51
|
Liang J, Gao Y, Feng Z, Zhang B, Na Z, Li D. Reactive oxygen species and ovarian diseases: Antioxidant strategies. Redox Biol 2023; 62:102659. [PMID: 36917900 PMCID: PMC10023995 DOI: 10.1016/j.redox.2023.102659] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
Reactive oxygen species (ROS) are mainly produced in mitochondria and are involved in various physiological activities of the ovary through signaling and are critical for regulating the ovarian cycle. Notably, the imbalance between ROS generation and the antioxidant defense system contributes to the development of ovarian diseases. These contradictory effects have critical implications for potential antioxidant strategies that aim to scavenge excessive ROS. However, much remains to be learned about how ROS causes various ovarian diseases to the application of antioxidant therapy for ovarian diseases. Here, we review the mechanisms of ROS generation and maintenance of homeostasis in the ovary and its associated physiological effects. Additionally, we have highlighted the pathological mechanisms of ROS in ovarian diseases and potential antioxidant strategies for treatment.
Collapse
Affiliation(s)
- Junzhi Liang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yingzhuo Gao
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Feng
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bowen Zhang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zhijing Na
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004, China; Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
52
|
Ra K, Park SC, Lee BC. Female Reproductive Aging and Oxidative Stress: Mesenchymal Stem Cell Conditioned Medium as a Promising Antioxidant. Int J Mol Sci 2023; 24:ijms24055053. [PMID: 36902477 PMCID: PMC10002910 DOI: 10.3390/ijms24055053] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
The recent tendency to delay pregnancy has increased the incidence of age-related infertility, as female reproductive competence decreases with aging. Along with aging, a lowered capacity of antioxidant defense causes a loss of normal function in the ovaries and uterus due to oxidative damage. Therefore, advancements have been made in assisted reproduction to resolve infertility caused by reproductive aging and oxidative stress, following an emphasis on their use. The application of mesenchymal stem cells (MSCs) with intensive antioxidative properties has been extensively validated as a regenerative therapy, and proceeding from original cell therapy, the therapeutic effects of stem cell conditioned medium (CM) containing paracrine factors secreted during cell culture have been reported to be as effective as that of direct treatment of source cells. In this review, we summarized the current understanding of female reproductive aging and oxidative stress and present MSC-CM, which could be developed as a promising antioxidant intervention for assisted reproductive technology.
Collapse
Affiliation(s)
- Kihae Ra
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Se Chang Park
- Laboratory of Aquatic Biomedicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (S.C.P.); (B.C.L.)
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (S.C.P.); (B.C.L.)
| |
Collapse
|
53
|
Kano F, Hashimoto N, Liu Y, Xia L, Nishihara T, Oki W, Kawarabayashi K, Mizusawa N, Aota K, Sakai T, Azuma M, Hibi H, Iwasaki T, Iwamoto T, Horimai N, Yamamoto A. Therapeutic benefits of factors derived from stem cells from human exfoliated deciduous teeth for radiation-induced mouse xerostomia. Sci Rep 2023; 13:2706. [PMID: 36792628 PMCID: PMC9932159 DOI: 10.1038/s41598-023-29176-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Radiation therapy for head and neck cancers is frequently associated with adverse effects on the surrounding normal tissue. Irreversible damage to radiation-sensitive acinar cells in the salivary gland (SG) causes severe radiation-induced xerostomia (RIX). Currently, there are no effective drugs for treating RIX. We investigated the efficacy of treatment with conditioned medium derived from stem cells from human exfoliated deciduous teeth (SHED-CM) in a mouse RIX model. Intravenous administration of SHED-CM, but not fibroblast-CM (Fibro-CM), prevented radiation-induced cutaneous ulcer formation (p < 0.0001) and maintained SG function (p < 0.0001). SHED-CM treatment enhanced the expression of multiple antioxidant genes in mouse RIX and human acinar cells and strongly suppressed radiation-induced oxidative stress. The therapeutic effects of SHED-CM were abolished by the superoxide dismutase inhibitor diethyldithiocarbamate (p < 0.0001). Notably, quantitative liquid chromatography-tandem mass spectrometry shotgun proteomics of SHED-CM and Fibro-CM identified eight proteins activating the endogenous antioxidant system, which were more abundant in SHED-CM than in Fibro-CM (p < 0.0001). Neutralizing antibodies against those activators reduced antioxidant activity of SHED-CM (anti-PDGF-D; p = 0.0001, anti-HGF; p = 0.003). Our results suggest that SHED-CM may provide substantial therapeutic benefits for RIX primarily through the activation of multiple antioxidant enzyme genes in the target tissue.
Collapse
Affiliation(s)
- Fumiya Kano
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Noboru Hashimoto
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Yao Liu
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Linze Xia
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Takaaki Nishihara
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Wakana Oki
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Keita Kawarabayashi
- grid.267335.60000 0001 1092 3579Department of Pediatric Dentistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Noriko Mizusawa
- grid.267335.60000 0001 1092 3579Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiko Aota
- grid.267335.60000 0001 1092 3579Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takayoshi Sakai
- grid.136593.b0000 0004 0373 3971Department of Oral-Facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masayuki Azuma
- grid.267335.60000 0001 1092 3579Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hideharu Hibi
- grid.27476.300000 0001 0943 978XDepartment of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonori Iwasaki
- grid.267335.60000 0001 1092 3579Department of Pediatric Dentistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tsutomu Iwamoto
- grid.265073.50000 0001 1014 9130Department of Pediatric Dentistry/Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Akihito Yamamoto
- Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504, Japan.
| |
Collapse
|
54
|
Yan K, Zheng J, Kluth MA, Li L, Ganss C, Yard B, Magdeburg R, Frank MH, Pallavi P, Keese M. ABCB5 + mesenchymal stromal cells therapy protects from hypoxia by restoring Ca 2+ homeostasis in vitro and in vivo. Stem Cell Res Ther 2023; 14:24. [PMID: 36759868 PMCID: PMC9912525 DOI: 10.1186/s13287-022-03228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Hypoxia in ischemic disease impairs Ca2+ homeostasis and may promote angiogenesis. The therapeutic efficacy of mesenchymal stromal cells (MSCs) in peripheral arterial occlusive disease is well established, yet its influence on cellular Ca2+ homeostasis remains to be elucidated. We addressed the influence of ATP-binding cassette subfamily B member 5 positive mesenchymal stromal cells (ABCB5+ MSCs) on Ca2+ homeostasis in hypoxic human umbilical vein endothelial cells (HUVECs) in vitro and in vivo. METHODS Hypoxia was induced in HUVECs by Cobalt (II) chloride (CoCl2) or Deferoxamine (DFO). Dynamic changes in the cytosolic- and endoplasmic reticulum (ER) Ca2+ and changes in reactive oxygen species were assessed by appropriate fluorescence-based sensors. Metabolic activity, cell migration, and tube formation were assessed by standard assays. Acute-on-chronic ischemia in Apolipoprotein E knock-out (ApoE-/-) mice was performed by double ligation of the right femoral artery (DFLA). ABCB5+ MSC cells were injected into the ischemic limb. Functional recovery after DFLA and histology of gastrocnemius and aorta were assessed. RESULTS Hypoxia-induced impairment of cytosolic and ER Ca2+ were restored by ABCB5+ MSCs or their conditioned medium. Similar was found for changes in intracellular ROS production, metabolic activity, migratory ability and tube formation. The restoration was paralleled by an increased expression of the Ca2+ transporter Sarco-/endoplasmic reticulum ATPase 2a (SERCA2a) and the phosphorylation of Phospholamban (PLN). In acute-on-chronic ischemia, ABCB5+ MSCs treated mice showed a higher microvascular density, increased SERCA2a expression and PLN phosphorylation relative to untreated controls. CONCLUSIONS ABCB5+ MSCs therapy can restore cellular Ca2+ homeostasis, which may beneficially affect the angiogenic function of endothelial cells under hypoxia in vitro and in vivo.
Collapse
Affiliation(s)
- Kaixuan Yan
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jiaxing Zheng
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Lin Li
- grid.7700.00000 0001 2190 4373Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany ,grid.7700.00000 0001 2190 4373European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Ganss
- TICEBA GmbH, Heidelberg, Germany ,grid.476673.7RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | - Benito Yard
- grid.7700.00000 0001 2190 4373V Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Magdeburg
- grid.411778.c0000 0001 2162 1728Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161 Mannheim, Germany
| | - Markus H. Frank
- grid.38142.3c000000041936754XDepartment of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XTransplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Stem Cell Institute, Harvard University, Cambridge, MA USA ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | - Prama Pallavi
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| | - Michael Keese
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Department for General and Visceral Surgery, Theresienkrankenhaus Mannheim, Mannheim, Germany. .,Department of Surgery, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68161, Mannheim, Germany.
| |
Collapse
|
55
|
Secretome of hESC-Derived MSC-like Immune and Matrix Regulatory Cells Mitigate Pulmonary Fibrosis through Antioxidant and Anti-Inflammatory Effects. Biomedicines 2023; 11:biomedicines11020463. [PMID: 36830999 PMCID: PMC9953085 DOI: 10.3390/biomedicines11020463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Oxidative stress and inflammation are major drivers in the pathogenesis and progression of pulmonary fibrosis (PF). The mesenchymal stem cell (MSC) secretome has regenerative potential and immunomodulatory functions. Human embryonic stem cell (hESC)-derived MSC-like immune and matrix regulatory cells (IMRCs) are manufacturable with large-scale good manufacturing practice (GMP) preparation. In the present study, the antioxidative and anti-inflammatory properties and the therapeutic effect of the secretome of hESC-MSC-IMRC-derived conditioned culture medium (CM) (hESC-MSC-IMRC-CM) were investigated. Results revealed the capacities of hESC-MSC-IMRC-CM to reduce bleomycin (BLM)-induced reactive oxygen species (ROS), extracellular matrix (ECM) deposition, and epithelial-mesenchymal transition (EMT) in A549 cells. The administration of concentrated hESC-MSC-IMRC-CM significantly alleviated the pathogenesis of PF in lungs of BLM-injured mice, as accessed by pathohistological changes and the expression of ECM and EMT. A mechanistic study further demonstrated that the hESC-MSC-IMRC-CM was able to inhibit BLM-induced ROS and pro-inflammatory cytokines, accompanied by a reduced expression of Nox4, Nrf2, Ho-1, and components of the Tlr4/MyD88 signaling cascade. These results provide a proof of concept for the hESC-MSC-IMRC-derived secretome treatment of PF, in part mediated by their antioxidative and anti-inflammatory effects. This study thus reinforces the development of ready-to-use, cell-free hESC-MSC-IMRC secretome biomedicine for the treatment of PF in clinical settings.
