Role of mesenchymal stem cells in sepsis and their therapeutic potential in sepsis‑associated myopathy (Review)
- Authors:
- Published online on: August 23, 2024 https://doi.org/10.3892/ijmm.2024.5416
- Article Number: 92
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Sepsis is a life-threatening condition characterized by the dysregulated response of the body to infection, leading to multiple organ dysfunction syndrome. It is a major factor contributing to death in patients admitted to the intensive care unit (ICU) (1-3). Sepsis-induced myopathy (SIM), also known as ICU-acquired weakness, is a common complication associated with sepsis. SIM is characterized by symmetrical atrophy, and weakness of the respiratory system and limb skeletal muscles, leading to prolonged mechanical ventilation, challenges in weaning patients off ventilators and limb dysfunction (4-6). This condition substantially affects the clinical course and recovery of patients, exacerbating their overall morbidity and prolonging their stay in the ICU (7,8).
The incidence of SIM is as high as 40% among critically ill patients in the ICU (5-7). SIM is associated with an increased risk of mortality and long-term functional impairment (9-12). Muscle wasting in sepsis occurs early and rapidly within the first 10 days of ICU admission (13). A meta-analysis of 10 cohort studies including 2,396 patients with sepsis found that patients with sepsis with sarcopenia had a significantly higher risk of early mortality than patients with sepsis without sarcopenia (8). Furthermore, critically ill patients who survive often experience reduced quality of life after discharge, primarily owing to the decline in physical function caused by skeletal muscle atrophy (14-16). Despite marked advancements in medicine and technology, developing effective treatments for SIM remains challenging.
Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from the mesoderm, and they are predominantly found in mesenchymal and connective tissues. MSCs can differentiate into bone, cartilage, muscle, fat and other tissues, and possess anti-inflammatory, immunoregulatory and paracrine properties. MSCs have shown efficacy in the treatment of cardiovascular diseases (17,18), respiratory diseases (19,20), motor system disorders (21) and sepsis (22). Furthermore, several studies have indicated that MSCs can enhance the function of organs affected by sepsis, including the heart, lungs, liver and kidneys (23-26). These findings suggest that MSCs may possess considerable potential in treating SIM. The present review aims to summarize the sources and biological characteristics of MSCs and their therapeutic potential in SIM.
Definition and properties of MSCs
Definition of MSCs
MSCs are a class of pluripotent stem cells originating from the mesoderm. They were first described by Friedenstein et al (27) in 1968 as adherent, fibroblast-like, non-hematopoietic precursor cells. In 2006, the International Society for Cellular Therapy established three primary criteria for defining MSCs: i) Adherence to plastic in standard culture conditions; ii) expression of the surface markers CD105, CD90 and CD73, along with the absence of HLA-DR, CD34, CD45, CD19 and CD11b; and iii) the ability to differentiate into osteoblasts, chondrocytes and adipocytes under appropriate culture conditions in vitro (28).
Origin and properties of MSCs
MSCs were initially isolated from the bone marrow, which remains their primary source; therefore, they are often referred to as bone marrow-derived mesenchymal stromal cells (BMSCs). In addition, MSCs are found in the adipose tissue, muscle, tendon, umbilical cord, placenta, spleen, peripheral blood and dental pulp (29-31). Some studies have identified perivascular cells as another source of MSCs. These supportive cells of the vessel wall exhibit chemotactic activity in response to inflammation and injury, and have an MSC-like phenotype (32).
Owing to their robust proliferative capacity and multidirectional differentiation potential, MSCs can differentiate into myocytes, osteoblasts, chondrocytes, adipocytes, hepatocytes, stromal cells and other cell types under suitable culture conditions (29-32). Furthermore, MSCs possess anti-inflammatory (33) and immunomodulatory properties (34,35). Upon injury or inflammation, MSCs migrate to the site of damage and interact with immune cells to secrete cytokines, scavenge pathogens, suppress inflammatory responses, and reduce oxidative stress and apoptosis (36). The mechanisms through which MSCs repair tissue damage include: i) Direct differentiation or cell fusion; ii) transfer of exosomes, cytokines or organelles to injured cells through tunnelling nanotubes (TNTs); and iii) promotion of tissue repair by releasing cytokines in a paracrine manner (37). MSCs are widely available, and easy to extract, culture and expand. The lack of major histocompatibility complex II or co-stimulatory factors endows MSCs with specific immune-privileged properties (38). These characteristics make MSC transplantation a viable treatment option for various diseases (39,40).
MSCs and organ failure in sepsis
In numerous preclinical studies (25,26,36,39,41), MSCs have been shown to serve an immunomodulatory and protective role in organ function impairment associated with sepsis, including in the brain, lungs, liver, kidneys, heart, intestines and blood vessels (Fig. 1).
MSCs and septic lung injury
The respiratory system is particularly vulnerable to sepsis (41). The primary pathological changes observed in sepsis include intrapulmonary inflammatory cell infiltration, release of inflammatory mediators, pulmonary capillary thrombosis, interstitial pulmonary edema and pulmonary fibrosis. These changes manifest primarily as progressive dyspnea and intractable hypoxemia in patients with acute lung injury secondary to sepsis (42). The activation and recruitment of inflammatory cells, such as neutrophils and macrophages, in pulmonary circulation serve a crucial role in the onset and progression of septic lung injury (43,44). Neutrophils activate Toll-like receptor (TLR)4 and CD14+ T cells, promoting the release of inflammatory mediators such as IL-1β, TNF-α, NO and reactive oxygen species (ROS). These mediators damage the alveolar-capillary interface, and the cellular barrier between the alveolar epithelium and endothelium, increasing the permeability of the alveolar-capillary membrane and leading to interstitial lung edema (45-47). Additionally, endotoxins and inflammatory factors stimulate the release of tissue factor in the lungs, activating the exogenous coagulation pathway, which releases thrombin, and leads to pulmonary capillary embolism and interstitial fibrosis. Thrombin further promotes neutrophil adhesion and activation, exacerbating lung injury (48-50).
Owing to their immunomodulatory properties, MSCs can inhibit the release of inflammatory cytokines in the lungs during sepsis, thereby alleviating lung injury. Studies have demonstrated that MSCs can reduce the number of inflammatory cells in the alveolar lavage fluid of septic mice and inhibit the release of inflammatory factors, such as IL-1β, IL-6 and TNF-α, thereby correcting the immune imbalance (51-53). Chen et al (54) revealed that MSCs used in combination with melatonin reduced the number of CD14+ and CD68+ T cells, and the levels of IL-6 in the lungs during acute lung injury in sepsis, thereby attenuating the inflammatory response. Moreover, MSCs secrete antimicrobial peptides (such as LL-37), complement components (such as C5a) and other antimicrobial substances to inhibit the proliferation of pathogenic microorganisms and promote macrophage polarization toward the M2 phenotype, enhancing the phagocytic activity of macrophages and reducing the intrapulmonary microbial load (55-59). Activation of the exogenous coagulation pathway and formation of microthrombi in pulmonary capillaries are other crucial causes of septic lung injury. Tan et al (60) validated that MSCs reduced thrombin levels in pulmonary microcirculation, alleviated microcirculation embolism and improved pulmonary blood circulation. Furthermore, MSCs can alleviate sepsis-induced lung injury by regulating the expression of microRNAs (miRNAs) in exosomes (61,62).
MSCs and myocardial injury in sepsis
Sepsis-induced myocardial dysfunction (SIMD) is one of the most common complications of sepsis. The pathogenic factors of SIMD include the release of myocardial depressant factor, upregulation of NO, impairment of myocardial calcium homeostasis and mitochondrial dysfunction. SIMD manifests primarily as myocardial ischemia and hypoxia, systolic myocardial dysfunction, and reduced left ventricular ejection fraction (EF), resulting in inadequate tissue and organ perfusion (63,64). TLRs on cardiomyocyte membranes activate downstream mTOR/NF-κB and mTOR/Akt signaling pathways by recognizing pathogen-associated molecular patterns and danger-associated molecular patterns, resulting in the release of inflammatory factors, such as TNF-α and IL-6, which directly contribute to myocardial injury (65-67). These inflammatory factors activate myocardial endothelial cells, leading to increased secretion of inducible NO synthase (iNOS) and excessive production of NO. Excess NO inhibits type I calcium channels and myocardial mitochondrial function, resulting in systolic dysfunction and decreased cardiac output (68-70). Additionally, the ratio of anti-apoptotic to pro-apoptotic proteins decreases in sepsis, leading to myocardial damage (71).
