regardless of whether they are experimental studies or clinical trials. By these means, donor factors that influence experimental or clinical findings can be made transparent and results are more comparable. Moreover, this knowledge can be used for study design to form a homogenous donor cohort by precise clinical history and physical examination.
Collapse
Affiliation(s)
| | | | | | | | - Paul Severin Wiggenhauser
- Klinikum der Universität München, Abteilung Handchirurgie, Plastische Chirurgie, Ästhetische Chirurgie
| |
Collapse
|
10
|
Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, Gargett CE. Endometrial and Menstrual Blood Mesenchymal Stem/Stromal Cells: Biological Properties and Clinical Application. Front Cell Dev Biol 2020; 8:497. [PMID: 32742977 PMCID: PMC7364758 DOI: 10.3389/fcell.2020.00497] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFβ receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.
Collapse
Affiliation(s)
- Mahmood Bozorgmehr
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Shohreh Nikoo
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobitechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Dewhurst RM, Scalzone A, Buckley J, Mattu C, Rankin KS, Gentile P, Ferreira AM. Development of Natural-Based Bone Cement for a Controlled Doxorubicin-Drug Release. Front Bioeng Biotechnol 2020; 8:754. [PMID: 32733869 PMCID: PMC7363953 DOI: 10.3389/fbioe.2020.00754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma (OS) accounts for 60% of all global bone cancer diagnoses. Intravenous administration of Doxorubicin Hydrochloride (DOXO) is the current form of OS treatment, however, systemic delivery has been linked to the onset of DOXO induced cardiomyopathy. Biomaterials including calcium phosphate cements (CPCs) and nanoparticles (NPs) have been tested as localized drug delivery scaffolds for OS cells. However, the tumor microenvironment is critical in cancer progression, with mesenchymal stem cells (MSCs) thought to promote OS metastasis and drug resistance. The extent of MSC assisted survival of OS cells in response to DOXO delivered by CPCs is unknown. In this study, we aimed at investigating the effect of DOXO release from a new formulation of calcium phosphate-based bone cement on the viability of OS cells cocultured with hMSC in vitro. NPs made of PLGA were loaded with DOXO and incorporated in the formulated bone cement to achieve local drug release. The inclusion of PLGA-DOXO NPs into CPCs was also proven to increase the levels of cytotoxicity of U2OS cells in mono- and coculture after 24 and 72 h. Our results demonstrate that a more effective localized DOXO delivery can be achieved via the use of CPCs loaded with PLGA-DOXO NPs compared to CPCs loaded with DOXO, by an observed reduction in metabolic activity of U2OS cells in indirect coculture with hMSCs. The presence of hMSCs offer a degree of DOXO resistance in U2OS cells cultured on PLGA-DOXO NP bone cements. The consideration of the tumor microenvironment via the indirect inclusion of hMSCs in this study can act as a starting point for future direct coculture and in vivo investigations.
Collapse
Affiliation(s)
- Rebecca Marie Dewhurst
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Annachiara Scalzone
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joseph Buckley
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Clara Mattu
- Department of Mechanical and Aerospace, Politecnico di Torino, Turin, Italy
| | - Kenneth S Rankin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
12
|
Domnina A, Ivanova J, Alekseenko L, Kozhukharova I, Borodkina A, Pugovkina N, Smirnova I, Lyublinskaya O, Fridlyanskaya I, Nikolsky N. Three-Dimensional Compaction Switches Stress Response Programs and Enhances Therapeutic Efficacy of Endometrial Mesenchymal Stem/Stromal Cells. Front Cell Dev Biol 2020; 8:473. [PMID: 32612993 PMCID: PMC7308716 DOI: 10.3389/fcell.2020.00473] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells are currently tested as a promising tool for the treatment of a wide range of human diseases. Enhanced therapeutic potential of spheroids formed from these cells has been proved in numerous studies, however, the fundamental basics of this effect are still being discussed. In this work, we showed that endometrial mesenchymal stem/stromal cells (eMSCs) assembled in spheroids possess a higher therapeutic efficacy compared to cells grown in monolayer in the treatment of the defects that are non-specific for eMSC tissue origin – skin wounds. With the purpose to elucidate the possible causes of superior spheroid potency, we compared the tolerance of eMSC cultivated in spheres and monolayer to the stress insults. Using genetically encoded hydrogen peroxide biosensor HyPer, we showed that three-dimensional configuration (3D) helped to shield the inner cell layers of spheroid from the external H2O2-induced oxidative stress. However, the viability of oxidatively damaged eMSCs in spheroids appeared to be much lower than that of monolayer cells. An extensive analysis, which included administration of heat shock and irradiation stress, revealed that cells in spheroids damaged by stress factors activate the apoptosis program, while in monolayer cells stress-induced premature senescence is developed. We found that basal down-regulation of anti-apoptotic and autophagy-related genes provides the possible molecular basis of the high commitment of eMSCs cultured in 3D to apoptosis. We conclude that predisposition to apoptosis provides the programmed elimination of damaged cells and contributes to the transplant safety of spheroids. In addition, to investigate the role of paracrine secretion in the wound healing potency of spheroids, we exploited the in vitro wound model (scratch assay) and found that culture medium conditioned by eMSC spheroids accelerates the migration of adherent cells. We showed that 3D eMSCs upregulate transcriptional activator, hypoxia-inducible factor (HIF)-1, and secret ten-fold more HIF-1-inducible pro-angiogenic factor VEGF (vascular endothelial growth factor) than monolayer cells. Taken together, these findings indicate that enhanced secretory activity can promote wound healing potential of eMSC spheroids and that cultivation in the 3D cell environment alters eMSC vital programs and therapeutic efficacy.