Collapse
|
56
|
Lan J, Zhou Y, Wang H, Tang J, Kang Y, Wang P, Liu X, Peng Y. Protective effect of human umbilical cord mesenchymal stem cell derived conditioned medium in a mutant TDP-43 induced motoneuron-like cellular model of ALS. Brain Res Bull 2023; 193:106-116. [PMID: 36563944 DOI: 10.1016/j.brainresbull.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/15/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a multi-factor neurodegenerative disease, characterized by the loss of motor neurons. TAR DNA-binding protein 43 (TDP-43) mutation, accumulation and aggregation, as well as oxidative stress are recognized as major pathological denominators and biochemical markers for ALS. Recently, human umbilical cord mesenchymal stem cell-derived conditioned medium (UC-CM) has been introduced to treat ALS patients. However, there is no research for the protective effect of UC-CM on the TDP-43 model of ALS. In this study, we evaluated the potential neuroprotective effect of UC-CM on a cellular ALS model expressing TDP-43mutant M337V, as well as its underlying mechanism. We found that 24 h UC-CM treatment could protect M337V expressing motor neurons by increasing cell viability and reducing LDH leakage. Furthermore, the aggregation of M337V, generation of ROS, malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), protein carbonyl and 8-OHdG were also reduced by UC-CM, indicating that UC-CM protected cells by reducing oxidative damage. Moreover, UC-CM significantly increased the expression of nuclear Nrf2 and its downstream enzyme HO1. The Nrf2 translocation inhibitor ML385 could inhibit the effect of UC-CM on the cell viability and aggregate of M337V. Our results suggest that UC-CM protect cells against M337V expression by its strong antioxidative effect via Nrf-2/HO-1 axis activation.
Collapse
Affiliation(s)
- Jiaqi Lan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yujun Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongyue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jingshu Tang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuying Kang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Peishen Wang
- Department of Cell Transplantation, The third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xuebin Liu
- Department of Cell Transplantation, The third Medical Center of Chinese PLA General Hospital, Beijing 100039, China; Beijing Zhongguang Tianyi Biotechnology Co., Ltd, Beijing 100026, China.
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
57
|
Savvidou MG, Georgiopoulou I, Antoniou N, Tzima S, Kontou M, Louli V, Fatouros C, Magoulas K, Kolisis FN. Extracts from Chlorella vulgaris Protect Mesenchymal Stromal Cells from Oxidative Stress Induced by Hydrogen Peroxide. PLANTS (BASEL, SWITZERLAND) 2023; 12:361. [PMID: 36679074 PMCID: PMC9866266 DOI: 10.3390/plants12020361] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Microalgae as unicellular eukaryotic organisms demonstrate several advantages for biotechnological and biological applications. Natural derived microalgae products demand has increased in food, cosmetic and nutraceutical applications lately. The natural antioxidants have been used for attenuation of mitochondrial cell damage caused by oxidative stress. This study evaluates the in vitro protective effect of Chlorella vulgaris bioactive extracts against oxidative stress in human mesenchymal stromal/stem cells (MSCs). The classical solid-liquid and the supercritical extraction, using biomass of commercially available and laboratory cultivated C. vulgaris, are employed. Oxidative stress induced by 300 μM H2O2 reduces cell viability of MSCs. The addition of C. vulgaris extracts, with increased protein content compared to carbohydrates, to H2O2 treated MSCs counteracted the oxidative stress, reducing reactive oxygen species levels without affecting MSC proliferation. The supercritical extraction was the most efficient extraction method for carotenoids resulting in enhanced antioxidant activity. Pre-treatment of MSCs with C. vulgaris extracts mitigates the oxidative damage ensued by H2O2. Initial proteomic analysis of secretome from licensed (TNFα-activated) MSCs treated with algal extracts reveals a signature of differentially regulated proteins that fall into clinically relevant pathways such as inflammatory signaling. The enhanced antioxidative and possibly anti-inflammatory capacity could be explored in the context of future cell therapies.
Collapse
Affiliation(s)
- Maria G. Savvidou
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Ioulia Georgiopoulou
- Laboratory of Thermodynamics and Transport Phenomena, School of Chemical Engineering, National Technical University of Athens, 9 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Nasia Antoniou
- TheraCell Advanced Biotechnologies, 14564 Kifisia, Greece
| | - Soultana Tzima
- Laboratory of Thermodynamics and Transport Phenomena, School of Chemical Engineering, National Technical University of Athens, 9 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Maria Kontou
- TheraCell Advanced Biotechnologies, 14564 Kifisia, Greece
| | - Vasiliki Louli
- Laboratory of Thermodynamics and Transport Phenomena, School of Chemical Engineering, National Technical University of Athens, 9 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | | | - Kostis Magoulas
- Laboratory of Thermodynamics and Transport Phenomena, School of Chemical Engineering, National Technical University of Athens, 9 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Fragiskos N. Kolisis
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| |
Collapse
|
58
|
Adipose-Derived Mesenchymal Stem Cells Inhibit JNK-Mediated Mitochondrial Retrograde Pathway to Alleviate Acetaminophen-Induced Liver Injury. Antioxidants (Basel) 2023; 12:antiox12010158. [PMID: 36671020 PMCID: PMC9854665 DOI: 10.3390/antiox12010158] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Acetaminophen (APAP) is the major cause of drug-induced liver injury, with limited treatment options. APAP overdose invokes excessive oxidative stress that triggers mitochondria-to-nucleus retrograde pathways, contributing to APAP-induced liver injury (AILI). Mesenchymal stem cell therapy is a promising tool for acute liver failure. Therefore, the purpose of this study was to investigate the beneficial effects of adipose-derived mesenchymal stem cell (AMSC) therapy on AILI and reveal the potential therapeutic mechanisms. C57BL/6 mice were used as the animal model and AML12 normal murine hepatocytes as the cellular model of APAP overdose. Immunohistochemical staining, Western blotting, immunofluorescence staining, and RNA sequencing assays were used for assessing the efficacy and validating mechanisms of AMSC therapy. We found AMSC therapy effectively ameliorated AILI, while delayed AMSC injection lost its efficacy related to the c-Jun N-terminal kinase (JNK)-mediated mitochondrial retrograde pathways. We further found that AMSC therapy inhibited JNK activation and mitochondrial translocation, reducing APAP-induced mitochondrial damage. The downregulation of activated ataxia telangiectasia-mutated (ATM) and DNA damage response proteins in AMSC-treated mouse liver indicated AMSCs blocked the JNK-ATM pathway. Overall, AMSCs may be an effective treatment for AILI by inhibiting the JNK-ATM mitochondrial retrograde pathway, which improves APAP-induced mitochondrial dysfunction and liver injury.
Collapse
|
59
|
Ibrahim D, Abozied N, Abdel Maboud S, Alzamami A, Alturki NA, Jaremko M, Alanazi MK, Alhuthali HM, Seddek A. Therapeutic potential of bone marrow mesenchymal stem cells in cyclophosphamide-induced infertility. Front Pharmacol 2023; 14:1122175. [PMID: 37033609 PMCID: PMC10073512 DOI: 10.3389/fphar.2023.1122175] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/26/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer is a deadly disease characterized by abnormal cell proliferation. Chemotherapy is one technique of cancer treatment. Cyclophosphamide (CYP) is the most powerful chemotherapy medication, yet it has serious adverse effects. It is an antimitotic medicine that regulates cell proliferation and primarily targets quickly dividing cells, and it has been related to varying levels of infertility in humans. In the current study, we assessed the biochemical, histological, and microscopic evaluations of testicular damage following cyclophosphamide administration. Further, we have explored the potential protective impact of mesenchymal stem cell (MSCs) transplantation. The biochemical results revealed that administration of cyclophosphamide increased serum concentrations of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), while it decreased serum concentrations of free testosterone hormone (TH), testicular follicle-stimulating hormone, luteinizing hormone, and free testosterone hormone concentrations, testicular total antioxidant capacity (TAC), and testicular activity of superoxide dismutase (SOD) enzyme. The histology and sperm examinations revealed that cyclophosphamide induced destruction to the architectures of several tissues in the testes, which drastically reduced the Johnsen score as well as the spermatogenesis process. Surprisingly, transplantation of mesenchymal stem cell after cyclophosphamide administration altered the deterioration effect of cyclophosphamide injury on the testicular tissues, as demonstrated by biochemical and histological analysis. Our results indicated alleviation of serum and testicular sex hormones, as well as testicular oxidative stress markers (total antioxidant capacity and superoxide dismutase activity), and nearly restored the normal appearance of the testicular tissues, Johnsen score, and spermatogenesis process. In conclusion, our work emphasizes the protective pharmacological use of mesenchymal stem cell to mitigate the effects of cyclophosphamide on testicular tissues that impair the spermatogenesis process following chemotherapy. These findings indicate that transferring mesenchymal stem cell to chemotherapy patients could significantly improve spermatogenesis.
Collapse
Affiliation(s)
- Dalia Ibrahim
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- *Correspondence: Dalia Ibrahim,
| | - Nadia Abozied
- The Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Samar Abdel Maboud
- The Department of Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah, Saudi Arabia
| | - Norah A. Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Maram Khalil Alanazi
- Pharm.D, Scientific Office and Regulatory Affair Department, Dallah Pharma Company, Riyadh, Saudi Arabia
| | - Hayaa M. Alhuthali
- Department of Clinical laboratory sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Asmaa Seddek
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
60
|
Farhoudi M, Sadigh-Eteghad S, Farjami A, Salatin S. Nanoparticle and Stem Cell Combination Therapy for the Management of Stroke. Curr Pharm Des 2023; 29:15-29. [PMID: 36515043 DOI: 10.2174/1381612829666221213113119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022]
Abstract
Stroke is currently one of the primary causes of morbidity and mortality worldwide. Unfortunately, the available treatments for stroke are still extremely limited. Indeed, stem cell (SC) therapy is a new option for the treatment of stroke that could significantly expand the therapeutic time window of stroke. Some proposed mechanisms for stroke-based SC therapy are the incorporation of SCs into the host brain to replace dead or damaged cells/tissues. Moreover, acute cell delivery can inhibit apoptosis and decrease lesion size, providing immunomudolatory and neuroprotection effects. However, several major SC problems related to SCs such as homing, viability, uncontrolled differentiation, and possible immune response, have limited SC therapy. A combination of SC therapy with nanoparticles (NPs) can be a solution to address these challenges. NPs have received considerable attention in regulating and controlling the behavior of SCs because of their unique physicochemical properties. By reviewing the pathophysiology of stroke and the therapeutic benefits of SCs and NPs, we hypothesize that combined therapy will offer a promising future in the field of stroke management. In this work, we discuss recent literature in SC research combined with NP-based strategies that may have a synergistic outcome after stroke incidence.
Collapse
Affiliation(s)
- Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Farjami
- Food and Drug Safety Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Salatin
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
61
|
Das M, Teli P, Vaidya A, Kale V. Secretome of Young Mesenchymal Stromal Cells Rejuvenates Aged Mesenchymal Stromal Cells by Normalizing Their Phenotype and Restoring Their Differentiation Profile. Stem Cells Dev 2023; 32:12-24. [PMID: 36453235 DOI: 10.1089/scd.2022.0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During aging, the proliferation and differentiation ability of mesenchymal stem/stromal cells (MSCs) gets affected, and hence, aged MSCs are not preferred for regenerative purposes. Rapid identification of aging-associated changes within MSCs and the mechanistic pathways involved are necessary to determine optimal cell sources to treat musculoskeletal disorders in older patients. In the present study, we have identified a set of phenotypic markers, namely downregulated expression of CD90 and upregulated expression of CD45, as age-defining markers for the bone marrow-derived MSCs. We also show that these phenotypic changes in aged MSCs correlate with their aging-mediated differentiation defects. We find that oxidative stress signaling leading to the activation of nuclear factor kappa B (NF-κB) plays an essential role in altering the phenotype and differentiation ability of the aged MSCs. We further show that treatment of aged MSCs with the conditioned medium (CM) derived from young MSCs (young-CM) restored their phenotype and differentiation potential to the young-like by ameliorating activation of NF-κB signaling in them. Similar changes could also be achieved by using an inhibitor of NF-κB signaling, showing that oxidative stress-induced NF-κB activation is the causative factor in the aging of MSCs. Additionally, we show that treating young MSCs with hydrogen peroxide mimics all the aging-mediated changes in them, underscoring the involvement of oxidative stress in the aging of MSCs. Overall, our data suggest that the altered expression of CD90 and CD45 surface markers can be used as a primary screen to identify the onset of aging in the MSCs, which can be quickly reversed by their in vitro treatment with young-CM or NF-κB inhibitor. Our study also puts the phenotypic characterization of MSCs in a clinical perspective.