Several studies have reported that MSCs attenuate inflammatory cell infiltration in cardiomyocytes during sepsis, alleviate myocardial injury, and improve myocardial contractility. Wu et al (72) reported that MSCs can markedly improve cardiac EF by reducing the expression of TLRs; inhibiting the NF-κB signaling pathway; and decreasing the levels of inflammatory factors, such as IL-1β, IL-6 and TNF-α, in the plasma and myocardium of septic mice. Weil et al (73) showed that, in addition to reducing the levels of myocardial inflammatory factors, MSCs can decrease the frequency of myocardial contraction, thereby improving myocardial function. Additionally, MSCs have been shown to secrete exosomes enriched with miRNA-223, which inhibits myocardial inflammatory responses and alleviates myocardial injury (74). Taken together, MSCs can attenuate the inflammatory response, inhibit NO release, improve myocardial calcium channel activity and mitochondrial function, and alleviate myocardial injury in sepsis by inhibiting inflammatory signaling pathways and secreting exosomes.
MSCs and septic liver injury
The liver exhibits the most robust inflammatory response to sepsis (75,76). Lipopolysaccharide in the peripheral blood stimulates Kupffer cells to secrete high-mobility group protein B1 (HMGB1); in the presence of HMGB1, immune cells release inflammatory factors, such as TNF-α and IL-6 (77,78). Studies have confirmed that TNF-α serves a crucial role in septic liver injury (77,78). Liver cells are rich in TNF-α receptors and TNF-α can directly cause liver injury. Additionally, TNF-α stimulates the release of other inflammatory factors, such as IL-6 and IL-1β; HMGB1 interacts with these factors, forming an inflammatory cascade that exacerbates liver injury. Moreover, TNF-α induces hepatocyte apoptosis, and recruits and activates inflammatory cells, such as neutrophils, further aggravating liver injury (77,78). Oxidative stress can increase the levels of ROS and NO in the liver, disrupting epithelial integrity, increasing hepatic permeability and triggering cholestasis (79).
Amplification of the inflammatory response is the primary cause of sepsis-induced liver injury. MSCs can inhibit the inflammatory response and the migration of inflammatory cells to the liver, thereby alleviating liver injury. Several studies have shown that MSCs can inhibit the release of TNF-α, IL-6 and monocyte chemotactic protein (MCP)-1, and upregulate the anti-inflammatory factors IL-10 and IL-4 to correct immune imbalance. In addition, MSCs can suppress the migration and activation of neutrophils in the liver, thereby alleviating liver damage caused by inflammation (80-82). Studies have demonstrated that adipose tissue-derived MSCs secrete TNF receptor 1, and attenuate apoptosis and inflammatory responses in the liver, thus improving the survival of septic mice (83,84). Furthermore, MSCs can promote macrophage polarization toward the M2 phenotype, clear pathogens, increase intrahepatic glycogen reserves, reduce hepatic oxidative stress, and decrease the plasma levels of aspartate aminotransferase and alanine aminotransferase (59).
MSCs and septic kidney injury
Sepsis-associated kidney injury (SAKI) involves the development of structural and functional abnormalities in the kidney following sepsis in patients without pre-existing kidney injury (85,86). Crucial factors involved in the pathogenesis of SAKI include inflammatory responses, abnormal intrarenal microcirculation, and altered bioenergetic processes of renal cells (85,86). The global incidence of SAKI in patients with sepsis ranges from 19 to 23%, whereas that in patients with septic shock is substantially higher, ranging from 51 to 66.9% (85,86). The excessive release of inflammatory factors in sepsis causes damage to glomerular capillary endothelial cells and tubular epithelial cells, resulting in increased glomerular capillary permeability and decreased glomerular filtration rate (GFR) (87,88). Furthermore, inflammatory factors can induce the release of tissue factor and activate the exogenous coagulation pathway, leading to the formation of microthrombi in renal microcirculation. Owing to ischemia and hypoxia, the production of ROS is increased in renal tissues, causing increased mitochondrial permeability and decreased mitochondrial membrane potential, leading to mitochondrial dysfunction and worsening of renal injury (89,90).
Studies have shown that MSCs can reduce the incidence of SAKI and attenuate renal tissue damage. Luo et al (91) showed that treatment with 106 MSCs within 3 h of induction of sepsis in mice reduced the incidence of SAKI when compared with control mice. After 24 h of induction, MSCs reduced the release of inflammatory factors, such as CXC, CCL and IL-17, and inhibited the migration of neutrophils to the kidney, improving renal tubular function and prolonging survival in mice with sepsis. Cóndor et al (92) demonstrated that umbilical cord Wharton's jelly-derived MSCs suppressed renal cell apoptosis, NF-κB expression, and the release of IL-1 and IL-6, thereby improving GFR and renal tubular function in sepsis. The combined use of adipose-derived MSCs (AMSCs) and exendin-4 (a glucagon-like peptide-1 analogue) can reduce renal oxidative stress and fibrosis in sepsis (93), and the combined use of MSCs and melatonin has been shown to substantially reduce NF-κB expression and the release of inflammatory factors (94). Additionally, MSCs can inhibit thrombosis in glomerular microcirculation, prevent the destruction of glomerular endothelial cells and restore renal function (90,92).
MSCs and sepsis-associated encephalopathy
Sepsisassociated encephalopathy (SAE) is characterized by confusion, coma and other changes in consciousness (95,96). Notably, 10-20% of patients with SAE experience long-term cognitive dysfunction worldwide (97). The pathogenesis of SAE primarily involves neuroinflammation, blood-brain barrier (BBB) impairment, cerebrovascular microcirculation disorder and mitochondrial dysfunction (95,96). MSCs can inhibit neuroinflammatory responses and promote the restoration of intracerebral microcirculation, thereby improving cognitive dysfunction following sepsis. Studies have demonstrated that MSCs injected via the internal jugular vein and peripheral vein can cross the BBB and colonize damaged sites in the brain (98,99). These infiltrating cells have been shown to inhibit the secretion of IL-6, IL-1β and TNF-α, and the proliferation and activation of microglia, consequently attenuating the inflammatory response (100-102). Tan et al (103) and Silva et al (104) demonstrated that MSCs improved cognitive function in septic mice by inhibiting neurological and peripheral inflammation. Studies have demonstrated that MSCs can secrete various trophic factors, including brain-derived neurotrophic factor, which promotes the differentiation of new neurons, and vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF), which promote local vascular regeneration and improve blood circulation in brain tissue (105-108). MSCs can transfer their mitochondria to damaged cells via membrane channels (TNTs), thereby continuing aerobic respiration and reducing ROS production. In addition, they can alleviate mitochondrial dysfunction and reduce oxidative stress (109). Liu et al (110) showed that olfactory mucosa-derived MSCs may promote the expression of UBIAD1, improving mitochondrial function. Cao et al (111) and Wang et al (112) demonstrated that MSCs could inhibit the expression of brain-iNOS and NADPH oxidase, and that MSC-derived exosomes contain antioxidant components, such as miRNAs, which may alleviate damage caused by oxidative stress (113).