Collapse
Affiliation(s)
- Alisa Domnina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Julia Ivanova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Larisa Alekseenko
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Irina Kozhukharova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Aleksandra Borodkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Natalia Pugovkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Irina Smirnova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Olga Lyublinskaya
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Irina Fridlyanskaya
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Nikolay Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
13
|
de la Torre P, Pérez-Lorenzo MJ, Alcázar-Garrido Á, Flores AI. Cell-Based Nanoparticles Delivery Systems for Targeted Cancer Therapy: Lessons from Anti-Angiogenesis Treatments. Molecules 2020; 25:E715. [PMID: 32046010 PMCID: PMC7038177 DOI: 10.3390/molecules25030715] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 02/05/2023] Open
Abstract
The main strategy of cancer treatment has focused on attacking the tumor cells. Some cancers initially responsive to chemotherapy become treatment-resistant. Another strategy is to block the formation of tumor vessels. However, tumors also become resistant to anti-angiogenic treatments, mostly due to other cells and factors present in the tumor microenvironment, and hypoxia in the central part of the tumor. The need for new cancer therapies is significant. The use of nanoparticle-based therapy will improve therapeutic efficacy and targeting, while reducing toxicity. However, due to inefficient accumulation in tumor sites, clearance by reticuloendothelial organs and toxicity, internalization or conjugation of drug-loaded nanoparticles (NPs) into mesenchymal stem cells (MSCs) can increase efficacy by actively delivering them into the tumor microenvironment. Nanoengineering MSCs with drug-loaded NPs can increase the drug payload delivered to tumor sites due to the migratory and homing abilities of MSCs. However, MSCs have some disadvantages, and exosomes and membranes from different cell types can be used to transport drug-loaded NPs actively to tumors. This review gives an overview of different cancer approaches, with a focus on hypoxia and the emergence of NPs as drug-delivery systems and MSCs as cellular vehicles for targeted delivery due to their tumor-homing potential.
Collapse
Affiliation(s)
| | | | | | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas 12), Avda. de Cordoba s/n, 28041 Madrid, Spain; (P.d.l.T.); (M.J.P.-L.)
