Abstract
Mesenchymal stem cells (multipotent stromal cells; MSCs) have been under investigation for the treatment of diverse diseases, with many promising outcomes achieved in animal models and clinical trials. The biological activity of MSC therapies has not been fully resolved which is critical to rationalizing their use and developing strategies to enhance treatment efficacy. Different paradigms have been constructed to explain their mechanism of action, including tissue regeneration, trophic/anti‐inflammatory secretion, and immunomodulation. MSCs rarely engraft and differentiate into other cell types after in vivo administration. Furthermore, it is equivocal whether MSCs function via the secretion of many peptide/protein ligands as their therapeutic properties are observed across xenogeneic barriers, which is suggestive of mechanisms involving mediators conserved between species. Oxidative stress is concomitant with cellular injury, inflammation, and dysregulated metabolism which are involved in many pathologies. Growing evidence supports that MSCs exert antioxidant properties in a variety of animal models of disease, which may explain their cytoprotective and anti‐inflammatory properties. In this review, evidence of the antioxidant effects of MSCs in in vivo and in vitro models is explored and potential mechanisms of these effects are discussed. These include direct scavenging of free radicals, promoting endogenous antioxidant defenses, immunomodulation via reactive oxygen species suppression, altering mitochondrial bioenergetics, and donating functional mitochondria to damaged cells. Modulation of the redox environment and oxidative stress by MSCs can mediate their anti‐inflammatory and cytoprotective properties and may offer an explanation to the diversity in disease models treatable by MSCs and how these mechanisms may be conserved between species.
Keywords: antioxidant, mesenchymal stem cell, mitochondria, multipotent stromal cell, oxidative stress, reactive oxygen species
MSCs exhibit antioxidant properties directly or indirectly. These antioxidant effects of MSCs associate with their therapeutic benefit in an array of pathologies.
Significance statement.
The role of mesenchymal stem cells (MSCs) in ameliorating oxidative and nitrosative injury has received considerable attention in recent years. The reduction‐oxidation (redox) environment regulates many physiological and pathophysiological mechanisms in cellular biology. Oxidative stress and redox imbalance are mediated by molecular constituents that are present in all living cells and share similar functions. The ability of MSCs to regulate these processes may offer an explanation to the diversity of disease models treatable by MSCs and to the effects of MSCs conserved between species. In this review, evidence of direct and indirect antioxidant mechanisms of MSC therapies is explored.
1. INTRODUCTION
Mesenchymal stem cells (multipotent stromal cells; MSCs) have been used as tools to treat a broad range of diseases in animal models due to their unique characteristics such as host immune evasion, rapid expansion, and their affluence in adult bone marrow and adipose tissue. The positive outcomes of these studies have driven hundreds of clinical trials into their application for diabetes, inflammatory disorders and various liver, kidney, lung, cardiovascular, musculoskeletal, neurological, and gastrointestinal diseases. 1 While several trials have demonstrated the therapeutic potential of MSCs, the failure to incorporate MSCs into current treatment regimens can be, in part, attributed to the lack of understanding pertaining to their biological mechanisms of action.
Initially, MSCs were explored as tools of regenerative medicine to replace damaged tissue. 2 However, administered MSCs were rarely observed to differentiate and effectively engraft into host tissues despite demonstrating favorable effects in many disease models. 3 Furthermore, the secretome of MSCs was identified to be therapeutic in many disease models in vitro and in vivo. Together, this resulted in a paradigm shift in recognition of the trophic actions of MSCs. 4 Despite extensive research investigating the anti‐inflammatory and trophic constituents of the MSC‐derived secretome, the therapeutic mechanisms of MSCs remain incompletely resolved. 5 MSCs demonstrate therapeutic attributes across xenogeneic barriers and, therefore, the therapeutic mechanisms of MSCs may be similar between species. There is strong evidence that the effects of MSCs are mediated via the secretion of protein/peptide ligands; however, it is equivocal whether these ligands are effective across xenogeneic barriers.
Recently, the role of MSCs in ameliorating oxidative and nitrosative injury has received considerable attention. The reduction‐oxidation (redox) environment regulates many physiological and pathophysiological mechanisms in cellular biology. Antioxidant effects of MSC therapy have been observed in various disease models such as diabetic injuries to the kidney, retina, sensory neurons, brain, and bone formation; chemotherapy‐ or radiation‐induced injury to the lungs, gonads, aorta, and brain; ischemic injury of the brain, heart, kidney, and liver; and traumatic injury to the spine and testis, cognitive disorders, gastrointestinal inflammation, septic injuries, and aging (Figure 1; Table 1). MSCs can directly reduce oxidative stress‐related injury in vitro in glial cells, neurons, cardiomyocytes, renal cells, endothelial cells, immune cells, hepatocytes, islet cells, fibroblasts, skeletal muscle, and other cells (Table 2). Oxidative stress is concomitant with cellular injury, inflammation, and dysregulated metabolism and, therefore, is a key pathophysiological mechanism of many diseases. Oxidative stress and redox imbalance are mediated by molecular constituents that are present in all living cells and share similar functions. Thus, the ability of MSCs to regulate these processes may offer an explanation to the diversity of disease models treatable by MSCs and to the effects of MSCs conserved between species.
FIGURE 1.
Antioxidant effects of MSC therapies. MSCs exhibit antioxidant properties directly by scavenging of free radicals and donating mitochondria or indirectly by upregulation antioxidant defenses in other cells and altering cellular bioenergetics. The immunosuppressive properties of MSCs can also avert the generation of reactive oxygen species (ROS). These mechanisms reduce oxidative stress, which associates with the therapeutic benefit of MSCs in an array of pathologies
TABLE 1.
Antioxidant activity of MSCs in disease models
Application | Model | MSCs used | Effects of MSC treatment | Antioxidant mechanisms | References |
---|---|---|---|---|---|
Aging | Aging‐related erectile dysfunction (rat) | Rat AT‐MSCs | ↑ Erectile response |
↓ Lipid peroxidation ↑ SOD activity |
62 |
Premature aging (Bmi−/−) (mouse) | Mouse amniotic membrane MSCs |
↑ Survival time ↓ Apoptosis in thymus and kidney ↑ Proliferation in thymus and kidney ↑ Mature immune cells ↑ Skeletal muscle growth ↓ Osteoporosis ↑ Bmi‐1 in liver, kidney, thymus, muscle, spleen, lung, and bone marrow |
↓ H2O2, ↑CAT, ↑SOD in the heart, liver, spleen, lung, kidney, BM and thymus, ↓ ROS in all except heart. ↓ DNA damage in cells of BM, spleen, lung and thymus MSCs secrete SOD (total) and CAT |
33 | |
Chemotherapy and radiation | Bleomycin‐induced pulmonary fibrosis (rat) | Rat mesenchymal stem cells (H4320‐1) | ↓ Fibrosis |
↓ NRF2, ↓ NQO1, ↓ HO‐1, ↓ γ‐GCS ↓ Lipid peroxidation, ↑ SOD activity
|
63 |
Bleomycin‐induced pulmonary fibrosis (mouse) | Human BM‐MSCs cell line Ue6E7T‐2 | ↓ Collagen |
↓ DNA oxidation ↓ ER stress marker BiP Effects negated by silencing STC‐1 and enhanced by STC‐1 over expression |
127 | |
Cisplatin‐induced acute kidney injury (rat) |
Human UC‐MSC exosomes |
↓ Blood urea nitrogen (MSC‐CM and fibroblast exosomes had no effect) ↓ Creatinine (MSC‐CM and fibroblast exosomes had no effect) ↓ Gross morphological damage ↓ Apoptosis (TUNEL) ↑ PCNA ↓ Bax, ↑ Bcl‐2 ↓ p38MAPK |
↓ DNA oxidation ↑ GSH ↓ Lipid peroxidation |
||
Cisplatin‐induced cognitive impairment (mice) | Mouse BM‐MSCs Intranasal delivery | ↑ Cognitive function |
↑ Maximal respiratory capacity and spare respiratory capacity of mitochondria ↓ Morphologically atypical mitochondria |
112 | |
Cisplatin‐induced gonadotoxicity (rat) | Rat BM‐MSCs |
↑ Testis weight and testosterone levels ↓ TNFα |
↓ Lipid peroxidation, ↑ SOD activity, ↑ GSH ↓ iNOS |
64 | |
Cisplatin‐induced renal injury (mouse) |
Mouse BM‐MSC‐CM |
↓ Weight loss ↓ Serum creatinine levels ↓ c‐caspase 3 expression ↓ Gross morphological damage |
|
94 | |
Lung radiation injury (mouse) | Mouse aorta‐derived and BM‐MSCs | ↓ Lung fibrosis |
↑ SOD1 expression in irradiated lung
|
59 | |
Paclitaxel‐induced neuropathy (rat) | Rat BM‐MSCs |
↑ Responses to thermal hyperalgesia and cold allodynia Sciatic nerve: ↑ NGF ↓ Pro‐inflammatory cytokines ↓ c‐caspase 3 |
Sciatic nerve: ↑ Total antioxidant capacity |
55 | |
Radiation‐induced aortic injury (mouse) | Human BM‐MSCs |
↓ Aorta thickness ↓ Collagen ↓ TGFβ ↓ TNFα ↓ ICAM ↓ Apoptosis |
↓ Nitrotyrosine ↓ Lipid peroxidation, ↑ HO‐1 ↑ CAT |
82 | |
Radiation‐induced neurological complication (mouse) | Human AT‐MSCs |
↑ Cognitive function ↓ Neuron loss ↓ Caspase 3 |
↓ Lipid peroxidation in hippocampus and brain lateral ventricle ↓ iNOS |
41 | |
Hyperglycemic injuries | Alloxan‐induced diabetes (rat) | Rat BM‐MSCs |
↑ Insulin ↓ Glucose levels ↓ Total cholesterol ↓ Triglycerides ↑ Vitamin E |
↑ GSH, ↑ GST, ↑ SOD ↓ NO ↓ lipid peroxidation |
88 |
Db/Db mouse model of type 2 diabetes | Mouse amniotic fluid MSCs |
↓ Weight gain ↓ Pro‐inflammatory cytokines↓ Apoptosis
|
↓ Lipid peroxidation ↓ 8‐isoprostane ↑ GSH ↓ GSSG |
85 | |
Diabetes‐induced cognitive impairment (mouse) | Rat BM‐MSC and exosomes |
↑ Cognitive function Hippocampus (CA1):
|
↓ Lipid peroxidation | 26 | |
Diabetic retinopathy (mouse) | Mouse AT‐MSCs Intravitreal injection |
↓ Retinal ganglion cell loss ↑ NGF, bFGF and GDNF ↑ TSP1 |
↓ ROS and lipid peroxidation | 28 | |
STZ‐induced diabetic osteoarthritis (mouse) | Mouse AT‐MSCs |
↑ Chondrocytes ↓ RAGE, NFκB |
↓ Lipid peroxidation | 128 | |
STZ‐induced diabetic nephropathy (rat) | Rat BM‐MSCs | ↓ Urinary albumin excretion and ameliorated glomerulosclerosis |
↓ Lipid peroxidation ↓ ROS ↑ SOD activity ↓ GLUT1 |
29 | |
STZ‐induced sensorial diabetic Neuropathy (mouse) |
Mouse BM‐MSCs |
↓ IL‐1β ↓TNFα ↑IL‐10 ↑ TGFβ ↓ Galectin‐3 |
↓ Lipid peroxidation ↓ Nitrite ↓ CAT ↓Gpx ↓ Nrf2 mRNA |
40 | |
STZ‐nicotinamide (diabetes)‐induced cardiac damage (rat) | Rat BM‐MSCs |
↓ c‐caspase 3, Bax and ↑ Bcl‐2 ↓ Cardiac fibrosis |
↓ Total oxidant content in serum ↑ Total antioxidant capacity in heart ↓ MPO ↓ iNOS |
56 | |
Inflammation | Caecal ligation‐puncture induced sepsis (rat) | Rat AT‐MSCs Administration of serum‐starved MSCs |
↑ Apoptosis in serum‐starved MSC↓ TNFα, NFkB in lungs and kidney ↓ Mitochondrial Bax and ↑ Bcl‐2 in lungs and kidney
|
↓ Protein oxidation in kidney ↑ NQO1 and HO‐1 in the lungs |
92 |
DSS‐induced colitis (mouse) | Mouse BM‐MSCs |
↓ Mucosal permeability: D‐lactic acid and Diamine oxidase ↑ E‐cadherin |
↓ Lipid peroxidation ↑ SOD |
65 | |
E coli‐induced Acute lung injury (mouse) |