MSCs and intestinal dysfunction in sepsis
Intestinal dysfunction can increase the global morbidity and mortality rates of patients with sepsis in the ICU to as high as 41.9% (114). MSCs can protect against sepsis-induced intestinal dysfunction by attenuating the intestinal inflammatory response, enhancing mitochondrial function and improving microbial diversity. Studies have demonstrated that MSCs may inhibit the production of the inflammatory factor TNF-α and promote the secretion of the anti-inflammatory factor IL-10 by modulating dendritic cell function (115,116). Chen et al (117) and Koliaraki et al (118) showed that MSCs can respond to TNF, inhibit the production of TNF-α and IL-12, and promote the secretion of IL-4 and IL-10. Parikh et al (119) and Zheng et al (120,121) showed that MSC-derived microvesicles could deliver mfn2 and PGC-1α to endothelial cells, promoting mitochondrial production and delivering the mitochondria directly to damaged endothelial cells to improve cellular energy metabolism and reduce oxidative stress-induced damage. Phinney et al (122) demonstrated that MSCs can promote mitochondrial autophagy. Additionally, MSCs have been shown to promote intestinal mucosal repair and increase the diversity of intestinal flora (123). For example, Valcz et al (124) showed that MSCs can colonize damaged intestinal mucosa and differentiate into intestinal mesenchymal or epithelial cells. Hayashi et al (125) also found that MSCs can secrete VEGF and TGF to promote the repair of damaged intestinal mucosa. Furthermore, MSCs may improve the diversity of intestinal flora, regulate the secretion of IgA and maintain intestinal symbiotic homeostasis (126).
MSCs and sepsis-induced coagulopathy (SIC)
SIC is characterized by an enhanced coagulation response and impairment of anticoagulation mechanisms, leading to extensive microthrombosis (127). The incidence of SIC in patients with sepsis is 50-70%, with ~35% of patients progressing to disseminated intravascular coagulation worldwide (128). The pathogenesis of SIC primarily includes the release of inflammatory mediators, endothelial cell injury, activation of the exogenous coagulation pathway, and inhibition of the anticoagulation and fibrinolytic systems (129). Studies have validated that MSCs possess potent anti-inflammatory and immunomodulatory functions. For example, Wang et al (130,131) and Miao et al (82) demonstrated that MSCs could inhibit the activity of the NLRP3 inflammasome, reduce the levels of IL-1β, IL-6 and TNF-α, and promote the secretion of IL-10. Furthermore, MSCs can alleviate vascular endothelial cell injury, thereby improving the anticoagulant function of protein C. Notably, Baudry et al (132) showed that MSCs pretreated with interferon-γ (IFNγ) reduced selectin-E levels and increased intercellular adhesion molecule-1 levels, thereby accelerating leukocyte flow in the blood, reducing leukocyte adhesion and attenuating vascular endothelial cell injury in septic mice (132). By improving the anticoagulant function of protein C, MSCs can substantially decrease the levels of von Willebrand factor and tissue factor in plasma, inhibit the exogenous coagulation pathway and reduce thrombin production, consequently ameliorating SIC (60).
Pathophysiological relationship between MSCs and skeletal muscle
MSCs possess a notable capacity for directed differentiation, which enables them to transform into skeletal muscle cells under appropriate conditions (133,134). In vitro studies have demonstrated that the transcription factor Pax-3 can induce the differentiation of MSCs to myogenic cells (135), whereas intracellular structural domain genes can drive the differentiation of MSCs to skeletal muscle cells (136). Additionally, mechanical traction can stimulate BMSCs to differentiate into skeletal muscle cells (137). An in vivo study validated that embryonic MSCs (EMSCs) promoted the repair of injured tibialis anterior muscle in mice, with >60% of EMSCs differentiating into skeletal myocytes (138).
In addition to their ability to differentiate into skeletal muscle cells, MSCs can enhance the functional recovery of injured skeletal muscles. In a previous study, BMSCs isolated from the tibia and femur of green fluorescent protein-expressing transgenic Sprague-Dawley rats were cultured and expanded in vitro. After these BMSCs were transplanted at the site of skeletal muscle injury, the contractile force of the injured muscle reached close to pre-injury levels after 1 month. By contrast, the contractile force in control rats recovered to only ~80% of the pre-injury levels (139). Another study showed that MSCs improved skeletal muscle function in a cell density-dependent manner, with the most significant treatment effect being observed when the number of transplanted MSCs was 10×106 (140). Notably, the treatment effect was not influenced by the timing of MSC transplantation or the sex of rats (141,142). Furthermore, MSCs have been shown to promote skeletal muscle angiogenesis and increase blood flow. In a previous study, transplantation of hypoxic MSCs in the hindlimb of mice resulted in a 2-fold increase in the number of skeletal muscle capillaries and a 7-fold increase in the number of vascular connections and branches when compared with the control group (139). Additionally, studies have shown that MSCs hold great promise in the treatment of various skeletal myopathies, such as Duchenne muscular dystrophy (143), skeletal muscle denervation atrophy (144) and traumatic skeletal muscle injury (145). These studies provide diverse avenues for further research on MSCs and the treatment of SIM using MSCs.
Potential mechanisms of MSCs in the treatment of SIM
Skeletal muscle, accounting for 35-45% of body weight in adult humans, is essential for maintaining all physiological activities. Promoting muscle regeneration and repair in patients with SIM is crucial for improving prognosis and quality of life. Factors that lead to skeletal muscle injury and dysfunction following sepsis include increased protein hydrolysis and decreased protein synthesis (146,147), oxidative stress (148), release of inflammatory mediators (149,150), skeletal muscle apoptosis (151), and skeletal muscle vascular damage. Skeletal muscle repair involves three stages, as follows: Inflammatory response, repair and remodeling. MSCs may enhance skeletal muscle function in SIM through numerous mechanisms (Fig. 2).
Myogenic differentiation
Owing to their multidirectional differentiation potential, MSCs can differentiate into skeletal muscle cells under appropriate conditions to directly repair muscle injury (152). Orlic et al (153) suggested that different environments lead to varying MSC differentiation rates, and that direct contact with injured organs or target cells is crucial for myogenic differentiation of MSCs. Egusa et al (154) showed that arranging BMSCs according to skeletal muscle fibers markedly improved the efficiency of the transformation of BMSCs to skeletal muscle. Studies have shown that treatment of MSCs with 5-azacytidine (5-AZA) may result in the formation of multinucleated myotubes expressing myosin after 7-11 days (155-157). Meligy et al (158) treated BMSCs, AMSCs and skeletal muscle-derived MSCs with 5-AZA and demonstrated that MSCs highly expressed myostatin after 1 week of treatment. In addition, microsatellite cells serve an essential role in the development and regeneration of skeletal muscles. These cells are dormant under physiological conditions but are activated upon muscle injury. Activated microsatellite cells can differentiate into myogenic cells that fuse to form myotubes (159,160). MSCs have been reported to secrete fibroblast growth factor (FGF), HGF and insulin-like growth factor-1 (IGF-1) to induce myogenic differentiation of microsatellite cells (161,162).
Homing of MSCs
Homing refers to the process by which MSCs are captured in the vascular system of the target tissue and subsequently migrate across the vascular endothelium to the target tissue (163). During inflammation or injury, various signaling molecules, such as chemokines, adhesion factors and growth factors, are locally released from the injured tissue, inducing MSCs to migrate to that tissue. Homing is crucial for the safe and effective clinical application of MSCs (164,165). The stromal cell-derived factor 1 (SDF-1)/CXCR-4 axis, comprising SDF-1 and its ligand CXCR-4, serves a vital role in MSC homing. Upon tissue injury, SDF-1α secretion increases, and MSCs expressing CXCR-4 migrate to the injury site along the SDF-1α concentration gradient to participate in tissue repair (166-169). Additionally, MCP (170,171), VEGF (172), HGF (173) and integrins (174) are essential for MSC homing. Ferrari et al (175) used BMSCs to treat injured muscles and found that BMSCs can migrate to the injury site, differentiating into skeletal muscle cells and promoting the regeneration of injured muscle fibers. Winkler et al (176) used MRI to visualize that labelled MSCs migrated to damaged muscle fibers and fused with them 24 h post-transplantation.
Anti-inflammatory effect of MSCs
In sepsis, the immune function becomes dysregulated, resulting in an uncontrolled inflammatory response, and the release of large amounts of TNF-α, IL-6 and other inflammatory factors. TNF-α promotes the secretion of iNOS, leading to muscle injury, whereas IL-6 has been shown to inhibit the myogenic differentiation of the C2C12 myotube cell line and the elongation of muscle protein peptide chains, causing muscle atrophy (149). The excessive release of inflammatory factors is a key driver of muscle injury (149). MSCs have powerful anti-inflammatory functions and can restore immune balance, an essential mechanism by which MSCs repair tissue damage (177). AMSCs have been shown to inhibit the expression of inflammatory factors, such as TNF-α, IL-6 and ROS, in injured gastrocnemius muscle while upregulating IL-4 and IL-10 levels to suppress the inflammatory response and repair muscle injury (178). BMSCs can reduce muscle fibrosis by inhibiting TGF-β1 expression. Moreover, MSCs can inhibit the activity of natural killer cells, preventing IFN-γ and TNF-α from exerting their effects, and can attenuate the muscle inflammatory response by inhibiting dendritic cell maturation.