Collapse
Affiliation(s)
- Madhurima Das
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India
| | - Prajakta Teli
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India.,Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
62
|
Martí‐Chillón G, Muntión S, Preciado S, Osugui L, Navarro‐Bailón A, González‐Robledo J, Sagredo V, Blanco JF, Sánchez‐Guijo F. Therapeutic potential of mesenchymal stromal/stem cells in critical-care patients with systemic inflammatory response syndrome. Clin Transl Med 2023; 13:e1163. [PMID: 36588089 PMCID: PMC9806020 DOI: 10.1002/ctm2.1163] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Despite notable advances in the support and treatment of patients admitted to the intensive care unit (ICU), the management of those who develop a systemic inflammatory response syndrome (SIRS) still constitutes an unmet medical need. MAIN BODY Both the initial injury (trauma, pancreatitis, infections) and the derived uncontrolled response promote a hyperinflammatory status that leads to systemic hypotension, tissue hypoperfusion and multiple organ failure. Mesenchymal stromal/stem cells (MSCs) are emerging as a potential therapy for severe ICU patients due to their potent immunomodulatory, anti-inflammatory, regenerative and systemic homeostasis-regulating properties. MSCs have demonstrated clinical benefits in several inflammatory-based diseases, but their role in SIRS needs to be further explored. CONCLUSION In the current review, after briefly overviewing SIRS physiopathology, we explore the potential mechanisms why MSC therapy could aid in the recovery of this condition and the pre-clinical and early clinical evidence generated to date.
Collapse
Affiliation(s)
| | - Sandra Muntión
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Silvia Preciado
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Lika Osugui
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Almudena Navarro‐Bailón
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Javier González‐Robledo
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Department of MedicineUniversity of SalamancaSalamancaSpain
| | | | - Juan F. Blanco
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
- Department of SurgeryUniversity of SalamancaSalamancaSpain
| | - Fermín Sánchez‐Guijo
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Department of MedicineUniversity of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| |
Collapse
|
63
|
Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and Mesenchymal Stem Cells: Basic Concepts, Challenges and Strategies. BIOLOGY 2022; 11:1678. [PMID: 36421393 PMCID: PMC9687158 DOI: 10.3390/biology11111678] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023]
Abstract
Aging and frailty are complex processes implicating multifactorial mechanisms, such as replicative senescence, oxidative stress, mitochondrial dysfunction, or autophagy disorder. All of these mechanisms drive dramatic changes in the tissue environment, such as senescence-associated secretory phenotype factors and inflamm-aging. Thus, there is a demand for new therapeutic strategies against the devastating effects of the aging and associated diseases. Mesenchymal stem cells (MSC) participate in a "galaxy" of tissue signals (proliferative, anti-inflammatory, and antioxidative stress, and proangiogenic, antitumor, antifibrotic, and antimicrobial effects) contributing to tissue homeostasis. However, MSC are also not immune to aging. Three strategies based on MSC have been proposed: remove, rejuvenate, or replace the senescent MSC. These strategies include the use of senolytic drugs, antioxidant agents and genetic engineering, or transplantation of younger MSC. Nevertheless, these strategies may have the drawback of the adverse effects of prolonged use of the different drugs used or, where appropriate, those of cell therapy. In this review, we propose the new strategy of "Exogenous Restitution of Intercellular Signalling of Stem Cells" (ERISSC). This concept is based on the potential use of secretome from MSC, which are composed of molecules such as growth factors, cytokines, and extracellular vesicles and have the same biological effects as their parent cells. To face this cell-free regenerative therapy challenge, we have to clarify key strategy aspects, such as establishing tools that allow us a more precise diagnosis of aging frailty in order to identify the therapeutic requirements adapted to each case, identify the ideal type of MSC in the context of the functional heterogeneity of these cellular populations, to optimize the mass production and standardization of the primary materials (cells) and their secretome-derived products, to establish the appropriate methods to validate the anti-aging effects and to determine the most appropriate route of administration for each case.
Collapse
Affiliation(s)
- Maria Fraile
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Arancha Martín
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Emergency, Hospital Universitario de Cabueñes, Los Prados, 395, 33394 Gijon, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Surgery, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| |
Collapse
|
64
|
Park JH, Koh EB, Seo YJ, Oh HS, Won JY, Hwang SC, Byun JH. Tiron Has Negative Effects on Osteogenic Differentiation via Mitochondrial Dysfunction in Human Periosteum-Derived Cells. Int J Mol Sci 2022; 23:ijms232214040. [PMID: 36430519 PMCID: PMC9693013 DOI: 10.3390/ijms232214040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/09/2022] [Accepted: 11/13/2022] [Indexed: 11/16/2022] Open
Abstract
Tiron is a potent antioxidant that counters the pathological effects of reactive oxygen species (ROS) production due to oxidative stress in various cell types. We examined the effects of tiron on mitochondrial function and osteoblastic differentiation in human periosteum-derived cells (hPDCs). Tiron increased mitochondrial activity and decreased senescence-associated β-galactosidase activity in hPDCs; however, it had a detrimental effect on osteoblastic differentiation by reducing alkaline phosphatase (ALP) activity and alizarin red-positive mineralization, regardless of H2O2 treatment. Osteoblast-differentiating hPDCs displayed increased ROS production compared with non-differentiating hPDCs, and treatment with tiron reduced ROS production in the differentiating cells. Antioxidants decreased the rates of oxygen consumption and ATP production, which are increased in hPDCs during osteoblastic differentiation. In addition, treatment with tiron reduced the levels of most mitochondrial proteins, which are increased in hPDCs during culture in osteogenic induction medium. These results suggest that tiron exerts negative effects on the osteoblastic differentiation of hPDCs by causing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
| | - Eun-Byeol Koh
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
| | - Young-Jin Seo
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
| | - Hye-Seong Oh
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
| | - Ju-Yeong Won
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52828, Korea
- Correspondence:
| |
Collapse
|
65
|
The Prescription of Oral Mucosal Mesenchymal Stem Cells post-Traumatic Brain Injury Improved the Kidney and Heart Inflammation and Oxidative Stress. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8235961. [PMID: 36408281 PMCID: PMC9671733 DOI: 10.1155/2022/8235961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022]
Abstract
Background In the last years, mesenchymal stem cells (MSCs) have been considered as a useful strategy to treat many diseases such as traumatic brain injury (TBI). The production of inflammatory agents by TBI elicits an inflammatory response directed to other systems of body, such as the heart and the kidneys. In this study, the efficacy of oral mucosal mesenchymal stem cells (OMSCs) prescription after TBI in inflammation and oxidative stress of the heart and kidneys was evaluated. Methods Twenty-four male rats were located in groups as follows: sham, TBI, vehicle (Veh), and stem cell (SC). OMSCs were injected intravenously 1 and 24 hours after TBI. Inflammatory, oxidative stress, and histopathological outcomes of the heart and kidney tissues were investigated 48 hours after TBI. Results TBI caused an increase in the level of interleukin-1β (IL-1β), interleukin-6 (IL-6), malondialdehyde (MDA), and carbonyl protein (PC) of the heart and kidney compared to the sham group. Superoxide dismutase (SOD), total antioxidant capacity (TAC), catalase (CAT), and interleukin-10 (IL-10) of the heart and kidney decreased after TBI. The use of OMSCs after TBI reduced the changes of these factors in both the heart and the kidney. Conclusion Application of OMSCs after TBI can decrease inflammation and oxidative stress of the heart and kidney tissues leading to the reduction of damage. Therefore, this method can be evaluated in the TBI patients in future studies.
Collapse
|
66
|
Effect of human mesenchymal stem cell secretome administration on morphine self-administration and relapse in two animal models of opioid dependence. Transl Psychiatry 2022; 12:462. [PMID: 36333316 PMCID: PMC9636200 DOI: 10.1038/s41398-022-02225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The present study investigates the possible therapeutic effects of human mesenchymal stem cell-derived secretome on morphine dependence and relapse. This was studied in a new model of chronic voluntary morphine intake in Wistar rats which shows classic signs of morphine intoxication and a severe naloxone-induced withdrawal syndrome. A single intranasal-systemic administration of MSCs secretome fully inhibited (>95%; p < 0.001) voluntary morphine intake and reduced the post-deprivation relapse intake by 50% (p < 0.02). Since several studies suggest a significant genetic contribution to the chronic use of many addictive drugs, the effect of MSCs secretome on morphine self-administration was further studied in rats bred as high alcohol consumers (UChB rats). Sub-chronic intraperitoneal administration of morphine before access to increasing concentrations of morphine solutions and water were available to the animals, led UChB rats to prefer ingesting morphine solutions over water, attaining levels of oral morphine intake in the range of those in the Wistar model. Intranasally administered MSCs secretome to UChB rats dose-dependently inhibited morphine self-administration by 72% (p < 0.001); while a single intranasal dose of MSC-secretome administered during a morphine deprivation period imposed on chronic morphine consumer UChB rats inhibited re-access morphine relapse intake by 80 to 85% (p < 0.0001). Both in the Wistar and the UChB rat models, MSCs-secretome administration reversed the morphine-induced increases in brain oxidative stress and neuroinflammation, considered as key engines perpetuating drug relapse. Overall, present preclinical studies suggest that products secreted by human mesenchymal stem cells may be of value in the treatment of opioid addiction.
Collapse
|
67
|
Bray ER, Kirsner RS, Badiavas EV. Mesenchymal Stem Cell-Derived Extracellular Vesicles as an Advanced Therapy for Chronic Wounds. Cold Spring Harb Perspect Biol 2022; 14:a041227. [PMID: 35817513 PMCID: PMC9524280 DOI: 10.1101/cshperspect.a041227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Chronic wounds are a significant challenge for patients, healthcare providers, and healthcare systems. Chronic wounds develop due to a complex interplay between chronic inflammation, tissue hypoxia, and oxidative stress, often occurring in the setting of advancing age. Ideally, new therapeutics should address all the components of chronic wound pathophysiology. Mesenchymal stem cell (MSC) therapies show significant promise to promote healing of chronic wounds. Extracellular vesicles (EVs) secreted by MSCs mediate many of their beneficial effects. We review the evidence demonstrating that MSC-EVs target the processes leading to chronic wounds. Additionally, we discuss how MSCs can be influenced to generate more potent wound healing EVs. Finally, we highlight the current state of EV clinical trials for wound healing and important preclinical studies that will lead to optimal use of MSC-EVs for patient care.