Mouse BM‐MSCs | ↓ Edema |
↓ MPO activity ↑ CAT, SOD, GPx, glutathione reductase and GSH ↑ Total antioxidant capacity ↓ Lipid peroxidation ↓ iNOS expression |
37 | |
| |||||
Endotoxin‐induced inflammation in plasma (mouse) | Mouse BM‐MSCs | ↑ Cys but ND to lung fibroblast
|
↑ GSH ND to glutathione disulfide (GSSG) or cysteine (CySS) ↓ GSH/GSSG redox potential ↓ Cys/CySS redox potential, ND to lung fibroblast |
51 | |
Freund's adjuvant‐induced arthritis (rat) | Rat BM‐MSCs |
↓ Antinuclear antibodies ↓ TNFα, IL‐9 and IL‐4 ↑ IFNγ and TGFβ ↓ Immune cell infiltration ↓ Cartilage and bone loss |
↓ Lipid peroxidation ↑ GSH↑ SOD activity |
66 | |
HOCl‐induced systemic sclerosis (mouse) | Mouse BM‐MSCs |
Serum: ↓ Systemic sclerosis biomarker (SCL‐70) Skin and lung: ↓ Collagen, ↓ αSMA ↓ TGFβ1 |
↓ Advanced oxidation protein products ↑ Total antioxidant capacity |
57 | |
IL‐10 −/− model of colitis (mouse) | Human BM‐MSCs |
↓ TNFα, IFNγ, IL‐4 and p‐NFκB |
↓ O2 .− and H2O2 ↓ Lipid peroxidation ↑ SOD1 and CAT |
34 | |
Immune complex‐mediated dermal vasculitis (mouse) | Human AT‐MSCs |
↓ Neutrophil accumulation ↓ Vascular permeability |
|
79 | |
LPS‐induced lung injury (rat) |
Rat BM‐MSCs |
↓ Lung edema ↓ Bronchoalveolar lavage protein ↓ Bronchoalveolar lavage cells ↓ Neutrophils ↓ NFκB, ↑ IL‐10 |
↓ MPO ↓ Lipid peroxidation ↓ FASL |
||
Sepsis‐induced brain injury (rat) |
Rat AT‐MSC exosomes |
↓ Apoptosis ↓ Inflammatory markers |
↓ Protein oxidation ↓ NOX1‐2 |
25 | |
Severe acute pancreatitis (rat) | Human BM‐MSCs |
↓ Serum amylase and lipase ↓ Pancreatic damage ↓ Inflammatory cells
↓ Pro‐inflammatory cytokines: TNFα, IL‐1β, IL‐6 |
↓ Lipid peroxidation ↑ SOD activity, ↑ GPx ↓ iNOS |
67 | |
Severe acute pancreatitis (rat) | Rat BM‐MSCs | ↓ Pancreatitis score
|
↑ HO‐1 ↑ CO ↓ MPO ↓ ROS and lipid peroxidation ↑ SOD activity and CAT expression |
30 | |
Kidney and bladder disease | Atherosclerosis‐induced chronic bladder ischemia (rat) | Human amniotic fluid‐derived MSCs |
↓ Bladder overactivity ↓ TNFα |
↓ DNA oxidation ↓ Lipid peroxidation |
129 |
Metabolic renovascular disease in swine |
Swine AT‐MSC extracellular vesicles |
↑ Capillary density |
Glutathione Peroxidase 1,4,6,7, GST Zeta 1, SOD1‐3, Peroxiredoxin 1‐6, Catalase, Cold Shock Domain Containing E1, Cytoglobin, Microsomal GST 3, Prostaglandin‐Endoperoxide Synthase 1, Peroxidasin, Albumin, Apolipoprotein E, Glutathione‐Disulfide Reductase, Thioredoxin Reductase 1‐2 ↓ 8‐isoprostane ↓ ROS ↓ Nitrotyrosine |
31 | |
Unilateral ureteral obstruction (rat) | Human UC‐MSC‐CM |
↓ Renal tubular damage ↓ Fibrosis ↓ Apoptosis ↑ Cell proliferation |
↓ ROS ↓ Lipid peroxidation ↑ GSH |
24 | |
Liver disease | Acetaminophen‐induced acute liver failure (mouse) | Human UC‐MSCs |
↑ Survival and liver weight ↓ Biomarkers of liver failure ↓ Apoptotic cells and necrotic tissue ↓ IL‐6 only observed with pretreatment |
↑ GSH ↑ SOD activity ↓ Lipid peroxidation only observed with pretreatment |
38 |
CCl4‐induced liver fibrosis (mouse) | Human BM‐MSCs | ↓ p47‐phox cells |
↓ Lipid peroxidation ↑ SOD activity, CAT and GSH |
68 | |
CCl4‐induced liver injury (mouse) | Allogeneic BM‐MSC |
↑ Serum albumin ↓ Serum ALT and AST ↓ Expression of TNFα, IL‐6, type 1 collagen and αSMA
|
↓ MPO ↑ SOD and catalase ↓ Lipid peroxidation |
||
CCl4–induced liver injury (rat) | Human BM‐MSC |
↓ Serum ALT and AST ↓ Liver fibrosis |
↑ GSH ↓ Lipid peroxidation |
89 | |
CCl4‐induced rat liver fibrosis | Human BM‐MSCs (cells and exosomes) |
↓ Fibrosis ↓ Pro‐inflammatory cytokines ↓Wnt signaling |
↓ Lipid peroxidation | 130 | |
N‐diethylnitrosamine‐induced hepatocarcinoma (rat) | Rat BM‐MSCs |
Administration of MSCs at early stage: ↓ Tumor incidence ↓ Tumor volume Administration of MSCs at late stage: ↑ Tumor volume |
↓ Lipid peroxidation ↓ Mitochondrial O2 .− ↑ Total antioxidative capacity ↓ DNA damage |
36 | |
Thioacetamide‐induced liver injury (mouse) | Canine BM‐MSCs |
↓ Lung injury ↓ Fibrosis |
↑ Total antioxidant capacity ↓ Lipid peroxidation |
58 | |
Lung diseases | Cigarette smoke‐induced chronic obstructive pulmonary disease (guinea pig) |
Guinea pig AT‐MSCs IV and intratracheal delivery |
|
↑ Thiol after IV administration ↓ Lipid peroxidation after IV and intratracheal delivery |
131 |
Mustard lung (human case study) | Human AT‐MSCs |
|
↑ GSH in sputum ↓ Lipid peroxidation |
49 | |
Ovalbumin and aluminum hydroxide‐induced asthma (mouse) | Human BM‐MSC |
↓ Mucin ↓ Collagen |
↓ Nitrotyrosine | 132 | |
Neural injury and cognition | APP/PS1 transgenic model of Alzheimer's disease (mouse) | Rat AT‐MSCs |
↑ Recognition in behavioral test ↑ Neurogenesis |
↓ ROS | 32 |
Chronic ethanol intake (rats) |
Human AT‐MSCs |
↓ETOH intake ↓Relapse after ETOH deprivation |
↓ Hippocampal GSSG/GSH | 86 | |
Collagenase induced‐intracerebral hemorrhage (rat) | Rat BM‐MSCs |
↓ Apoptosis ↓ Edema ↓ Blood–brain barrier permeability ↓ Pro‐inflammatory cytokines ↑ TSG6, TGFβ1 and IL‐10 |
↓ iNOS ↓ ONOO− ↓ MPO |
39 | |
Pilocarpine induction of temporal lobe epilepsy (rat) | Rat BM‐MSCs |
↓ Caspase 3 ↓ Glutamate ↑GABA↓TNFα ↓ IL‐1β |
↑ GSH ↓ Lipid peroxidation ↑ Paraoxonase‐1 |
90 | |
Spontaneous stroke (rat) |
Rat BM‐MSCs |
↓ Apoptosis |
↓ O2 .− ↓ Lipid peroxidation |
35 | |
Tg2576 mice (Alzheimer's disease) | Human UC‐MSCs |
Effects on hippocampus:
↑Neurogenesis |
↓ Lipid peroxidation ↑ SOD activity ↑ nNOS ↑total NO |
43 | |
YG8 transgenic model of Friedreich's ataxia (mouse) | Mouse BM‐MSCs |
↑ BDNF, NT3, and NT4 in dorsal root ganglia (DRG) ↑ GFAP, Tuj1, and MAP2 in DRG ↑ Bcl‐2 ↑ Frataxin |
↓ SOD2 and SOD3↑ CAT and GPx1 | 81 | |
Oxygen tension injuries | Acute ischemic stroke (rat) | Swine AT‐MSCs |
↓ Infarct area ↓ Inflammatory cytokines ↓ c‐caspase 3 ↓ c‐PARP ↓ γ‐H2AX ↓cytosolic cytochrome c |
↓ NOX1 and NOX2 ↓ Protein oxidation |
102 |
Acute myocardial infarction (swine) |
Swine BM‐MSCs Autologous |
↓ Bax, c‐caspase 3 and c‐PARP ↓ Inflammation ↓ Infarct area
|
↓ Oxidized protein ↓ NOX1 and NOX2 |
103 | |
Hepatic I/R injury (rat) |
Human UC‐MSC extracellular vesicles |
↓ Necrotic area ↓ c‐caspase3 |
↓ ROS
SOD2 mimetic restored effects of MSC |
80 | |
I/R model of laparoscopic partial hepatectomy (swine) | Swine AT‐MSCs |
|
↓ Lipid peroxidation ↓ MPO ↑ SOD activity |
69 | |
Kidney from acute I/R injury (rat) | Rat AT‐MSCs (cells and exosomes) |
↓ Histological injury score ↓ Pro‐inflammatory cytokines ↓ TGFβ↓ c‐caspase 3, c‐PARP, mitochondrial Bax, cytochrome c translocation
|
↓ NOX1 and NOX2 ↓ Oxidized protein ↓ MPO ↓ γ‐H2AX DNA damage |
104 | |
Kidney ischemia (rat) |
Human WJ‐MSC Extracellular vesicles |
↓ Renal injury score ↓ Apoptosis ↑ Nuclear NRF2
|
↓ Lipid peroxidation ↓ DNA oxidation ↑ SOD activity ↑ HO‐1 expression |
52 | |
Myocardial I/R injury (mouse) | HuES9.E1 derived MSC exosomes |
↓ Infarct size in vivo and ex vivo
↑ Contraction and relaxation ↓ End‐diastolic pressure ↑ pAkt, pGSK3 alpha/beta and ↓ pJnk ↓ Peripheral leukocytes |
↑ ATP/ADP NADH/NAD+ ratios after reperfusion ↓ Protein oxidation |
101 | |
Neonatal hyperoxic lung injury (rat) | Human UCB‐derived MSCs |
↓ Apoptosis ↓ Pro‐inflammatory cytokines
|
↓ P47phox ↓ MPO |
133 | |
O2‐induced bronchopulmonary dysplasia (rat) | Rat BM‐MSC‐CM |
↓ Pulmonary hypertension ↓ Right ventricular hypertrophy ↓ Pulmonary arterial medial wall thickness ↓ Gross morphological damage to alveoli ↑ Therapeutic value from the CM of hyperoxic MCSs compared to normoxic MSCs
|
↑ hydroxyl radical antioxidant capacity in MSCs compared with lung fibroblast | 61 | |
Renal I/R injury (rat) | Rat BM‐MSCs |
↓ Cellular degeneration (histopathology) ↑ EGF MSCs had no effect on pERK1/2
|
↑ HO‐1 ↓ DNA oxidation |
93 | |
Renal I/R injury (rat) | Human WJ‐MSC micro vesicles |
↑ Cell proliferation ↓ Apoptosis ↓ Fibrosis
|
↓ ROS, lipid peroxidation and protein oxidation ↓ NOX2 |
134 | |
Semaxanib/hypoxia‐induced pulmonary arterial hypertension (rat) | Human BM‐MSCs |
↓Hypertension In pulmonary artery: ↑ Metabolites TCA cycle‐associated metabolites ↓ Fructose and sorbitol—glycolysis associated |
↑ GSH/GSSG and Cys/CySS ratios | 87 | |
Small bowel I/R injury (rat) | Rat AT‐MSCs |
↓ Intestinal permeability ↓ TNFα and NFκB ↓ Protein oxidation and lipid peroxidation ↓ Cytosolic cytochrome c, c‐caspase 3, c‐caspase 9, c‐PARP, APAF‐1 and mitochondrial Bax ↓ Cell proliferation ↓ Apoptosis ↓ Immune cells |
↓ MPO and iNOS ↓ NOX1 and NOX2 ↑ NQO1, glutathione reductase, GPx1 ↑ HO‐1 cells |
84 | |
Tissue/cell engraftment | Ovarian tissue autograft (mouse) | Mouse AT‐MSCs |
↑ Graft efficacy ↑ IL‐10 ↓TNFα and IL‐6 ↓ Apoptosis |
↑ SOD activity ↓ Lipid peroxidation |
70 |
Traumatic injuries | Spinal cord injury (canine) | Canine AT‐MSCs |
↑ Motor function ↓ Hemorrhagic area ↓ Microglia ↓ TNFα, IL‐6 and COX2 |
↓ Lipid peroxidation and protein oxidation | 42 |
Testicular torsion injury (rat) | Rat AT‐MSCs | ↓ Apoptosis | ↓ Lipid peroxidation | 135 |
Abbreviations: ALT, alanine aminotransferase; APAF‐1, apoptotic protease activating factor 1; AST, aspartate aminotransferase; AT‐MSC, adipose tissue‐derived MSC; BAX, Bcl‐2‐associated X protein; Bcl‐2, B‐cell lymphoma 2; BDNF, brain‐derived neurotrophic factor; bFGF, basic fibroblast growth factor; BM‐MSCs, bone marrow‐derived MSC; CAT, catalase; c‐caspase 3, cleaved‐caspase 3; CM, conditioned medium; CO, carbon monoxide; COX2, cyclooxygenase‐2; c‐PARP, cleaved poly (ADP‐ribose) polymerase; Cys, cysteine; DRG, dorsal root ganglion; DSS, dextran sulfate sodium; EGF, epidermal growth factor; ER stress, endoplasmic reticulum stress; GABA, gamma‐aminobutyric acid; GDNF, glial cell‐derived neurotrophic factor; GFAP, glial fibrillary acidic protein; GLUT1, glucose transporter 1; GPx, glutathione peroxidase; GSH, glutathione; H2O2, hydrogen peroxide; HO‐1, heme oxygenase‐1; I/R, Ischemia / reperfusion; ICAM, Intercellular adhesion molecule; IFNγ, interferon gamma; IL, interleukin; iNOS, inducible nitric oxide synthase; IV, intravenous; LPS, lipopolysaccharide; MAP2, microtubule associated protein‐2; MPO, myeloperoxidase; NAD(P)H, nicotinamide adenine dinucleotide phosphate hydrogen; ND, No difference; NFκB, Nuclear factor κB; NGF, Nerve growth factor; nNOS, neuronal nitric oxide synthase; NO, nitric oxide; NOX, NAD(P)H oxidase; NQO1, NAD(P)H quinone dehydrogenase 1; NRF2, nuclear factor erythroid 2‐related factor 2; NT3 and 4, neurotrophin 3 and 4; O2−, superoxide; ONOO−, peroxynitrite; p38MAPK, p38 mitogen‐activated protein kinases; pAkt, phosphorylated protein kinase B; PCNA, proliferating cell nuclear antigen; pGSK3, phosphorylated glycogen synthase kinase 3 beta; pJnk, phosphorylated c‐Jun N‐terminal kinases; pERK1/2, phosphorylated extracellular signal‐regulated kinases 1/2; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species; SOD, superoxide dismutase; STC‐1, stanniocalcin‐1; STZ, streptozocin; TCA cycle, tricarboxylic acid cycle; TGFβ, transforming growth factor beta; TNFα, tumor necrosis factor alpha; TSG6, TNFα‐stimulated gene‐6; TSP1, thrombospondin 1; Tuj1, neuron‐specific class III beta‐tubulin; UC‐MSC, umbilical cord‐derived MSC; αSMA, alpha‐smooth muscle actin; γ‐GCS, gamma‐glutamylcysteine synthetase; γ‐H2AX, gamma‐H2A histone family member X; Δψm, mitochondrial membrane potential; CySS, cystine (disulfide form of cysteine); GSSG, glutathione disulfide.