MSCs modulate macrophage phenotype
Macrophages are heterogeneous immune cells that can be classified into two phenotypes: M1 and M2, based on their function and markers. M1-type macrophages secrete inflammatory factors, such as TNF-α, IL-1α, IL-6 and IL-12, which can inhibit the myogenic differentiation of C2C12 myotubes and lead to skeletal muscle injury (179-181). Conversely, M2-type macrophages can secrete anti-inflammatory factors, remove pathogens and apoptotic cells, promote myogenic differentiation of C2C12 cells, attenuate the skeletal muscle inflammatory response and facilitate muscle repair (179-181). When MSCs are co-cultured with macrophages in vitro, the inflammatory factors secreted by M1-type macrophages have been shown to activate MSCs. In turn, MSCs secrete anti-inflammatory factors, such as IL-10, IL-4 and TGF-β, which can promote the conversion of M1-type macrophages to M2-type macrophages. The release of these mediators is crucial for mediating M2-type macrophage polarization (182). MSCs can also induce M2-type macrophage polarization through the exosome pathway. MSC-derived exosomes have been reported to promote the conversion of M1-type macrophages to M2-type macrophages in infarcted myocardium, thereby reducing the local inflammatory response in a mouse model (182). Exosomal miRNA sequencing has revealed that miRNA-182 in MSC exosomes may mediate M2-type macrophage polarization (183-185). Additionally, MSCs can facilitate M2-type macrophage polarization via the regulatory T cell pathway (186).
Paracrine effects of MSCs
Paracrine secretion underpins the application of stem cells in tissue regeneration and constitutes a critical mechanism by which MSCs repair skeletal muscle injury. MSCs regulate skeletal muscle repair by releasing various growth factors and exosomes through the paracrine pathway. Studies have confirmed that MSCs secrete IGF-1, VEGF, sphingosine 1-phosphate (S1P) and other cellular growth factors involved in skeletal muscle injury repair (122,137). VEGF has been shown to promote capillary endothelial cell proliferation and the myogenic differentiation of C2C12 cells (187), while IGF-1 can enhance endothelial cell migration to the injury site and repair local blood circulation (188). S1P reduces skeletal muscle apoptosis, promotes myogenic proliferation, and stimulates satellite cell proliferation and differentiation (189). Moreover, MSC-derived exosomes serve an essential role in skeletal muscle repair. It has been found that BMSC-derived exosomes contain FGF-2, platelet-derived growth factor-BB and recombinant granulocyte colony-stimulating factor, which are associated with skeletal muscle regeneration (137,188). Exosomes are also enriched with miRNAs and proteins crucial for skeletal muscle injury repair. For example, miRNA-21 in exosomes inhibits apoptosis, and miRNA-494 promotes MSC myogenic differentiation and vascular regeneration (122,190). Studies have demonstrated that exosomes are enriched in annexin A1, which is essential for myofilament repair after skeletal muscle injury (191,192). Similarly, it has been shown that exosomes contain various proteins related to skeletal muscle injury repair, such as filament proteins and myosins, which can promote the proliferation of skeletal muscle satellite cells and muscle repair (193-195).
MSCs promote skeletal muscle vascular regeneration
Vascular regeneration has a crucial role in repairing skeletal muscle injury, and studies have shown that MSCs can promote this process by secreting cytokines. In 2000, Oswald et al (196) first applied VEGF to induce BMSCs to differentiate into vascular endothelial cells, although the differentiation efficiency was low. Since then, researchers have significantly increased the efficiency of vascular endothelial cell differentiation by adding VEFG, IGF-1, FGF and S1P to MSC culture media (197,198). Sassoli et al (187) found that MSCs secrete large amounts of VEGF, which promotes skeletal muscle vascular regeneration by activating the Notch-1 signaling pathway. MSCs also activate the TLR-2/TLR-6 pathway to enhance vascular regeneration through paracrine secretion. Grote et al (199) significantly increased the secretion of growth factors and cytokines, such as VEGF and granulocyte-macrophage colony-stimulating factor, by adding the TLR-2/TLR-6 agonist MLP-2 to MSC culture media. Additionally, Leroux et al (200) found that hypoxia-pretreated MSCs transplanted into skeletal muscle showed increased Wnt4 expression, and a significant increase in skeletal muscle vascular regeneration and blood flow, suggesting that MSCs promote vascular regeneration through the Wnt4 signaling pathway.
Limitations of MSCs therapy
MSCs have significant differentiation potential and can repair skeletal muscle injury through various mechanisms. However, several limitations hinder the clinical application of MSCs in treating skeletal muscle injuries.
Firstly, the optimal tissue source of MSCs remains undetermined. MSCs are primarily derived from bone marrow, adipose tissue, cord blood, peripheral blood and dental pulp, with bone marrow being the most common source. However, acquiring BMSCs requires invasive procedures, the isolation rate is low (0.001-0.01%), and the multi-directional differentiation ability of BMSCs decreases with age. Peripheral blood-derived MSCs are easy to obtain and possess the most potent immunosuppressive function, but they take longer to cultivate in vitro (201,202). AMSCs have a similar immunomodulatory capacity to dental pulp-derived MSCs (DPMSCs); AMSCs are easy to obtain and have a high cell isolation rate (3%), whereas DPMSCs have strong proliferative capacity (203). However, cord blood-derived MSCs exhibit a higher potential for multi-directional differentiation (203).
Secondly, the administration route for MSCs, whether local or systemic, remains controversial in terms of effectiveness. Local application at the injury site is fast-acting but carries a risk of bleeding (204). Systemic administration methods include subcutaneous, intramuscular, intravenous and intraperitoneal injections, each with varying efficacy. Castelo-Branco et al (205) reported that intraperitoneal but not intravenous administration of MSCs exhibited better homing and anti-inflammatory effects, while Gonçalves et al (206) reported conflicting findings. Roux et al (207) preferred intraperitoneal injection due to the lower probability of pulmonary embolism. Intramuscular and subcutaneous injections can prolong the duration of MSCs in vivo, with intramuscular injections showing retention for up to 100 days; however, data on intramuscular injections remain insufficient (204,208).
Finally, the optimal therapeutic dose of MSCs is not well-defined. Although studies have shown that administering 12×109 MSCs is safe, it does not imply that higher doses are more effective. MSCs possess anti-apoptotic, self-replicating and growth-regulating abilities, similar to tumor cells, and administering large doses may pose safety risks (209,210). Some studies have indicated that the effective minimum dose ranges from 10×108 to 15×108, with an optimal dose for aging patients at 10×108. Dose response data showing differential efficacy for improved outcomes were reported in three trials, which indicated that there was no significant difference between the therapeutic effects of 10×108 and 20×108 (211-213). Therefore, further studies are needed to validate the optimal dose for treating skeletal muscle diseases.
Discussion of the therapeutic potential of MSCs in SIM
Although advancements in medical technology have improved the early diagnosis and treatment of sepsis, multiple organ failure remains a leading cause of death in patients with sepsis (214). MSCs have emerged as a promising therapeutic option owing to their immunomodulatory, anti-inflammatory and regenerative properties (60,82,104,121). The present review highlights the therapeutic role of MSCs in sepsis-induced organ failure, focusing on lung injury, myocardial injury, kidney injury, encephalopathy and intestinal dysfunction. Existing studies have suggested that MSCs possess marked potential in the treatment of sepsis-induced organ failure. An in-depth understanding of the mechanisms of MSCs and a comparison of their efficacy across different organs may expand their application, eventually improving the outcomes and reducing the mortality rate of sepsis.
