1
|
Ali SR, Ahmad W, Salim A, Durrieu MC, Khan I. Xenogeneic Stem Cell–Induced Cardiac Progenitor Cells Regenerated Infarcted Myocardium in Rat Model. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:110-125. [DOI: 10.1007/s40883-023-00311-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/04/2023] [Accepted: 06/22/2023] [Indexed: 09/11/2024]
|
2
|
Zhang Q, Wang J, Zhang J, Liu F. Potential functions and therapeutic implications of glioma-resident mesenchymal stem cells. Cell Biol Toxicol 2023; 39:853-866. [PMID: 37138122 DOI: 10.1007/s10565-023-09808-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023]
Abstract
Mesenchymal stem cells (MSCs) are emerging crucial regulators in the tumor microenvironment (TME), which contributes to tumor progression and therapeutic resistance. MSCs are considered to be the stromal components of several tumors, their ultimate contribution to tumorigenesis and their potential to drive tumor stem cells, especially in the unique microenvironment of gliomas. Glioma-resident MSCs (GR-MSCs) are non-tumorigenic stromal cells. The phenotype of GR-MSCs is similar to that of prototype bone marrow-MSCs and GR-MSCs enhance the GSCs tumorigenicity via the IL-6/gp130/STAT3 pathway. The higher percentage of GR-MSCs in TME results in the poor prognosis of glioma patients and illuminate the tumor-promoting roles for GR-MSCs by secreting specific miRNA. Furthermore, the GR-MSC subpopulations associated with CD90 expression determine their different functions in glioma progression and CD90low MSCs generate therapeutic resistance by increasing IL-6-mediated FOXS1 expression. Therefore, it is urgent to develop novel therapeutic strategies targeting GR-MSCs for GBM patients. Despite that several functions of GR-MSCs have been confirmed, their immunologic landscapes and deeper mechanisms associated with the functions are not still expounded. In this review, we summarize the progress and potential function of GR-MSCs, as well as highlight their therapeutic implications based on GR-MSCs in GBM patients.
Collapse
Affiliation(s)
- Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Laboratory of Biomedical Materials, Beijing, China.
| |
Collapse
|
3
|
El-Husseiny HM, Mady EA, Helal MAY, Tanaka R. The Pivotal Role of Stem Cells in Veterinary Regenerative Medicine and Tissue Engineering. Vet Sci 2022; 9:648. [PMID: 36423096 PMCID: PMC9698002 DOI: 10.3390/vetsci9110648] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 07/30/2023] Open
Abstract
The introduction of new regenerative therapeutic modalities in the veterinary practice has recently picked up a lot of interest. Stem cells are undifferentiated cells with a high capacity to self-renew and develop into tissue cells with specific roles. Hence, they are an effective therapeutic option to ameliorate the ability of the body to repair and engineer damaged tissues. Currently, based on their facile isolation and culture procedures and the absence of ethical concerns with their use, mesenchymal stem cells (MSCs) are the most promising stem cell type for therapeutic applications. They are becoming more and more well-known in veterinary medicine because of their exceptional immunomodulatory capabilities. However, their implementation on the clinical scale is still challenging. These limitations to their use in diverse affections in different animals drive the advancement of these therapies. In the present article, we discuss the ability of MSCs as a potent therapeutic modality for the engineering of different animals' tissues including the heart, skin, digestive system (mouth, teeth, gastrointestinal tract, and liver), musculoskeletal system (tendons, ligaments, joints, muscles, and nerves), kidneys, respiratory system, and eyes based on the existing knowledge. Moreover, we highlighted the promises of the implementation of MSCs in clinical use in veterinary practice.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Mahmoud A. Y. Helal
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Animal Medicine, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi 183-8509, Tokyo, Japan
| |
Collapse
|
4
|
Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev 2022; 188:114449. [PMID: 35835353 DOI: 10.1016/j.addr.2022.114449] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/03/2022] [Accepted: 07/06/2022] [Indexed: 12/20/2022]
Abstract
The Enhanced Permeability and Retention (EPR) effect has been recognized as the central paradigm in tumor-targeted delivery in the last decades. In the wake of this concept, nanotechnologies have reached phenomenal levels in research. However, clinical tumors display a poor manifestation of EPR effect. Factors including tumor heterogeneity, complicating tumor microenvironment, and discrepancies between laboratory models and human tumors largely contribute to poor efficiency in tumor-targeted delivery and therapeutic failure in clinical translation. In this article, approaches for evaluation of EPR effect in human tumor were overviewed as guidance to employ EPR effect for cancer treatment. Strategies to augment EPR-mediated tumoral delivery are discussed in different dimensions including enhancement of vascular permeability, depletion of tumor extracellular matrix and optimization of nanoparticle design. Besides, the recent development in alternative tumor-targeted delivery mechanisms are highlighted including transendothelial pathway, endogenous cell carriers and non-immunogenic bacteria-mediated delivery. In addition, the emerging preclinical models better reflect human tumors are introduced. Finally, more rational applications of EPR effect in other disease and field are proposed. This article elaborates on fundamental reasons for the gaps between theoretical expectation and clinical outcomes, attempting to provide some perspective directions for future development of cancer nanomedicines in this still evolving landscape.
Collapse
Affiliation(s)
- Yixuan Zi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Kaiyun Yang
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zimei Wu
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
5
|
Cabanillas Stanchi KM, Böhringer J, Strölin M, Groeschel S, Lenglinger K, Treuner C, Kehrer C, Laugwitz L, Bevot A, Kaiser N, Schumm M, Lang P, Handgretinger R, Krägeloh-Mann I, Müller I, Döring M. Hematopoietic stem cell transplantation with mesenchymal stromal cells in children with metachromatic leukodystrophy. Stem Cells Dev 2022; 31:163-175. [PMID: 35323019 DOI: 10.1089/scd.2021.0352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder primarily affecting the white matter of the nervous system that results from a deficiency of the arylsulfatase A (ARSA). Mesenchymal stem cells (MSCs) are able to secrete ARSA and have shown beneficial effects in MLD patients. In this retrospective analysis, 10 pediatric MLD patients (MSCG) underwent allogeneic hematopoietic stem cell transplantation (HSCT) and received two applications of 2 x 106 MSCs/kg bodyweight at day +30 and +60 after HSCT between 2007 and 2018. MSC safety, occurrence of graft-versus-host disease (GvHD), blood ARSA levels, chimerism, cell regeneration and engraftment, MRI changes, and the gross motor function were assessed within the first year of HSCT. The long-term data included clinical outcomes and safety aspects of MSCs. Data were compared to a control cohort of seven pediatric MLD patients (CG) who underwent HSCT only. The application of MSC in pediatric MLD patients after allogeneic HSCT was safe and well tolerated and long-term potentially MSC-related adverse effects up to 13.5 years after HSCT were not observed. Patients achieved significantly higher ARSA levels (CG: median 1.03 nmol∙10-6, range 0.41-1.73 | MSCG: median 1.58 nmol∙10-6, range 0.44-2.6; p<0.05), as well as significantly higher leukocyte (p<0.05) and thrombocyte (p<0.001) levels within 365 days of MSC application compared to CG patients. Statistically significant effects on acute GvHD, regeneration of immune cells, engraftment, MRI changes, gross motor function, and clinical outcomes were not detected. In conclusion, the application of MSCs in pediatric MLD patients after allogeneic HSCT was safe and well tolerated. The two applications of 2 x 106/kg allogeneic MSCs were followed by improved engraftment and hematopoiesis within the first year after HSCT. Larger, prospective trials are necessary to evaluate the impact of MSC application on engraftment and hematopoietic recovery.
Collapse
Affiliation(s)
| | - Judith Böhringer
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Manuel Strölin
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Samuel Groeschel
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Katrin Lenglinger
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Claudia Treuner
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Christiane Kehrer
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Lucia Laugwitz
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Andrea Bevot
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Nadja Kaiser
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Michael Schumm
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Peter Lang
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Rupert Handgretinger
- Children's University Hospital, Hematology/Oncology, Hoppe-Seyler-Str. 1, Tuebingen, Germany, 72076;
| | | | - Ingo Müller
- University Medical Center Hamburg-Eppendorf, 37734, Department of Pediatric Hematology and Oncology, Hamburg, Hamburg, Germany;
| | - Michaela Döring
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| |
Collapse
|
6
|
Khalil Alyahya H, Subash-Babu P, Mohammad Salamatullah A, Hayat K, Albader N, Alkaltham MS, Ahmed MA, Arzoo S, Bourhia M. Quantification of Chlorogenic Acid and Vanillin from Coffee Peel Extract and its Effect on α-Amylase Activity, Immunoregulation, Mitochondrial Oxidative Stress, and Tumor Suppressor Gene Expression Levels in H 2O 2-Induced Human Mesenchymal Stem Cells. Front Pharmacol 2021; 12:760242. [PMID: 34795590 PMCID: PMC8593645 DOI: 10.3389/fphar.2021.760242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Polyphenols and flavonoid-rich foods help in arresting reactive oxygen species development and protecting DNA from oxidative damage. Coffee peel (CP) preparations are consumed as beverages, and their total polyphenol or flavonoid content and their effect on oxidative stress-induced human mesenchymal stem cells (hMSCs) are poorly understood. Method: We prepared hot water extracts of CP (CPE) and quantified the amount of total polyphenol and flavonoid using HPLC analysis. In addition, CPE have been studied for their α-amylase inhibitory effect and beneficial effects in oxidative stress-induced hMSCs. Results: The obtained results show that the availability of chlorogenic acid, vanillin, and salicylic acid levels in CPE is more favorable for enhancing cell growth, nuclear integrity, and mitochondrial efficiency which is confirmed by propidium iodide staining and JC-1 staining. CPE treatment to hMSCs for 48 h reduced oxidative stress by decreasing mRNA expression levels of LPO and NOX-4 and in increasing antioxidant CYP1A, GSH, GSK-3β, and GPX mRNA expressions. Decreased pro-inflammatory (TNF-α, NF-κβ, IL-1β, TLR-4) and increased tumor suppressor genes (except Bcl-2) such as Cdkn2A, p53 expressions have been observed. Conclusions: The availability of CGA in CPs effectively reduced mitochondrial oxidative stress, reduced pro-inflammatory cytokines, and increased tumor suppressor genes.