TABLE 2.
Antioxidant effects of MSCs in in vitro models
Cell types | Model | MSCs used | Antioxidant and other effects of MSCs | References | |
---|---|---|---|---|---|
Cardiomyocytes and endothelial cells | Glucose‐deprived hypoxia‐reoxygenated H9c2 cardiomyocytes (rat) | Rat BM‐MSCs Direct coculture with GFP+ MSCs |
↓ Apoptosis ↓ Bax ↑ Bcl‐2 ↓ Caspase 3 ↓ Δψm
|
121 | |
H2O2‐treated RL14 cardiomyocytes and human umbilical vein endothelial cells (HUVEC) | Human AT‐MSCs |
|
53 | ||
I/R of ventricular myocytes (mouse) in vitro | Mouse BM‐MSC‐CM |
↓ Cell loss ↓ Early afterdepolarization of myocytes ↓ Excessive depolarization of Δψm after reperfusion ↓ Exaggerated hyperpolarization of Δψm after acute reperfusion—effect prevented by PI3K, Akt, and Ik,ATP inhibition
↓ Δψm hyperpolarization ↓ Mitochondrial O2 .−
|
54 | ||
Oxygen glucose deprivation and reoxygenation of human umbilical vein endothelial cells (HUVEC) | Human BM‐MSCs |
↓ Cell death ↑ Oxygen consumption rate and ↓ extracellular acidification rate
|
136 | ||
Cytarabine‐treated human umbilical cord vein endothelial cells (HUVEC) | Human BM‐MSCs |
↓ Apoptosis ↑ Capillary formation |
122 | ||
tert‐Butyl hydroperoxide‐treated umbilical endothelial cells (human) | Human placental MSC‐CM |
↓ ROS ↓ Apoptosis
↑ SOD2 mRNA and protein
|
77 | ||
Fibroblasts | tert‐Butyl hydroperoxide‐treated human dermal fibroblasts |
Human AT‐MSC‐CM |
↑ Antioxidant capacity over normal culture media ↑ Cell survival ↓ Morphological damage ↑ SOD activity in human dermal fibroblasts ↑ GPx activity in human dermal fibroblasts |
||
UV‐exposed fibroblasts (human) | Human UC‐MSC‐CM |
↑ Cell viability ↑ SOD activity |
73 | ||
Glial cells and neurons |
Activated microglia and NO‐induced neuronal death (rat) |
Human BM‐MSCs |
↓ Neuronal loss from activated microglia ↓ Neuronal loss from NO
|
71 | |
Amyloid‐β oligomer‐induced damage to hippocampal neurons (rat) |
Rat BM‐MSCs Transwell coculture and exosomes |
↓ ROS
|
83 | ||
Glucose‐deprived hypoxia‐reoxygenated primary astrocytes (human) |
Human dental pulp‐derived and BM‐MSCs Transwell and CM |
↑ Viability of astrocytes ↓ ROS ↓ IL‐1β after MSC‐CM ↓ Astrogliosis |
137 | ||
Glucose‐deprived scratch injured T98G glioblastoma cells (human) | Human AT‐MSC‐CM |
↑ Wound closure ↑ Viability ↓ ROS |
138 | ||
H2O2‐treated cortex‐derived neural stem cells (rat) | Rat BM‐MSC‐CM |
↓ Apoptosis ↓ Lipid peroxidation ↑ SOD activity |
74 | ||
H2O2−treated motor neurons (NSC‐34) expressing human mutant SOD1 (ALS) | Mouse AT‐MSC exosomes | ↑ Cell viability of naïve cells and SOD1 mutant cells | 139 | ||
H2O2‐treated retinal ganglion cells (RGCs) (rat) |
Rat BM‐MSCs Transwell |
↓ Apoptosis ↓ Lipid peroxidation and ↑SOD activity in RGCs ↓ IL‐1β and TNFα in supernatant ↑ BDNF and CNTF in RGCs |
75 | ||
H2O2‐treated SH‐SY5Y neuroblastoma cells (human) |
Human AT‐MSC CM |
↑ Viability ↓ Antioxidant capacity ↓ ROS
|
140 | ||
Sevoflurane‐induced apoptosis in human neuroglioma H4 cells | Rat BM‐MSCs |
↓ c‐caspase 3 and Bax ↓ ROS ↓ Cytochrome c translocation ↑ ATP |
141 | ||
Hepatocytes | Acetaminophen and H2O2‐treated human hepatocytes (HepG2) | Rat BM‐MSC‐CM |
↑ Cell viability ↓ ROS |
142 | |
H2O2‐treated AML12 hepatocytes (murine) | Mouse BM‐MSC extracellular vesicles |
↓ ROS ↓ Pro‐inflammatory cytokines |
143 | ||
H2O2‐treated human fetal hepatocytes (LO2 cells) | Human UC‐MSCs extracellular vesicles |
↓ ROS ↓ Mitochondrial O2 .− ↓ Apoptosis
Silencing of SOD2 in MSCs inhibits therapeutic effect of exosomes |
80 | ||
Immune cells | Cytarabine or methotrexate‐treated immortalized human T lymphocytes (Jurkat cells) | Human BM‐MSCs |
MSC direct coculture: ↓ Apoptosis ↓ Mitochondrial O2 .−
|
120 | |
LPS‐stimulated blood‐derived monocytes (human) | Human AT‐MSCs |
↓ TNFα ↓ Nitrite ↓ COX2 ↓ MPO ↓ ROS |
110 | ||
LPS‐treated human monocyte‐derived macrophages |
Human BM‐MSC CM and extracellular vesicles |
↑ oxygen consumption rate ↑ phagocytic phenotype
|
114 | ||
LPS‐treated neutrophils (human) |
Human UC‐MSCs Transwell and extracellular vesicles |
↓ Lipid peroxidation ↓ ROS ↓ MPO activity
|
80 | ||
Macrophages in vitro (human and mouse) | Human BM‐MSCs |
↓ ROS‐associated with NRLP3 inflammasome activation ↓ NRLP3 associated caspase 1 activation ↓ NRLP3 associated IL‐1β and IL‐18 secretion ↓ TNFα and IL‐6 transcription
|
111 | ||
PMA‐activated neutrophils (mouse and human) | Human AT‐MSCs |
↓ Respiratory burst (ROS) dependent on SOD3 expression by MSCs ↓ Apoptosis ↓ MPO protein and activity |
79 | ||
Islet cells | Cytokine cocktail‐exposed islet cells (rat) | Human BM‐MSCs |
↑ Insulin secretion ↑ SOD1, ↑ NQO1, ↑HO‐1 ↑Ferritin H |
78 | |
Hypoxia (1% O2) exposed porcine islet cells |
Human UC‐MSC CM and exosomes |
↓ Apoptosis ↓ ROS ↓ Mitochondrial O2 . − ↑ GSH, ↑ GPx activity
|
144 | ||
Hypoxia‐exposed neonatal porcine islet cell clusters (porcine) |
Human UC‐MSC CM and exosomes |
↓ Apoptosis ↑ Oxygen consumption rate
|
113 | ||
Normoxia‐ and hypoxia‐exposed WJ‐MSC engineered islet‐like cells (human) |
Human WJ‐MSCs |
↑ Proliferation ↓apoptosis and ↓ROS in both conditions ↓ NO and O2 .− in hypoxia |
145 | ||
Primary islet cells (mouse) |
Mouse BM‐MSCs Transwell coculture |
↑ GSTM1 | 91 | ||
Keratinocytes | High glucose and LPS‐treated primary keratinocytes (rat) | Rat BM‐MSC‐CM |
↑ Viability ↑ Wound assay closure ↓ ROS
|
146 | |
Lung epithelial cells | H2O2‐treated human alveolar basal epithelial adenocarcinoma cells (A549) | Human BM‐MSCs |
↑ Cell viability ↑ Transcription and protein expression of STC‐1 in H2O2‐treated MSCs ↓ Cell viability with Anti‐STC‐1 ↑ Cell viability with recombinant STC‐1
↓ Cell viability with STC‐1 siRNA MSCs ↑ ROS with STC‐1 siRNA MSCs ↑ mRNA expression of uncoupling protein 2 in A549 ↓ mRNA expression of uncoupling protein 2 with anti‐STC‐1 |
100 | |
Osteocytes | Mitochondrial DNA (mtDNA)‐depleted 143B osteosarcoma cells (human) | Human WJ‐MSCs |
MSCs and mitochondria‐depleted cells removed via auxotrophic restriction
|
124 | |
Renal cells | Cisplatin‐treated renal proximal tubular cells (rat) | Human UC‐MSC exosomes |
↓ Δψm ↑ PCNA ↓ Oxidized DNA ↓ Lipid peroxidation ↑ GSH ↓ Bax, ↑ Bcl‐2 |
||
H2O2‐treated renal tubular epithelial cells (rat) in vitro | Rat BM‐MSCs |
↓ Apoptosis ↓ Cell loss ↑ Mitosis ↓ Bax expression ↑ p‐ERK1/2 |
93 | ||
High glucose‐treated glomerular mesangial cells (rat) | Rat BM‐MSC‐CM |
↓ ROS ↓ GLUT1
|
29 | ||
Hypoxia reoxygenation of rat kidney epithelial cells (NRK‐52E) |
Human WJ‐MSC extracellular vesicles |
↓ ROS ↑ Activated NRF2 ↑ ARE activity ↑ HO‐1 |
52 | ||
Oxalate and calcium oxalate monohydrate‐treated human proximal tubular epithelial (HK‐2) |
Human UC‐MSC exosomes |
↓ Apoptosis ↓ Lipid peroxidation ↓ H2O2 ↓ ROS ↓ LDH ↓ Mesenchymal markers ↓ Migration |
147 | ||
Skeletal muscle cells |
Dexamethasone‐induced muscle atrophy in L6 rat skeletal muscle cells |
Human UC‐MSC‐CM |
↑ Muscle related gene expression (myogenin, desmin) ↑ SOD activity ↓ ROS generation ↑ CAT, SOD1, GPx‐1 in L6 cells |
76 | |
Dexamethasone‐induced muscle atrophy in L6 rat skeletal muscle cells |
Human UC‐MSC (isolated mitochondria) Centrifugal delivery of exogenous mitochondria |
↑ Cell proliferation ↑ Δψm ↑ ATP content ↓ Mitochondrial O2 .− |
125 | ||
Trophoblasts | Hypoxia (1% O2) trophoblast cells (mouse) |
Mouse BM‐MSCs Transwell |
↑ Mitofusin‐2 ↑ β‐HCG and progesterone ↑ ATP levels ↓ Caspase 3 and 9 ↓ Bax, ↑Bcl‐2 ↓ Apoptosis |
148 |
Abbreviations: ARE, antioxidant response element; AT‐MSC, adipose tissue‐derived MSC; BAX, Bcl‐2‐associated X protein; BDNF, brain‐derived neurotrophic factor; BM‐MSCs, bone marrow‐derived MSC; c‐caspase 3, cleaved‐caspase 3; CM, conditioned medium; CNTF, ciliary neurotrophic factor; COX2, cyclooxygenase‐2; ERK, extracellular signal‐regulated kinases; GLUT1, glucose transporter 1; GPx, glutathione peroxidase; GST, glutathione S‐transferase; H2O2, hydrogen peroxide; HGF, hepatocyte growth factor; HO‐1, heme oxygenase‐1; I/R, ischemia/reperfusion; ICAM, intercellular adhesion molecule; IFNγ, interferon gamma; IL, interleukin; LDH, lactate dehydrogenase; LPS, lipopolysaccharide; M2, type‐2 macrophages; MPO, myeloperoxidase; NAC, N‐acetyl cysteine; NAD(P)H, nicotinamide adenine dinucleotide phosphate hydrogen; NO, Nitric oxide; NQO1, NAD(P)H quinone dehydrogenase 1; NRF2, nuclear factor erythroid 2‐related factor 2; NRLP3, nod‐like receptor protein‐3; O2 .−, superoxide; pERK1/2, phosphorylated extracellular signal‐regulated kinases 1/2; PRDX1‐6, peroxiredoxin; RGCs, retinal ganglion cell; ROS, reactive oxygen species; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription 3; STC‐1, stanniocalcin‐1; TNFα, tumor necrosis factor alpha; TNT, tunneling nanotube; TXN, thioredoxin; UC‐MSC, umbilical cord‐derived MSC; β‐HCG, β‐human chorionic gonadotropin; Δψm, mitochondrial membrane potential.
Oxidative stress refers to a deviation from the physiological redox state and an increase in pro‐oxidants, or free radicals, that structurally change lipids, proteins, and DNA in a way that causes pathology or damage to a cell or tissue. 6 The most widely studied free radicals are reactive oxygen species (ROS), which can also include reactive molecules that have a stable charge. The three major endogenous ROS include the superoxide anion (O2 .−), hydroxyl radical (•OH), and hydrogen peroxide (H2O2). 7 , 8 O2 .− is predominantly generated by nicotinamide adenine dinucleotide phosphate reduced (NADPH)‐oxidase (NOX) family enzymes or, by the mitochondria, as a by‐product of oxidative phosphorylation. 9 The level of mitochondria‐derived O2 .− depends on metabolic substrates, cytosolic Ca2+ levels, pH, and oxygen tension. 10 O2 .− generated from complexes of the electron transport chain (ETC) are highly reactive and can damage the mitochondrion. 11 The detoxification of O2 .− into H2O2 is mediated by superoxide dismutase (SOD). 9 However, H2O2 can also be generated in various metabolic processes and by dual oxidases (DUOX). 12 While H2O2 is more stable than O2 .−, its detoxification is crucial as it possesses a weak peroxide bond that makes it susceptible to reacting with metals, such as Fe2+, to generate reactive •OH through the Fenton reaction. 13 Both, H2O2 and O2 .−, are diffusible across cell membranes and can promote cell death and inflammatory signaling. 14 , 15 Several studies have demonstrated that MSCs can reduce ROS and biomarkers of oxidative stress. In this review, evidence of direct and indirect antioxidant mechanisms of MSC therapies is explored.