Collapse
Affiliation(s)
- Eric R Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | - Evangelos V Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
68
|
Asgari Taei A, Khodabakhsh P, Nasoohi S, Farahmandfar M, Dargahi L. Paracrine Effects of Mesenchymal Stem Cells in Ischemic Stroke: Opportunities and Challenges. Mol Neurobiol 2022; 59:6281-6306. [PMID: 35922728 DOI: 10.1007/s12035-022-02967-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 07/17/2022] [Indexed: 10/16/2022]
Abstract
It is well acknowledged that neuroprotective effects of transplanted mesenchymal stem cells (MSCs) in ischemic stroke are attributed to their paracrine-mediated actions or bystander effects rather than to cell replacement in infarcted areas. This therapeutic plasticity is due to MSCs' ability to secrete a broad range of bioactive molecules including growth factors, trophic factors, cytokines, chemokines, and extracellular vesicles, overall known as the secretome. The secretome derivatives, such as conditioned medium (CM) or purified extracellular vesicles (EVs), exert remarkable advantages over MSC transplantation in stroke treating. Here, in this review, we used published information to provide an overview on the secretome composition of MSCs, underlying mechanisms of therapeutic effects of MSCs, and preclinical studies on MSC-derived products application in stroke. Furthermore, we discussed current advantages and challenges for successful bench-to-bedside translation.
Collapse
Affiliation(s)
- Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pariya Khodabakhsh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Farahmandfar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
69
|
Eiro N, Fraile M, González-Jubete A, González LO, Vizoso FJ. Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges. Int J Mol Sci 2022; 23:ijms23168905. [PMID: 36012170 PMCID: PMC9408403 DOI: 10.3390/ijms23168905] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are an example of chronic diseases affecting 40% of the population, which involved tissue damage and an inflammatory process not satisfactorily controlled with current therapies. Data suggest that mesenchymal stem cells (MSC) may be a therapeutic option for these processes, and especially for IBD, due to their multifactorial approaches such as anti-inflammatory, anti-oxidative stress, anti-apoptotic, anti-fibrotic, regenerative, angiogenic, anti-tumor, or anti-microbial. However, MSC therapy is associated with important limitations as safety issues, handling difficulties for therapeutic purposes, and high economic cost. MSC-derived secretome products (conditioned medium or extracellular vesicles) are therefore a therapeutic option in IBD as they exhibit similar effects to their parent cells and avoid the issues of cell therapy. In this review, we proposed further studies to choose the ideal tissue source of MSC to treat IBD, the implementation of new standardized production strategies, quality controls and the integration of other technologies, such as hydrogels, which may improve the therapeutic effects of derived-MSC secretome products in IBD.
Collapse
Affiliation(s)
- Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| | - Maria Fraile
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | | | - Luis O. González
- Department of Anatomical Pathology, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Department of Surgery, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| |
Collapse
|
70
|
Liu B, Kong Y, Shi W, Kuss M, Liao K, Hu G, Xiao P, Sankarasubramanian J, Guda C, Wang X, Lei Y, Duan B. Exosomes derived from differentiated human ADMSC with the Schwann cell phenotype modulate peripheral nerve-related cellular functions. Bioact Mater 2022; 14:61-75. [PMID: 35310346 PMCID: PMC8892082 DOI: 10.1016/j.bioactmat.2021.11.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration remains a significant clinical challenge due to the unsatisfactory functional recovery and public health burden. Exosomes, especially those derived from mesenchymal stem cells (MSCs), are promising as potential cell-free therapeutics and gene therapy vehicles for promoting neural regeneration. In this study, we reported the differentiation of human adipose derived MSCs (hADMSCs) towards the Schwann cell (SC) phenotype (hADMSC-SCs) and then isolated exosomes from hADMSCs with and without differentiation (i.e., dExo vs uExo). We assessed and compared the effects of uExo and dExo on antioxidative, angiogenic, anti-inflammatory, and axon growth promoting properties by using various peripheral nerve-related cells. Our results demonstrated that hADMSC-SCs secreted more neurotrophic factors and other growth factors, compared to hADMSCs without differentiation. The dExo isolated from hADMSC-SCs protected rat SCs from oxidative stress and enhanced HUVEC migration and angiogenesis. Compared to uExo, dExo also had improved performances in downregulating pro-inflammatory gene expressions and cytokine secretions and promoting axonal growth of sensory neurons differentiated from human induced pluripotent stem cells. Furthermore, microRNA (miRNA) sequencing analysis revealed that exosomes and their parent cells shared some similarities in their miRNA profiles and exosomes displayed a distinct miRNA signature. Many more miRNAs were identified in dExo than in uExo. Several upregulated miRNAs, like miRNA-132-3p and miRNA-199b-5p, were highly related to neuroprotection, anti-inflammation, and angiogenesis. The dExo can effectively modulate various peripheral nerve-related cellular functions and is promising for cell-free biological therapeutics to enhance neural regeneration. Exosomes were isolated from hADMSCs with and without differentiation towards SC phenotype (i.e., dExo vs uExo). hADMSC-SCs secreted more growth factors compared to hADMSCs without differentiation. The dExo protected rat SCs from oxidative stress and enhanced endothelial cell migration and angiogenesis. dExo promoted axonal growth of sensory neurons differentiated from hiPSCs. miRNA sequencing analysis unveiled and compared the exosomal and cellular miRNA profiles.
Collapse
|
71
|
Gergin ÖÖ, Pehlivan SS, Ulger M, Mat OC, Bayram A, Gönen ZB, Gökdemir NS, Biçer C, Yildiz K, Yay AH. Efficacy of stem cell-based therapies for colistin-induced nephrotoxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 94:103933. [PMID: 35863655 DOI: 10.1016/j.etap.2022.103933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/18/2022] [Accepted: 07/10/2022] [Indexed: 06/09/2023]
Abstract
The increase in infections with multidrug resistant bacteria has forced to return to the use of colistin, antibiotic with known nephrotoxicity. Mesenchymal stem cells (MSCs) are being extensively investigated for their potential in regenerative medicine. This study aimed to investigate the possible protective mechanisms of the MSCs against kidney injury induced by colistin. Forty adult female albino rats were randomly classified into 4 equal groups; the control group, the MSC-treated group (a single dose of 1 ×106 /ml MSCs through the tail vein), the colistin-treated group (36 mg/kg/day colistin was given for 7 days), and the both colistin and MSC group (36 mg/kg/day colistin and 1 ×106 /ml MSCs). Main outcome measures were histopathological alterations, kidney malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT) and immunohistological autophagy evaluation. MSC repressed the progression of colistin-induced kidney injury as evidenced by the improvement of histopathological alterations and the substantial increase MDA, and decrease SOD and CAT in serum levels. Moreover, MSC resulted in a profound reduction in oxidative stress as manifested by decreased MDA and increased SOD in serum. Notably, MSC suppressed colistin-induced autophagy; it reduced renal levels of Beclin-1, P62 and LC3A/B. Furthermore, MSC decreased renal levels of eNOS. Lastly, MSC efficiently decreased expression of the TUNEL positive cell number. MSC confers protection against colistin-induced kidney injury by alleviating oxidative stress, nitric oxide synthase besides modulating reducing autophagy and apoptosis.
Collapse
Affiliation(s)
- Özlem Öz Gergin
- Department of Anaesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey.
| | - Sibel Seckin Pehlivan
- Department of Anaesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey.
| | - Menekse Ulger
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, 38039 Kayseri, Turkey.
| | - Ozge Cengiz Mat
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, 38039 Kayseri, Turkey.
| | - Adnan Bayram
- Department of Anaesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey.
| | | | - Nur Seda Gökdemir
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.
| | - Cihangir Biçer
- Department of Anaesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey.
| | - Karamehmet Yildiz
- Department of Anaesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey.
| | - Arzu Hanım Yay
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, 38039 Kayseri, Turkey; Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.
| |
Collapse
|
72
|
Miller CL, O JM, Allan JS, Madsen JC. Novel approaches for long-term lung transplant survival. Front Immunol 2022; 13:931251. [PMID: 35967365 PMCID: PMC9363671 DOI: 10.3389/fimmu.2022.931251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Allograft failure remains a major barrier in the field of lung transplantation and results primarily from acute and chronic rejection. To date, standard-of-care immunosuppressive regimens have proven unsuccessful in achieving acceptable long-term graft and patient survival. Recent insights into the unique immunologic properties of lung allografts provide an opportunity to develop more effective immunosuppressive strategies. Here we describe advances in our understanding of the mechanisms driving lung allograft rejection and highlight recent progress in the development of novel, lung-specific strategies aimed at promoting long-term allograft survival, including tolerance.
Collapse
Affiliation(s)
- Cynthia L. Miller
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
| | - Jane M. O
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
| | - James S. Allan
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Joren C. Madsen
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA, United States
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
73
|
Macrophage bioassay standardization to assess the anti-inflammatory activity of mesenchymal stromal cell-derived small extracellular vesicles. Cytotherapy 2022; 24:999-1012. [PMID: 35798638 DOI: 10.1016/j.jcyt.2022.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND AIMS Owing to the lack of biological assays, determining the biological activity of extracellular vesicles has proven difficult. Here the authors standardized an in vitro assay to assess the anti-inflammatory activity of mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) based on their ability to prevent acquisition of the M1 phenotype in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Induction of tumor necrosis factor alpha, IL-1β, IL-6 and inducible nitric oxide synthase (iNOS) characterizes the M1 phenotype. Nitric oxide released by iNOS turns into nitrite, which can be easily quantitated in culture media by Griess reaction. METHODS The authors first tested different assay conditions in 96-well plates, including two seeding densities (2 × 104 cells/well and 4 × 104 cells/well), four LPS doses (1 ng/mL, 10 ng/mL, 100 ng/mL and 1000 ng/mL) and two time points (16 h and 24 h), in order to determine the best set-up to accurately measure nitrite concentration as an index of M1 macrophage polarization. RESULTS The authors found that seeding 2 × 104 cells/well and stimulating with 10 ng/mL LPS for 16 h allowed the inhibition of nitrite production by 60% with the use of dexamethasone. Using these established conditions, the authors were able to test different MSC-sEV preparations and generate dose-response curves. Moreover, the authors fully analytically validated assay performance and fulfilled cross-validation against other M1 markers. CONCLUSIONS The authors standardized a quick, cheap and reproducible in vitro macrophage assay that allows for the evaluation and estimation of the anti-inflammatory activity of MSC-sEVs.