| |
Collapse
|
14
|
Fallah M, Mohammadi H, Shaki F, Hosseini-Khah Z, Moloudizargari M, Dashti A, Ziar A, Mohammadpour A, Mirshafa A, Modanloo M, Shokrzadeh M. Doxorubicin and liposomal doxorubicin induce senescence by enhancing nuclear factor kappa B and mitochondrial membrane potential. Life Sci 2019; 232:116677. [DOI: 10.1016/j.lfs.2019.116677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/20/2019] [Accepted: 07/20/2019] [Indexed: 01/01/2023]
|
15
|
Manley H, Sprinks J, Breedon P. Menstrual Blood-Derived Mesenchymal Stem Cells: Women's Attitudes, Willingness, and Barriers to Donation of Menstrual Blood. J Womens Health (Larchmt) 2019; 28:1688-1697. [PMID: 31397634 DOI: 10.1089/jwh.2019.7745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Menstrual blood contains mesenchymal stem cells (MenSC), considered a potential "off-the-shelf" treatment for a range of diseases and medical conditions. Samples of menstrual blood can be collected painlessly, inexpensively, and as frequently as every month for cell therapy. While there has been considerable previous research into the clinical advantages of MenSC, there is currently little understanding of potential donors' attitudes regarding menstrual blood donation and MenSC. Methods: One hundred women 18 years of age or over were surveyed to understand attitudes and potential barriers to menstrual blood donation. The questionnaire assessed participant age and brief medical history (giving birth, donating blood, donating stem cells), menstrual experience (period rating, preferred menstrual hygiene products), and whether participants would donate MenSC or accept MenSC therapy. Results: MenSC was met with a generally positive response, with 78% of menstruating women willing to donate menstrual blood. No significant relationship was recognized between willingness to donate menstrual blood with age, history of childbirth or blood donation, menstruation perception, and preferred menstrual hygiene product. Women rated their period experience better after being made aware of the ability to donate menstrual blood, meaning MenSC therapy can be beneficial for donors as well as patients. Conclusions: Considering women's attitudes to MenSC and donation of menstrual blood, the future of MenSC therapy is positive; women are generally willing to donate menstrual blood, independent of age, perception of periods, and history of childbirth and blood donation.
Collapse
Affiliation(s)
- Hannah Manley
- Medical Engineering Design Research Group, Nottingham Trent University, Nottingham, United Kingdom
| | - James Sprinks
- Medical Engineering Design Research Group, Nottingham Trent University, Nottingham, United Kingdom
| | - Philip Breedon
- Medical Engineering Design Research Group, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
16
|
Wei Y, Elahy M, Friedhuber AM, Wong JY, Hughes JD, Doschak MR, Dass CR. Triple-threat activity of PEDF in bone tumors: Tumor inhibition, tissue preservation and cardioprotection against doxorubicin. Bone 2019; 124:103-117. [PMID: 31028961 DOI: 10.1016/j.bone.2019.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is known for its osteogenic properties, but its effects against primary and secondary bone tumors have not comprehensively been demonstrated. We show the ubiquitous expression of PEDF in murine embryonic tissue. Continuous administration of PEDF in pregnant mice for five days did not adversely affect foetal health, despite PEDF's known potent antiangiogenic properties. In the case of the devastating childhood bone cancer osteosarcoma, PEDF has direct anticancer activity per se, and protects against the toxicity of doxorubicin in the heart, small intestine and testes. PEDF demonstrated anti-proliferative and pro-apoptotic effects against human prostate and breast cancer cells, tumors which are known to metastasize to bone as the preferred secondary site. Caspase-2 was activated in both tumor cell types by PEDF. In models of prostate and breast cancer in bone, PEDF significantly reduced tumor volumes. When combined with zoledronic acid, continuously-administered PEDF significantly reduced breast tumor volume at the bone, and was able to preserve the quality of bone better than the combination therapy. These multiple positive findings make PEDF an ideal endogenous and safe biological for possible future clinical testing.
Collapse
Affiliation(s)
- Yongzhong Wei
- Department of Orthopaedics, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Mina Elahy
- School of Medical Sciences, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | - Anna M Friedhuber
- Department of Pathology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Jia Y Wong
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Jeffery D Hughes
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Michael R Doschak
- Department of Biomedical Engineering, University of Alberta, Alberta T6G 2E1, Canada; Department of Dentistry, University of Alberta, Alberta T6G 2E1, Canada
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Perth, WA 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; College of Health and Biomedicine, Victoria University, St Albans, Melbourne, VIC 3021, Australia.