2. MSCs ARE RESISTANT AND RESPOND TO OXIDATIVE STRESS
The therapeutic properties of MSCs have been explored in many models of disease associated with high levels of ROS and biomarkers of oxidative injury. MSCs must survive these volatile environments to exert their therapeutic effects, which can present as a challenge for their engraftment after administration. Nonetheless, several studies have demonstrated that MSCs are highly resistant to oxidative insult. The oxidative effects of ionizing radiation are limited on MSCs which have been attributed to their ability to directly scavenge free radicals. 16 It has been demonstrated that MSCs are resistant to oxidative and nitrosative stimuli in vitro which is associated with constitutively expressed antioxidant enzymes SOD1, SOD2, catalase (CAT), and glutathione peroxidase (GPx), in addition to high levels of the antioxidant glutathione (GSH). 17 Depletion of GSH results in a loss of tolerance to oxidative stress. MSCs also constitutively express heat‐shock protein 70 (HSP70) and sirtuin (SIRT)3, 18 which may also play a role in the resistance of MSCs to oxidative/nitrosative injury. SIRT1 is also required for MSC survival against H2O2 and its overexpression has a protective effect. 19 Likewise, SIRT6 has been suggested to confer resistance to oxidative insult and basal ROS production in MSCs via downstream production of antioxidants including heme oxygenase‐1 (HO‐1). 20 Overexpression of HO‐1 ameliorates elevations in ROS and cellular senescence in SIRT6‐null MSCs and, therefore, appears to be a critical component of the survival mechanism of MSCs in oxidative environment. 20
In addition to wielding constitutive antioxidants, MSCs are also capable of significant adaptions in response to redox stress. MSCs exposed to lipopolysaccharide (LPS) produce oxidative and nitrosative free radicals. 18 In parallel, several adaptive processes are observed including the upregulation and/or nuclear translocation of redox‐sensitive factors (nuclear factor kappa‐B [NFκB], thioredoxin [TRX1], apurinic/apyrimidinic endonuclease redox effector factor‐1 [APE1/Ref‐1], nuclear factor erythroid 2‐related factor 2 [NRF2], forkhead box O3 [FOXO3]. and HO‐1), as well as mitochondrial remodeling and autophagy. Similarly, MSCs exposed to hypoxic conditions (1.5%‐2% O2) exhibit increased intracellular ROS and cells respond by upregulating the expression of hypoxia‐inducible factor 1 alpha (HIF‐1α), erythropoietin receptor, CAT, HO‐1, and antiapoptotic Bcl‐2 family of proteins. 21 Exposure of MSCs to hypoxia during in vitro culture can enhance their anti‐inflammatory, antioxidative, and cytoprotective properties. 21 , 22 , 23 This suggests that the ability of MSCs to tolerate and respond to oxidative environment may be critical to their engraftment and therapeutic efficacy at sites of tissue injury.
3. EFFECT OF MSCs ON OXIDATIVE STRESS BIOMARKERS AND ROS
In disease models, oxidative stress is typically quantified via biomarkers of oxidation to DNA and proteins or lipid peroxidation. Administration of MSCs has been demonstrated to reduce levels of one or more of these markers in a variety of animal models associated with oxidative stress (Table 1). Injection of MSCs themselves may not be critical to their antioxidant effects as administration of their conditioned medium (CM) also reduced lipid peroxidation in a model of ureteral obstruction‐induced kidney injury. 24 Administration of MSC‐derived exosomes was also effective to rescue protein oxidation and lipid peroxidation in animal models of septic and hyperglycemic brain injury and cognitive impairment. 25 , 26 Likewise, DNA oxidation and lipid peroxidation caused by cisplatin‐induced kidney damage are alleviated by exosomes from human umbilical cord‐derived MSCs (UC‐MSCs); these results were confirmed in vitro with renal proximal tubular cells. 27 Treatments with MSCs were also demonstrated to reduce levels of ROS in animal models of diabetic retinopathy and nephropathy, severe acute pancreatitis, ureteral obstruction‐induced kidney damage, Alzheimer's disease, and metabolic renovascular disease. 24 , 28 , 29 , 30 , 31 , 32 Specifically, MSC treatments have been shown to reduce levels of H2O2 in intestinal inflammation and several organs in a model of premature aging. 33 , 34 MSCs also reduced levels of O2 .− in colitis and spontaneous stroke. 34 , 35
Typically, the reduction of oxidative stress markers by MSC treatments is associated with functional recovery and positive outcomes in animal models. The exception to this is the diversity of responses at various stages of hepatocarcinoma whereby antioxidant effects of MSCs reduce tumor burden at the early stages of disease by protecting the integrity of DNA but increase tumor progression at the late stages of the disease possibly by reducing ROS‐associated cell death. 36 The timing of treatments may also affect MSCs ability to attenuate oxidative stress as pretreatment with MSCs is more effective to prevent oxidative stress in septic lung injury and acute liver failure. 37 , 38
Several studies have also investigated the therapeutic properties of MSCs on nitrosative stress which is particularly of interest in neural diseases. MSCs reduced the volatile peroxynitrite (ONOO−) in a model of intracerebral hemorrhage and nitrite levels in diabetic sensory neuropathy. 39 , 40 In a model of radiation‐induced neurological complications, intranasal delivery of MSCs reduced inducible nitric oxide synthase (iNOS) expression and oxidative stress biomarkers, which are associated with improved cognitive performance and neuronal survival. 41 In a model of spinal cord injury, adipose tissue‐derived MSCs (AT‐MSCs) also demonstrated antioxidant activity with a reduction in lipid peroxidation and protein oxidation; however, no significant effects were observed for nitrosylation. 42 Furthermore, MSCs have been associated with increased nitric oxide (NO) in a model of Alzheimer's disease which may have been driven by preventing the loss of neuronal nitric oxide synthase (nNOS). 43 Alternatively, several studies observed a reduction in inflammation‐induced iNOS and nNOS expression after MSC treatment. 37 , 44 , 45 , 46 , 47 Overall, while the majority of studies support antioxidative effects of MSCs, their antinitrosative effects are unclear and likely to be disease and tissue specific. Furthermore, NO can be produced by nonhuman MSCs which is thought to be critical to their immunomodulatory function which may also explain these inconsistent effects on nitrosative stress. 42 , 48
Although studies in cells and animal models unequivocally demonstrate that MSC treatments reduce levels of oxidative stress, albeit limited data exist from human studies. Nonetheless, favorable outcomes in a case study utilizing MSCs to treat the lungs of a subject previously exposed to sulfur mustard gas were attributed to the antioxidant properties of MSCs as evidenced by reduced lipid peroxidation levels in the sputum. 49
The antioxidant effects of MSC treatments are likely to be a specific property of these cells as they are more efficacious than hematopoietic stem cells and fibroblast at reducing oxidative stress in carbon tetrachloride‐induced‐liver injury and sepsis, respectively. 50 , 51 Likewise, fibroblast exosomes have no effect on kidney injury‐induced by ischemia and chemotherapy. 27 , 52
The alleviation of oxidative stress in animal models is associated with decreased pro‐inflammatory cytokines and markers of cellular death highlighting the close association between these processes. The precise mechanisms of in vivo MSC treatments are difficult to determine as cell death, inflammation, and oxidative stress occur concomitantly and perpetuate each other. However, a growing body of evidence suggests that MSCs have a direct role on suppressing oxidative stress and ROS production which may mediate their antiapoptotic and anti‐inflammatory effects.
4. ANTIOXIDATIVE MECHANISMS OF MSCs
The potential for MSCs to attenuate oxidative injury is unequivocally demonstrated by the reduction in ROS and biomarkers of oxidative stress in many disease models. Evidence form in vitro models suggests that MSCs directly protect cells from oxidative stimuli (Table 2). This is often associated with a reduction in ROS suggesting that MSCs avert the negative effects of oxidative stress by reducing the oxidative stimuli. The antioxidative effects of MSCs often occur in a paracrine manner in vitro and the administration of MSC‐conditioned medium (CM) can also reduce oxidative stress in vivo suggesting a paracrine component to their mechanism. 24 Nevertheless, others report that the antioxidant effects of MSCs can be cell contact dependent 53 ; albeit, these mechanisms could be disease and tissue dependent. Currently, MSCs have been proposed to reduce oxidative injury via scavenging free radicals, enhancing host antioxidant defenses, modulating the inflammatory response, augmenting cellular respiration and mitochondrial functions, or donating their mitochondria to protect damaged cells (Figure 1). 37 , 50 , 51 , 53 , 54
4.1. Antioxidant defense and scavenging
To maintain redox homeostasis and prevent excessive production of free radicals, cells rely on a complement of enzymatic antioxidants, including SODs, CAT, GPx, and small nonenzymatic antioxidants, such as GSH. After MSC treatments, the total antioxidant capacity of tissues are enhanced as observed in models of chemotherapy‐induced neuropathy, hyperglycemia‐induced cardiac damage, systemic sclerosis, hepatocarcinoma, and acute liver injury. 36 , 55 , 56 , 57 , 58 MSCs are receptive to oxidative stimuli and exhibit all necessary machinery to efficiently process ROS. 18 , 59 Furthermore, media conditioned by MSCs have potent antioxidant capacity indicating that MSCs actively secrete antioxidants. 60 MSC‐CM has more effective antioxidant properties than CM from lung fibroblasts. 61 In several models of disease, MSC treatments upregulate the expression of antioxidant defense enzymes in vivo (Table 1). Therefore, the antioxidant effects of MSCs may be explained by their ability to directly scavenge free radicals and by enhancing antioxidant defenses in host tissues through upregulation of antioxidant enzymes.
Volatile O2 − is produced during cellular respiration by the mitochondria and NOX enzymes during tissue inflammation; O2 − is eliminated by SOD which catalyzes its conversion to H2O2. Antioxidant effects of MSC treatments have been associated with enhanced SOD activity or the expression of SODs in models of aging, age‐related erectile dysfunction, chemotherapy‐induced pulmonary fibrosis or gonadotoxicity colitis, pancreatitis, septic lung injury, arthritis, hepatotoxicity and hepatic ischemia reperfusion injury, Alzheimer's disease, and ovarian autografts. 33 , 34 , 37 , 38 , 43 , 50 , 62 , 63 , 64 , 65 , 66 , 67 , 68 , 69 , 70 MSCs secrete all isoforms of SOD including SOD1 and SOD2, which are archetypically not released extracellularly. 33 , 59 , 71 , 72 Thus, it is difficult to interpret whether enhanced SOD activity and/or SOD expression is due to MSCs or host‐tissue‐derived SOD. In in vitro studies, MSC‐CM or MSCs in transwell cocultures promote SOD activity in tert‐Butyl hydroperoxide or UV‐exposed fibroblasts, H2O2‐treated neural stem cells or retinal ganglion cells, and dexamethasone‐induced muscle atrophy model. 60 , 73 , 74 , 75 , 76 These studies suggest that MSCs enhance SOD activity in cells exposed to oxidative stimuli. Likewise, MSC‐CM increases SOD1 expression in islet cells exposed to pro‐inflammatory cytokines and SOD2 in tert‐Butyl hydroperoxide‐treated umbilical endothelial cells. 77 , 78 In endothelial cells, increased SOD2 expression was regulated by signal transducer and activator of transcription (STAT3) signaling and knockdown of either SOD2 or STAT3 decreased the antiapoptotic effects of the MSC‐CM. 77 These findings suggest that MSC upregulation of SOD in host tissues may be critical to their antioxidant effects. Alternatively, MSCs stimulated with TNF‐α and IFN‐γ were found to secrete high levels of SOD3, which was a major contributor to the antioxidant properties of MSCs in the amelioration of NO‐induced neuronal death in vitro. 71 Likewise, SOD3 expression in MSCs was necessary to suppress neutrophil respiratory burst and the accumulation in immune complex‐mediated dermal vasculitis. 79 Similarly, silencing of SOD2 in MSCs inhibits the antioxidant properties and therapeutic efficacy of their exosomes in hepatic I/R injury in vivo and H2O2‐treated human fetal hepatocytes which can be recovered by the addition of a SOD2 mimetic. 80
CAT and GPx are responsible for detoxifying H2O2 by its conversion to oxygen and water. MSCs secrete CAT and upregulation of CAT expression is associated with the therapeutic properties of MSCS in models of Friedreich's ataxia, radiation‐induced aortic injury, septic lung injury, and hepatoxicity. 33 , 37 , 50 , 68 , 81 , 82 Likewise, upregulation of CAT associates with the antioxidant properties of MSCs in aging and colitis which parallels measurable reductions in H2O2. 33 , 34 Exosomes derived from MSCs also express functional CAT. 31 , 83 Inhibition of CAT suppresses their protective effects on amyloid‐β oligomer‐induced damage to hippocampal neurons indicating that this enzyme may also mediate antioxidant effects of MSCs. 83 MSC treatments have also been demonstrated to upregulate the expression of GPx in septic lung injury, severe acute pancreatitis, small bowel ischemia/reperfusion (I/R) injury, and Friedreich's ataxia. 37 , 67 , 81 , 84 MSC‐CM can also enhance GPx activity in fibroblasts under oxidative stimuli. 60 Therefore, both CAT and GPx may play a role in MSC treatments in reducing oxidative stress by the elimination of H2O2 which was commonly used as a cell death inducing oxidative stimulus in in vitro studies.