Collapse
|
74
|
Clinical Application of Induced Hepatocyte-like Cells Produced from Mesenchymal Stromal Cells: A Literature Review. Cells 2022; 11:cells11131998. [PMID: 35805080 PMCID: PMC9265349 DOI: 10.3390/cells11131998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
Liver disease is a leading cause of mortality worldwide, resulting in 1.3 million deaths annually. The vast majority of liver disease is caused by metabolic disease (i.e., NASH) and alcohol-induced hepatitis, and to a lesser extent by acute and chronic viral infection. Furthermore, multiple insults to the liver is becoming common due to the prevalence of metabolic and alcohol-related liver diseases. Despite this rising prevalence of liver disease, there are few treatment options: there are treatments for viral hepatitis C and there is vaccination for hepatitis B. Aside from the management of metabolic syndrome, no direct liver therapy has shown clinical efficacy for metabolic liver disease, there is very little for acute alcohol-induced liver disease, and liver transplantation remains the only effective treatment for late-stage liver disease. Traditional pharmacologic interventions have failed to appreciably impact the pathophysiology of alcohol-related liver disease or end-stage liver disease. The difficulties associated with developing liver-specific therapies result from three factors that are common to late-stage liver disease arising from any cause: hepatocyte injury, inflammation, and aberrant tissue healing. Hepatocyte injury results in tissue damage with inflammation, which sensitizes the liver to additional hepatocyte injury and stimulates hepatic stellate cells and aberrant tissue healing responses. In the setting of chronic liver insults, there is progressive scarring, the loss of hepatocyte function, and hemodynamic dysregulation. Regenerative strategies using hepatocyte-like cells that are manufactured from mesenchymal stromal cells may be able to correct this pathophysiology through multiple mechanisms of action. Preclinical studies support their effectiveness and recent clinical studies suggest that cell replacement therapy can be safe and effective in patients with liver disease for whom there is no other option.
Collapse
|
75
|
Hamid OIA, Domouky AM, El-Fakharany YM. Molecular evidence of the amelioration of toluene induced encephalopathy by human breast milk mesenchymal stem cells. Sci Rep 2022; 12:9194. [PMID: 35654991 PMCID: PMC9163168 DOI: 10.1038/s41598-022-13173-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022] Open
Abstract
Toluene was widely used volatile organic compound that accumulates in tissues with high lipid content. Stem cells have been proposed as an increasingly attractive approach for repair of damaged nervous system, we aimed to evaluate the ability of breast milk mesenchymal stem cells (MSc) to ameliorate toluene-induced encephalopathy. Sixty adult male albino rats were assigned to 3 groups, control, toluene, and toluene/breast milk-MSc. Neurological assessment was evaluated as well as serum levels of glial fibrillary acidic protein (GFAP), tumor necrosis factor-alpha (TNF-α), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), tissue dopamine and oxidative markers. Gene expression of peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ), nuclear factor-kappaB (NF-kB), and interleukin-6 (IL-6) were evaluated. Moreover, histological and immunohistochemical investigation were done. Results revealed that toluene caused cerebral injury, as evidenced by a significant increase in serum GFAP, TNF-α, malondialdehyde (MDA) and nitric oxide (NO), a significant decrease in serum NGF, tissue dopamine and oxidative markers, besides, a non-significant change in VEGF. Toluene also caused changes in normal cerebral structure and cellular degeneration, including a significant decrease in the total number of neurons and thickness of frontal cortex. Meninges showing signs of inflammation with inflammatory cell infiltration and exudation, a significant decrease in MBP immunoreactivity, and increase in the percent of high motility group box protein-1 (HMGB1) positive cells. PPAR- ɣ, NF-kB, and IL-6 gene expression were all considerably elevated by toluene. These changes were greatly improved by breast milk MSc. Therefore, we conclude that breast milk MSc can attenuate toluene-induced encephalopathy.
Collapse
Affiliation(s)
- Omaima I Abdel Hamid
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Alsharquiah, 44519, Egypt
| | - Ayat M Domouky
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Alsharquiah, 44519, Egypt.
| | - Yara M El-Fakharany
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Alsharquiah, 44519, Egypt
| |
Collapse
|
76
|
Sanmartin MC, Borzone FR, Giorello MB, Yannarelli G, Chasseing NA. Mesenchymal Stromal Cell-Derived Extracellular Vesicles as Biological Carriers for Drug Delivery in Cancer Therapy. Front Bioeng Biotechnol 2022; 10:882545. [PMID: 35497332 PMCID: PMC9046597 DOI: 10.3389/fbioe.2022.882545] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death worldwide, with 10.0 million cancer deaths in 2020. Despite advances in targeted therapies, some pharmacological drawbacks associated with anticancer chemo and immunotherapeutic agents include high toxicities, low bioavailability, and drug resistance. In recent years, extracellular vesicles emerged as a new promising platform for drug delivery, with the advantage of their inherent biocompatibility and specific targeting compared to artificial nanocarriers, such as liposomes. Particularly, mesenchymal stem/stromal cells were proposed as a source of extracellular vesicles for cancer therapy because of their intrinsic properties: high in vitro self-renewal and proliferation, regenerative and immunomodulatory capacities, and secretion of extracellular vesicles that mediate most of their paracrine functions. Moreover, extracellular vesicles are static and safer in comparison with mesenchymal stem/stromal cells, which can undergo genetic/epigenetic or phenotypic changes after their administration to patients. In this review, we summarize currently reported information regarding mesenchymal stem/stromal cell-derived extracellular vesicles, their proper isolation and purification techniques - from either naive or engineered mesenchymal stem/stromal cells - for their application in cancer therapy, as well as available downstream modification methods to improve their therapeutic properties. Additionally, we discuss the challenges associated with extracellular vesicles for cancer therapy, and we review some preclinical and clinical data available in the literature.
Collapse
Affiliation(s)
- María Cecilia Sanmartin
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
77
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
78
|
Alexander JF, Mahalingam R, Seua AV, Wu S, Arroyo LD, Hörbelt T, Schedlowski M, Blanco E, Kavelaars A, Heijnen CJ. Targeting the Meningeal Compartment to Resolve Chemobrain and Neuropathy via Nasal Delivery of Functionalized Mitochondria. Adv Healthc Mater 2022; 11:e2102153. [PMID: 35007407 PMCID: PMC9803615 DOI: 10.1002/adhm.202102153] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/03/2022] [Indexed: 01/03/2023]
Abstract
Cognitive deficits (chemobrain) and peripheral neuropathy occur in ∼75% of patients treated for cancer with chemotherapy and persist long-term in >30% of survivors. Without preventive or curative interventions and with increasing survivorship rates, the population debilitated by these neurotoxicities is rising. Platinum-based chemotherapeutics, including cisplatin, induce neuronal mitochondrial defects leading to chemobrain and neuropathic pain. This study investigates the capacity of nasally administered mesenchymal stem cell-derived mitochondria coated with dextran-triphenylphosphonium polymer (coated mitochondria) to reverse these neurotoxicities. Nasally administered coated mitochondria are rapidly detectable in macrophages in the brain meninges but do not reach the brain parenchyma. The coated mitochondria change expression of >2400 genes regulating immune, neuronal, endocrine and vascular pathways in the meninges of mice treated with cisplatin. Nasal administration of coated mitochondria reverses cisplatin-induced cognitive deficits and resolves neuropathic pain at a >55-times lower dose compared to uncoated mitochondria. Reversal of these neuropathologies is associated with resolution of cisplatin-induced deficits in myelination, synaptosomal mitochondrial integrity and neurogenesis. These findings demonstrate that nasally administered coated mitochondria promote resolution of chemobrain and peripheral neuropathy, thereby identifying a novel facile strategy for clinical application of mitochondrial donation and treating central and peripheral nervous system pathologies by targeting the brain meninges.
Collapse
Affiliation(s)
- Jenolyn F. Alexander
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Alexandre V. Seua
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Luis D. Arroyo
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Tina Hörbelt
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Corresponding Author
| |
Collapse
|
79
|
The protective effect of N-acetylcysteine on antimycin A-induced respiratory chain deficiency in mesenchymal stem cells. Chem Biol Interact 2022; 360:109937. [DOI: 10.1016/j.cbi.2022.109937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022]
|
80
|
Bone Marrow Mesenchymal Stem Cells and Their Derived Extracellular Vesicles Attenuate Non-Alcoholic Steatohepatitis-Induced Cardiotoxicity via Modulating Cardiac Mechanisms. Life (Basel) 2022; 12:life12030355. [PMID: 35330106 PMCID: PMC8952775 DOI: 10.3390/life12030355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular-disease (CVD)-related mortality has been fueled by the upsurge of non-alcoholic steatohepatitis (NASH). Mesenchymal stem cells (MSCs) were extensively studied for their reparative power in ameliorating different CVDs via direct and paracrine effects. Several reports pointed to the importance of bone marrow mesenchymal stem cells (BM-MSCs) as a reliable therapeutic approach for several CVDs. Nevertheless, their therapeutic potential has not yet been investigated in the cardiotoxic state that is induced by NASH. Thus, this study sought to investigate the molecular mechanisms associated with cardiotoxicity that accompany NASH. Besides, we aimed to comparatively study the therapeutic effects of bone-marrow mesenchymal-stem-cell-derived extracellular vesicles (BM-MSCs-EV) and BM-MSCs in a cardiotoxic model that is induced by NASH in rats. Rats were fed with high-fat diet (HFD) for 12 weeks. At the seventh week, BM-MSCs-EV were given a dose of 120 µg/kg i.v., twice a week for six weeks (12 doses per 6 weeks). Another group was treated with BM-MSCs at a dose of 1 × 106 cell i.v., per rat once every 2 weeks for 6 weeks (3 doses per 6 weeks). BM-MSCs-EV demonstrated superior cardioprotective effects through decreasing serum cardiotoxic markers, cardiac hypoxic state (HIF-1) and cardiac inflammation (NF-κB p65, TNF-α, IL-6). This was accompanied by increased vascular endothelial growth factor (VEGF) and improved cardiac histopathological alterations. Both BM-MSCs-EV and BM-MSCs restored the mitochondrial antioxidant state through the upregulation of UCP2 and MnSOD genes. Besides, mitochondrial Parkin-dependent and -independent mitophagies were regained through the upregulation of (Parkin, PINK1, ULK1, BNIP3L, FUNDC1) and (LC3B). These effects were mediated through the regulation of pAKT, PI3K, Hypoxia, VEGF and NF-κB signaling pathways by an array of secreted microRNAs (miRNAs). Our findings unravel the potential ameliorative effects of BM-MSCs-EV as a comparable new avenue for BM-MSCs for modulating cardiotoxicity that is induced by NASH.