Collapse
Affiliation(s)
- Heba Khalil Alyahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Pandurangan Subash-Babu
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad Mohammad Salamatullah
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Khizar Hayat
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nawal Albader
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Saeed Alkaltham
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Asif Ahmed
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Shaista Arzoo
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Chemistry-Biochemistry, Environment, Nutrition and Health, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| |
Collapse
|
7
|
Kenyon NS, Willman MA, Han D, Leeman RS, Rabassa A, Diaz WL, Geary JC, Poumian-Ruiz E, Griswold AJ, Van Booven DJ, Thompson R, Ordoukhanian P, Head SR, Kenyon NM, McHenry KG, Salomon DR, Bartholomew AM, Berman DM. Extended survival versus accelerated rejection of nonhuman primate islet allografts: Effect of mesenchymal stem cell source and timing. Am J Transplant 2021; 21:3524-3537. [PMID: 34008325 PMCID: PMC9034438 DOI: 10.1111/ajt.16693] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 01/25/2023]
Abstract
Mesenchymal stem cells (MSC) have been shown to be immunomodulatory, tissue regenerative, and graft promoting; however, several questions remain with regard to ideal MSC source and timing of administration. In this study, we utilized a rigorous preclinical model of allogeneic islet cell transplantation, incorporating reduced immune suppression and near to complete mismatch of major histocompatibility antigens between the diabetic cynomolgus monkey recipient and the islet donor, to evaluate both the graft promoting impact of MSC source, that is, derived from the islet recipient, the islet donor or an unrelated third party as well as the impact of timing. Co-transplant of MSC and islets on post-operative day 0, followed by additional IV MSC infusions in the first posttransplant month, resulted in prolongation of rejection free and overall islet survival and superior metabolic control for animals treated with recipient as compared to donor or third-party MSC. Immunological analyses demonstrated that infusion of MSC from either source did not prevent alloantibody formation to the islet or MSC donor; however, treatment with recipient MSC resulted in significant downregulation of memory T cells, decreased anti-donor T cell proliferation, and a trend toward increased Tregulatory:Tconventional ratios.
Collapse
Affiliation(s)
- Norma S. Kenyon
- Diabetes Research Institute, University of Miami, Miami, Florida, USA,Department of Surgery, University of Miami, Miami, Florida, USA,Department of Microbiology and Immunology, University of Miami, Miami, Florida, USA,Department of Biomedical Engineering, University of Miami, Miami, Florida, USA
| | | | - Dongmei Han
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Rachel S. Leeman
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Alex Rabassa
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Waldo L. Diaz
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - James C. Geary
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Ena Poumian-Ruiz
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Anthony J. Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA,The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, USA
| | - Derek J. Van Booven
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Ryan Thompson
- The Scripps Research Institute, La Jolla, California, USA
| | - Philip Ordoukhanian
- The Scripps Research Institute, La Jolla, California, USA,The Scripps Research Institute Genomics Core Facility, La Jolla, California, USA
| | - Steven R. Head
- The Scripps Research Institute, La Jolla, California, USA,The Scripps Research Institute Genomics Core Facility, La Jolla, California, USA
| | - Norman M. Kenyon
- Diabetes Research Institute, University of Miami, Miami, Florida, USA,Department of Surgery, University of Miami, Miami, Florida, USA
| | - Kenton G. McHenry
- National Center for Supercomputing Applications, University of Illinois, Urbana-Champaign, Chicago, Illinois, USA
| | | | | | - Dora M. Berman
- Diabetes Research Institute, University of Miami, Miami, Florida, USA,Department of Surgery, University of Miami, Miami, Florida, USA
| |
Collapse
|
8
|
Subash-Babu P, Al-Saran N, M Alshammari G, Naif Al-Harbi L, Hussain Alhussain M, Shamlan G, Abdulaziz AlSedairy S, Abdullah Alshatwi A. Evaluation of Biosafety, Antiobesity, and Endothelial Cells Proliferation Potential of Basil Seed Extract Loaded Organic Solid Lipid Nanoparticle. Front Pharmacol 2021; 12:722258. [PMID: 34671255 PMCID: PMC8521050 DOI: 10.3389/fphar.2021.722258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/25/2021] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to synthesize solid lipid nanoparticles to enhance liposome-assisted intracellular uptake of basil seed active components in adipocytes and vascular smooth muscle cells to attain increased bioavailability. To obtain solid lipid nanoparticle (SLNp), the water phase containing basil seed extract (BSE) was encapsulated with lipid matrix containing chia seed phospholipids using homogenization and cold ultra-sonication method. The physicochemical characterization of BSE loaded solid lipid nanoparticles (BSE-SLNp) has been analyzed using Zetasizer, FT-IR, and TEM. The BSE-SLNp showed an average diameter of 20-110 nm on the day of preparation and it remains the same after 60 days of storage. The cytotoxicity assay confirmed that the BSE-SLNp did not produce toxicity in hMSCs, preadipocytes, or human umbilical vein endothelial cells (HUVECs) until the tested higher dose up to 64 μg/ml. During effective dose determination, 4 μg/ml of BSE-SLNp confirmed non-toxic and enhanced metabolic function in hMSCs, preadipocytes, and HUVECs. Biosafety assay confirmed normal nuclear morphology in PI staining and high mitochondrial membrane potential in JC-1 assay within 48 h in hMSCs. The maturing adipocyte treated with 4 μg/ml of BSE-SLNp significantly increased the mitochondrial efficiency and fatty acid beta-oxidation (PPARγC1α, UCP-1, and PRDM-16) related gene expression levels. Oxidative stress induced HUVECs treated with 4 μg/ml of BSE-SLNp potentially enhanced antioxidant capacity, cell growth, and microtubule development within 48 h H2O2 induced oxidative stressed HUVECs have shown 39.8% viable cells, but treatment with BSE-SLNp has shown 99% of viable cells within 48 h confirmed by Annexin-V assay. In addition, mitochondrial membrane potential (Δψm) increased to 89.4% confirmed by JC-1 assay. The observed DNA integrity, cell viability was confirmed by increased antioxidant and tumor suppressor-related gene expression levels. VEGF expression has been significantly increased and pro-inflammation-related mRNA levels were decreased in BSE-SLNp treated cells. In conclusion, enhanced adipocyte fatty acid oxidation is directly associated with decreased adipocytokine secretion which arrests obesity-associated comorbidities. In addition, suppressing vascular cell oxidative stress and metabolic inflammation supports vascular cell proliferation and arrests ageing-related vascular diseases.
Collapse
Affiliation(s)
- Pandurangan Subash-Babu
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nada Al-Saran
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghedeir M Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Maha Hussain Alhussain
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghalia Shamlan
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sahar Abdulaziz AlSedairy
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ali Abdullah Alshatwi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Arjmand B, Abedi M, Arabi M, Alavi-Moghadam S, Rezaei-Tavirani M, Hadavandkhani M, Tayanloo-Beik A, Kordi R, Roudsari PP, Larijani B. Regenerative Medicine for the Treatment of Ischemic Heart Disease; Status and Future Perspectives. Front Cell Dev Biol 2021; 9:704903. [PMID: 34568321 PMCID: PMC8461329 DOI: 10.3389/fcell.2021.704903] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular disease is now the leading cause of adult death in the world. According to new estimates from the World Health Organization, myocardial infarction (MI) is responsible for four out of every five deaths due to cardiovascular disease. Conventional treatments of MI are taking aspirin and nitroglycerin as intermediate treatments and injecting antithrombotic agents within the first 3 h after MI. Coronary artery bypass grafting and percutaneous coronary intervention are the most common long term treatments. Since none of these interventions will fully regenerate the infarcted myocardium, there is value in pursuing more innovative therapeutic approaches. Regenerative medicine is an innovative interdisciplinary method for rebuilding, replacing, or repairing the missed part of different organs in the body, as similar as possible to the primary structure. In recent years, regenerative medicine has been widely utilized as a treatment for ischemic heart disease (one of the most fatal factors around the world) to repair the lost part of the heart by using stem cells. Here, the development of mesenchymal stem cells causes a breakthrough in the treatment of different cardiovascular diseases. They are easily obtainable from different sources, and expanded and enriched easily, with no need for immunosuppressing agents before transplantation, and fewer possibilities of genetic abnormality accompany them through multiple passages. The production of new cardiomyocytes can result from the transplantation of different types of stem cells. Accordingly, due to its remarkable benefits, stem cell therapy has received attention in recent years as it provides a drug-free and surgical treatment for patients and encourages a more safe and feasible cardiac repair. Although different clinical trials have reported on the promising benefits of stem cell therapy, there is still uncertainty about its mechanism of action. It is important to conduct different preclinical and clinical studies to explore the exact mechanism of action of the cells. After reviewing the pathophysiology of MI, this study addresses the role of tissue regeneration using various materials, including different types of stem cells. It proves some appropriate data about the importance of ethical problems, which leads to future perspectives on this scientific method.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Arabi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahdieh Hadavandkhani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Kordi
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar Roudsari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Al-Harbi LN, Pandurangan SB, Al-Dossari AM, Shamlan G, Salamatullah AM, Alshatwi AA, Alotiby AA. Beta vulgaris rubra L. (Beetroot) Peel Methanol Extract Reduces Oxidative Stress and Stimulates Cell Proliferation via Increasing VEGF Expression in H 2O 2 Induced Oxidative Stressed Human Umbilical Vein Endothelial Cells. Genes (Basel) 2021; 12:genes12091380. [PMID: 34573361 PMCID: PMC8466581 DOI: 10.3390/genes12091380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
The antioxidant capacity of polyphenols and flavonoids present in dietary agents aids in arresting the development of reactive oxygen species (ROS) and protecting endothelial smooth muscle cells from oxidative stress/induced necrosis. Beetroot (Beta vulgaris var. rubra L.; BVr) is a commonly consumed vegetable representing a rich source of antioxidants. Beetroot peel’s bioactive compounds and their role in human umbilical vein endothelial cells (HUVECs) are still under-researched. In the present study, beetroot peel methanol extract (BPME) was prepared, and its effect on the bio-efficacy, nuclear integrity, mitochondrial membrane potential and vascular cell growth, and immunoregulation-related gene expression levels in HUVECs with induced oxidative stress were analysed. Gas chromatography–mass spectroscopy (GC-MS) results confirmed that BPME contains 5-hydroxymethylfurfural (32.6%), methyl pyruvate (15.13%), furfural (9.98%), and 2,3-dihydro-3,5-dihydroxy-6-methyl-4H-Pyran-4-one (12.4%). BPME extract effectively enhanced cell proliferation and was confirmed by MTT assay; the nuclear integrity was confirmed by propidium iodide (PI) staining assay; the mitochondrial membrane potential (Δψm) was confirmed by JC-1 staining assay. Annexin V assay confirmed that BPME-treated HUVECs showed 99% viable cells, but only 39.8% viability was shown in HUVECs treated with H2O2 alone. In addition, BPME treatment of HUVECs for 48 h reduced mRNA expression of lipid peroxide (LPO) and increased NOS-3, Nrf-2, GSK-3β, GPX, endothelial nitric oxide synthase (eNOS) and vascular cell growth factor (VEGF) mRNA expression levels. We found that BPME treatment decreased proinflammatory (nuclear factor-κβ (F-κβ), tissue necrosis factor-α (TNF-α), toll-like receptor-4 (TLR-4), interleukin-1β (IL-1β)) and vascular inflammation (intracellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), EDN1, IL-1β)-related mRNA expressions. In conclusion, beetroot peel treatment effectively increased vascular smooth cell growth factors and microtubule development, whereas it decreased vascular inflammatory regulators. BPME may be beneficial for vascular smooth cell regeneration, tissue repair and anti-ageing potential.