The glutathione system is critical to scavenging ROS in animals, plants and fungi. GSH exerts its antioxidant effects by reducing free radicals and peroxides. During this process, glutathione disulfide (GSSG) is generated which is converted back to GSH by NADPH and is catalyzed by glutathione reductase. Increased levels of GSSG to GSH are suggestive of an oxidative redox state. Conversely, higher GSH‐to‐GSSG ratio is observed after MSC treatment in models of type 2 diabetes, in the hippocampus after chronic ethanol intake and hypoxia‐induced hypertension. 85 , 86 , 87 Likewise, MSCs improve GSH‐to‐GSSG ratio in sepsis which is not achieved by fibroblast. 51 Similarly, MSCs were found to increase GSH levels in diabetic models, septic lung injury, arthritis, hepatotoxicity, and epilepsy. 37 , 38 , 66 , 68 , 88 , 89 , 90 These effects were also observed by MSC‐CM in a model of unilateral ureteral obstruction which may suggest GSH was host‐tissue‐derived. 24 The effects of MSCs on GSH levels may be mediated by their ability to upregulate glutathione reductase which was observed in septic lung injury, acute pancreatitis, and I/R injury of the small bowel. 37 , 67 , 84 Notably, MSCs have also been shown to upregulate the expression of glutathione S‐transferases (GSTs) in cells and tissues which detoxify many damaging molecules that arise from redox imbalance such as peroxidized lipids. 88 , 91 Glutathione reductase and GSTs are secreted by MSCs via exosomes. 31 While GSH has been the best studied scavenger in MSC treatments, they have also been demonstrated to upregulate the expression of the enzyme NAD(P)H quinone oxidoreductase 1 (NQO1) which detoxify quinones that contribute to the generation of ROS in septic lung injury, small bowel I/R injury in vivo, and cytokine‐exposed islet cell in vitro. 78 , 84 , 92
The multifunctional antioxidant HO‐1 has also been implicated in the therapeutic effects of MSC treatments. HO‐1's antioxidant and therapeutic mechanisms have been attributed to its ability to degrade heme, which is pro‐oxidative and the scavenging abilities of its products, biliverdin and bilirubin. HO‐1 expression is inducible in response to oxidative stress via nuclear factor erythroid 2‐related factor 2 (NRF2) which appears to be important in the adaptive responses of MSCs to inflammation and ROS. 18 Downregulation of NRF2 and HO‐1 has been observed after MSC treatment of chemotherapy‐induced pulmonary fibrosis and diabetic sensory neuropathy which may be explained by a reduction in oxidative stimuli. 40 , 63 Conversely, the antioxidant effects of MSCs in several other models associate with upregulation of HO‐1 such as radiation‐induced aortic injury, septic lung injury, pancreatitis, and renal injuries caused by altered oxygen tensions and cisplatin. 30 , 82 , 92 , 93 , 94 HO‐1 was determined to partially contribute to the effects of MSCs in pancreatitis as its inhibition with zinc protoporphyrin negated some of the effects of MSC treatments, including upregulation of CAT and increased SOD activity, which may indicate that these processes are downstream of HO‐1 activity. 30 After MSC treatment of small bowel I/R injury, a larger number of HO‐1 expressing cells were observed which did not appear to completely colocalize with engrafted MSCs suggesting that treatments may increase HO‐1 expression in cells of the host. 84 It has been demonstrated that overexpression of HO‐1 in MSCs enhances their therapeutic activity in septic lung injury which was attributed to its pro‐survival properties. 95 Others have reported that MSCs still respond efficiently to oxidative insult with silenced HO‐1 by upregulating GSH pathway enzymes. 96 Nevertheless, the CM of MSCs derived from HO‐1−/− mice are unable to attenuate cisplatin‐induced renal injury and therefore HO‐1 appears to have an important role in the antioxidant properties of the MSC secretome. 94
Together, these studies demonstrate that antioxidants secreted by MSCs and their ability to upregulate host antioxidant defenses contribute to the suppression of oxidative stress. The exosomes derived from MSCs appear to be particularly rich in machinery to process ROS and can include, but not limited to, GPx, GSTs, SOD1‐3, peroxiredoxin 1‐6, CAT, cytoglobin, prostaglandin‐endoperoxide synthase 1, peroxidasin, albumin, apolipoprotein E, glutathione‐disulfide reductase, and thioredoxin reductase 1‐2. 31 , 80 Notwithstanding, recombinant application of factors secreted by MSCs, such as hepatocyte growth factor (HGF) and basic fibroblast growth factors (bFGF), has been demonstrated to upregulate Gpx1, CAT, and SOD activity via SIRT1 and FOXO1 during age‐related loss of ovarian function. 97 Mechanisms of antioxidant defense mediated by scavenging of ROS by MSCs and the host could occur independently or simultaneously and are likely to be disease‐specific.
4.2. Antioxidant effects on inflammation
Immune function is regulated by free radicals and the redox system; leukocytes and pro‐inflammatory mediators enhance the formation of free radicals and perturb the redox environment creating a positive feedback cycle. 98 The immunomodulatory action of MSCs is a well‐documented phenomenon; however, their role in the interactions between the immune system and oxidative stress is not fully understood. Oxidative and/or nitrosative free radicals unequivocally play a role in all grades of acute and chronic inflammation. At physiological levels, they act as cellular signals modifying function and initiating necessary cell death programs. However, excessive generation of free radicals and/or inadequate scavenging results in protein oxidation, lipid peroxidation, and DNA damage that can be detrimental both intrinsically to the cell and the surrounding microenvironment. The ROS and reactive nitrogen species involved can take many forms and be generated from a variety of sources. Large amounts of the highly reactive O2 − anion are generated from NOX expressed by innate leukocytes. 99 SOD catalyzes the conversion of O2 − to H2O2 which phagocytes and neutrophils use to generate hypochlorous acid (HOCl) via myeloperoxidase (MPO). 99 Collectively, this is referred to as respiratory burst, a crucial element of the bactericidal response and inflammatory signaling. Nonetheless, MPO activity and O2 − are also associated with various inflammatory diseases. In inflammatory bouts, there is often a parallel increase in the expression of iNOS in leukocytes and, thus, subsequent generation of the free radical NO. Nonimmune cells such as epithelial cells are also capable of expressing iNOS and NOX to generate NO and O2 −.
MSC treatments reduce inflammation and oxidative stress in colitis, pancreatitis, arthritis, sepsis, vasculitis, stroke, myocardial infarction, hyperoxic lung injury, and I/R injury of the kidneys and bowel (Table 1). These effects have included a reduction in inflammatory cytokines TNFα, IFNγ, interleukin (IL)‐1β, IL‐6, IL‐9, and IL‐4; decreased expression of ROS producing enzymes NOX, MPO, and iNOS; as well as a net reduction in the infiltration of immune cells such as neutrophils (Table 1). The anti‐inflammatory effects of MSCs in pancreatitis were partially dependent on their expression of the antioxidant pathway enzyme HO‐1. 30 Previously, it was demonstrated in a model of sepsis that MSC treatments can reduce pro‐inflammatory cytokines in the serum and normalize thiol/disulfide redox pairings responsible for free radical scavenging. 51 Decreased levels of IL‐1β and TNFα superseded restoration of redox homeostasis. This suggests that the aversion of oxidative injury was secondary to the immunomodulation of pro‐inflammatory signaling, at least in acute septic inflammation. Conversely, MSCs can directly reduce oxidative injury in many cell types in vitro; thus, it is likely that MSCs may also reduce oxidative stress in tissues by mechanism other than suppressing the immune system. 27 , 54 , 60 , 75 , 77 , 100 This is highlighted in in vivo and organotypic ex vivo models of myocardial I/R injury where MSC‐derived exosomes ameliorated infarction injury without altering leukocyte recruitment. 101 In in vivo experiments, protein oxidation was reduced by MSC‐derived exosomes after 1 hour; neutrophils were yet to infiltrate into the tissue. After 24 hours, MSCs reduced peripheral blood leukocyte numbers and neutrophil infiltration into the myocardium; thus, the antioxidative activity of MSCs preceded signals recruiting leukocytes. 101 This suggests that MSCs can attenuate oxidative stress‐induced tissue injury first, which can limit the recruitment of immune cells and subsequent inflammation in this model. This may be mediated by their ability to suppress NOX1 and 2 on resident cells which are downregulated by MSC treatments in acute myocardial infarction, sepsis‐induced brain injury, acute ischemic stroke, I/R injury to kidneys, and small bowel which were all associated with reduced inflammation. 25 , 84 , 102 , 103 , 104
Neutrophils appear to be key mediators of oxidative stress in inflammation. These cells harbor an abundance of MPO, a major catalyst for hypochlorite and NO‐derived oxidants. 105 , 106 MSCs attenuate the infiltration of neutrophils and reduce MPO levels in several disease models. 50 , 107 , 108 MSCs can also directly dampen the respiratory burst in neutrophils and suppress MPO activity required to produce free radical required for their pro‐inflammatory function which was dependent on SOD3 and occurs in a paracrine manner. 79 , 80 , 109 Likewise, MSCs can also directly decrease ROS and MPO in stimulated monocytes and macrophages which suppress their pro‐inflammatory phenotype. 110 , 111 These data suggest that MSCs not only suppress the immune system to prevent oxidative injury, but also that their mechanism of immunosuppression is reliant on their antioxidant properties.
4.3. Cellular bioenergetics
Free radicals are produced by several metabolic processes and the mitochondria during cellular respiration. Dysfunction in mitochondria can cause cellular injury which is mediated through the generation of O2 .− and proteins that initiate cellular apoptosis. Depolarization of the mitochondrial membrane potential (Δψm) is a hallmark of mitochondrial dysfunction leading to cell death. Hyperglycemia can also cause oxidative stress via several mechanisms including the formation of free radical as by‐products of glucose auto‐oxidation that deplete antioxidant defense and advanced glycation end products that induce cellular stress. In models of hyperglycemia, MSC treatments can reduce the expression of glucose and fatty acid transports in kidney and cardiac tissue cells, which prevents glucose transport and ROS generation. 29 , 56 Therefore, the antioxidant effects of MSC treatments in models of diabetes may be downstream of glycemic control. Conversely, regulation of mitochondrial function and oxidative phosphorylation by MSCs has been implicated in several disease models. MSC treatments improve chemotherapy‐induced cognitive impairment, which associates with enhanced respiratory capacity of the mitochondria. 112 The effects of MSCs on the mitochondria appear to occur in a paracrine manner as MSC‐CM increases the oxygen consumption rate of hypoxia‐exposed neonatal porcine islet cells and LPS‐treated macrophages. 113 , 114 Similarly, MSC‐derived extracellular vesicles suppress mitochondrial O2 .− levels in H2O2‐treated human fetal hepatocytes, which is associated with a reduction in apoptosis. 80
The potential for MSCs to directly attenuate mitochondrial dysfunction has been demonstrated in an in vitro model of I/R injury in mouse ventricular myocytes. 54 Within 5 minutes of reperfusion, cells exhibited an exaggerated Δψm hyperpolarization, which was reduced by conditioning the reperfusion solution with MSCs. The exaggerated hyperpolarization was followed by a continuous depolarization in controls after 15 minutes which was also attenuated by the paracrine secretion of MSCs. Decay of the Δψm was likely a result of the mitochondrial permeability transition pore opening. The exaggerated hyperpolarization of the Δψm was also averted by a mitochondrial ROS scavenger which simultaneously decreased mitochondrial O2 − generation demonstrating the close relationship between these events. Similarly, MSC secretion decreased mitochondrial O2 −, which led to the suggestion that MSCs may also attenuate Δψm dysfunction via scavenging of O2 −.
Depolarization of the Δψm in cisplatin‐treated renal proximal tubular cells has also been reportedly attenuated using exosomes derived from UC‐MSCs. 27 In vitro, BM‐MSCs were demonstrated to upregulate uncoupling protein 2 (UCP2) transcription in H2O2‐treated alveolar basal epithelial adenocarcinoma cells which reduces the formation of mitochondria‐derived O2 − by lowering the proton‐motive force across the mitochondrial membrane and provides another potential mechanism for the alleviation of mitochondrial dysfunction. 100 This was regulated by the paracrine secretion of stanniocalcin‐1 by MSCs, which enhanced UCP2, correlating with cell survival and decreased ROS generation. MSCs secreted stanniocalcin‐1 may also attenuate inflammation as it decreases mitochondrial ROS and subsequent activation of the nucleotide‐binding oligomerization domain (NOD)‐like receptor protein 3 (NLRP3) inflammasome. 111 BM‐MSCs inhibited the activity of the NLRP3 inflammasome in primed macrophages which is responsible for recognizing damage‐associated molecular patterns and initiating the inflammatory cascade through activation and secretion of IL‐1β. 115 , 116 MSCs have also been demonstrated to secrete the redox‐sensitive protein DJ‐1, which has established roles in maintaining mitochondrial biogenesis and respiratory chain efficiency and could potentially mediate the neuroprotective effects of the MSC secretome as shown in Parkinson's disease models. 117 Collectively, these studies demonstrate that MSCs can ameliorate mitochondrial dysfunction in a paracrine manner with diverse therapeutic outcomes.
4.4. Mitochondrial donation
Recently, a concept has emerged that MSCs may be able to alter oxidative phosphorylation and ROS generation in cells through donation of mitochondria themselves. Islam et al 118 observed mitochondrial transfer from human BM‐MSCs to alveolar epithelium in a mouse model of LPS‐induced lung injury. BM‐MSC administration attenuated decreased intracellular ATP in the alveoli caused by lung injury; notably, ATP (visualized by a molecular probe) was predominantly restored at the site of mitochondrial transfer and immediately surrounding alveoli. MSCs with a mutation in connexin 43, a protein involved in the formation of gap junctions, were unable to transfer mitochondria despite being functionally competent and subsequently did not restore ATP, surfactant secretion, or reduce leukocyte infiltration. This phenomenon only occurred in LPS‐exposed lungs indicating that mitochondrial transfer is dependent on stimulants from damaged tissues.