Collapse
|
81
|
Miceli V, Bertani A. Mesenchymal Stromal/Stem Cells and Their Products as a Therapeutic Tool to Advance Lung Transplantation. Cells 2022; 11:cells11050826. [PMID: 35269448 PMCID: PMC8909054 DOI: 10.3390/cells11050826] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Lung transplantation (LTx) has become the gold standard treatment for end-stage respiratory failure. Recently, extended lung donor criteria have been applied to decrease the mortality rate of patients on the waiting list. Moreover, ex vivo lung perfusion (EVLP) has been used to improve the number/quality of previously unacceptable lungs. Despite the above-mentioned progress, the morbidity/mortality of LTx remains high compared to other solid organ transplants. Lungs are particularly susceptible to ischemia-reperfusion injury, which can lead to graft dysfunction. Therefore, the success of LTx is related to the quality/function of the graft, and EVLP represents an opportunity to protect/regenerate the lungs before transplantation. Increasing evidence supports the use of mesenchymal stromal/stem cells (MSCs) as a therapeutic strategy to improve EVLP. The therapeutic properties of MSC are partially mediated by secreted factors. Hence, the strategy of lung perfusion with MSCs and/or their products pave the way for a new innovative approach that further increases the potential for the use of EVLP. This article provides an overview of experimental, preclinical and clinical studies supporting the application of MSCs to improve EVLP, the ultimate goal being efficient organ reconditioning in order to expand the donor lung pool and to improve transplant outcomes.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), 90127 Palermo, Italy
- Correspondence: (V.M.); (A.B.); Tel.: +39-091-21-92-430 (V.M.); +39-091-21-92-111 (A.B.)
| | - Alessandro Bertani
- Thoracic Surgery and Lung Transplantation Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
- Correspondence: (V.M.); (A.B.); Tel.: +39-091-21-92-430 (V.M.); +39-091-21-92-111 (A.B.)
| |
Collapse
|
82
|
Chiang MC, Chern E. Current Development, Obstacle and Futural Direction of Induced Pluripotent Stem Cell and Mesenchymal Stem Cell Treatment in Degenerative Retinal Disease. Int J Mol Sci 2022; 23:ijms23052529. [PMID: 35269671 PMCID: PMC8910526 DOI: 10.3390/ijms23052529] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Degenerative retinal disease is one of the major causes of vision loss around the world. The past several decades have witnessed emerging development of stem cell treatment for retinal disease. Nevertheless, sourcing stem cells remains controversial due to ethical concerns and their rarity. Furthermore, induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are both isolated from patients’ mature tissues; thus, issues such as avoiding moral controversy and adverse events related to immunosuppression and obtaining a large number of cells have opened a new era in regenerative medicine. This review focuses on the current application and development, clinical trials, and latest research of stem cell therapy, as well as its limitations and future directions.
Collapse
|
83
|
Recent research on the mechanism of mesenchymal stem cells in the treatment of bronchopulmonary dysplasia. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:108-114. [PMID: 35177185 PMCID: PMC8802385 DOI: 10.7499/j.issn.1008-8830.2109166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease due to impaired pulmonary development and is one of the main causes of respiratory failure in preterm infants. Preterm infants with BPD have significantly higher complication and mortality rates than those without BPD. At present, comprehensive management is the main intervention method for BPD, including reasonable respiratory and circulatory support, appropriate enteral nutrition and parenteral nutrition, application of caffeine/glucocorticoids/surfactants, and out-of-hospital management after discharge. The continuous advances in stem cell medicine in recent years provide new ideas for the treatment of BPD. Various pre-clinical trials have confirmed that stem cell therapy can effectively prevent lung injury and promote lung growth and damage repair. This article performs a comprehensive analysis of the mechanism of mesenchymal stem cells in the treatment of BPD, so as to provide a basis for clinical applications.
Collapse
|
84
|
Gallo A, Cuscino N, Contino F, Bulati M, Pampalone M, Amico G, Zito G, Carcione C, Centi C, Bertani A, Conaldi PG, Miceli V. Changes in the Transcriptome Profiles of Human Amnion-Derived Mesenchymal Stromal/Stem Cells Induced by Three-Dimensional Culture: A Potential Priming Strategy to Improve Their Properties. Int J Mol Sci 2022; 23:ijms23020863. [PMID: 35055049 PMCID: PMC8778321 DOI: 10.3390/ijms23020863] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are believed to function in vivo as a homeostatic tool that shows therapeutic properties for tissue repair/regeneration. Conventionally, these cells are expanded in two-dimensional (2D) cultures, and, in that case, MSCs undergo genotypic/phenotypic changes resulting in a loss of their therapeutic capabilities. Moreover, several clinical trials using MSCs have shown controversial results with moderate/insufficient therapeutic responses. Different priming methods were tested to improve MSC effects, and three-dimensional (3D) culturing techniques were also examined. MSC spheroids display increased therapeutic properties, and, in this context, it is crucial to understand molecular changes underlying spheroid generation. To address these limitations, we performed RNA-seq on human amnion-derived MSCs (hAMSCs) cultured in both 2D and 3D conditions and examined the transcriptome changes associated with hAMSC spheroid formation. We found a large number of 3D culture-sensitive genes and identified selected genes related to 3D hAMSC therapeutic effects. In particular, we observed that these genes can regulate proliferation/differentiation, as well as immunomodulatory and angiogenic processes. We validated RNA-seq results by qRT-PCR and methylome analysis and investigation of secreted factors. Overall, our results showed that hAMSC spheroid culture represents a promising approach to cell-based therapy that could significantly impact hAMSC application in the field of regenerative medicine.
Collapse
Affiliation(s)
- Alessia Gallo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Nicola Cuscino
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Flavia Contino
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Matteo Bulati
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Mariangela Pampalone
- Fondazione Ri.MED, 90127 Palermo, Italy; (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giandomenico Amico
- Fondazione Ri.MED, 90127 Palermo, Italy; (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giovanni Zito
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | | | - Claudio Centi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Alessandro Bertani
- Thoracic Surgery and Lung Transplantation Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy;
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
- Correspondence: ; Tel.: +39-09-1219-2430
| |
Collapse
|
85
|
Conditioned Medium from Bone Marrow Mesenchymal Stem Cells Restored Oxidative Stress-Related Impaired Osteogenic Differentiation. Int J Mol Sci 2021; 22:ijms222413458. [PMID: 34948255 PMCID: PMC8706339 DOI: 10.3390/ijms222413458] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress from high levels of intracellular reactive oxygen species (ROS) has been linked to various bone diseases. Previous studies indicate that mesenchymal stem cells (MSC) secrete bioactive factors (conditioned medium (MSC-CM)) that have antioxidant effects. However, the antioxidant role of MSC-CM on osteogenesis has not been fully studied. We aimed to identify antioxidant proteins in MSC-CM using mass spectrometry-based proteomics and to explore their effects on osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSC) exposed to oxidative stress induced by hydrogen peroxide (H2O2). Our analysis revealed that MSC-CM is comprised of antioxidant proteins that are involved in several biological processes, including negative regulation of apoptosis and positive regulation of cell proliferation. Then, hBMSC exposed to H2O2 were treated with MSC-CM, and the effects on their osteogenic differentiation were evaluated. MSC-CM restored H2O2-induced damage to hBMSC by increasing the antioxidant enzyme-SOD production and the mRNA expression level of the anti-apoptotic BCL-2. A decrease in ROS production and cellular apoptosis was also shown. MSC-CM also modulated mRNA expression levels of osteogenesis-related genes, runt-related transcription factor 2, collagen type I, bone morphogenic protein 2, and osteopontin. Furthermore, collagen type I protein secretion, alkaline phosphatase activity, and in vitro mineralization were increased. These results indicate that MSC-CM contains several proteins with antioxidant and anti-apoptotic properties that restored the impaired hBMSC osteogenic differentiation associated with oxidative stress.
Collapse
|
86
|
Franck T, Ceusters J, Graide H, Mouithys-Mickalad A, Serteyn D. Muscle Derived Mesenchymal Stem Cells Inhibit the Activity of the Free and the Neutrophil Extracellular Trap (NET)-Bond Myeloperoxidase. Cells 2021; 10:3486. [PMID: 34943996 PMCID: PMC8700239 DOI: 10.3390/cells10123486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/21/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to migrate to tissue injury sites to participate in immune modulation, tissue remodelling and wound healing, reducing tissue damage. Upon neutrophil activation, there is a release of myeloperoxidase (MPO), an oxidant enzyme. But little is known about the direct role of MSCs on MPO activity. The aim of this study was to investigate the effect of equine mesenchymal stem cells derived from muscle microinvasive biopsy (mdMSC) on the oxidant response of neutrophils and particularly on the activity of the myeloperoxidase released by stimulated equine neutrophils. After specific treatment (trypsin and washings in phosphate buffer saline), the mdMSCs were exposed to isolated neutrophils. The effect of the suspended mdMSCs was studied on the ROS production and the release of total and active MPO by stimulated neutrophils and specifically on the activity of MPO in a neutrophil-free model. Additionally, we developed a model combining adherent mdMSCs with neutrophils to study total and active MPO from the neutrophil extracellular trap (NET). Our results show that mdMSCs inhibited the ROS production, the activity of MPO released by stimulated neutrophils and the activity of MPO bound to the NET. Moreover, the co-incubation of mdMSCs directly with MPO results in a strong inhibition of the peroxidase activity of MPO, probably by affecting the active site of the enzyme. We confirm the strong potential of mdMSCs to lower the oxidant response of neutrophils. The novelty of our study is an evident inhibition of the activity of MPO by MSCs. The results indicated a new potential therapeutic approach of mdMSCs in the inhibition of MPO, which is considered as a pro-oxidant actor in numerous chronic and acute inflammatory pathologies.
Collapse
Affiliation(s)
- Thierry Franck
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Justine Ceusters
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Hélène Graide
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
| | - Ange Mouithys-Mickalad
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
| | - Didier Serteyn
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (J.C.); (H.G.); (A.M.-M.); (D.S.)
- Research Unit FARAH, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| |
Collapse
|
87
|
The therapeutic potential and efficiency of Intracerebroventricular transplantation and intravenous injection of Mesenchymal stem cells in relieving Aß hallmarks and improving cognitive dysfunction in AD induced model. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
88
|
Kulus M, Sibiak R, Stefańska K, Zdun M, Wieczorkiewicz M, Piotrowska-Kempisty H, Jaśkowski JM, Bukowska D, Ratajczak K, Zabel M, Mozdziak P, Kempisty B. Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues-Origins, Characteristics, Signaling Pathways, and Clinical Trials. Cells 2021; 10:cells10123278. [PMID: 34943786 PMCID: PMC8699543 DOI: 10.3390/cells10123278] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Rafał Sibiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
| | - Maciej Zdun
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Hanna Piotrowska-Kempisty
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Kornel Ratajczak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Correspondence:
| |
Collapse
|
89
|
Mazher KM, Ahmed OM, Abdallah Sayed H, Nabil TM. The Role of Bone Marrow-Derived Mesenchymal Stromal Cells and Hesperidin in Ameliorating Nephrotoxicity Induced by Cisplatin in Male Wistar Rats. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:133-146. [PMID: 34703797 PMCID: PMC8496246 DOI: 10.22088/ijmcm.bums.10.2.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/11/2021] [Indexed: 11/26/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) and antioxidants opened the way for many effective therapeutic experiments against damaged organs like kidneys. Nephrotoxicity is the main complication of chemotherapeutic drugs. Therefore, the present study aimed to investigate the efficacy of BM-MSCs and hesperidin to treat cisplatin-induced nephrotoxicity in rats. Fifty rats were divided into five equal groups of 10 each. Group-I served as a control group, group-II received a single dose of cisplatin (7.5 mg/kg) intraperitoneally to induce nephrotoxicity, group-III received a daily dose of hesperidin (40 mg/kg) orally for four weeks, and on the 5th day cisplatin was administered an hour before hesperidin administration. Group-IV consisted of cisplatin-treated rats that were intravenously injected with 1х106 BM-MSCs cells/rat once per week. Group V contained cisplatin-treated rats that received a combination of hesperidin and BM-MSCs with the same dosage regimes. After four weeks, serum and kidney samples were collected for biochemical, histological, and immunohistochemical examinations were performed. Cisplatin administered rats showed deteriorated biochemical parameters and severe degenerative changes in renal tissue. Both single and combined hesperidin and BM-MSCs treatments restored the renal biochemical parameters. Histologically, the renal tissues significantly improved in the BM-MSCs treated group in comparison with the hesperidin treated group. Moreover, combined treatment (i.e., group V) showed complete restoration of the normal architecture in the renal tissue. Our data suggest that the combined treatment of BM-MSCs and hesperidin has a potent renoprotective efficacy against cisplatin-induced nephrotoxicity rather than the single treatment.