SIM results from a complex interplay of inflammation, oxidative stress and metabolic dysregulation (4-6,215,216). It not only results from sepsis but also influences the progression and outcomes of the disease (215). It contributes to malnutrition and negative nitrogen balance, weakening the overall condition and immune response of patients, thus making it more challenging to combat the infection (8,217). Additionally, atrophy of respiratory muscles, such as the diaphragm, can impair breathing and increase the likelihood of respiratory failure (218), necessitating prolonged mechanical ventilation, which in turn increases morbidity and mortality (218). Therefore, SIM is closely associated with the progression of sepsis in a cycle that worsens clinical outcomes. Addressing skeletal muscle atrophy through early intervention, nutritional support, physical rehabilitation and novel therapeutic approaches can help disrupt this cycle, and improve recovery and survival rates in patients with sepsis.
MSCs possess several characteristics that make them promising candidates for treating SIM. First, MSCs exert anti-inflammatory and immunomodulatory effects, which can attenuate the excessive inflammatory response caused by sepsis (111). Second, MSCs can secrete various growth factors and cytokines that promote tissue repair and regeneration (219). They can enhance the repair and regeneration of skeletal muscles by secreting exosomes and microvesicles that deliver beneficial molecules to the damaged site (220). Third, the antioxidant activity of MSCs can alleviate oxidative stress-induced damage in muscle cells. In particular, MSCs decrease the production of free radicals by improving the intracellular redox balance, thereby protecting muscle cells from oxidative stress-induced injury (221). Although preclinical studies have strongly supported the potential benefits of MSCs in the treatment of sepsis, further clinical research is warranted to translate the findings into effective treatments. A better understanding of the use of MSCs may help improve clinical outcomes and reduce the long-term impact of sepsis on skeletal muscle health.
Outlook and conclusion
SIM is a common complication in patients with sepsis, significantly affecting prognosis and quality of life, with a lack of effective therapeutic measures. MSCs have emerged as a novel treatment for certain refractory diseases, and have demonstrated good clinical efficacy in treating tissue repair, skeletal muscle diseases and sepsis-related conditions. The present review specifically focused on the mechanisms and therapeutic potential of MSCs in SIM. While there have been some studies on the application of MSCs in sepsis, a focused review on SIM is still lacking. Secondly, this review integrated the latest experimental and clinical data, providing comprehensive evidence for the application of MSCs in SIM. By synthesizing these data, this review not only highlighted the therapeutic potential of MSCs but also detailed their mechanisms of action. This may help to fill the current gaps in the literature, and offers valuable guidance for future research and clinical practice. Finally, a detailed comparison of the efficacy of MSCs in treating damage across different organs was conducted, including the lungs, heart, kidneys, brain and intestines. This comparative analysis offers a new perspective, demonstrating the role of MSCs in multi-organ protection. By elucidating how MSCs can mitigate damage in various organs, the present study provides a holistic view of their therapeutic potential, which is crucial for developing effective treatment strategies for SIM. Therefore, the present study may improve the understanding and application of MSCs in SIM, and their underlying mechanisms. However, skeletal muscle repair is a highly complex biological process. The mechanisms, efficiency and safety of MSCs in treating SIM have not yet been fully elucidated, necessitating further research to clarify these aspects. More clinical studies are required to obtain sufficient data to verify the safety and therapeutic effects of MSCs in treating SIM. As research on MSCs progresses, there is reason to believe that MSCs will become an effective treatment for SIM in the near future.
Availability of data and materials
Not applicable.
Authors' contributions
All authors contributed to the design of the study and writing of the manuscript. DW, SQ, LX, YL and CW contributed to the literature collection, analysis and interpretation for writing this review. DW and YW wrote the main manuscript text and prepared the figures. ZL, XB and YL revised the article critically for important intellectual content. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
This study was supported by grants from the National Natural Science Foundation of China (grant nos. 82002101 and 82002096).
References
Tupchong K, Koyfman A and Foran M: Sepsis, severe sepsis, and septic shock: A review of the literature. Afr J Emerg Med. 5:127–135. 2015. View Article : Google Scholar | |
Evans L, Rhodes A, Alhazzani W, Antonelli M, Coopersmith CM, French C, Machado FR, Mcintyre L, Ostermann M, Prescott HC, et al: Surviving sepsis campaign: International guidelines for management of sepsis and septic shock 2021. Crit Care Med. 49:e1063–e1143. 2021. View Article : Google Scholar : PubMed/NCBI | |
Matthaeus-Kraemer CT, Thomas-Rueddel DO, Schwarzkopf D, Rueddel H, Poidinger B, Reinhart K and Bloos F: Crossing the handover chasm: Clinicians' perceptions of barriers to the early detection and timely management of severe sepsis and septic shock. J Crit Care. 36:85–91. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Xu L, Yang Z, Wang D, Li T, Yang F, Li Z, Bai X and Wang Y: Gut-muscle axis and sepsis-induced myopathy: The potential role of gut microbiota. Biomed Pharmacother. 163:1148372023. View Article : Google Scholar : PubMed/NCBI | |
Mankowski RT, Laitano O, Clanton TL and Brakenridge SC: Pathophysiology and treatment strategies of acute myopathy and muscle wasting after sepsis. J Clin Med. 10:18742021. View Article : Google Scholar : PubMed/NCBI | |
Callahan LA and Supinski GS: Sepsis-induced myopathy. Crit Care Med. 37(10 Suppl): S354–S367. 2009. View Article : Google Scholar | |
Schefold JC, Bierbrauer J and Weber-Carstens S: Intensive care unit-acquired weakness (ICUAW) and muscle wasting in critically ill patients with severe sepsis and septic shock. J Cachexia Sarcopenia Muscle. 1:147–157. 2010. View Article : Google Scholar | |
Liu W, Hu C and Zhao S: Sarcopenia and mortality risk of patients with sepsis: A meta-analysis. Int J Clin Pract. 2022:49744102022. View Article : Google Scholar : PubMed/NCBI | |
Panahi A, Malekmohammad M, Soleymani F and Hashemian SM: The prevalence and outcome of intensive care unit acquired weakness (ICUAW). Tanaffos. 19:250–255. 2020. | |
Dinglas VD, Aronson Friedman L, Colantuoni E, Mendez-Tellez PA, Shanholtz CB, Ciesla ND, Pronovost PJ and Needham DM: Muscle weakness and 5-year survival in acute respiratory distress syndrome survivors. Crit Care Med. 45:446–453. 2017. View Article : Google Scholar : PubMed/NCBI | |
Meyer-Frießem CH, Malewicz NM, Rath S, Ebel M, Kaisler M, Tegenthoff M, Schildhauer TA, Pogatzki-Zahn EM, Maier C and Zahn PK: Incidence, time course and influence on quality of life of intensive care unit-acquired weakness symptoms in long-term intensive care survivors. J Intensive Care Med. 36:1313–1322. 2021. View Article : Google Scholar | |