MSCs cultured in hyperoxic (21% O2: normoxic atmosphere) conditions produce high levels of mitochondrial O2 .−, depolarize Δψm, and induce mitophagy. 119 Mitochondria are loaded into phagosomes and shuttled to the plasma membrane. 119 These effects were reduced by culturing MSCs closer to a normoxic oxygen concentration (5% O2: hypoxic atmosphere). Macrophages have been observed to phagocytose these vesicles containing the partially depolarized mitochondria, which can fuse with endogenous mitochondria in macrophages. This protects silica‐exposed macrophages by increasing their oxygen consumption rate and decreasing mitochondrial O2 .− production. These effects could not be elicited when MSCs were substituted by human fibroblasts. 119 This suggests that mitochondrial transfer may be stimulated by oxidative stress in MSCs. Conversely, MSCs have also been demonstrated to engulf mitochondria from other somatic cells exposed to H2O2. MSCs degraded the engulfed mitochondria which stimulated HO‐1 expression, mitochondrial biogenesis in MSCs, and the transfer of functional MSCs to damaged cells. 53 Inhibition of mitophagy negated the cytoprotective effects of MSCs in other somatic cells, which suggests that MSC sensing of damaged mitochondria may mediate their therapeutic responses. 53 Supporting this, the cell contact‐dependent transfer of mitochondria from chemotherapy‐treated T lymphocytes to MSCs was also determined to be critical to their ability to decrease mitochondrial O2 .− production and cell death in T lymphocytes. 120 Nonetheless, in this study, mitochondrial transfer appeared to be predominately unidirectional and very few MSC‐derived mitochondria were observed in T cells. This suggests that MSC sensing of mitochondria can promote therapeutic mechanisms other than mitochondrial donation. 120
Although extracellular vesicles can contain whole mitochondria, several studies suggest that the donation of mitochondria may be contact‐dependent. MSCs have been found to transfer mitochondria via tunneling nanotubes (TNT) to glucose‐deprived and hypoxia‐reoxygenated cardiomyocytes which prevented Δψm depolarization and cell apoptosis. Albeit inhibition of TNT formation only partially reversed the effects of MSCs indicating other cytoprotective mechanisms were still active. 121 Similarly, MSCs transfer mitochondria to chemotherapy‐treated endothelial cells, which appears to occur in a unidirectional manner, unlike in T lymphocytes. 120 , 122 Miro1 is important to TNT formation and its overexpression in MSCs can enhance mitochondrial transfer. 123 The effects of MSC mitochondrial donation are sufficient to rescue cellular respiration, proliferation, and motility in mitochondria‐depleted osteosarcoma cells. 124 These effects can be maintained for 45 passages, which highlights the therapeutic potential of MSC‐derived mitochondria. 124 Exogenous application of mitochondria isolated from MSCs may also offer therapeutic benefit and are able to protect dexamethasone‐treated muscle cells form oxidative stress in vitro. 125 Albeit, delivery of MSC‐derived mitochondria in in vivo poses a challenge. While several studies have demonstrated contact‐dependent transfer of mitochondria between MSCs and other cells, MSCs have also been reported to donate mitochondria to LPS‐treated macrophages via secreted extracellular vesicles. 114 Exposure of macrophages to MSC‐derived exosomes promoted their indication to the type 2 phenotype, which exerted anti‐inflammatory effects after adoptive transfer in septic lung injury. 114 The effects of MSCs were dependent on enhancing mitochondrial function in macrophages and were inhibited by damaging mitochondria in MSCs and blocking extracellular vesicles. 114 The therapeutic use of MSCs to deliver functional mitochondria to damaged tissue is an intriguing concept and warrants further study; however, another recent advancement reported by Panfoli et al 126 suggests that the exosomes of MSCs are capable of oxidative phosphorylation independent of the mitochondria. Subsets of MSC‐derived exosomes isolated from the umbilical cord of term newborns were discovered to contain complexes of the ETC embedded in the membrane. These exosomes possessed an electrochemical membrane potential, consumed O2, and produced ATP. The therapeutic application of these exosomes is yet to be investigated; nonetheless, this may present a viable tool to restore dysfunctional oxidative phosphorylation and ATP synthesis in damaged cells.
5. CONCLUSION
The presented studies evidently demonstrate that MSCs exhibit antioxidant potential either directly via scavenging of ROS and donating mitochondria or indirectly by upregulation antioxidant defenses in other cells and altering cellular bioenergetics. These effects can occur in combination with the previously recognized trophic and vesicular components of the MSC secretome acting directly on regenerative pathways. Likewise, antioxidant and trophic pathways appear to mediate the cytoprotective effect of MSC treatments which are ROS dependent. MSCs have frequently been utilized in inflammatory diseases to modulate the immune response. In this context, immunosuppression can avert ROS generation which is generated by MPO and NOX enzymes as a part of the inflammatory response. However, MSCs have now been shown to exert immunosuppressive effects by dampening ROS production and enhancing mitochondrial function in macrophages and neutrophils. Therefore, the antioxidant properties confer a role in the trophic and anti‐inflammatory mechanisms of MSC therapy. Considering that oxidative stress is implicated in almost every disease, these antioxidant properties, along with regenerative capacity of MSC seretome, may explain why MSC treatments are useful for such a spectrum of seemingly unlinked pathologies (Figure 1). Future studies should seek to clarify disease‐specific nuances of the antioxidative mechanisms of MSCs and MSC‐derived products. Likewise, improving the antioxidant effects of MSCs by enhancing the expression of antioxidant enzymes or promoting mitochondrial donation may be useful to optimize MSC‐based therapies and improve outcomes.
CONFLICT OF INTEREST
The authors declared no potential conflicts of interest.
AUTHOR CONTRIBUTIONS
R.S.: conception and design, manuscript writing, final approval of manuscript. K.N.: conception and design, manuscript writing, financial support, final approval of manuscript.
Stavely R, Nurgali K. The emerging antioxidant paradigm of mesenchymal stem cell therapy. STEM CELLS Transl Med. 2020;9:985–1006. 10.1002/sctm.19-0446
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
REFERENCES
- 1. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 2016;25(5):829‐848. [DOI] [PubMed] [Google Scholar]
- 2. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med. 2001;226(6):507‐520. [DOI] [PubMed] [Google Scholar]
- 3. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076‐1084. [DOI] [PubMed] [Google Scholar]
- 4. van Poll D, Parekkadan B, Rinkes IB, et al. Mesenchymal stem cell therapy for protection and repair of injured vital organs. Cell Mol Bioeng. 2008;1(1):42‐50. [Google Scholar]
- 5. Gao F, Chiu SM, Motan DAL, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7(1):e2062‐e2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Sies H. Oxidative stress: a concept in redox biology and medicine. Redox Biol. 2015;4:180‐183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ J. 2012;5(1):9‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol. 2014;24(10):R453‐R462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Dan Dunn J, Alvarez LAJ, Zhang X, Soldati T. Reactive oxygen species and mitochondria: a nexus of cellular homeostasis. Redox Biol. 2015;6:472‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Aon MA, Cortassa S, O'Rourke B. Redox‐optimized ROS balance: a unifying hypothesis. Biochim Biophys Acta. 2010;1797(6–7):865‐877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Chen Y, Azad MB, Gibson SB. Superoxide is the major reactive oxygen species regulating autophagy. Cell Death Differ. 2009;16(7):1040‐1052. [DOI] [PubMed] [Google Scholar]
- 12. De Deken X, Corvilain B, Dumont JE, et al. Roles of DUOX‐mediated hydrogen peroxide in metabolism, host defense, and signaling. Antioxid Redox Signal. 2014;20(17):2776‐2793. [DOI] [PubMed] [Google Scholar]
- 13. Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82:969‐974. [DOI] [PubMed] [Google Scholar]
- 14. Fisher AB. Redox signaling across cell membranes. Antioxid Redox Signal. 2009;11(6):1349‐1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Bienert GP, Schjoerring JK, Jahn TP. Membrane transport of hydrogen peroxide. Biochim Biophys Acta (BBA)—Biomembr. 2006;1758(8):994‐1003. [DOI] [PubMed] [Google Scholar]
- 16. Chen M‐F, Lin C‐T, Chen W‐C, et al. The sensitivity of human mesenchymal stem cells to ionizing radiation. Intern J Radiat Oncol Biol Phys. 2006;66(1):244‐253. [DOI] [PubMed] [Google Scholar]
- 17. Valle‐Prieto A, Conget PA. Human mesenchymal stem cells efficiently manage oxidative stress. Stem Cells Dev. 2010;19(12):1885‐1893. [DOI] [PubMed] [Google Scholar]
- 18. Gorbunov NV, Garrison BR, McDaniel DP, et al. Adaptive redox response of mesenchymal stromal cells to stimulation with lipopolysaccharide inflammagen: mechanisms of remodeling of tissue barriers in sepsis. Oxid Med Cell Longev. 2013;2013:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Liu T, Ma X, Ouyang T, et al. SIRT1 reverses senescence via enhancing autophagy and attenuates oxidative stress‐induced apoptosis through promoting p53 degradation. Int J Biol Macromol. 2018;117:225‐234. [DOI] [PubMed] [Google Scholar]
- 20. Pan H, Guan D, Liu X, et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res. 2016;26(2):190‐205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Lan YW, Choo KB, Chen CM, et al. Hypoxia‐preconditioned mesenchymal stem cells attenuate bleomycin‐induced pulmonary fibrosis. Stem Cell Res Ther. 2015;6:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Qin HH, Filippi C, Sun S, Lehec S, Dhawan A, Hughes RD. Hypoxic preconditioning potentiates the trophic effects of mesenchymal stem cells on co‐cultured human primary hepatocytes. Stem Cell Res Ther. 2015;6(1):237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Kim Y, Jin HJ, Heo J, et al. Small hypoxia‐primed mesenchymal stem cells attenuate graft‐versus‐host disease. Leukemia. 2018;32(12):2672‐2684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Liu B, Ding F‐X, Liu Y, et al. Human umbilical cord‐derived mesenchymal stem cells conditioned medium attenuate interstitial fibrosis and stimulate the repair of tubular epithelial cells in an irreversible model of unilateral ureteral obstruction. Nephrol Ther. 2018;23(8):728‐736. [DOI] [PubMed] [Google Scholar]
- 25. Chang C‐L, Chen H‐H, Chen K‐H, et al. Adipose‐derived mesenchymal stem cell‐derived exosomes markedly protected the brain against sepsis syndrome induced injury in rat. Am J Transl Res. 2019;11(7):3955‐3971. [PMC free article] [PubMed] [Google Scholar]
- 26. Nakano M, Nagaishi K, Konari N, et al. Bone marrow‐derived mesenchymal stem cells improve diabetes‐induced cognitive impairment by exosome transfer into damaged neurons and astrocytes. Sci Rep. 2016;6:24805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Zhou Y, Xu H, Xu W, et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin‐induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther. 2013;4(2):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Ezquer M, Urzua CA, Montecino S, Leal K, Conget P, Ezquer F. Intravitreal administration of multipotent mesenchymal stromal cells triggers a cytoprotective microenvironment in the retina of diabetic mice. Stem Cell Res Ther. 2016;7:42‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Lv S, Cheng J, Sun A, et al. Mesenchymal stem cells transplantation ameliorates glomerular injury in streptozotocin‐induced diabetic nephropathy in rats via inhibiting oxidative stress. Diabetes Res Clin Pract. 2014;104(1):143‐154. [DOI] [PubMed] [Google Scholar]
- 30. Ma Z, Song G, Zhao D, et al. Bone marrow‐derived mesenchymal stromal cells ameliorate severe acute pancreatitis in rats via hemeoxygenase‐1‐mediated anti‐oxidant and anti‐inflammatory effects. Cytotherapy. 2019;21(2):162‐174. [DOI] [PubMed] [Google Scholar]
- 31. Eirin A, Zhu X‐Y, Jonnada S, Lerman A, van Wijnen AJ, Lerman LO. Mesenchymal stem cell‐derived extracellular vesicles improve the renal microvasculature in metabolic renovascular disease in swine. Cell Transplant. 2018;27(7):1080‐1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Yan Y, Ma T, Gong K, Ao Q, Zhang X, Gong Y. Adipose‐derived mesenchymal stem cell transplantation promotes adult neurogenesis in the brains of Alzheimer's disease mice. Neural Regen Res. 2014;9(8):798‐805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Xie C, Jin J, Lv X, Tao J, Wang R, Miao D. Anti‐aging effect of transplanted amniotic membrane mesenchymal stem cells in a premature aging model of Bmi‐1 deficiency. Sci Rep. 2015;5(1):13975. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 34. Jung K‐J, Lee KW, Park CH, et al. Mesenchymal stem cells decrease oxidative stress in the bowels of Interleukin‐10 knockout mice. Gut Liver. 2020;14(1):100‐107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Calió ML, Marinho DS, Ko GM, et al. Transplantation of bone marrow mesenchymal stem cells decreases oxidative stress, apoptosis, and hippocampal damage in brain of a spontaneous stroke model. Free Radic Biol Med. 2014;70:141‐154. [DOI] [PubMed] [Google Scholar]