Collapse
Affiliation(s)
- Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
- Correspondence:
| | - Subash-Babu Pandurangan
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
| | - Alhanouf Mohammed Al-Dossari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
| | - Ghalia Shamlan
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
| | - Ahmad Mohammad Salamatullah
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
| | - Ali A Alshatwi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (S.-B.P.); (A.M.A.-D.); (G.S.); (A.M.S.); (A.A.A.)
| | - Amna Abdullah Alotiby
- Department of Haematology and Immunology, Faculty of Medicine, Umm Alqura University, Makkah 24237, Saudi Arabia;
| |
Collapse
|
11
|
Prządka P, Buczak K, Frejlich E, Gąsior L, Suliga K, Kiełbowicz Z. The Role of Mesenchymal Stem Cells (MSCs) in Veterinary Medicine and Their Use in Musculoskeletal Disorders. Biomolecules 2021; 11:1141. [PMID: 34439807 PMCID: PMC8391453 DOI: 10.3390/biom11081141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Regenerative medicine is a dynamically developing field of human and veterinary medicine. The animal model was most commonly used for mesenchymal stem cells (MSCs) treatment in experimental and preclinical studies with a satisfactory therapeutic effect. Year by year, the need for alternative treatments in veterinary medicine is increasing, and other applications for promising MSCs and their biological derivatives are constantly being sought. There is also an increase in demand for other methods of treating disease states, of which the classical treatment methods did not bring the desired results. Cell therapy can be a realistic option for treating human and animal diseases in the near future and therefore additional research is needed to optimize cell origins, numbers, or application methods in order to standardize the treatment process and assess its effects. The aim of the following work was to summarize available knowledge about stem cells in veterinary medicine and their possible application in the treatment of chosen musculoskeletal disorders in dogs and horses.
Collapse
Affiliation(s)
- Przemysław Prządka
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| | - Krzysztof Buczak
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| | - Ewelina Frejlich
- 2nd Department of General Surgery and Surgical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Ludwika Gąsior
- Vets & Pets Veterinary Clinic, Zakladowa 11N, 50-231 Wroclaw, Poland;
| | - Kamil Suliga
- Student Veterinary Surgical Society “LANCET”, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwaldzki 51, 50-366 Wroclaw, Poland;
| | - Zdzisław Kiełbowicz
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| |
Collapse
|
12
|
Hassan SA, Elghait ATA, Abdelqader ZS, Meligy FY. Therapeutic efficiency of adipose-derived mesenchymal stem cells in healing of experimentally induced gastric ulcers in rats. Anat Cell Biol 2021; 54:361-374. [PMID: 34290152 PMCID: PMC8493023 DOI: 10.5115/acb.21.034] [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: 02/18/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/27/2022] Open
Abstract
Gastric (peptic) ulcer is a major gastrointestinal disorder with high morbidity and mortality. While several drugs have been used to treat gastric ulcers, such as proton pump inhibitor-based triple therapy for Helicobacter pylori eradication, but hey result in adverse side effects. Therefore, development of new alternative therapies is desirable. Many recent studies have shown that mesenchymal stem cells (MSCs) might have an enhancing effect on the ulcerated gastric mucosa. The aim of this study is to evaluate the efficacy of MSCs in the treatment of indomethacin-induced gastric ulcer, and to compare it with the normal ulcer autohealing. This work was performed on 36 adult male albino rats, divided into four groups: Group I (control group), Group II (ulcer group), Group III (autohealing group), and Group IV (stem cells-treated group). The histological changes of gastric mucosa were examined in sections stained with H&E using light microscope for expression of vascular endothelial growth factors (VEGF) and proliferating cell nuclear antigen (PCNA) in immunohistochemical stained sections using image analyzer. The results from MSCs-treated group revealed restoration of the normal architecture of the gastric mucosa with comparison to the autohealing group which showed excessive granulation tissue and heavy cellular infiltration with disorganized architecture of the fundic mucosa. Immunohistochemical examination showed strong expression of both VEGF and PCNA in the MSCs-treated group. So it was concluded that MSCs accelerate gastric ulcer healing when injected intraperitoneally, compared to autohealing process which showed delayed healing.
Collapse
Affiliation(s)
- Safaa A Hassan
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Taha Abou Elghait
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt.,Histology and Cell Biology Department, Sphinx University, Assiut, Egypt
| | - Zainab S Abdelqader
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma Y Meligy
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
13
|
Aithal AP, Bairy LK, Seetharam RN. Safety and therapeutic potential of human bone marrow-derived mesenchymal stromal cells in regenerative medicine. Stem Cell Investig 2021; 8:10. [PMID: 34124233 DOI: 10.21037/sci-2020-036] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/24/2021] [Indexed: 12/20/2022]
Abstract
Regenerative medicine is considered as an alternative approach to healthcare. Owing to their pluripotent abilities and their relative lack of ethical and legal issues, adult stem cells are considered as optimal candidates for use in the treatment of various diseases. Bone marrow-derived mesenchymal stem cells are among the most promising candidates for clinical applications as they have expressed a higher degree of plasticity in vitro. Many investigators have begun to examine how bone marrow stem cells might be used to rebuild damaged tissues. The systemic administration of cells for therapeutic applications requires efficient migration and homing of cells to the target site. Cell adhesion molecules and their ligands, chemokines, extracellular matrix components and specialized bone marrow niches all participate in the proper regulation of this process. MSCs suppress the pathophysiological process that is mediated by chronic inflammation and contributes to a modification of the microenvironment and tissue regeneration. Due to the intricacy of the mesenchymal stem cell, there is ever-increasing amount of data emerging about their migration and regenerative mechanisms. Many factors influence MSC mobilization and their homing to injured tissues. This review summarizes the current clinical and pre-clinical data available in literature regarding the use of MSC in tissue repair and their prospective therapeutic role in various diseases and the underlying repair mechanisms will be discussed.
Collapse
Affiliation(s)
- Ashwini P Aithal
- Department of Anatomy, Melaka Manipal Medical College (Manipal Campus), Manipal Academy of Higher Education, Manipal, India
| | - Laxminarayana K Bairy
- Department of Pharmacology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | | |
Collapse
|
14
|
Denu RA, Hematti P. Optimization of oxidative stress for mesenchymal stromal/stem cell engraftment, function and longevity. Free Radic Biol Med 2021; 167:193-200. [PMID: 33677063 DOI: 10.1016/j.freeradbiomed.2021.02.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are multipotent cells that possess great potential as a cellular therapeutic based on their ability to differentiate to different lineages and to modulate immune responses. However, their potential is limited by their low tissue abundance, and thus the need for robust ex vivo expansion prior to their application. This creates its own issues, namely replicative senescence, which could lead to reduced MSC functionality and negatively impact their engraftment. Ex vivo expansion and MSC aging are associated with greater oxidative stress. Therefore, there is great need to identify strategies to reduce oxidative stress in MSCs. This review summarizes the achievements made to date in addressing oxidative stress in MSCs and speculates about interesting avenues of future investigation to solve this critical problem.
Collapse
Affiliation(s)
- Ryan A Denu
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peiman Hematti
- Departments of Medicine, Pediatrics, Surgery and Biomedical Engineering, Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
15
|
Potential Use of Mesenchymal Multipotent Cells for Hemopoietic Stem Cell Transplantation: Pro and Contra. J Pediatr Hematol Oncol 2021; 43:90-94. [PMID: 33560076 DOI: 10.1097/mph.0000000000002065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/06/2020] [Indexed: 12/25/2022]
Abstract
The potential of mesenchymal multipotent (stem) cells (MSC) to modify immune reactions and mediate hematopoiesis boosted great interest for their use in allogeneic hemopoietic stem cell transplantation. Because of MSC production of a wide range of cytokines and growth factors, these cells are included in the therapy of graft-versus-host disease (GVHD). A number of clinical studies have demonstrated safety and efficacy of MSC-based therapy in acute GVHD. Japan and some other countries approved biomedical cell products on the base of allogeneic bone marrow (BM) MSCs as medical agents for acute GVHD treatment. Besides, MSCs may form BM stroma and improve hematopoiesis. Simultaneous transplantation of hematopoietic stem cells and MSCs effectively improved engraftment and prevented GVHD in transplantation of umbilical cord blood and human leukocyte antigens-incompatible BM stem cells. The review presents the analysis of clinical studies of MSCs in allogeneic hematopoietic stem cell transplantation and discusses different approaches for improvement of MSC-based GVHD treatment and prophylaxis.