Appleton RT, Kinsella J and Quasim T: The incidence of intensive care unit-acquired weakness syndromes: A systematic review. J Intensive Care Soc. 16:126–136. 2015. View Article : Google Scholar : PubMed/NCBI | |
de Andrade-Junior MC, de Salles ICD, de Brito CMM, Pastore-Junior L, Righetti RF and Yamaguti WP: Skeletal muscle wasting and function impairment in intensive care patients with severe COVID-19. Front Physiol. 12:6409732021. View Article : Google Scholar : PubMed/NCBI | |
Herridge MS, Tansey CM, Matté A, Tomlinson G, Diaz-Granados N, Cooper A, Guest CB, Mazer CD, Mehta S, Stewart TE, et al: Functional disability 5 years after acute respiratory distress syndrome. N Engl J Med. 364:1293–1304. 2011. View Article : Google Scholar : PubMed/NCBI | |
Odden AJ, Rohde JM, Bonham C, Kuhn L, Malani PN, Chen LM, Flanders SA and Iwashyna TJ: Functional outcomes of general medical patients with severe sepsis. BMC Infect Dis. 13:5882013. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Huang Y, Chen Y, Shen X, Pan H and Yu W: Impact of muscle mass on survival in patients with sepsis: A systematic review and meta-analysis. Ann Nutr Metab. 77:330–336. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yan W, Chen Y, Guo Y, Xia Y, Li C, Du Y, Lin C, Xu X, Qi T, Fan M, et al: Irisin promotes cardiac homing of intravenously delivered MSCs and protects against ischemic heart injury. Adv Sci (Weinh). 9:e21036972022. View Article : Google Scholar : PubMed/NCBI | |
Gnecchi M, Danieli P and Cervio E: Mesenchymal stem cell therapy for heart disease. Vascul Pharmacol. 57:48–55. 2012. View Article : Google Scholar : PubMed/NCBI | |
Tzouvelekis A, Toonkel R, Karampitsakos T, Medapalli K, Ninou I, Aidinis V, Bouros D and Glassberg MK: Mesenchymal stem cells for the treatment of idiopathic pulmonary fibrosis. Front Med (Lausanne). 5:1422018. View Article : Google Scholar : PubMed/NCBI | |
Sinclair K, Yerkovich ST and Chambers DC: Mesenchymal stem cells and the lung. Respirology. 18:397–411. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ye T, Chen Z, Zhang J, Luo L, Gao R, Gong L, Du Y, Xie Z, Zhao B, Li Q and Wang Y: Large extracellular vesicles secreted by human iPSC-derived MSCs ameliorate tendinopathy via regulating macrophage heterogeneity. Bioact Mater. 21:194–208. 2022.PubMed/NCBI | |
He X, Ai S, Guo W, Yang Y, Wang Z, Jiang D and Xu X: Umbilical cord-derived mesenchymal stem (stromal) cells for treatment of severe sepsis: Aphase 1 clinical trial. Transl Res. 199:52–61. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cribbs SK and Martin GS: Stem cells in sepsis and acute lung injury. Am J Med Sci. 341:325–332. 2011. View Article : Google Scholar | |
Walter J, Ware LB and Matthay MA: Mesenchymal stem cells: Mechanisms of potential therapeutic benefit in ARDS and sepsis. Lancet Respir Med. 2:1016–1026. 2014. View Article : Google Scholar : PubMed/NCBI | |
Khosrojerdi A, Soudi S, Hosseini AZ, Eshghi F, Shafiee A and Hashemi SM: Immunomodulatory and therapeutic effects of mesenchymal stem cells on organ dysfunction in sepsis. Shock. 55:423–440. 2021. View Article : Google Scholar | |
Ho MSH, Mei SHJ and Stewart DJ: The immunomodulatory and therapeutic effects of mesenchymal stromal cells for acute lung injury and sepsis. J Cell Physiol. 230:2606–2617. 2015. View Article : Google Scholar : PubMed/NCBI | |
Friedenstein AJ, Petrakova KV, Kurolesova AI and Frolova GP: Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation. 6:230–247. 1968. View Article : Google Scholar : PubMed/NCBI | |
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, Deans R, Keating A, Prockop D and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8:315–317. 2006. View Article : Google Scholar : PubMed/NCBI | |
Bianco P: 'Mesenchymal' stem cells. Annu Rev Cell Dev Biol. 30:677–704. 2014. View Article : Google Scholar | |
Rankin S: Mesenchymal stem cells. Thorax. 67:565–566. 2012. View Article : Google Scholar : PubMed/NCBI | |
Jiang W and Xu J: Immune modulation by mesenchymal stem cells. Cell Prolif. 53:e127122020. View Article : Google Scholar : | |
Yianni V and Sharpe PT: Perivascular-derived mesenchymal stem cells. J Dent Res. 98:1066–1072. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zimmermann JA, Hettiaratchi MH and McDevitt TC: Enhanced immunosuppression of T cells by sustained presentation of bioactive interferon-γ within three-dimensional mesenchymal stem cell constructs. Stem Cells Transl Med. 6:223–237. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yarygin KN, Lupatov AY and Sukhikh GT: Modulation of immune responses by mesenchymal stromal cells. Bull Exp Biol Med. 161:561–565. 2016. View Article : Google Scholar : PubMed/NCBI | |
Glenn JD and Whartenby KA: Mesenchymal stem cells: Emerging mechanisms of immunomodulation and therapy. World J Stem Cells. 6:526–539. 2014. View Article : Google Scholar : PubMed/NCBI | |
Saeedi P, Halabian R and Fooladi AAI: Antimicrobial effects of mesenchymal stem cells primed by modified LPS on bacterial clearance in sepsis. J Cell Physiol. 234:4970–4986. 2019. View Article : Google Scholar | |
Spees JL, Lee RH and Gregory CA: Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 7:1252016. View Article : Google Scholar : PubMed/NCBI | |
Le Blanc K, Tammik C, Rosendahl K, Zetterberg E and Ringdén O: HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 31:890–896. 2003. View Article : Google Scholar : PubMed/NCBI | |
Uccelli A, Moretta L and Pistoia V: Mesenchymal stem cells in health and disease. Nat Rev Immunol. 8:726–736. 2008. View Article : Google Scholar | |
Minguell JJ, Conget P and Erices A: Biology and clinical utilization of mesenchymal progenitor cells. Braz J Med Biol Res. 33:881–887. 2000. View Article : Google Scholar : PubMed/NCBI | |
Kim WY and Hong SB: Sepsis and acute respiratory distress syndrome: Recent update. Tuberc Respir Dis (Seoul). 79:53–57. 2016. View Article : Google Scholar : PubMed/NCBI | |
Martin GS and Bernard GR: Airway and lung in sepsis. Intensive Care Med. 27(Suppl 1): S63–S79. 2001. View Article : Google Scholar : PubMed/NCBI | |
Davis C: Risk factors for the development of acute lung injury in patients with septic shock: An observational cohort study. J Emerg Med. 36:P982009. View Article : Google Scholar | |
Schmidt EP, Yang Y, Janssen WJ, Gandjeva A, Perez MJ, Barthel L, Zemans RL, Bowman JC, Koyanagi DE, Yunt ZX, et al: The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 18:1217–1223. 2012. View Article : Google Scholar : PubMed/NCBI | |
Lee WL and Slutsky AS: Sepsis and endothelial permeability. N Engl J Med. 363:689–691. 2010. View Article : Google Scholar : PubMed/NCBI | |
Lomas-Neira J, Perl M, Venet F, Chung CS and Ayala A: The role and source of tumor necrosis factor-α in hemorrhage-induced priming for septic lung injury. Shock. 37:611–620. 2012. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Taneja R, Razavi HM, Law C, Gillis C and Mehta S: Specific role of neutrophil inducible nitric oxide synthase in murine sepsis-induced lung injury in vivo. Shock. 37:539–547. 2012. View Article : Google Scholar : PubMed/NCBI | |
Grover SP and Mackman N: Tissue factor: An essential mediator of hemostasis and trigger of thrombosis. Arterioscler Thromb Vasc Biol. 38:709–725. 2018. View Article : Google Scholar : PubMed/NCBI | |
Witkowski M, Landmesser U and Rauch U: Tissue factor as a link between inflammation and coagulation. Trends Cardiovasc Med. 26:297–303. 2016. View Article : Google Scholar : PubMed/NCBI | |