- 36. Zong C, Zhang H, Yang X, et al. The distinct roles of mesenchymal stem cells in the initial and progressive stage of hepatocarcinoma. Cell Death Dis. 2018;9(3):345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Shalaby SM, Amal S, Abd‐Allah SH, et al. Mesenchymal stromal cell injection protects against oxidative stress in Escherichia coli‐induced acute lung injury in mice. Cytotherapy. 2014;16(6):764‐775. [DOI] [PubMed] [Google Scholar]
- 38. Liu Z, Meng F, Li C, et al. Human umbilical cord mesenchymal stromal cells rescue mice from acetaminophen‐induced acute liver failure. Cytotherapy. 2014;16(9):1207‐1219. [DOI] [PubMed] [Google Scholar]
- 39. Chen M, Li X, Zhang X, et al. The inhibitory effect of mesenchymal stem cell on blood‐brain barrier disruption following intracerebral hemorrhage in rats: contribution of TSG‐6. J Neuroinflammation. 2015;12:61‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Evangelista AF, Vannier‐Santos MA, de Assis Silva GS, et al. Bone marrow‐derived mesenchymal stem/stromal cells reverse the sensorial diabetic neuropathy via modulation of spinal neuroinflammatory cascades. J Neuroinflammation. 2018;15(1):189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Soria B, Martin‐Montalvo A, Aguilera Y, et al. Human mesenchymal stem cells prevent neurological complications of radiotherapy. Front Cell Neurosci. 2019;13:204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kim Y, Jo S‐H, Kim WH, et al. Antioxidant and anti‐inflammatory effects of intravenously injected adipose derived mesenchymal stem cells in dogs with acute spinal cord injury. Stem Cell Res Ther. 2015;6:229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Cui Y, Ma S, Zhang C, et al. Human umbilical cord mesenchymal stem cells transplantation improves cognitive function in Alzheimer's disease mice by decreasing oxidative stress and promoting hippocampal neurogenesis. Behav Brain Res. 2017;320:291‐301. [DOI] [PubMed] [Google Scholar]
- 44. Stavely R, Robinson AM, Miller S, et al. Allogeneic Guinea pig mesenchymal stem cells ameliorate neurological changes in experimental colitis. Stem Cell Res Ther. 2015;6(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Stavely R, Robinson AM, Miller S, et al. Human adult stem cells derived from adipose tissue and bone marrow attenuate enteric neuropathy in the Guinea‐pig model of acute colitis. Stem Cell Res Ther. 2015;6(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Robinson AM, Miller S, Payne N, Boyd R, Sakkal S, Nurgali K. Neuroprotective potential of mesenchymal stem cell‐based therapy in acute stages of TNBS‐induced colitis in Guinea‐pigs. PloS One. 2015;10(9):e0139023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Song WJ, Li Q, Ryu MO, et al. TSG‐6 released from intraperitoneally injected canine adipose tissue‐derived mesenchymal stem cells ameliorate inflammatory bowel disease by inducing M2 macrophage switch in mice. Stem Cell Res Ther. 2018;9(1):91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Ren G, Su J, Zhang L, et al. Species variation in the mechanisms of mesenchymal stem cell‐mediated immunosuppression. Stem Cells. 2009;27(8):1954‐1962. [DOI] [PubMed] [Google Scholar]
- 49. Nejad‐Moghaddam A, Ajdari S, Tahmasbpour E, Goodarzi H, Panahi Y, Ghanei M. Adipose‐derived mesenchymal stem cells for treatment of airway injuries in a patient after long‐term exposure to sulfur mustard. Cell J. 2017;19(1):117‐126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Pulavendran S, Vignesh J, Rose C. Differential anti‐inflammatory and anti‐fibrotic activity of transplanted mesenchymal vs. hematopoietic stem cells in carbon tetrachloride‐induced liver injury in mice. Int Immunopharmacol. 2010;10(4):513‐519. [DOI] [PubMed] [Google Scholar]
- 51. Iyer SS, Torres‐Gonzalez E, Neujahr DC, et al. Effect of bone marrow‐derived mesenchymal stem cells on endotoxin‐induced oxidation of plasma cysteine and glutathione in mice. Stem Cells Intern. 2010;2010:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zhang G, Zou X, Huang Y, et al. Mesenchymal stromal cell‐derived extracellular vesicles protect against acute kidney injury through anti‐oxidation by enhancing Nrf2/ARE activation in rats. Kidney Blood Pressure Res. 2016;41(2):119‐128. [DOI] [PubMed] [Google Scholar]
- 53. Mahrouf‐Yorgov M, Augeul L, Da Silva CC, et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017;24(7):1224‐1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. DeSantiago J, Bare DJ, Banach K. Ischemia/reperfusion injury protection by mesenchymal stem cell derived antioxidant capacity. Stem Cells Dev. 2013;22(18):2497‐2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Al‐Massri KF, Ahmed LA, El‐Abhar HS. Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel‐induced neuropathy in rats: role of Notch1 receptor and JAK/STAT signaling pathway. Behav Brain Res. 2019;360:303‐311. [DOI] [PubMed] [Google Scholar]
- 56. Hamza AA, Fikry EM, Abdallah W, Amin A. Mechanistic insights into the augmented effect of bone marrow mesenchymal stem cells and thiazolidinediones in streptozotocin‐nicotinamide induced diabetic rats. Sci Rep. 2018;8(1):9827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Maria AT, Toupet K, Bony C, et al. Antifibrotic, antioxidant, and immunomodulatory effects of mesenchymal stem cells in HOCl‐induced systemic sclerosis. Arthritis Rheumatol. 2016;68(4):1013‐1025. [DOI] [PubMed] [Google Scholar]
- 58. Quintanilha LF, Takami T, Hirose Y, et al. Canine mesenchymal stem cells show antioxidant properties against thioacetamide‐induced liver injury in vitro and in vivo. Hepatol Res. 2014;44(10):E206‐E217. [DOI] [PubMed] [Google Scholar]
- 59. Klein D, Steens J, Wiesemann A, et al. Mesenchymal stem cell therapy protects lungs from radiation‐induced endothelial cell loss by restoring superoxide dismutase 1 expression. Antioxid Redox Signal. 2017;26(11):563‐582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Kim W‐S, Park B‐S, Kim H‐K, et al. Evidence supporting antioxidant action of adipose‐derived stem cells: protection of human dermal fibroblasts from oxidative stress. J Dermatol Sci. 2008;49(2):133‐142. [DOI] [PubMed] [Google Scholar]
- 61. Waszak P, Alphonse R, Vadivel A, Ionescu L, Eaton F, Thébaud B. Preconditioning enhances the paracrine effect of mesenchymal stem cells in preventing oxygen‐induced neonatal lung injury in rats. Stem Cells Dev. 2012;21(15):2789‐2797. [DOI] [PubMed] [Google Scholar]
- 62. Yang J, Zhang Y, Zang G, et al. Adipose‐derived stem cells improve erectile function partially through the secretion of IGF‐1, bFGF, and VEGF in aged rats. Andrology. 2018;6(3):498‐509. [DOI] [PubMed] [Google Scholar]
- 63. Ni S, Wang D, Qiu X, Pang L, Song Z, Guo K. Bone marrow mesenchymal stem cells protect against bleomycin‐induced pulmonary fibrosis in rat by activating Nrf2 signaling. Int J Clin Exp Pathol. 2015;8(7):7752‐7761. [PMC free article] [PubMed] [Google Scholar]
- 64. Sherif IO, Sabry D, Abdel‐Aziz A, Sarhan OM. The role of mesenchymal stem cells in chemotherapy‐induced gonadotoxicity. Stem Cell Res Ther. 2018;9(1):196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Sun T, Gao GZ, Li RF, et al. Bone marrow‐derived mesenchymal stem cell transplantation ameliorates oxidative stress and restores intestinal mucosal permeability in chemically induced colitis in mice. Am J Transl Res. 2015;7(5):891‐901. [PMC free article] [PubMed] [Google Scholar]
- 66. Abd‐Elhalem SS, Haggag NZ, El‐Shinnawy NA. Bone marrow mesenchymal stem cells suppress IL‐9 in adjuvant‐induced arthritis. Autoimmunity. 2018;51(1):25‐34. [DOI] [PubMed] [Google Scholar]
- 67. Jung KH, Yi T, Son MK, Song SU, Hong SS. Therapeutic effect of human clonal bone marrow‐derived mesenchymal stem cells in severe acute pancreatitis [journal article]. Arch Pharm Res. 2015;38(5):742‐751. [DOI] [PubMed] [Google Scholar]
- 68. Qiao H, Zhou Y, Qin X, Cheng J, He Y, Jiang Y. NADPH oxidase signaling pathway mediates mesenchymal stem cell‐induced inhibition of hepatic stellate cell activation. Stem Cells Intern. 2018;2018:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Ge Y, Zhang Q, Jiao Z, Li H, Bai G, Wang H. Adipose‐derived stem cells reduce liver oxidative stress and autophagy induced by ischemia‐reperfusion and hepatectomy injury in swine. Life Sci. 2018;214:62‐69. [DOI] [PubMed] [Google Scholar]
- 70. Shojafar E, Mehranjani MS, Shariatzadeh SMA. Adipose‐derived mesenchymal stromal cell transplantation at the graft site improves the structure and function of autografted mice ovaries: a stereological and biochemical analysis. Cytotherapy. 2018;20(11):1324‐1336. [DOI] [PubMed] [Google Scholar]
- 71. Kemp K, Gray E, Mallam E, Scolding N, Wilkins A. Inflammatory cytokine induced regulation of superoxide dismutase 3 expression by human mesenchymal stem cells. Stem Cell Rev Rep. 2010;6(4):548‐559. [DOI] [PubMed] [Google Scholar]
- 72. Mitchell R, Mellows B, Sheard J, et al. Secretome of adipose‐derived mesenchymal stem cells promotes skeletal muscle regeneration through synergistic action of extracellular vesicle cargo and soluble proteins. Stem Cell Res Ther. 2019;10(1):116‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Liu Q, Luo Z, He S, et al. Conditioned serum‐free medium from umbilical cord mesenchymal stem cells has anti‐photoaging properties. Biotechnol Lett. 2013;35(10):1707‐1714. [DOI] [PubMed] [Google Scholar]
- 74. Niu Y, Xia X, Song P, et al. Bone mesenchymal stem cell‐conditioned medium attenuates the effect of oxidative stress injury on NSCs by inhibiting the Notch1 signaling pathway. Cell Biol Int. 2019;43(11):1267‐1275. [DOI] [PubMed] [Google Scholar]
- 75. Cui Y, Xu N, Xu W, Xu G. Mesenchymal stem cells attenuate hydrogen peroxide‐induced oxidative stress and enhance neuroprotective effects in retinal ganglion cells. In Vitro Cell Develop Biol—Anim. 2017;53(4):328‐335. [DOI] [PubMed] [Google Scholar]
- 76. Park C‐M, Kim MJ, Kim S‐M, Park JH, Kim ZH, Choi YS. Umbilical cord mesenchymal stem cell‐conditioned media prevent muscle atrophy by suppressing muscle atrophy‐related proteins and ROS generation. In Vitro Cell Develop Biol—Anim. 2016;52(1):68‐76. [DOI] [PubMed] [Google Scholar]
- 77. Liu S‐H, Huang J‐P, Lee RK‐K, et al. Paracrine factors from human placental multipotent mesenchymal stromal cells protect endothelium from oxidative injury via STAT3 and manganese superoxide dismutase activation. Biol Reprod. 2010;82(5):905‐913. [DOI] [PubMed] [Google Scholar]
- 78. Laporte C, Tubbs E, Cristante J, et al. Human mesenchymal stem cells improve rat islet functionality under cytokine stress with combined upregulation of heme oxygenase‐1 and ferritin. Stem Cell Res Ther. 2019;10(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Jiang D, Muschhammer J, Qi Y, et al. Suppression of neutrophil‐mediated tissue damage—a novel skill of mesenchymal stem cells. Stem Cells. 2016;34(9):2393‐2406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Yao J, Zheng J, Cai J, et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia‐reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J. 2018;33(2):1695‐1710. [DOI] [PubMed] [Google Scholar]
- 81. Jones J, Estirado A, Redondo C, et al. Mesenchymal stem cells improve motor functions and decrease neurodegeneration in ataxic mice. Mol Ther. 2015;23(1):130‐138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Shen Y, Jiang X, Meng L, et al. Transplantation of bone marrow mesenchymal stem cells prevents radiation‐induced artery injury by suppressing oxidative stress and inflammation. Oxid Med Cell Longev. 2018;2018:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. de Godoy MA, Saraiva LM, de Carvalho LR, et al. Mesenchymal stem cells and cell‐derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid‐β oligomers. J Biol Chem. 2018;293(6):1957‐1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Chang C‐L, Sung P‐H, Sun C‐K, et al. Protective effect of melatonin‐supported adipose‐derived mesenchymal stem cells against small bowel ischemia‐reperfusion injury in rat. J Pineal Res. 2015;59(2):206‐220. [DOI] [PubMed] [Google Scholar]
- 85. Feng J, Lu C, Dai Q, Sheng J, Xu M. SIRT3 facilitates amniotic fluid stem cells to repair diabetic nephropathy through protecting mitochondrial homeostasis by modulation of mitophagy. Cell Physiol Biochem. 2018;46(4):1508‐1524. [DOI] [PubMed] [Google Scholar]
- 86. Ezquer F, Quintanilla ME, Morales P, et al. Activated mesenchymal stem cell administration inhibits chronic alcohol drinking and suppresses relapse‐like drinking in high‐alcohol drinker rats. Addict Biol. 2019;24(1):17‐27. [DOI] [PubMed] [Google Scholar]
- 87. Hogan SE, Salazar MPR, Cheadle J, et al. Mesenchymal stromal cell‐derived exosomes improve mitochondrial health in pulmonary arterial hypertension. Am J Physiol—Lung Cell Mol Physiol. 2019;316(5):L723‐L737. [DOI] [PubMed] [Google Scholar]
- 88. El‐Tantawy WH, Al Haleem ENA. Therapeutic effects of stem cell on hyperglycemia, hyperlipidemia, and oxidative stress in alloxan‐treated rats. Mol Cell Biochem. 2014;391(1–2):193‐200. [DOI] [PubMed] [Google Scholar]
- 89. Ayatollahi M, Hesami Z, Jamshidzadeh A, Gramizadeh B. Antioxidant effects of bone marrow mesenchymal stem cell against carbon tetrachloride‐induced oxidative damage in rat livers. Intern J Organ Transplant Med. 2014;5(4):166‐173. [PMC free article] [PubMed] [Google Scholar]
- 90. Salem NA, El‐Shamarka M, Khadrawy Y, et al. New prospects of mesenchymal stem cells for ameliorating temporal lobe epilepsy. Inflammopharmacology. 2018;26(4):963‐972. [DOI] [PubMed] [Google Scholar]