Collapse
Affiliation(s)
- Khalid Mohamed Mazher
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Beni-Suef University, 62511, Beni-Suef, Egypt
| | - Osama Mohamed Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, 62521, Beni-Suef, Egypt
| | | | - Taghreed Mohamed Nabil
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Beni-Suef University, 62511, Beni-Suef, Egypt
| |
Collapse
|
90
|
Bhalerao A, Raut S, Noorani B, Mancuso S, Cucullo L. Molecular Mechanisms of Multi-Organ Failure in COVID-19 and Potential of Stem Cell Therapy. Cells 2021; 10:2878. [PMID: 34831101 PMCID: PMC8616204 DOI: 10.3390/cells10112878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023] Open
Abstract
As the number of confirmed cases and deaths occurring from Coronavirus disease 2019 (COVID-19) surges worldwide, health experts are striving hard to fully comprehend the extent of damage caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 primarily manifests itself in the form of severe respiratory distress, it is also known to cause systemic damage to almost all major organs and organ systems within the body. In this review, we discuss the molecular mechanisms leading to multi-organ failure seen in COVID-19 patients. We also examine the potential of stem cell therapy in treating COVID-19 multi-organ failure cases.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Salvatore Mancuso
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
91
|
Cheng YH, Cheng SJ, Chen HH, Hsu WC. Development of injectable graphene oxide/laponite/gelatin hydrogel containing Wharton's jelly mesenchymal stem cells for treatment of oxidative stress-damaged cardiomyocytes. Colloids Surf B Biointerfaces 2021; 209:112150. [PMID: 34656814 DOI: 10.1016/j.colsurfb.2021.112150] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/13/2021] [Accepted: 10/03/2021] [Indexed: 11/08/2022]
Abstract
In the initial stage of myocardial infarction (MI), cardiomyocyte necrosis activates aninflammatory response and increases the reactive oxygen species (ROS) content. Graphene oxide (GO) possesses potential antioxidant properties and can provide the adequate mechanical support for cell growth. The clinical studies showed that direct injection of Wharton's jelly mesenchymal stem cells (WJ-MSCs) into infarcted areas of myocardium can reduce apoptosis and fibrosis. Gelatin is a natural polymer and can promote cell attachment. Nanoclay laponite with shear-thinning properties can be injected and gelled in-situ without chemical triggers. In the study, injectable GO/laponite/gelatin (GO-LG) hydrogel was developed and characterized. The results of cell viability showed that the optimal concentration of GO flasks (200 to 300 nm) to treat cells was 100 μg/ml. Addition of nanosized GO to the laponite/gelatin (LG) hydrogel could increase the mechanical strength and have both hemocompatibility and cytocompatibility. The release of GO from LG hydrogel could inhibit the H2O2-induced oxidative stress. The GO-LG hydrogel containing WJ-MSCs could decrease inflammation and apoptosis level and increase the cell viability of cardiomyocytes under oxidative stress. We believe that utilizing this newly developed GO-LG hydrogel containing WJ-MSCs may have potential applications in the future for treatment of MI.
Collapse
Affiliation(s)
- Yung-Hsin Cheng
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.
| | - Shih-Jen Cheng
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Hsin-Ho Chen
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Wei-Chia Hsu
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|
92
|
Rowe G, Tracy E, Beare JE, LeBlanc AJ. Cell therapy rescues aging-induced beta-1 adrenergic receptor and GRK2 dysfunction in the coronary microcirculation. GeroScience 2021; 44:329-348. [PMID: 34608562 DOI: 10.1007/s11357-021-00455-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/03/2021] [Indexed: 01/08/2023] Open
Abstract
Our past study showed that coronary arterioles isolated from adipose-derived stromal vascular fraction (SVF)-treated rats showed amelioration of the age-related decrease in vasodilation to beta-adrenergic receptor (β-AR) agonist and improved β-AR-dependent coronary flow and microvascular function in a model of advanced age. We hypothesized that intravenously (i.v.) injected SVF improves coronary microvascular function in aged rats by re-establishing the equilibrium of the negative regulators of the internal adrenergic signaling cascade, G-protein receptor kinase 2 (GRK2) and G-alpha inhibitory (Gαi) proteins, back to youthful levels. Female Fischer-344 rats aged young (3 months, n = 24), old (24 months, n = 26), and old animals that received 1 × 107 green fluorescent protein (GFP+) SVF cells (O + SVF, n = 11) 4 weeks prior to sacrifice were utilized. Overnight urine was collected prior to sacrifice for catecholamine measurements. Cardiac samples were used for western blotting while coronary arterioles were isolated for pressure myography studies, immunofluorescence staining, and RNA sequencing. Coronary microvascular levels of the β1 adrenergic receptor are decreased with advancing age, but this decreased expression was rescued by SVF treatment. Aging led to a decrease in phosphorylated GRK2 in cardiomyocytes vs. young control with restoration of phosphorylation status by SVF. In vessels, there was no change in genetic transcription (RNAseq) or protein expression (immunofluorescence); however, inhibition of GRK2 (paroxetine) led to improved vasodilation to norepinephrine in the old control (OC) and O + SVF, indicating greater GRK2 functional inhibition of β1-AR in aging. SVF works to improve adrenergic-mediated vasodilation by restoring the β1-AR population and mitigating signal cascade inhibitors to improve vasodilation.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA
| | - Evan Tracy
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40292, USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
93
|
Galderisi U, Peluso G, Di Bernardo G. Clinical Trials Based on Mesenchymal Stromal Cells are Exponentially Increasing: Where are We in Recent Years? Stem Cell Rev Rep 2021; 18:23-36. [PMID: 34398443 PMCID: PMC8365566 DOI: 10.1007/s12015-021-10231-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs), present in the stromal component of several tissues, include multipotent stem cells, progenitors, and differentiated cells. MSCs have quickly attracted considerable attention in the clinical field for their regenerative properties and their ability to promote tissue homeostasis following injury. In recent years, MSCs mainly isolated from bone marrow, adipose tissue, and umbilical cord—have been utilized in hundreds of clinical trials for the treatment of various diseases. However, in addition to some successes, MSC-based therapies have experienced several failures. The number of new trials with MSCs is exponentially growing; still, complete results are only available for a limited number of trials. This dearth does not help prevent potentially inefficacious and unnecessary clinical trials. Results from unsuccessful studies may be useful in planning new therapeutic approaches to improve clinical outcomes. In order to bolster critical analysis of trial results, we reviewed the state of art of MSC clinical trials that have been published in the last six years. Most of the 416 published trials evaluated MSCs’ effectiveness in treating cardiovascular diseases, GvHD, and brain and neurological disorders, although some trials sought to treat immune system diseases and wounds and to restore tissue. We also report some unorthodox clinical trials that include unusual studies.
Collapse
Affiliation(s)
- Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA.,Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy. .,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
94
|
Mesenchymal Stem Cell-Based Therapy as an Alternative to the Treatment of Acute Respiratory Distress Syndrome: Current Evidence and Future Perspectives. Int J Mol Sci 2021; 22:ijms22157850. [PMID: 34360616 PMCID: PMC8346146 DOI: 10.3390/ijms22157850] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) represents a current challenge for medicine due to its incidence, morbidity and mortality and, also, the absence of an optimal treatment. The COVID-19 outbreak only increased the urgent demand for an affordable, safe and effective treatment for this process. Early clinical trials suggest the therapeutic usefulness of mesenchymal stem cells (MSCs) in acute lung injury (ALI) and ARDS. MSC-based therapies show antimicrobial, anti-inflammatory, regenerative, angiogenic, antifibrotic, anti-oxidative stress and anti-apoptotic actions, which can thwart the physiopathological mechanisms engaged in ARDS. In addition, MSC secretome and their derived products, especially exosomes, may reproduce the therapeutic effects of MSC in lung injury. This last strategy of treatment could avoid several safety issues potentially associated with the transplantation of living and proliferative cell populations and may be formulated in different forms. However, the following diverse limitations must be addressed: (i) selection of the optimal MSC, bearing in mind both the heterogeneity among donors and across different histological origins, (ii) massive obtention of these biological products through genetic manipulations of the most appropriate MSC, (iii) bioreactors that allow their growth in 3D, (iv) ideal culture conditions and (v) adequate functional testing of these obtaining biological products before their clinical application.
Collapse
|
95
|
Neuroprotective Potential of Bone Marrow-Derived Mesenchymal Stem Cells Following Chemotherapy. Biomedicines 2021; 9:biomedicines9070750. [PMID: 34209542 PMCID: PMC8301303 DOI: 10.3390/biomedicines9070750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin (CP) is extensively used in the medical oncology field for malignancy treatment, but its use is associated with neurological side effects that compromise the patients' quality of life. Cytotherapy is a new treatment strategy for tissue damage that has recently emerged. The use of bone marrow-derived mesenchymal stem cells (BM-MSCs) was investigated for its therapeutic potential against CP-induced chemobrain as well as various models of brain damage. This study was carried out to elucidate, for the first time, the role of the intravenous injection (IV) of BM-MSCs against CP-induced neurotoxicity in a rat model through investigation of the parameters of oxidative stress, inflammation, and apoptosis in brain tissue. A rat model of neurotoxicity was generated by intraperitoneal injection of 7.5 mg/kg CP while 2 × 106 BM-MSCs was given by IV as a therapeutic dose. Injection of CP led to a significant rise in malondialdehyde and nitric oxide levels accompanied by a marked depletion of superoxide dismutase and reduced glutathione content in brain tissue in comparison to the normal control (NC) rats. Furthermore, a remarkable rise in the brain levels of inflammatory cytokines interleukin (IL)-1β and IL-6, together with the expression of apoptotic marker caspase-3, and the downregulation of the brain expression of proliferating marker Ki-67 in brain tissue were detected in the CP group compared to the NC group. Histopathological alterations were observed in the brain tissue of the CP group. BM-MSCs mitigated the biochemical and histopathological alterations induced by CP without affecting brain cell proliferation. BM-MSCs could be used as a promising neuroprotective agent against CP-induced neurotoxicity.