Evans CE and Zhao YY: Impact of thrombosis on pulmonary endothelial injury and repair following sepsis. Am J Physiol Lung Cell Mol Physiol. 312:L441–L451. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhao Y, Yang C, Wang H, Li H, Du J, Gu W and Jiang J: Therapeutic effects of bone marrow-derived mesenchymal stem cells on pulmonary impact injury complicated with endotoxemia in rats. Int Immunopharmacol. 15:246–253. 2013. View Article : Google Scholar : PubMed/NCBI | |
Lee FY, Chen KH, Wallace CG, Sung PH, Sheu JJ, Chung SY, Chen YL, Lu HI, Ko SF, Sun CK, et al: Xenogeneic human umbilical cord-derived mesenchymal stem cells reduce mortality in rats with acute respiratory distress syndrome complicated by sepsis. Oncotarget. 8:45626–45642. 2017. View Article : Google Scholar : PubMed/NCBI | |
Asami T, Ishii M, Namkoong H, Yagi K, Tasaka S, Asakura T, Suzuki S, Kamo T, Okamori S, Kamata H, et al: Anti-inflammatory roles of mesenchymal stromal cells during acute Streptococcus pneumoniae pulmonary infection in mice. Cytotherapy. 20:302–313. 2017. View Article : Google Scholar | |
Chen HH, Chang CL, Lin KC, Sung PH, Chai HT, Zhen YY, Chen YC, Wu YC, Leu S, Tsai TH, et al: Melatonin augments apoptotic adipose-derived mesenchymal stem cell treatment against sepsis-induced acute lung injury. Am J Transl Res. 6:439–458. 2014.PubMed/NCBI | |
Li W, Chen W, Huang S, Tang X, Yao G and Sun L: Mesenchymal stem cells enhance pulmonary antimicrobial immunity and prevent following bacterial infection. Stem Cells Int. 2020:31694692020. View Article : Google Scholar : PubMed/NCBI | |
Krasnodembskaya A, Song Y, Fang X, Gupta N, Serikov V, Lee JW and Matthay MA: Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells. 28:2229–2238. 2010. View Article : Google Scholar : PubMed/NCBI | |
Rabani R, Volchuk A, Jerkic M, Ormesher L, Garces-Ramirez L, Canton J, Masterson C, Gagnon S, Tatham KC, Marshall J, et al: Mesenchymal stem cells enhance NOX2-dependent reactive oxygen species production and bacterial killing in macrophages during sepsis. Eur Respir J. 51:17020212018. View Article : Google Scholar : PubMed/NCBI | |
Yao M, Cui B, Zhang W, Ma W, Zhao G and Xing L: Exosomal miR-21 secreted by IL-1β-primed-mesenchymal stem cells induces macrophage M2 polarization and ameliorates sepsis. Life Sci. 264:1186582021. View Article : Google Scholar | |
Krasnodembskaya A, Samarani G, Song Y, Zhuo H, Su X, Lee JW, Gupta N, Petrini M and Matthay MA: Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am J Physiol Lung Cell Mol Physiol. 302:L1003–L1013. 2012. View Article : Google Scholar : PubMed/NCBI | |
Tan L, Huang Y, Pan X, Quan S, Xu S, Li D, Song L, Zhang X, Chen W and Pan J: Administration of bone marrow stromal cells in sepsis attenuates sepsis-related coagulopathy. Ann Med. 48:235–245. 2016. View Article : Google Scholar : PubMed/NCBI | |
Dos Santos CC, Amatullah H, Vaswani CM, Maron-Gutierrez T, Kim M, Mei SHJ, Szaszi K, Monteiro APT, Varkouhi AK, Herreroz R, et al: Mesenchymal stromal (stem) cell therapy modulates miR-193b-5p expression to attenuate sepsis-induced acute lung injury. Eur Respir J. 59:20042162022. View Article : Google Scholar | |
Younes N, Zhou L, Amatullah H, Mei SHJ, Herrero R, Lorente JA, Stewart DJ, Marsden P, Liles WC, Hu P and Dos Santos CC: Mesenchymal stromal/stem cells modulate response to experimental sepsis-induced lung injury via regulation of miR-27a-5p in recipient mice. Thorax. 75:556–567. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bi CF, Liu J, Yang LS and Zhang JF: Research progress on the mechanism of sepsis induced myocardial injury. J Inflamm Res. 15:4275–4290. 2022. View Article : Google Scholar : PubMed/NCBI | |
Aneman A and Vieillard-Baron A: Cardiac dysfunction in sepsis. Intensive Care Med. 42:2073–2076. 2016. View Article : Google Scholar : PubMed/NCBI | |
Rudiger A and Singer M: Mechanisms of sepsis-induced cardiac dysfunction. Crit Care Med. 35:1599–1608. 2007. View Article : Google Scholar : PubMed/NCBI | |
Kumar A, Thota V, Dee L, Olson J, Uretz E and Parrillo JE: Tumor necrosis factor alpha and interleukin 1beta are responsible for in vitro myocardial cell depression induced by human septic shock serum. Resuscitation. 32:P1661996. View Article : Google Scholar | |
Zhang X, Lu C, Gao M, Cao X, Ha T, Kalbfleisch JH, Williams DL, Li C and Kao RL: Toll-like receptor 4 plays a central role in cardiac dysfunction during trauma hemorrhage shock. Shock. 42:31–37. 2014. View Article : Google Scholar : PubMed/NCBI | |
Sato R and Nasu M: A review of sepsis-induced cardiomyopathy. J Intensive Care. 3:482015. View Article : Google Scholar : PubMed/NCBI | |
Stengl M, Bartak F, Sykora R, Chvojka J, Benes J, Krouzecky A, Novak I, Sviglerova J, Kuncova J and Matejovic M: Reduced L-type calcium current in ventricular myocytes from pigs with hyperdynamic septic shock. Crit Care Med. 38:579–587. 2010. View Article : Google Scholar | |
Kumar A, Brar R, Wang P, Dee L, Skorupa G, Khadour F, Schulz R and Parrillo JE: Role of nitric oxide and cGMP in human septic serum-induced depression of cardiac myocyte contractility. Am J Physiol. 276:R265–R276. 1999.PubMed/NCBI | |
Lv X and Wang H: Pathophysiology of sepsis-induced myocardial dysfunction. Mil Med Res. 3:302016.PubMed/NCBI | |
Wu Y, Zhou J, Bi L, Huang M, Han Y, Zhang Q, Zhu D and Zhou S: Effects of bone marrow mesenchymal stem cells on the cardiac function and immune system of mice with endotoxemia. Mol Med Rep. 13:5317–5325. 2016. View Article : Google Scholar : PubMed/NCBI | |
Weil BR, Herrmann JL, Abarbanell AM, Manukyan MC, Poynter JA and Meldrum DR: Intravenous infusion of mesenchymal stem cells is associated with improved myocardial function during endotoxemia. Shock. 36:235–241. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Huang W, Wang Y and Fan GC: Abstract 12290: Exosomal miR-223 contributes to mesenchymal stem cell-elicited cardio-protection in polymicrobial sepsis. Circulation. 1322015. | |
Giovannini I, Chiarla C, Giuliante F, Vellone M, Ardito F and Nuzzo G: Sepsis-induced cholestasis. Hepatology. 47:3612008. View Article : Google Scholar | |
Woźnica EA, Inglot M, Woźnica RK and Łysenko L: Liver dysfunction in sepsis. Adv Clin Exp Med. 27:547–551. 2018. View Article : Google Scholar | |
Gaddam RR, Fraser R, Badiei A, Chambers S, Cogger VC, Le Couteur DG and Bhatia M: Differential effects of kupffer cell inactivation on inflammation and the liver sieve following caecal-ligation and puncture-induced sepsis in mice. Shock. 47:480–490. 2017. View Article : Google Scholar | |
Wang H and Liu D: Baicalin inhibits high-mobility group box 1 release and improves survival in experimental sepsis. Shock. 41:324–330. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Yin Y and Yao Y: Advances in sepsis-associated liver dysfunction. Burns Trauma. 2:97–105. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yagi H, Soto-Gutierrez A, Kitagawa Y, Tilles AW, Tompkins RG and Yarmush ML: Bone marrow mesenchymal stromal cells attenuate organ injury induced by LPS and burn. Cell Transplant. 19:823–830. 2010. View Article : Google Scholar : PubMed/NCBI | |
Wu KH, Wu HP, Chao WR, Lo WY, Tseng PC, Lee CJ, Peng CT, Lee MS and Chao YH: Time-series expression of toll-like receptor 4 signaling in septic mice treated with mesenchymal stem cells. Shock. 45:634–640. 2016. View Article : Google Scholar | |