Collapse
|
16
|
You J, Bragin A, Liu H, Li L. Preclinical studies of transcranial photobiomodulation in the neurological diseases. TRANSLATIONAL BIOPHOTONICS 2021. [DOI: 10.1002/tbio.202000024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Jing You
- Department of Biomedical Engineering University of North Texas Denton Texas USA
| | - Anatol Bragin
- Department of Neurology University of California Los Angeles Los Angeles California USA
- Brain Research Institute University of California Los Angeles Los Angeles California USA
| | - Hanli Liu
- Department of Bioengineering University of Texas at Arlington Arlington Texas USA
| | - Lin Li
- Department of Biomedical Engineering University of North Texas Denton Texas USA
- Department of Neurology University of California Los Angeles Los Angeles California USA
| |
Collapse
|
17
|
Mallis P, Michalopoulos E, Chatzistamatiou T, Stavropoulos-Giokas C. Mesenchymal stromal cells as potential immunomodulatory players in severe acute respiratory distress syndrome induced by SARS-CoV-2 infection. World J Stem Cells 2020; 12:731-751. [PMID: 32952855 PMCID: PMC7477656 DOI: 10.4252/wjsc.v12.i8.731] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/10/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 and the related coronavirus disease-19 (COVID-19) is a worldwide emerging situation, which was initially reported in December 2019 in Wuhan, China. Currently, more than 7258842 new cases, and more than 411879 deaths have been reported globally. This new highly transmitted coronavirus is responsible for the development of severe acute respiratory distress syndrome. Due to this disorder, a great number of patients are hospitalized in the intensive care unit followed by connection to extracorporeal membrane oxygenation for breath supporting and survival. Severe acute respiratory distress syndrome is mostly accompanied by the secretion of proinflammatory cytokines, including interleukin (IL)-2, IL-6, IL-7, granulocyte colony-stimulating factor (GSCF), interferon-inducible protein 10 (IP10), monocyte chemotactic protein-1 (MCP1), macrophage inflammatory protein 1A (MIP1A), and tumor necrosis factor alpha (TNF-α), an event which is known as "cytokine storm". Further disease pathology involves a generalized modulation of immune responses, leading to fatal multiorgan failure. Currently, no specific treatment or vaccination against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been developed. Mesenchymal stromal cells (MSCs), which are known for their immunosuppressive actions, could be applied as an alternative co-therapy in critically-ill COVID-19 patients. Specifically, MSCs can regulate the immune responses through the conversion of Th1 to Th2, activation of M2 macrophages, and modulation of dendritic cells maturation. These key immunoregulatory properties of MSCs may be exerted either by produced soluble factors or by cell-cell contact interactions. To date, several clinical trials have been registered to assess the safety, efficacy, and therapeutic potential of MSCs in COVID-19. Moreover, MSC treatment may be effective for the reversion of ground-glass opacity of damaged lungs and reduce the tissue fibrosis. Taking into account the multifunctional properties of MSCs, the proposed stem-cell-based therapy may be proven significantly effective in critically-ill COVID-19 patients. The current therapeutic strategy may improve the patient's overall condition and in parallel may decrease the mortality rate of the current disease.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece.
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | | | | |
Collapse
|
18
|
Kim YG, Choi J, Kim K. Mesenchymal Stem Cell‐Derived Exosomes for Effective Cartilage Tissue Repair and Treatment of Osteoarthritis. Biotechnol J 2020; 15:e2000082. [DOI: 10.1002/biot.202000082] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Young Guk Kim
- Department of Chemical and Biochemical Engineering Dongguk University 30 Pildong‐ro 1‐gil Seoul 04620 Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering Chung‐Ang University 47 Heukseok‐ro Seoul 06911 Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering Dongguk University 30 Pildong‐ro 1‐gil Seoul 04620 Republic of Korea
| |
Collapse
|
19
|
Shammaa R, El-Kadiry AEH, Abusarah J, Rafei M. Mesenchymal Stem Cells Beyond Regenerative Medicine. Front Cell Dev Biol 2020; 8:72. [PMID: 32133358 PMCID: PMC7040370 DOI: 10.3389/fcell.2020.00072] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are competent suitors of cellular therapy due to their therapeutic impact on tissue degeneration and immune-based pathologies. Additionally, their homing and immunomodulatory properties can be exploited in cancer malignancies to transport pharmacological entities, produce anti-neoplastic agents, or induce anti-tumor immunity. Herein, we create a portfolio for MSC properties, showcasing their distinct multiple therapeutic utilities and successes/challenges thereof in both animal studies and clinical trials. We further highlight the promising potential of MSCs not only in cancer management but also in instigating tumor-specific immunity - i.e., cancer vaccination. Finally, we reflect on the possible reasons impeding the clinical advancement of MSC-based cancer vaccines to assist in contriving novel methodologies from which a therapeutic milestone might emanate.
Collapse
Affiliation(s)
- Riam Shammaa
- Canadian Centre for Regenerative Therapy, Toronto, ON, Canada.,IntelliStem Technologies Inc., Toronto, ON, Canada.,Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Abed El-Hakim El-Kadiry
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Jamilah Abusarah
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada.,Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
20
|
Black L, Zorina T. Cell-based immunomodulatory therapy approaches for type 1 diabetes mellitus. Drug Discov Today 2020; 25:380-391. [DOI: 10.1016/j.drudis.2019.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/11/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
|
21
|
Conditioned Medium from Adipose-Derived Stem Cell Inhibits Jurkat Cell Proliferation through TGF- β1 and p38/MAPK Pathway. Anal Cell Pathol (Amst) 2020; 2019:2107414. [PMID: 31934530 PMCID: PMC6942699 DOI: 10.1155/2019/2107414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/10/2019] [Accepted: 08/26/2019] [Indexed: 02/05/2023] Open
Abstract
Background Since the first report on the immunomodulatory and immunosuppressive properties of Adipose-Derived Stem Cells (ADSCs), many studies have elucidated the underlying molecular mechanism of their suppressive activity on mixed lymphocyte reaction (MLR). However, a gap exists in our understanding of the molecular mechanism of ADSC-conditioned medium (ADSC-CM) on MLR. Methods ADSCs were isolated from Human Adipose Tissues, and Enzyme-linked Immunosorbent Assay (ELISA) was used to identify the concentration of transforming growth factor β1 (TGF-β1) in ADSC-CM. The transcript abundance of TGF-β1, as well as that of insulin-like growth factor binding protein 3 (IGF-BP3), was evaluated using qRT-PCR on Jurkat cells cultured in ADSC-CM for 24 hours. The proliferation of the Jurkat cells was assessed using cell cycle assay. Western blotting was performed to identify potential signaling molecules involved in the ADSC-CM-induced inhibition of Jurkat cell proliferation. Results The findings confirm that the isolated ADSCs demonstrate classic ADSC characteristics. The level of TGF-β1 was found to be low in ADSC-CM, as assessed by ELISA. Jurkat cells grown in ADSC-CM show reduced gene expression of TGF-β1 and IGF-BP3 compared with that of the control group. Furthermore, western blotting of ADSC-CM grown Jurkat cells that were blocked at the G0/G1 stage indicates that ADSC-CM decreases the protein expression of pP38 in a dose-dependent manner. Conclusion ADSC-CM can inhibit Jurkat cell proliferation through the TGF-β1-p38 signaling pathway.
Collapse
|
22
|
Ciciarello M, Corradi G, Loscocco F, Visani G, Monaco F, Cavo M, Curti A, Isidori A. The Yin and Yang of the Bone Marrow Microenvironment: Pros and Cons of Mesenchymal Stromal Cells in Acute Myeloid Leukemia. Front Oncol 2019; 9:1135. [PMID: 31709192 PMCID: PMC6823864 DOI: 10.3389/fonc.2019.01135] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have, for a long time, been recognized as pivotal contributors in the set up and maintenance of the hematopoietic stem cell (HSC) niche, as well as in the development and differentiation of the lympho-hematopoietic system. MSCs also have a unique immunomodulatory capacity, which makes them able to affect, both in vitro and in vivo, the function of immune cells. These features, namely the facilitation of stem cell engraftment and the inhibition of lymphocyte responses, have both proven essential for successful allogeneic stem cell transplantation (allo-SCT), which remains the only curative option for several hematologic malignancies. For example, in steroid-refractory acute graft-vs. host disease developing after allo-SCT, MSCs have produced significant results and are now considered a treatment option. However, more recently, the other side of the MSC coin has been unveiled, because of their emerging role in creating a protective and immune-tolerant microenvironment able to support the survival of leukemic cells and affect the response to therapies. In this light, it has been proposed that the failure of current treatments to efficiently override the stroma-mediated protection of leukemic cells accounts for the high rate of relapse in acute myeloid leukemia, at least in part. In this review, we will focus on emerging microenvironment-driven mechanisms conferring a survival advantage to leukemic cells overt physiological HSCs. This body of evidence increasingly highlights the opportunity to consider tumor-microenvironment interactions when designing new therapeutic strategies.
Collapse
Affiliation(s)
- Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giulia Corradi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Federica Monaco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Antonio Curti
- Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| |
Collapse
|
23
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
24
|
Tompkins BA, Balkan W, Winkler J, Gyöngyösi M, Goliasch G, Fernández-Avilés F, Hare JM. Preclinical Studies of Stem Cell Therapy for Heart Disease. Circ Res 2019; 122:1006-1020. [PMID: 29599277 DOI: 10.1161/circresaha.117.312486] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As part of the TACTICS (Transnational Alliance for Regenerative Therapies in Cardiovascular Syndromes) series to enhance regenerative medicine, here, we discuss the role of preclinical studies designed to advance stem cell therapies for cardiovascular disease. The quality of this research has improved over the past 10 to 15 years and overall indicates that cell therapy promotes cardiac repair. However, many issues remain, including inability to provide complete cardiac recovery. Recent studies question the need for intact cells suggesting that harnessing what the cells release is the solution. Our contribution describes important breakthroughs and current directions in a cell-based approach to alleviating cardiovascular disease.