| |
Collapse
|
17
|
Abushouk AI, Salem AMA, Saad A, Afifi AM, Afify AY, Afify H, Salem HSE, Ghanem E, Abdel-Daim MM. Mesenchymal Stem Cell Therapy for Doxorubicin-Induced Cardiomyopathy: Potential Mechanisms, Governing Factors, and Implications of the Heart Stem Cell Debate. Front Pharmacol 2019; 10:635. [PMID: 31258475 PMCID: PMC6586740 DOI: 10.3389/fphar.2019.00635] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
Over the past decades, researchers have reported several mechanisms for doxorubicin (DOX)-induced cardiomyopathy, including oxidative stress, inflammation, and apoptosis. Another mechanism that has been suggested is that DOX interferes with the cell cycle and induces oxidative stress in C-kit+ cells (commonly known as cardiac progenitor cells), reducing their regenerative capacity. Cardiac regeneration through enhancing the regenerative capacity of these cells or administration of other stem cells types has been the axis of several studies over the past 20 years. Several experiments revealed that local or systemic injections with mesenchymal stem cells (MSCs) were associated with significantly improved cardiac function, ameliorated inflammatory response, and reduced myocardial fibrosis. They also showed that several factors can affect the outcome of MSC treatment for DOX cardiomyopathy, including the MSC type, dose, route, and timing of administration. However, there is growing evidence that the C-kit+ cells do not have a cardiac regenerative potential in the adult mammalian heart. Similarly, the protective mechanisms of MSCs against DOX-induced cardiomyopathy are not likely to include direct differentiation into cardiomyocytes and probably occur through paracrine secretion, antioxidant and anti-inflammatory effects. Better understanding of the involved mechanisms and the factors governing the outcomes of MSCs therapy are essential before moving to clinical application in patients with DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
| | | | - Anas Saad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed M Afifi
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Hesham Afify
- Wake Forest University, Winston-Salem, NC, United States
| | | | - Esraa Ghanem
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
18
|
Kornienko JS, Smirnova IS, Pugovkina NA, Ivanova JS, Shilina MA, Grinchuk TM, Shatrova AN, Aksenov ND, Zenin VV, Nikolsky NN, Lyublinskaya OG. High doses of synthetic antioxidants induce premature senescence in cultivated mesenchymal stem cells. Sci Rep 2019; 9:1296. [PMID: 30718685 PMCID: PMC6361906 DOI: 10.1038/s41598-018-37972-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Stress-induced premature senescence program is known to be activated in cells by various genotoxic stressors, and oxidative stress is considered to be the main of those. To this end, many studies discover antioxidants as protective anti-aging agents. In the current study, we examined the effects of different antioxidants (Tempol, resveratrol, NAC, DPI) on the mesenchymal stem cells maintained in normal physiological conditions. We used high, but non-cytotoxic antioxidant doses which are widely used in laboratory practice to protect cells from oxidative damage. We show that these substances induce reversible block of cell proliferation and do not cause any genotoxic effects when applied to the quiescent cells. However, the same doses of the same substances, when applied to the proliferating cells, can induce irreversible cell cycle arrest, DNA strand breaks accumulation and DNA damage response activation. As a consequence, antioxidant-induced DNA damage results in the stress-induced premature senescence program activation. We conclude that high doses of antioxidants, when applied to the proliferating cells that maintain physiological levels of reactive oxygen species, can cause DNA damage and induce premature senescence which suggests to re-estimate believed unconditional anti-aging antioxidant properties.
Collapse
Affiliation(s)
- Ju S Kornienko
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - I S Smirnova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N A Pugovkina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - Ju S Ivanova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - M A Shilina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - T M Grinchuk
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - A N Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N D Aksenov
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - V V Zenin
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - N N Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia
| | - O G Lyublinskaya
- Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky pr. 4, St.Petersburg, 194064, Russia.
| |
Collapse
|
19
|
Quiescent Human Mesenchymal Stem Cells Are More Resistant to Heat Stress than Cycling Cells. Stem Cells Int 2018; 2018:3753547. [PMID: 30675168 PMCID: PMC6323451 DOI: 10.1155/2018/3753547] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/20/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Quiescence is the prevailing state of many cell types under homeostatic conditions. Yet, surprisingly, little is known about how quiescent cells respond to environmental challenges. The aim of the present study is to compare stress responses of cycling and quiescent mesenchymal stem cells (MSC). Human endometrial mesenchymal cells (eMSС) were employed as adult stem cells. eMSC quiescence was modeled by serum starvation. Sublethal heat shock (HS) was used as a stress factor. Both quiescent and cycling cells were heated at 45°C for 30 min and then returned to standard culture conditions for their recovery. HS response was monitored by DNA damage response, stress-induced premature senescence (SIPS), cell proliferation activity, and oxidative metabolism. It has been found that quiescent cells repair DNA more rapidly, resume proliferation, and undergo SIPS less than proliferating cells. HS-enforced ROS production in heated cycling cells was accompanied with increased expression of genes regulating redox-active proteins. Quiescent cells exposed to HS did not intensify the ROS production, and genes involved in antioxidant defense were mostly silent. Altogether, the results have shown that quiescent cells are more resistant to heat stress than cycling cells. Next-generation sequencing (NGS) demonstrates that HS-survived cells retain differentiation capacity and do not exhibit signs of spontaneous transformation.
Collapse
|