Collapse
|
96
|
Tseng WC, Lee PY, Tsai MT, Chang FP, Chen NJ, Chien CT, Hung SC, Tarng DC. Hypoxic mesenchymal stem cells ameliorate acute kidney ischemia-reperfusion injury via enhancing renal tubular autophagy. Stem Cell Res Ther 2021; 12:367. [PMID: 34183058 PMCID: PMC8240301 DOI: 10.1186/s13287-021-02374-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is an emerging global healthcare issue without effective therapy yet. Autophagy recycles damaged organelles and helps maintain tissue homeostasis in acute renal ischemia-reperfusion (I/R) injury. Hypoxic mesenchymal stem cells (HMSCs) represent an innovative cell-based therapy in AKI. Moreover, the conditioned medium of HMSCs (HMSC-CM) rich in beneficial trophic factors may serve as a cell-free alternative therapy. Nonetheless, whether HMSCs or HMSC-CM mitigate renal I/R injury via modulating tubular autophagy remains unclear. METHODS Renal I/R injury was induced by clamping of the left renal artery with right nephrectomy in male Sprague-Dawley rats. The rats were injected with either PBS, HMSCs, or HMSC-CM immediately after the surgery and sacrificed 48 h later. Renal tubular NRK-52E cells subjected to hypoxia-reoxygenation (H/R) injury were co-cultured with HMSCs or treated with HMSC-CM to assess the regulatory effects of HSMCs on tubular autophagy and apoptosis. The association of tubular autophagy gene expression and renal recovery was also investigated in patients with ischemic AKI. RESULT HMSCs had a superior anti-oxidative effect in I/R-injured rat kidneys as compared to normoxia-cultured mesenchymal stem cells. HMSCs further attenuated renal macrophage infiltration and inflammation, reduced tubular apoptosis, enhanced tubular proliferation, and improved kidney function decline in rats with renal I/R injury. Moreover, HMSCs suppressed superoxide formation, reduced DNA damage and lipid peroxidation, and increased anti-oxidants expression in renal tubular epithelial cells during I/R injury. Co-culture of HMSCs with H/R-injured NRK-52E cells also lessened tubular cell death. Mechanistically, HMSCs downregulated the expression of pro-inflammatory interleukin-1β, proapoptotic Bax, and caspase 3. Notably, HMSCs also upregulated the expression of autophagy-related LC3B, Atg5 and Beclin 1 in renal tubular cells both in vivo and in vitro. Addition of 3-methyladenine suppressed the activity of autophagy and abrogated the renoprotective effects of HMSCs. The renoprotective effect of tubular autophagy was further validated in patients with ischemic AKI. AKI patients with higher renal LC3B expression were associated with better renal recovery. CONCLUSION The present study describes that the enhancing effect of MSCs, and especially of HMCSs, on tissue autophagy can be applied to suppress renal tubular apoptosis and attenuate renal impairment during renal I/R injury in the rat. Our findings provide further mechanistic support to HMSCs therapy and its investigation in clinical trials of ischemic AKI.
Collapse
Affiliation(s)
- Wei-Cheng Tseng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao-Tung University, Hsinchu, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Ying Lee
- Holistic Education Center, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Pang Chang
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Microbiology and Immunology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chiang-Ting Chien
- Department of Life Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shih-Chieh Hung
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Integrative Stem Cell Center, Department of Orthopedics, and Institute of New Drug Development, New Drug Development Center, China Medical University, Taichung, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, 128, Section 2, Academia Road, Taipei, 11529, Taiwan.
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan. .,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao-Tung University, Hsinchu, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao-Tung University, Hsinchu, Taiwan.
| |
Collapse
|
97
|
Jung H, Son GM, Lee JJ, Park HS. Therapeutic Effects of Tonsil-derived Mesenchymal Stem Cells in an Atopic Dermatitis Mouse Model. In Vivo 2021; 35:845-857. [PMID: 33622877 DOI: 10.21873/invivo.12325] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Mesenchymal stem cells (MSCs) have been suggested as an alternative therapeutic option in atopic dermatitis. Palatine tonsils are lymphoepithelial tissue located around the oropharynx and have been proposed as one of the important alternative sources of MSCs. The purpose of this study was to evaluate the protective and therapeutic effects of tonsil-derived MSCs (TMSCs) in a 2,4-dinitrofluorobenzene (DNFB)-induced mouse model of atopic dermatitis (AD). MATERIALS AND METHODS The effect of TMSCs was evaluated in 20 C57BL/6J mice that were randomly divided into four groups (normal, DNFB-PBS, DNFB-TMSC7, and DNFB-TMSC16 group). TMSCs were subcutaneously injected into DNFB-sensitized mice on day 7 (DNFB-TMSC7 group) and day 16 (DNFB-TMSC16 group). Several parameters of inflammation were assessed. RESULTS Subcutaneously injected TMSCs significantly improved the inflammatory symptoms in a DNFB-induced AD model mice, particularly showing therapeutic effects rather than protective effects. TMSC treatment inhibited T-cell-mediated inflammatory responses by decreasing the levels of IL-6, IL-1β, TNF-α (Th1 cell marker), IL-4 (Th2 cell marker), and B-cell-mediated serum IgE. In contrast, TMSCs enhanced the anti-inflammatory cytokine TGF-β. CONCLUSION In vitro and in vivo results suggest that TMSC treatment improved inflammatory skin lesions in the DNFB-induced AD mice model via immunomodulatory effects of the TMSCs. TMSCs inhibit T-cell and B-cell mediated responses, and enhance the anti-inflammatory responses.
Collapse
Affiliation(s)
- Harry Jung
- Institute of New Frontier Research Team, Hallym University, Hallym Clinical and Translation Science Institute, Chuncheon, Republic of Korea
| | - Gil Myeong Son
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae Jun Lee
- Department of Anesthesiology and Pain Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hae Sang Park
- Institute of New Frontier Research Team, Hallym University, Hallym Clinical and Translation Science Institute, Chuncheon, Republic of Korea; .,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
98
|
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: 4.0] [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
|
99
|
Planat-Benard V, Varin A, Casteilla L. MSCs and Inflammatory Cells Crosstalk in Regenerative Medicine: Concerted Actions for Optimized Resolution Driven by Energy Metabolism. Front Immunol 2021; 12:626755. [PMID: 33995350 PMCID: PMC8120150 DOI: 10.3389/fimmu.2021.626755] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are currently widely used in cell based therapy regarding to their remarkable efficacy in controlling the inflammatory status in patients. Despite recent progress and encouraging results, inconstant therapeutic benefits are reported suggesting that significant breakthroughs in the understanding of MSCs immunomodulatory mechanisms of action remains to be investigated and certainly apprehended from original point of view. This review will focus on the recent findings regarding MSCs close relationship with the innate immune compartment, i.e. granulocytes and myeloid cells. The review will also consider the intercellular mechanism of communication involved, such as factor secretion, cell-cell contact, extracellular vesicles, mitochondria transfer and efferocytosis. Immune-like-properties of MSCs supporting part of their therapeutic effect in the clinical setting will be discussed, as well as their potentials (immunomodulatory, anti-bacterial, anti-inflammatory, anti-oxidant defenses and metabolic adaptation…) and effects mediated, such as cell polarization, differentiation, death and survival on various immune and tissue cell targets determinant in triggering tissue regeneration. Their metabolic properties in term of sensing, reacting and producing metabolites influencing tissue inflammation will be highlighted. The review will finally open to discussion how ongoing scientific advances on MSCs could be efficiently translated to clinic in chronic and age-related inflammatory diseases and the current limits and gaps that remain to be overcome to achieving tissue regeneration and rejuvenation.
Collapse
Affiliation(s)
- Valerie Planat-Benard
- RESTORE, University of Toulouse, UMR 1031-INSERM, 5070-CNRS, Etablissement Français du Sang-Occitanie (EFS), Université Paul Sabatier, Toulouse, France
| | - Audrey Varin
- RESTORE, University of Toulouse, UMR 1031-INSERM, 5070-CNRS, Etablissement Français du Sang-Occitanie (EFS), Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- RESTORE, University of Toulouse, UMR 1031-INSERM, 5070-CNRS, Etablissement Français du Sang-Occitanie (EFS), Université Paul Sabatier, Toulouse, France
| |
Collapse
|
100
|
Hassen MT, Mohamed HK, Montaser MM, El-Sharnouby ME, Awad N, Ebiya RA. Molecular, Immunomodulatory, and Histopathological Role of Mesenchymal Stem Cells and Beetroot Extract on Cisplatin Induced Testicular Damage in Albino Rats. Animals (Basel) 2021; 11:ani11041142. [PMID: 33923635 PMCID: PMC8074130 DOI: 10.3390/ani11041142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The chemotherapeutic agent Cisplatin (Cis) has testicular damage as a side effect. Therefore, efforts are being done by scientists to get over this effect. The current experiment was done to utilize bone marrow-derived stem cells (BM-MSCs) and beetroot extract (BRE) in reducing the Cis testicular damage in rats. In the current study, Cis reduced the sperm count, plasma testosterone level, the testicular activity of alkaline phosphatase beside a marked inhabitation of succinate dehydrogenase activity. Also, it significantly increased malondialdehyde and along with a marked decrease in testis reduced glutathione content and total antioxidant capacity. At the same time, Cis administration resulted in a marked elevation in interleukine-6 and the iNOS and caspase-3 genes, however it decreased the expression of steroidogenic acute regulatory protein (StAR). Stem cell therapy (BM-MSCs) was accompanied with the use of herbal therapy (BRE) resulted in great improvement of all previous parameters. These results were confirmed by histopathological and immunohistochemical examination. In conclusion the current study recommends the use of beetroot as natural food in combination with stem cell therapy for the patient suffering from the testicular side effect of cisplatin chemotherapy. Abstract Cisplatin (Cis) a drug commonly used as a chemotherapeutic agent to treat various types of cancer, inducing testicular damage. The present study aimed to investigate the inhibitory potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) and beetroot extract (BRE) in albino rats after testicular toxicity induced by cisplatin. Thirty adult male albino rats were grouped into: the control group, Cis group receiving a single dose of 7 mg/kg i.p. (intraperitoneal) to induce testicular toxicity, Cis plus BM-MSCs injected Cis followed by 2 × 106 of BM-MSCs; Cis plus BRE group receiving Cis followed by 300 mg/kg body weight/day of BRE, and Cis plus BM-MSCs and BRE group. In the current study, Cis reduced sperm count, serum testosterone level, and testicular activity of alkaline phosphatase (AKP), besides a marked inhibition of succinate dehydrogenase (SDH) activity. In addition, it significantly increased malondialdehyde (MDA) and along with a marked decrease in testis reduced glutathione content and total antioxidant capacity (TAC). At the same time, Cis administration resulted in a marked elevation in interleukine-6 and the iNOS and caspase-3 genes; however, it decreased the expression of steroidogenic acute regulatory protein (StAR). Combined treatment with BM-MSCs and BRE resulted in great improvement of all previous parameters. These results were also confirmed by histopathological and immunohistochemical examination. In conclusion, both MSCs and BRE were found to have potent potentials to inhibit testicular damage induced by cisplatin.
Collapse
Affiliation(s)
- Marwa T. Hassen
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt; (M.T.H.); (H.K.M.); (R.A.E.)
| | - Hanaa K. Mohamed
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt; (M.T.H.); (H.K.M.); (R.A.E.)
| | - Metwally M. Montaser
- Science and Technology Department, University College of Ranyah, Taif University, Ranyah 21975, Saudi Arabia
- Correspondence:
| | | | - Nabil Awad
- Department of Genetics, Faculty of Agriculture and Natural Resources, Aswan University, Aswan 81528, Egypt;
- College of Biotechnology, Misr University for Science and Technology, Giza 12563, Egypt
| | - Rasha A. Ebiya
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt; (M.T.H.); (H.K.M.); (R.A.E.)
| |
Collapse
|