Collapse
Affiliation(s)
- Bryon A Tompkins
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Wayne Balkan
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Johannes Winkler
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Mariann Gyöngyösi
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Georg Goliasch
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Francisco Fernández-Avilés
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.).
| |
Collapse
|
25
|
Segaliny AI, Cheng JL, Farhoodi HP, Toledano M, Yu CC, Tierra B, Hildebrand L, Liu L, Liao MJ, Cho J, Liu D, Sun L, Gulsen G, Su MY, Sah RL, Zhao W. Combinatorial targeting of cancer bone metastasis using mRNA engineered stem cells. EBioMedicine 2019; 45:39-57. [PMID: 31281099 PMCID: PMC6642316 DOI: 10.1016/j.ebiom.2019.06.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bone metastases are common and devastating to cancer patients. Existing treatments do not specifically target the disease sites and are therefore ineffective and systemically toxic. Here we present a new strategy to treat bone metastasis by targeting both the cancer cells ("the seed"), and their surrounding niche ("the soil"), using stem cells engineered to home to the bone metastatic niche and to maximise local delivery of multiple therapeutic factors. METHODS We used mesenchymal stem cells engineered using mRNA to simultaneously express P-selectin glycoprotein ligand-1 (PSGL-1)/Sialyl-Lewis X (SLEX) (homing factors), and modified versions of cytosine deaminase (CD) and osteoprotegerin (OPG) (therapeutic factors) to target and treat breast cancer bone metastases in two mouse models, a xenograft intratibial model and a syngeneic model of spontaneous bone metastasis. FINDINGS We first confirmed that MSC engineered using mRNA produced functional proteins (PSGL-1/SLEX, CD and OPG) using various in vitro assays. We then demonstrated that mRNA-engineered MSC exhibit enhanced homing to the bone metastatic niche likely through interactions between PSGL-1/SLEX and P-selectin expressed on tumour vasculature. In both the xenograft intratibial model and syngeneic model of spontaneous bone metastasis, engineered MSC can effectively kill tumour cells and preserve bone integrity. The engineered MSC also exhibited minimal toxicity in vivo, compared to its non-targeted chemotherapy counterpart (5-fluorouracil). INTERPRETATION Our combinatorial targeting of both the cancer cells and the niche represents a simple, safe and effective way to treat metastatic bone diseases, otherwise difficult to manage with existing strategies. It can also be applied to other cell types (e.g., T cells) and cargos (e.g., genome editing components) to treat a broad range of cancer and other complex diseases. FUND: National Institutes of Health, National Cancer Institute of the National Institutes of Health, Department of Defense, California Institute of Regenerative Medicine, National Science Foundation, Baylx Inc., and Fondation ARC pour la recherche sur le cancer.
Collapse
Affiliation(s)
- Aude I Segaliny
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jason L Cheng
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Henry P Farhoodi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael Toledano
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Chih Chun Yu
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Beatrice Tierra
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Leanne Hildebrand
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Linan Liu
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael J Liao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jaedu Cho
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Dongxu Liu
- Department of Civil and Environmental Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Lizhi Sun
- Department of Civil and Environmental Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Gultekin Gulsen
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert L Sah
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
26
|
Chen L, Shi K, Andersen TL, Qiu W, Kassem M. KIAA1199 is a secreted molecule that enhances osteoblastic stem cell migration and recruitment. Cell Death Dis 2019; 10:126. [PMID: 30755597 PMCID: PMC6372631 DOI: 10.1038/s41419-018-1202-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
Factors mediating mobilization of osteoblastic stem and progenitor cells from their bone marrow niche to be recruited to bone formation sites during bone remodeling are poorly known. We have studied secreted factors present in the bone marrow microenvironment and identified KIAA1199 (also known as CEMIP, cell migration inducing hyaluronan binding protein) in human bone biopsies as highly expressed in osteoprogenitor reversal cells (Rv.C) recruited to the eroded surfaces (ES), which are the future bone formation sites. In vitro, KIAA1199 did not affect the proliferation of human osteoblastic stem cells (also known as human bone marrow skeletal or stromal stem cells, hMSCs); but it enhanced cell migration as determined by scratch assay and trans-well migration assay. KIAA1199 deficient hMSCs (KIAA1199down) exhibited significant changes in cell size, cell length, ratio of cell width to length and cell roundness, together with reduction of polymerization actin (F-actin) and changes in phos-CFL1 (cofflin1), phos-LIMK1 (LIM domain kinase 1) and DSTN (destrin), key factors regulating actin cytoskeletal dynamics and cell motility. Moreover, KIAA1199down hMSC exhibited impaired Wnt signaling in TCF-reporter assay and decreased expression of Wnt target genes and these effects were rescued by KIAA1199 treatment. Finally, KIAA1199 regulated the activation of P38 kinase and its associated changes in Wnt-signaling. Thus, KIAA1199 is a mobilizing factor that interacts with P38 and Wnt signaling, and induces changes in actin cytoskeleton, as a mechanism mediating recruitment of hMSC to bone formation sites.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
| | - Kaikai Shi
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Weimin Qiu
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
- The Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark.
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
27
|
|
28
|
Sultan N, Amin LE, Zaher AR, Scheven BA, Grawish ME. Dental pulp stem cells: Novel cell-based and cell-free therapy for peripheral nerve repair. World J Stomatol 2019; 7:1-19. [DOI: 10.5321/wjs.v7.i1.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/15/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
The regeneration of peripheral nerves comprises complicated steps involving a set of cellular and molecular events in distal nerve stumps with axonal sprouting and remyelination. Stem cell isolation and expansion for peripheral nerve repair (PNR) can be achieved using a wide diversity of prenatal and adult tissues, such as bone marrow or brain tissues. The ability to obtain stem cells for cell-based therapy (CBT) is limited due to donor site morbidity and the invasive nature of the harvesting process. Dental pulp stem cells (DPSCs) can be relatively and simply isolated from the dental pulps of permanent teeth, extracted for surgical or orthodontic reasons. DPSCs are of neural crest origin with an outstanding ability to differentiate into multiple cell lineages. They have better potential to differentiate into neural and glial cells than other stem cell sources through the expression and secretion of certain markers and a range of neurotropic factors; thus, they should be considered a good choice for PNR using CBT. In addition, these cells have paracrine effects through the secretion of neurotrophic growth factors and extracellular vesicles, which can enhance axonal growth and remyelination by decreasing the number of dying cells and activating local inhabitant stem cell populations, thereby revitalizing dormant or blocked cells, modulating the immune system and regulating inflammatory responses. The use of DPSC-derived secretomes holds great promise for controllable and manageable therapy for peripheral nerve injury. In this review, up-to-date information about the neurotrophic and neurogenic properties of DPSCs and their secretomes is provided.
Collapse
Affiliation(s)
- Nessma Sultan
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Laila E Amin
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed R Zaher
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Ben A Scheven
- School of Dentistry, Oral Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, United Kingdom
| | - Mohammed E Grawish
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
29
|
Mesenchymal Stromal Cells: Role in the BM Niche and in the Support of Hematopoietic Stem Cell Transplantation. Hemasphere 2018; 2:e151. [PMID: 31723790 PMCID: PMC6745957 DOI: 10.1097/hs9.0000000000000151] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/21/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are key elements in the bone marrow (BM) niche where they interact with hematopoietic stem progenitor cells (HSPCs) by offering physical support and secreting soluble factors, which control HSPC maintenance and fate. Although necessary for their maintenance, MSCs are a rare population in the BM, they are plastic adherent and can be ex vivo expanded to reach numbers adequate for clinical use. In light of HSPC supportive properties, MSCs have been employed in phase I/II clinical trials of hematopoietic stem cell transplantation (HSCT) to facilitate engraftment of hematopoietic stem cells (HSCs). Moreover, they have been utilized to expand ex vivo HSCs before clinical use. The available clinical evidence from these trials indicate that MSC administration is safe, as no acute and long-term adverse events have been registered in treated patients, and may be efficacious in promoting hematopoietic engraftment after HSCT. In this review, we critically discuss the role of MSCs as component of the BM niche, as recent advances in defining different mesenchymal populations in the BM have considerably increased our understanding of this complex environment. Moreover, we will revise published literature on the use of MSCs to support HSC engraftment and expansion, as well as consider potential new MSC application in the clinical context of ex vivo gene therapy with autologous HSC.
Collapse
|
30
|
Tao Z, Tan S, Chen W, Chen X. Stem Cell Homing: a Potential Therapeutic Strategy Unproven for Treatment of Myocardial Injury. J Cardiovasc Transl Res 2018; 11:403-411. [PMID: 30324254 DOI: 10.1007/s12265-018-9823-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Despite advances in the prevention and therapeutic modalities of ischemic heart disease, morbidity and mortality post-infarction heart failure remain big challenges in modern society. Stem cell therapy is emerging as a promising therapeutic strategy. Stem cell homing, the ability of stem cells to find their destination, is receiving more attention. Identification of specific cues and understanding the signaling pathways that direct stem cells to targeted destination will improve stem cell homing efficiency. This review discusses the cellular and molecular mechanism of stem cell homing at length in the light of literature and analyzes the problem and considerations of this approach as a treatment strategy for the treatment of ischemic heart disease clinically.
Collapse
Affiliation(s)
- Zhonghao Tao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Shihua Tan
- National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
31
|
Fu Y, Karbaat L, Wu L, Leijten J, Both SK, Karperien M. Trophic Effects of Mesenchymal Stem Cells in Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 23:515-528. [PMID: 28490258 DOI: 10.1089/ten.teb.2016.0365] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are considered to hold great therapeutic value for cell-based therapy and for tissue regeneration in particular. Recent evidence indicates that the main underlying mechanism for MSCs' beneficial effects in tissue regeneration is based on their capability to produce a large variety of bioactive trophic factors that stimulate neighboring parenchymal cells to start repairing damaged tissues. These new findings could potentially replace the classical paradigm of MSC differentiation and cell replacement. These bioactive factors have diverse actions like modulating the local immune system, enhancing angiogenesis, preventing cell apoptosis, and stimulating survival, proliferation, and differentiation of resident tissue specific cells. Therefore, MSCs are referred to as conductors of tissue repair and regeneration by secreting trophic mediators. In this review article, we have summarized the studies that focused on the trophic effects of MSC within the context of tissue regeneration. We will also highlight the various underlying mechanisms used by MSCs to act as trophic mediators. Besides the secretion of growth factors, we discuss two additional mechanisms that are likely to mediate MSC's beneficial effects in tissue regeneration, namely the production of extracellular vesicles and the formation of membrane nanotubes, which can both connect different cells and transfer a variety of trophic factors varying from proteins to mRNAs and miRNAs. Furthermore, we postulate that apoptosis of the MSCs is an integral part of the trophic effect during tissue repair.