- 91. Yoshimatsu G, Sakata N, Tsuchiya H, et al. The co‐transplantation of bone marrow derived mesenchymal stem cells reduced inflammation in intramuscular islet transplantation. PloS One. 2015;10(2):e0117561‐e0117561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sung P‐H, Chang C‐L, Tsai T‐H, et al. Apoptotic adipose‐derived mesenchymal stem cell therapy protects against lung and kidney injury in sepsis syndrome caused by cecal ligation puncture in rats. Stem Cell Res Ther. 2013;4(6):155‐155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Liu H, McTaggart SJ, Johnson DW, et al. Original article anti‐oxidant pathways are stimulated by mesenchymal stromal cells in renal repair after ischemic injury. Cytotherapy. 2012;14(2):162‐172. [DOI] [PubMed] [Google Scholar]
- 94. Zarjou A, Kim J, Traylor AM, et al. Paracrine effects of mesenchymal stem cells in cisplatin‐induced renal injury require heme oxygenase‐1. Am J Physiol—Renal Physiol. 2011;300(1):F254‐F262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Chen X, Wu S, Tang L, et al. Mesenchymal stem cells overexpressing heme oxygenase‐1 ameliorate lipopolysaccharide‐induced acute lung injury in rats. J Cell Physiol. 2019;234(5):7301‐7319. [DOI] [PubMed] [Google Scholar]
- 96. Nowak WN, Taha H, Kachamakova‐Trojanowska N, et al. Murine bone marrow mesenchymal stromal cells respond efficiently to oxidative stress despite the low level of heme oxygenases 1 and 2. Antioxid Redox Signal. 2018;29(2):111‐127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Ding C, Zou Q, Wang F, et al. HGF and BFGF secretion by human adipose‐derived stem cells improves ovarian function during natural aging via activation of the SIRT1/FOXO1 signaling pathway. Cell Physiol Biochem. 2018;45(4):1316‐1332. [DOI] [PubMed] [Google Scholar]
- 98. Kim YW, West XZ, Byzova TV. Inflammation and oxidative stress in angiogenesis and vascular disease. J Mol Med (Berlin, Germany). 2013;91(3):323‐328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Robinson JM. Reactive oxygen species in phagocytic leukocytes. Histochem Cell Biol. 2008;130(2):281‐297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Ohkouchi S, Block GJ, Katsha AM, et al. Mesenchymal stromal cells protect cancer cells from ROS‐induced apoptosis and enhance the Warburg effect by secreting STC1. Mol Ther. 2012;20(2):417‐423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Arslan F, Lai RC, Smeets MB, et al. Mesenchymal stem cell‐derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013;10(3):301‐312. [DOI] [PubMed] [Google Scholar]
- 102. Chen K‐H, Chen C‐H, Wallace CG, et al. Intravenous administration of xenogenic adipose‐derived mesenchymal stem cells (ADMSC) and ADMSC‐derived exosomes markedly reduced brain infarct volume and preserved neurological function in rat after acute ischemic stroke. Oncotarget. 2016;7(46):74537‐74556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Sheu J‐J, Lee F‐Y, Yuen C‐M, et al. Combined therapy with shock wave and autologous bone marrow‐derived mesenchymal stem cells alleviates left ventricular dysfunction and remodeling through inhibiting inflammatory stimuli, oxidative stress & enhancing angiogenesis in a swine myocardial infarction model. Int J Cardiol. 2015;193:69‐83. [DOI] [PubMed] [Google Scholar]
- 104. Lin K‐C, Yip H‐K, Shao P‐L, et al. Combination of adipose‐derived mesenchymal stem cells (ADMSC) and ADMSC‐derived exosomes for protecting kidney from acute ischemia–reperfusion injury. Int J Cardiol. 2016;216:173‐185. [DOI] [PubMed] [Google Scholar]
- 105. Zhang R, Brennan M‐L, Shen Z, et al. Myeloperoxidase functions as a major enzymatic catalyst for initiation of lipid peroxidation at sites of inflammation. J Biol Chem. 2002;277(48):46116‐46122. [DOI] [PubMed] [Google Scholar]
- 106. Pattison DI, Davies MJ. Reactions of myeloperoxidase‐derived oxidants with biological substrates: gaining chemical insight into human inflammatory diseases. Curr Med Chem. 2006;13(27):3271‐3290. [DOI] [PubMed] [Google Scholar]
- 107. El‐Attar S, Elsayed L, Rashed L. Role of stem cells and antioxidant on modulation of body defense mechanism in lipopolysaccharide‐induced acute lung injury in rats. Med J Cairo Univ. 2012;80(2):559‐573. [Google Scholar]
- 108. Gonzalez‐Rey E, Anderson P, González MA, et al. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut. 2009;58(7):929‐939. [DOI] [PubMed] [Google Scholar]
- 109. Raffaghello L, Bianchi G, Bertolotto M, et al. Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells. 2008;26(1):151‐162. [DOI] [PubMed] [Google Scholar]
- 110. Guillen MI, Platas J, Perez Del Caz MD, et al. Paracrine anti‐inflammatory effects of adipose tissue‐derived mesenchymal stem cells in human monocytes. Front Physiol. 2018;9:661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Oh JY, Ko JH, Lee HJ, et al. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells. 2014;32(6):1553‐1563. [DOI] [PubMed] [Google Scholar]
- 112. Chiu GS, Boukelmoune N, Chiang ACA, et al. Nasal administration of mesenchymal stem cells restores cisplatin‐induced cognitive impairment and brain damage in mice. Oncotarget. 2018;9(85):35581‐35597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Nie W, Ma X, Yang C, et al. Human mesenchymal‐stem‐cells‐derived exosomes are important in enhancing porcine islet resistance to hypoxia. Xenotransplantation. 2018;25(5):e12405. [DOI] [PubMed] [Google Scholar]
- 114. Morrison TJ, Jackson MV, Cunningham EK, et al. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am J Respir Crit Care Med. 2017;196(10):1275‐1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Liu L, Dong Y, Ye M, et al. The pathogenic role of NLRP3 inflammasome activation in inflammatory bowel diseases of both mice and humans. J Crohn's Colitis. 2017;11(6):737‐750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Lazaridis L‐D, Pistiki A, Giamarellos‐Bourboulis EJ, et al. Activation of NLRP3 inflammasome in inflammatory bowel disease: differences between Crohn's disease and ulcerative colitis. Dig Dis Sci. 2017;62(9):2348‐2356. [DOI] [PubMed] [Google Scholar]
- 117. Vilaça‐Faria H, Salgado AJ, Teixeira FG. Mesenchymal stem cells‐derived exosomes: a new possible therapeutic strategy for Parkinson's disease? Cell. 2019;8(2):118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Islam MN, Das SR, Emin MT, et al. Mitochondrial transfer from bone‐marrow‐derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759‐765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Phinney DG, Di Giuseppe M, Njah J, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs [article]. Nat Commun. 2015;6:8472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Wang J, Liu X, Qiu Y, et al. Cell adhesion‐mediated mitochondria transfer contributes to mesenchymal stem cell‐induced chemoresistance on T cell acute lymphoblastic leukemia cells. J Hematol Oncol. 2018;11(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Han H, Hu J, Yan Q, et al. Bone marrow‐derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol Med Rep. 2016;13(2):1517‐1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Feng Y, Zhu R, Shen J, et al. Human bone marrow mesenchymal stem cells rescue endothelial cells experiencing chemotherapy stress by mitochondrial transfer via tunneling nanotubes. Stem Cells Dev. 2019;28(10):674‐682. [DOI] [PubMed] [Google Scholar]
- 123. Babenko V, Silachev D, Popkov V, et al. Miro1 enhances mitochondria transfer from multipotent mesenchymal stem cells (MMSC) to neural cells and improves the efficacy of cell recovery. Molecules. 2018;23(3):687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Lin H‐Y, Liou C‐W, Chen S‐D, et al. Mitochondrial transfer from Wharton's jelly‐derived mesenchymal stem cells to mitochondria‐defective cells recaptures impaired mitochondrial function. Mitochondrion. 2015;22:31‐44. [DOI] [PubMed] [Google Scholar]
- 125. Kim MJ, Hwang JW, Yun C‐K, Lee Y, Choi YS. Delivery of exogenous mitochondria via centrifugation enhances cellular metabolic function. Sci Rep. 2018;8(1):3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Panfoli I, Ravera S, Podesta M, et al. Exosomes from human mesenchymal stem cells conduct aerobic metabolism in term and preterm newborn infants. FASEB J. 2016;30(4):1416‐1424. [DOI] [PubMed] [Google Scholar]
- 127. Ono M, Ohkouchi S, Kanehira M, et al. Mesenchymal stem cells correct inappropriate epithelial–mesenchyme relation in pulmonary fibrosis using Stanniocalcin‐1. Mol Ther. 2015;23(3):549‐560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Dubey NK, Wei H‐J, Yu S‐H, et al. Adipose‐derived stem cells attenuates diabetic osteoarthritis via inhibition of glycation‐mediated inflammatory Cascade. Aging Dis. 2019;10(3):483‐496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Liang CC, Shaw SWS, Lin YH, Lee TH. Amniotic fluid stem cells ameliorate bladder dysfunction induced by chronic bladder ischemia in rat. Neurourol Urodyn. 2018;37(1):123‐131. [DOI] [PubMed] [Google Scholar]
- 130. Rong X, Liu J, Yao X, Jiang T, Wang Y, Xie F. Human bone marrow mesenchymal stem cells‐derived exosomes alleviate liver fibrosis through the Wnt/β‐catenin pathway. Stem Cell Res Ther. 2019;10(1):98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Ghorbani A, Feizpour A, Hashemzahi M, et al. The effect of adipose derived stromal cells on oxidative stress level, lung emphysema and white blood cells of Guinea pigs model of chronic obstructive pulmonary disease. J Facult Pharm Tehran Univ Med Sci. 2014;22(1):26‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Malaquias M, Oyama L, Jericó P, et al. Effects of mesenchymal stromal cells play a role the oxidant/antioxidant balance in a murine model of asthma. Allergol Immunopathol. 2018;46(2):136‐143. [DOI] [PubMed] [Google Scholar]
- 133. Chang YS, Choi SJ, Ahn SY, et al. Timing of umbilical cord blood derived mesenchymal stem cells transplantation determines therapeutic efficacy in the neonatal hyperoxic lung injury. PloS One. 2013;8(1):e52419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Zhang G, Zou X, Miao S, et al. The anti‐oxidative role of micro‐vesicles derived from human Wharton‐jelly mesenchymal stromal cells through NOX2/gp91(phox) suppression in alleviating renal ischemia‐reperfusion injury in rats. PloS One. 2014;9(3):e92129‐e92129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Hsiao C‐H, Ji AT‐Q, Chang C‐C, Cheng CJ, Lee LM, Ho JHC. Local injection of mesenchymal stem cells protects testicular torsion‐induced germ cell injury. Stem Cell Res Ther. 2015;6(1):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Liu K, Ji K, Guo L, et al. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia–reperfusion model via tunneling nanotube like structure‐mediated mitochondrial transfer. Microvasc Res. 2014;92:10‐18. [DOI] [PubMed] [Google Scholar]
- 137. Song M, Jue S‐S, Cho Y‐A, Kim EC. Comparison of the effects of human dental pulp stem cells and human bone marrow‐derived mesenchymal stem cells on ischemic human astrocytes in vitro. J Neurosci Res. 2015;93(6):973‐983. [DOI] [PubMed] [Google Scholar]
- 138. Torrente D, Avila M, Cabezas R, et al. Paracrine factors of human mesenchymal stem cells increase wound closure and reduce reactive oxygen species production in a traumatic brain injury in vitro model. Hum Exp Toxicol. 2014;33(7):673‐684. [DOI] [PubMed] [Google Scholar]
- 139. Bonafede R, Scambi I, Peroni D, et al. Exosome derived from murine adipose‐derived stromal cells: neuroprotective effect on in vitro model of amyotrophic lateral sclerosis. Exp Cell Res. 2016;340(1):150‐158. [DOI] [PubMed] [Google Scholar]
- 140. Palomares T, Cordero M, Bruzos‐Cidon C, Torrecilla M, Ugedo L, Alonso‐Varona A. The neuroprotective effect of conditioned medium from human adipose‐derived mesenchymal stem cells is impaired by N‐acetyl cysteine supplementation [journal article]. Mol Neurobiol. 2018;55(1):13‐25. [DOI] [PubMed] [Google Scholar]
- 141. Cheng Y, Jiang Y, Zhang L, et al. Mesenchymal stromal cells attenuate sevoflurane‐induced apoptosis in human neuroglioma H4 cells. BMC Anesthesiol. 2018;18(1):84‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Damania A, Jaiman D, Teotia AK, Kumar A. Mesenchymal stromal cell‐derived exosome‐rich fractionated secretome confers a hepatoprotective effect in liver injury. Stem Cell Res Ther. 2018;9(1):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Haga H, Yan IK, Borrelli DA, et al. Extracellular vesicles from bone marrow‐derived mesenchymal stem cells protect against murine hepatic ischemia/reperfusion injury. Liver Transpl. 2017;23(6):791‐803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Tan Y, Nie W, Chen C, et al. Mesenchymal stem cells alleviate hypoxia‐induced oxidative stress and enhance the pro‐survival pathways in porcine islets. Exp Biol Med. 2019;244(9):781‐788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Chandravanshi B, Bhonde RR. Shielding engineered islets with mesenchymal stem cells enhance survival under hypoxia. J Cell Biochem. 2017;118(9):2672‐2683. [DOI] [PubMed] [Google Scholar]
- 146. Li M, Zhao Y, Hao H, et al. Mesenchymal stem cell‐conditioned medium improves the proliferation and migration of keratinocytes in a diabetes‐like microenvironment. Int J Low Extrem Wounds. 2015;14(1):73‐86. [DOI] [PubMed] [Google Scholar]
- 147. Li D, Zhang D, Tang B, et al. Exosomes from human umbilical cord mesenchymal stem cells reduce damage from oxidative stress and the epithelial‐mesenchymal transition in renal epithelial cells exposed to oxalate and calcium oxalate monohydrate. Stem Cells Intern. 2019;2019:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Wang L, Xu X, Kang L, Xiang W. Bone marrow mesenchymal stem cells attenuate mitochondria damage induced by hypoxia in mouse trophoblasts. PloS One. 2016;11(4):e0153729. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.