Collapse
Affiliation(s)
- Yao Fu
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Lisanne Karbaat
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Ling Wu
- 2 Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, Los Angeles, California
| | - Jeroen Leijten
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Sanne K Both
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Marcel Karperien
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| |
Collapse
|
32
|
Macrin D, Joseph JP, Pillai AA, Devi A. Eminent Sources of Adult Mesenchymal Stem Cells and Their Therapeutic Imminence. Stem Cell Rev Rep 2018; 13:741-756. [PMID: 28812219 DOI: 10.1007/s12015-017-9759-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the recent times, stem cell biology has garnered the attention of the scientific fraternity and the general public alike due to the immense therapeutic potential that it holds in the field of regenerative medicine. A breakthrough in this direction came with the isolation of stem cells from human embryo and their differentiation into cell types of all three germ layers. However, the isolation of mesenchymal stem cells from adult tissues proved to be advantageous over embryonic stem cells due to the ethical and immunological naivety. Mesenchymal Stem Cells (MSCs) isolated from the bone marrow were found to differentiate into multiple cell lineages with the help of appropriate differentiation factors. Furthermore, other sources of stem cells including adipose tissue, dental pulp, and breast milk have been identified. Newer sources of stem cells have been emerging recently and their clinical applications are also being studied. In this review, we examine the eminent sources of Mesenchymal Stem Cells (MSCs), their immunophenotypes, and therapeutic imminence.
Collapse
Affiliation(s)
- Dannie Macrin
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India
| | - Joel P Joseph
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India
| | | | - Arikketh Devi
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
33
|
Park BN, Lim TS, Yoon JK, An YS. In vivo tracking of intravenously injected mesenchymal stem cells in an Alzheimer's animal model. Cell Transplant 2018; 27:1203-1209. [PMID: 30008224 PMCID: PMC6434469 DOI: 10.1177/0963689718788067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose: The purpose of this study was to investigate how intravenously injected bone marrow-derived mesenchymal stem cells (BMSCs) are distributed in the body of an Alzheimer’s disease (AD) animal model. Methods: Stem cells were collected from bone marrow of mice and labeled with Indium-111 (111In). The 111In-labeled BMSCs were infused intravenously into 3×Tg-AD mice in the AD group and non-transgenic mice (B6129SF2/J) as controls. Biodistribution was evaluated with a gamma counter and gamma camera 24 and 48 h after injecting the stem cells. Results: A gamma count of the brain showed a higher distribution of labeled cells in the AD model than in the control group at 24 (p = .0004) and 48 h (p = .0016) after injection of the BMSCs. Similar results were observed by gamma camera imaging (i.e., brain uptake in the AD model was significantly higher than that in the control group). Among the other organs, uptake by the spleen was the highest in both groups. More BMSCs were found in the lungs of the control group than in those of the AD group. Conclusions: These results suggest that more intravenously infused BMSCs reached the brain in the AD model than in the control group, but the numbers of stem cells reaching the brain was very small.
Collapse
Affiliation(s)
- Bok-Nam Park
- 1 Department of Nuclear Medicine and Molecular Imaging, Suwon, Korea
| | - Tae Sung Lim
- 2 Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Joon-Kee Yoon
- 1 Department of Nuclear Medicine and Molecular Imaging, Suwon, Korea
| | - Young-Sil An
- 1 Department of Nuclear Medicine and Molecular Imaging, Suwon, Korea
| |
Collapse
|
34
|
Approaches to the removal of T-lymphocytes to minimize graft-versus-host disease in patients with primary immunodeficiencies who do not have a matched sibling donor. Curr Opin Allergy Clin Immunol 2018; 17:414-420. [PMID: 28968273 DOI: 10.1097/aci.0000000000000402] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Since the advent of T-lymphocyte depletion in hematopoietic stem cell transplantation (HSCT) for primary immunodeficiency, survival following this procedure has remained poor compared to results when using matched sibling or matched unrelated donors, over the last 40 years. However, three new techniques are radically altering the approach to HSCT for those with no matched donor, particularly those with primary immunodeficiencies which are not severe combined immunodeficiency. RECENT FINDINGS Three main techniques of T-lymphocyte depletion are altering donor choice for patients with primary immunodeficiencies and have improved transplant survival for primary immunodeficiencies to over 90%, equivalent to that for matched sibling and matched unrelated donor transplants. CD3 T cell receptor (TCR)αβ CD19 depletion, CD45RA depletion and use of posttransplant cyclophosphamide give similar overall survival of 90%, although viral reactivation remains a concern. Further modification of CD3 TCRαβ CD19 depletion by adding back inducible caspase-9 suicide gene-modified CD3 TCRαβ T-lymphocytes may further improve outcomes for patients with systemic viral infection. SUMMARY Over the last 5 years, the outcomes of HSCT using new T-lymphocyte depletion methods have improved to the extent that they are equivalent to outcomes of matched sibling donors and may be preferred in the absence of a fully matched sibling donor, over an unrelated donor to reduce the risk of graft versus host disease.
Collapse
|
35
|
Mohammadi M, Mohammadi M, Rezaee MA, Ghadimi T, Abolhasani M, Rahmani MR. Effect of gestational age on migration ability of the human umbilical cord vein mesenchymal stem cells. Adv Med Sci 2018; 63:119-126. [PMID: 29120852 DOI: 10.1016/j.advms.2017.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/11/2017] [Accepted: 08/22/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Migration ability of mesenchymal stem cells (MSCs) towards chemotactic mediators is a determinant factor in cell therapy. MSCs derived from different sources show different properties. Here we compared the migration ability of the term and the pre-term human umbilical cord vein MSCs (hUCV-MSCs). MATERIALS/METHODS MSCs were isolated from term and pre-term umbilical cord vein, and cultured to passage 3-4. Migration rate of both groups was assessed in the presence of 10% FBS using chemotaxis assay. Surface expression of CXCR4 was measured by flow cytometery. The relative gene expression of CXCR4, IGF1-R, PDGFRα, MMP-2, MMP-9, MT1-MMP and TIMP-2 were evaluated using real time PCR. RESULTS The isolation rate of the pre-term hUCV-MSCs was higher than the term hUCV-MSCs. Phenotype characteristics and differentiation ability of the term and pre-term hUCV-MSCs were not different. The migration rate of the pre-term hUCV-MSCs was more than the term hUCV-MSCs. Gene and surface expressions of the CXCR4 were both significantly higher in the pre-term hUCV-MSCs (P≤0.05). The mRNA levels of PDGFRα, MMP-2, MMP-9, MT1-MMP and TIMP-2 showed no significant difference between the two groups. CONCLUSION Our results showed that the gestational age can affect the migration ability of the hUCV-MSCs.
Collapse
Affiliation(s)
- Mobin Mohammadi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology and Hematology, Faculty of medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Mohammadi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Rezaee
- Zoonosis Research center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Medical Laboratory Sciences, Faculty of Paramedicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Tayyeb Ghadimi
- Department of Surgery, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Massume Abolhasani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology and Hematology, Faculty of medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Reza Rahmani
- Department of Immunology and Hematology, Faculty of medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran; Zoonosis Research center, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
36
|
Human Adipose Stromal Cells Increase Survival and Mesenteric Perfusion Following Intestinal Ischemia and Reperfusion Injury. Shock 2018; 46:75-82. [PMID: 26796571 DOI: 10.1097/shk.0000000000000571] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Intestinal ischemia can quickly escalate to bowel necrosis and perforation. Transplantation of stem cells presents a novel treatment modality for this problem. We hypothesized that: human adipose-derived stromal cells (hASCs) would increase survival and mesenteric perfusion to a greater degree compared with differentiated cellular controls following ischemic intestinal injury, and improved outcomes with hASC therapy would be associated with preservation of intestinal histological and tight junction architecture, and lower levels of systemic inflammation following intestinal injury. METHODS hASCs and keratinocytes (differentiated cellular control) were cultured on polystyrene flasks at 37°C in 5% CO2 in air. Adult male C57Bl6J mice were anesthetized and a midline laparotomy performed. The intestines were eviscerated, the small bowel mesenteric root identified, and intestinal ischemia was established by temporarily occluding the superior mesenteric artery for 60 min with a noncrushing vascular clamp. Following ischemia, the clamp was removed, and the intestines were returned to the abdominal cavity. Before abdominal closure, 2 million hASCs or keratinocytes in 250 μL of phosphate-buffered saline (carrier for cells and control solution) were infused into the peritoneum. Animals were allowed to recover for 12 or 24 h (perfusion, histology, cytokine, and immunofluoresence studies), or 7 days (survival studies). Intestinal perfusion was assessed by laser Doppler imaging. Intestinal tissue segments were stained with hematoxylin and eosin, as well as antibodies for the tight junction protein claudin-1. Separate aliquots of intestine, liver, and lung tissue were homogenized and assessed for inflammatory cytokines via multiplex beaded assay. RESULTS Animals administered hASCs following intestinal ischemia and reperfusion (I/R) injury had significantly greater 7-day survival and better postischemic recovery of mesenteric perfusion compared with vehicle or keratinocyte therapy. hASCs also abated intestinal mucosal destruction, facilitated preservation of intestinal tight junctions, and decreased the systemic inflammatory response to injury. CONCLUSIONS Human adipose-derived stromal cells improved survival and mesenteric perfusion and attenuated the mucosal damage associated with intestinal I/R injury. hASCs should be considered as a plausible cell source for novel cellular treatment plans following intestinal ischemia.
Collapse
|
37
|
Bogdanowicz DR, Lu HH. Designing the stem cell microenvironment for guided connective tissue regeneration. Ann N Y Acad Sci 2018; 1410:3-25. [PMID: 29265419 DOI: 10.1111/nyas.13553] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/20/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022]
Abstract
Adult mesenchymal stem cells (MSCs) are an attractive cell source for regenerative medicine because of their ability to self-renew and their capacity for multilineage differentiation and tissue regeneration. For connective tissues, such as ligaments or tendons, MSCs are vital to the modulation of the inflammatory response following acute injury while also interacting with resident fibroblasts to promote cell proliferation and matrix synthesis. To date, MSC injection for connective tissue repair has yielded mixed results in vivo, likely due to a lack of appropriate environmental cues to effectively control MSC response and promote tissue healing instead of scar formation. In healthy tissues, stem cells reside within a complex microenvironment comprising cellular, structural, and signaling cues that collectively maintain stemness and modulate tissue homeostasis. Changes to the microenvironment following injury regulate stem cell differentiation, trophic signaling, and tissue healing. Here, we focus on models of the stem cell microenvironment that are used to elucidate the mechanisms of stem cell regulation and inspire functional approaches to tissue regeneration. Recent studies in this frontier area are highlighted, focusing on how microenvironmental cues modulate MSC response following connective tissue injury and, more importantly, how this unique cell environment can be programmed for stem cell-guided tissue regeneration.
Collapse
Affiliation(s)
- Danielle R Bogdanowicz
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, New York
| | - Helen H Lu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
38
|
Affiliation(s)
- H T Hassan
- Institute of Medical Sciences, University of Lincoln, UK.
| | | |
Collapse
|
39
|
Sui BD, Hu CH, Liu AQ, Zheng CX, Xuan K, Jin Y. Stem cell-based bone regeneration in diseased microenvironments: Challenges and solutions. Biomaterials 2017; 196:18-30. [PMID: 29122279 DOI: 10.1016/j.biomaterials.2017.10.046] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/21/2017] [Accepted: 10/28/2017] [Indexed: 12/17/2022]
Abstract
Restoration of extensive bone loss and defects remain as an unfulfilled challenge in modern medicine. Given the critical contributions to bone homeostasis and diseases, mesenchymal stem cells (MSCs) have shown great promise to jumpstart and facilitate bone healing, with immense regenerative potential in both pharmacology-based endogenous MSC rescue/mobilization in skeletal diseases and emerging application of MSC transplantation in bone tissue engineering and cytotherapy. However, efficacy of MSC-based bone regeneration was not always achieved; particularly, fulfillment of MSC-mediated bone healing in diseased microenvironments of host comorbidities remains as a major challenge. Indeed, impacts of diseased microenvironments on MSC function rely not only on the dynamic regulation of resident MSCs by surrounding niche to convoy pathological signals of bone, but also on the profound interplay between transplanted MSCs and recipient components that mediates and modulates therapeutic effects on skeletal conditions. Accordingly, novel solutions have recently been developed, including improving resistance of MSCs to diseased microenvironments, recreating beneficial microenvironments to guarantee MSC-based regeneration, and usage of subcellular vesicles of MSCs in cell-free therapies. In this review, we summarize state-of-the-art knowledge regarding applications and challenges of MSC-mediated bone healing, further offering principles and effective strategies to optimize MSC-based bone regeneration in aging and diseases.
Collapse
Affiliation(s)
- Bing-Dong Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - An-Qi Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Kun Xuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
40
|
|
41
|
Erices AA, Allers CI, Conget PA, Rojas CV, Minguell JJ. Human Cord Blood-Derived Mesenchymal Stem Cells Home and Survive in the Marrow of Immunodeficient Mice after Systemic Infusion. Cell Transplant 2017; 12:555-61. [PMID: 14579923 DOI: 10.3727/000000003108747154] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone marrow is the residence site of mesenchymal stem cells (MSC), which upon commitment and maturation develop into several mesenchymal phenotypes. Recently, we have described the presence of MSC in human cord blood (cbMSC) and informed that their properties are the same as those for MSC obtained from adult bone marrow. In this study we have investigated the capability of transplanted cbMSC to home and survive in the marrow of unconditioned nude mice. cbMSC utilized for transplantation studies were characterized by morphology, differentiation potential, and immunophenotype. After transplantation by systemic infusion, human DNA (as detected by PCR amplification of human-specific β-globin gene) was detected in the marrow of recipients as well as in ex vivo-expanded stromal cells prepared from the marrow of transplanted animals. These results demonstrate homing and survival of cbMSC into the recipient marrow and also suggest a mesenchymal-orientated fate of engrafted cells, because human DNA was also detected in cells of other recipient tissues, like cardiac muscle, teeth, and spleen.
Collapse
Affiliation(s)
- Alejandro A Erices
- Programa Terapias Génicas y Celulares, INTA, Universidad de Chile, Santiago, Chile.
| | | | | | | | | |
Collapse
|
42
|
Ouyang HW, Goh JCH, Lee EH. Viability of Allogeneic Bone Marrow Stromal Cells following Local Delivery into Patella Tendon in Rabbit Model. Cell Transplant 2017; 13:649-658. [DOI: 10.3727/000000004783983549] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bone marrow stromal cells are potentially useful for tendon repair and regeneration. To provide lasting benefits, the seeded cells must survive implantation at local tendon sites. Our objective was to determine the in vivo fate of allogeneic bone marrow stromal cells (bMSCs) at different time points after implantation into patella tendon defects (i.e., at 2, 3, 5, and 8 weeks). The protocol involved the labeling of bMSCs with green fluorescent protein (GFP) or carboxyfluorescein diacetate (CFDA) before implantation. A window defect (5 × 5 mm) was created at the central portion of rabbit patella tendon and subsequently treated with GFP- or CFDA-marked bMSCs. The marked bMSCs were loaded into the window defect with fibrin glue. Upon sacrifice of the rabbits at the different time points, the implant site of the patellar tendon was immediately retrieved and the viability of the labeled cells was assessed under confocal microscopy. The results showed that the seeded bMSCs remained viable within the tendon wound site for at least 8 weeks after implantation. The cell morphology was changed from a round shape at 2 weeks to a spindle shape at 5 weeks after implantation. This study demonstrated that the bMSCs remained viable for prolonged periods after implantation and therefore have the potential to influence the formation and remodeling of neotendon tissue after tendon repair.
Collapse
Affiliation(s)
- Hong Wei Ouyang
- Department of Orthopaedic Surgery, National University of Singapore, Singapore 119260
| | - James C. H. Goh
- Department of Orthopaedic Surgery, National University of Singapore, Singapore 119260
- Division of Bioengineering, National University of Singapore, Singapore 119260
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, National University of Singapore, Singapore 119260
| |
Collapse
|
43
|
Li F, Guo X, Chen SY. Function and Therapeutic Potential of Mesenchymal Stem Cells in Atherosclerosis. Front Cardiovasc Med 2017; 4:32. [PMID: 28589127 PMCID: PMC5438961 DOI: 10.3389/fcvm.2017.00032] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/01/2017] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a complicated disorder and largely attributable to dyslipidaemia and chronic inflammation. Despite therapeutic advances over past decades, atherosclerosis remains the leading cause of mortality worldwide. Due to their capability of immunomodulation and tissue regeneration, mesenchymal stem cells (MSCs) have evolved as an attractive therapeutic agent in various diseases including atherosclerosis. Accumulating evidences support the protective role of MSCs in all stages of atherosclerosis. In this review, we highlight the current understanding of MSCs including their characteristics such as molecular markers, tissue distribution, migratory property, immune-modulatory competence, etc. We also summarize MSC functions in animal models of atherosclerosis. MSC transplantation is able to modulate cytokine and chemokine secretion, reduce endothelial dysfunction, promote regulatory T cell function, decrease dyslipidemia, and stabilize vulnerable plaques during atherosclerosis development. In addition, MSCs may migrate to lesions where they develop into functional cells during atherosclerosis formation. Finally, the perspectives of MSCs in clinical atherosclerosis therapy are discussed.
Collapse
Affiliation(s)
- Feifei Li
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA.,The Department of Cardiovascular Surgery, Union Hospital, Wuhan, China
| | - Xia Guo
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| |
Collapse
|
44
|
Intravenous mesenchymal stromal cell therapy for inflammatory bowel disease: Lessons from the acute graft versus host disease experience. Cytotherapy 2017; 19:655-667. [PMID: 28433516 DOI: 10.1016/j.jcyt.2017.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/01/2017] [Accepted: 03/06/2017] [Indexed: 12/18/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are primitive, supportive cells of the bone marrow with tri-lineage potential to differentiate into bone, cartilage, fat and muscle. These cells possess both in vitro and in vivo immunomodulatory and wound-healing properties. Several studies have demonstrated efficacy of intravenously administered BMSCs in treating acute graft-versus-host disease (GvHD). Use of intravenous (IV) BMSCs in inflammatory bowel diseases (IBD) in humans has been limited to small studies in adults, but results have been promising. There remain many unanswered questions regarding safety, tolerability, effectiveness and optimal use of BMSCs to treat IBD, particularly in immunocompromised patients. This article reviews the evidence for using BMSCs to treat acute GvHD and how this experience may inform the potential use of BMSCs as a treatment for IBD.
Collapse
|
45
|
Grégoire C, Lechanteur C, Briquet A, Baudoux É, Baron F, Louis E, Beguin Y. Review article: mesenchymal stromal cell therapy for inflammatory bowel diseases. Aliment Pharmacol Ther 2017; 45:205-221. [PMID: 27878827 DOI: 10.1111/apt.13864] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/21/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are chronic relapsing diseases in which pro-inflammatory immune cells and cytokines induce intestinal tissue damage and disability. Mesenchymal stromal cells (MSCs) exert powerful immunomodulatory effects and stimulate tissue repair. AIM To review the current data on mesenchymal stromal cell therapy in IBD. METHOD We searched PubMed and 'ClinicalTrials.gov' databases using the terms 'mesenchymal stromal cells', 'mesenchymal stem cell transplantation', 'inflammatory bowel diseases', 'Crohn disease' and 'colitis, ulcerative'. Additional publications were identified from individual article reference lists. RESULTS MSCs include inhibition of Th1/Th17 lymphocytes and recruitment of regulatory T lymphocytes, induction of antigen-presenting cells into a regulatory-like profile, and stimulation of epithelial cell differentiation and proliferation. More than 200 patients with refractory fistulas have been treated with local injections of MSCs, resulting in complete response in more than half, and in overall response in approximately two thirds of patients. In refractory luminal Crohn's disease, 49 cases of systemic MSC infusions have been reported, while trials with autologous MSCs resulted in mitigated responses, studies using allogeneic MSCs were promising, with around 60% of patients experiencing a response and around 40% achieving clinical remission. CONCLUSIONS Mesenchymal stromal cells might represent a promising therapy for IBD, especially for Crohn's disease. There remain many unsolved questions concerning the optimal origin and source of mesenchymal stromal cells, dosage and modalities of administration. Moreover, mesenchymal stromal cells still need to prove their effectiveness compared with conventional treatments in randomised controlled trials.
Collapse
Affiliation(s)
- C Grégoire
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - C Lechanteur
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - A Briquet
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - É Baudoux
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - F Baron
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - E Louis
- Department of Gastroenterology, CHU of Liège, University of Liège, Liège, Belgium
| | - Y Beguin
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium.,Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| |
Collapse
|
46
|
Marfy‐Smith SJ, Clarkin CE. Are Mesenchymal Stem Cells So Bloody Great After All? Stem Cells Transl Med 2016; 6:3-6. [PMID: 28170195 PMCID: PMC5442748 DOI: 10.5966/sctm.2016-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 07/15/2016] [Indexed: 12/26/2022] Open
Abstract
This Perspective discusses some activities of mesenchymal stem cells (MSCs) in the context of angiogenesis, focusing on contrasting effects that could call into question the extent to which MSCs can be used clinically in the future. We report on the antiangiogenic/antiproliferative effects of specific MSC populations (including bone marrow MSCs), their paracrine activity, tissue heterogeneity, and endothelial cell interactions. Also discussed are what could lead to contrasting effects of the influence of MSCs in regulating angiogenesis, pointing to some negative effects of these cells. In conclusion, this article highlights important aspects of MSC behavior within the perspective of translational medicine applications. Stem Cells Translational Medicine2017;6:3–6
Collapse
Affiliation(s)
| | - Claire E. Clarkin
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
47
|
Characterization and Expression of Senescence Marker in Prolonged Passages of Rat Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:8487264. [PMID: 27579045 PMCID: PMC4989133 DOI: 10.1155/2016/8487264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
The present study is aimed at optimizing the in vitro culture protocol for generation of rat bone marrow- (BM-) derived mesenchymal stem cells (MSCs) and characterizing the culture-mediated cellular senescence. The initial phase of generation and characterization was conducted using the adherent cells from Sprague Dawley (SD) rat's BM via morphological analysis, growth kinetics, colony forming unit capacity, immunophenotyping, and mesodermal lineage differentiation. Mesenchymal stem cells were successfully generated and characterized as delineated by the expressions of CD90.1, CD44H, CD29, and CD71 and lack of CD11b/c and CD45 markers. Upon induction, rBM-MSCs differentiated into osteocytes and adipocytes and expressed osteocytes and adipocytes genes. However, a decline in cell growth was observed at passage 4 onwards and it was further deciphered through apoptosis, cell cycle, and senescence assays. Despite the enhanced cell viability at later passages (P4-5), the expression of senescence marker, β-galactosidase, was significantly increased at passage 5. Furthermore, the cell cycle analysis has confirmed the in vitro culture-mediated cellular senescence where cells were arrested at the G0/G1 phase of cell cycle. Although the currently optimized protocols had successfully yielded rBM-MSCs, the culture-mediated cellular senescence limits the growth of rBM-MSCs and its potential use in rat-based MSC research.
Collapse
|
48
|
Sui B, Hu C, Zhang X, Zhao P, He T, Zhou C, Qiu X, Chen N, Zhao X, Jin Y. Allogeneic Mesenchymal Stem Cell Therapy Promotes Osteoblastogenesis and Prevents Glucocorticoid-Induced Osteoporosis. Stem Cells Transl Med 2016; 5:1238-46. [PMID: 27365487 DOI: 10.5966/sctm.2015-0347] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/07/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED : Gene-modified mesenchymal stem cell (MSC)-like cells with enhanced bone marrow homing and osteogenesis have been used in treating glucocorticoid-induced murine osteoporosis (GIOP). Recent preclinical studies have further demonstrated the immunomodulatory and anticatabolic potential of allogeneic MSCs in treating osteoporosis under inflammatory and autoimmune conditions. In this study, we investigated whether systemic infusion of allogeneic MSCs without genetic manipulation could prevent GIOP, whether anabolic and anticatabolic effects existed, and whether homing or immunomodulation underlay the putative therapeutic effects. Allogeneic bone marrow-derived MSCs (BMMSCs) were isolated, identified, and systemically infused into mice treated with excessive dexamethasone. We revealed that allogeneic MSC transplantation prevented the reduction of bone mass and strength in GIOP. Bone histomorphometric analyses of bone remodeling demonstrated the maintenance of bone formation and osteoblast survival after MSC therapy. Using green fluorescent protein (GFP)-labeled BMMSCs, we showed that donor BMMSCs(GFP) homed and inhabited recipient bone marrow for at least 4 weeks and prevented recipient bone marrow cell apoptosis, as shown by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. Furthermore, donor BMMSCs(GFP) committed to Osterix (Osx)(+) osteoblast progenitors and induced recipient osteoblastogenesis, as exhibited by GFP-Osx double-labeling immunofluorescence analysis. No anticatabolic effects or systemic immunomodulatory effects of infused BMMSCs were detected. These findings demonstrated that allogeneic MSC therapy prevented GIOP by inhabiting and functioning in recipient bone marrow, which promoted osteoblastogenesis, which in turn maintained bone formation. Our findings provide important information regarding cell-based anabolic therapy for GIOP and uncover MSC behaviors following the homing event. SIGNIFICANCE This study revealed the therapeutic potential of systemically infused, genetically unmodified allogeneic MSCs in glucocorticoid-induced osteoporosis. The donor MSCs inhabited recipient bone marrow and promoted osteoblastogenesis. The therapeutic effects were based on maintenance of bone formation. These results provide important information regarding cell-based anabolic therapy for glucocorticoid-induced osteoporosis and uncover previously unrecognized mesenchymal stem cell behaviors following a homing event. The current study also indicates that minimizing the time of cell culture confers an advantage for increasing transplanted mesenchymal stem cells to the targeted organ to promote therapeutic effects.
Collapse
Affiliation(s)
- Bingdong Sui
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China State Key Laboratory of Military Stomatology, Department of Dental Materials, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Department of Dental Materials, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chenghu Hu
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xinyi Zhang
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Pan Zhao
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Tao He
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Cuihong Zhou
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Nan Chen
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xinyi Zhao
- State Key Laboratory of Military Stomatology, Department of Dental Materials, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Department of Dental Materials, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Yan Jin
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
49
|
Effects of Oxidative Stress on Mesenchymal Stem Cell Biology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2989076. [PMID: 27413419 PMCID: PMC4928004 DOI: 10.1155/2016/2989076] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/29/2016] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) are multipotent stem cells present in most fetal and adult tissues. Ex vivo culture-expanded MSCs are being investigated for tissue repair and immune modulation, but their full clinical potential is far from realization. Here we review the role of oxidative stress in MSC biology, as their longevity and functions are affected by oxidative stress. In general, increased reactive oxygen species (ROS) inhibit MSC proliferation, increase senescence, enhance adipogenic but reduce osteogenic differentiation, and inhibit MSC immunomodulation. Furthermore, aging, senescence, and oxidative stress reduce their ex vivo expansion, which is critical for their clinical applications. Modulation of sirtuin expression and activity may represent a method to reduce oxidative stress in MSCs. These findings have important implications in the clinical utility of MSCs for degenerative and immunological based conditions. Further study of oxidative stress in MSCs is imperative in order to enhance MSC ex vivo expansion and in vivo engraftment, function, and longevity.
Collapse
|
50
|
Oron A, Oron U. Low-Level Laser Therapy to the Bone Marrow Ameliorates Neurodegenerative Disease Progression in a Mouse Model of Alzheimer's Disease: A Minireview. Photomed Laser Surg 2016; 34:627-630. [PMID: 27294393 DOI: 10.1089/pho.2015.4072] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE This communication reviews the ability of low-level laser therapy (LLLT) to stimulate mesenchymal stem cells (MSCs) in autologous bone marrow (BM) to enhance the capacity of MSCs to infiltrate the brain, clear β-amyloid, and improve cognition. BACKGROUND We recently reported that LLLT applied to the BM enhanced the proliferation of MSCs and their mobilization toward the ischemic heart region, suggesting a possible application of this approach in regenerative medicine and neurodegenerative diseases. It was also shown that circulating monocytes can infiltrate the brain and reduce brain amyloid load in an Alzheimer's disease (AD) mouse model. METHODS AND RESULTS MSCs from wild-type mice stimulated with LLLT demonstrated an increased ability to maturate toward a monocyte lineage and to increase phagocytosis of soluble Aβ in vitro. Furthermore, weekly LLLT for 2 months to the BM, starting at 4 months of age (progressive stage of the disease in these 5XFAD transgenic male mice), improved memory and spatial learning, compared to a sham-treated AD mouse model. Histology revealed a significant reduction in Aβ brain burden in the laser-treated mice compared to the nonlaser-treated ones. CONCLUSIONS The application of LLLT to the BM is suggested as a therapeutic approach in progressive stages of AD, and its potential role in mediating MSC therapy in brain amyloidogenic disease is implied.
Collapse
Affiliation(s)
- Amir Oron
- 1 Department of Orthopedic Surgery, Kaplan Medical Center , Rehovot, Israel
| | - Uri Oron
- 2 Department of Zoology, George S. Wise Faculty of Life Sciences and Sagol School of Neuroscience, Tel Aviv University , Tel Aviv, Israel
| |
Collapse
|