Miao CM, Jiang XW, He K, Li PZ, Liu ZJ, Cao D, Ou ZB, Gong JP, Liu CA and Cheng Y: Bone marrow stromal cells attenuate LPS-induced mouse acute liver injury via the prostaglandin E 2-dependent repression of the NLRP3 inflammasome in Kupffer cells. Immunol Lett. 179:102–113. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yagi H, Soto-Gutierrez A, Navarro-Alvarez N, Nahmias Y, Goldwasser Y, Kitagawa Y, Tilles AW, Tompkins RG, Parekkadan B and Yarmush ML: Reactive bone marrow stromal cells attenuate systemic inflammation via sTNFR1. Mol Ther. 18:1857–1864. 2010. View Article : Google Scholar : PubMed/NCBI | |
Liang H, Ding X, Yu Y, Zhang H, Wang L, Kan Q, Ma S, Guan F and Sun T: Adipose-derived mesenchymal stem cells ameliorate acute liver injury in rat model of CLP induced-sepsis via sTNFR1. Exp Cell Res. 383:1114652019. View Article : Google Scholar : PubMed/NCBI | |
Umbro I, Gentile G, Tinti F, Muiesan P and Mitterhofer AP: Recent advances in pathophysiology and biomarkers of sepsis-induced acute kidney injury. J Infect. 72:131–142. 2016. View Article : Google Scholar | |
Gómez H and Kellum JA: Sepsis-induced acute kidney injury. Curr Opin Crit Care. 22:546–553. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zarjou A and Agarwal A: Sepsis and acute kidney injury. J Am Soc Nephrol. 22:999–1006. 2011. View Article : Google Scholar : PubMed/NCBI | |
Yoshimoto K, Komaru Y, Iwagami M and Doi K: Acute kidney injury in sepsis: Evidence from Asia. Semin Nephrol. 40:489–497. 2020. View Article : Google Scholar : PubMed/NCBI | |
Manrique-Caballero CL, Del Rio-Pertuz G and Gomez H: Sepsis-associated acute kidney injury. Crit Care Clin. 37:279–301. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bellomo R, Kellum JA, Ronco C, Wald R, Martensson J, Maiden M, Bagshaw SM, Glassford NJ, Lankadeva Y, Vaara ST and Schneider A: Acute kidney injury in sepsis. Intensive Care Med. 43:816–828. 2017. View Article : Google Scholar : PubMed/NCBI | |
Luo CJ, Zhang FJ, Zhang L, Geng YQ, Li QG, Hong Q, Fu B, Zhu F, Cui SY, Feng Z, et al: Mesenchymal stem cells ameliorate sepsis-associated acute kidney injury in mice. Shock. 41:123–129. 2014. View Article : Google Scholar | |
Cóndor JM, Rodrigues CE, Sousa Moreira RD, Canale D, Volpini RA, Shimizu MH, Camara NO, Noronha Ide L and Andrade L: Treatment with human Wharton's Jelly-derived mesenchymal stem cells attenuates sepsis-induced kidney injury, liver injury, and endothelial dysfunction. Stem Cells Transl Med. 5:1048–1057. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chen CH, Cheng BC, Chen KH, Shao PL, Sung PH, Chiang HJ, Yang CC, Lin KC, Sun CK, Sheu JJ, et al: Combination therapy of exendin-4 and allogenic adipose-derived mesenchymal stem cell preserved renal function in a chronic kidney disease and sepsis syndrome setting in rats. Oncotarget. 8:100002–100020. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen HH, Lin KC, Wallace CG, Chen YT, Yang CC, Leu S, Chen YC, Sun CK, Tsai TH, Chen YL, et al: Additional benefit of combined therapy with melatonin and apoptotic adipose-derived mesenchymal stem cell against sepsis-induced kidney injury. J Pineal Res. 57:16–32. 2014. View Article : Google Scholar : PubMed/NCBI | |
Polito A, Eischwald F, Maho AL, Polito A, Azabou E, Annane D, Chrétien F, Stevens RD, Carlier R and Sharshar T: Pattern of brain injury in the acute setting of human septic shock. Crit Care. 17:R2042013. View Article : Google Scholar : PubMed/NCBI | |
Catarina AV, Branchini G, Bettoni L, De Oliveira JR and Nunes FB: Sepsis-associated encephalopathy: From pathophysiology to progress in experimental studies. Mol Neurobiol. 58:2770–2779. 2021. View Article : Google Scholar : PubMed/NCBI | |
Prescott HC and Angus DC: Enhancing recovery from sepsis: A review. JAMA. 319:62–75. 2018. View Article : Google Scholar : PubMed/NCBI | |
Oh SH, Choi C, Chang DJ, Shin DA, Lee N, Jeon I, Sung JH, Lee H, Hong KS, Ko JJ and Song J: Early neuroprotective effect with lack of long-term cell replacement effect on experimental stroke after intra-arterial transplantation of adipose-derived mesenchymal stromal cells. Cytotherapy. 17:1090–1103. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jiang W, Liang G, Li X, Li Z, Gao X, Feng S, Wang X, Liu M and Liu Y: Intracarotid transplantation of autologous adipose-derived mesenchymal stem cells significantly improves neurological deficits in rats after MCAo. J Mater Sci Mater Med. 25:1357–1366. 2014. View Article : Google Scholar : PubMed/NCBI | |
Huang P, Gebhart N, Richelson E, Brott TG, Meschia JF and Zubair AC: Mechanism of mesenchymal stem cell-induced neuron recovery and anti-inflammation. Cytotherapy. 16:1336–1344. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yoo SW, Chang DY, Lee HS, Kim GH, Park JS, Ryu BY, Joe EH, Lee YD, Kim SS and Suh-Kim H: Immune following suppression mesenchymal stem cell transplantation in the ischemic brain is mediated by TGF-β. Neurobiol Dis. 58:249–257. 2013. View Article : Google Scholar : PubMed/NCBI | |
Redondo-Castro E, Cunningham C, Miller J, Martuscelli L, Aoulad-Ali S, Rothwell NJ, Kielty CM, Allan SM and Pinteaux E: Interleukin-1 primes human mesenchymal stem cells towards an anti-inflammatory and pro-trophic phenotype in vitro. Stem Cell Res Ther. 8:792017. View Article : Google Scholar : PubMed/NCBI | |
Tan L, Cheng Y, Wang H, Tong J and Qin X: Peripheral transplantation of mesenchymal stem cells at sepsis convalescence improves cognitive function of sepsis surviving mice. Oxid Med Cell Longev. 2022:68977652022. View Article : Google Scholar : PubMed/NCBI | |
Silva AYO, Amorim ÉA, Barbosa-Silva MC, Lima MN, Oliveira HA, Granja MG, Oliveira KS, Fagundes PM, Neris RLS, Campos RMP, et al: Mesenchymal stromal cells protect the blood-brain barrier, reduce astrogliosis, and prevent cognitive and behavioral alterations in surviving septic mice. Crit Care Med. 48:e290–e298. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li X, Zheng W, Bai H, Wang J, Wei R, Wen H and Ning H: Intravenous administration of adipose tissue-derived stem cells enhances nerve healing and promotes BDNF expression via the TrkB signaling in a rat stroke model. Neuropsychiatr Dis Treat. 12:1287–1293. 2016.PubMed/NCBI | |
Han C, Song L, Liu Y, Zou W, Jiang C and Liu J: Rat cortex and hippocampus-derived soluble factors for the induction of adipose-derived mesenchymal stem cells into neuron-like cells. Cell Biol Int. 38:768–776. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gutiérrez-Fernández M, Rodriguez-Frutos B, Ramos-Cejudo J, Otero-Ortega L, Fuentes B, Vallejo-Cremades MT, Sanz-Cuesta BE and Díez-Tejedor E: Comparison between xenogeneic and allogeneic adipose mesenchymal stem cells in the treatment of acute cerebral infarct: Proof of concept in rats. J Transl Med. 13:462015. View Article : Google Scholar : PubMed/NCBI | |
Ribeiro CA, Fraga JS, Grãos M, Neves NM, Reis RL, Gimble JM, Sousa N and Salgado AJ: The secretome of stem cells isolated from the adipose tissue and Wharton jelly acts differently on central nervous system derived cell populations. Stem Cell Res Ther. 3:182012. View Article : Google Scholar : PubMed/NCBI | |
Mahrouf-Yorgov M, Augeul L, Da Silva CC, Jourdan M, Rigolet M, Manin S, Ferrera R, Ovize M, Henry A, Guguin A, et al: Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 24:1224–1238. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Huang Y, He J, Zhuo Y, Chen W, Ge L, Duan D, Lu M and Hu Z: Olfactory mucosa mesenchymal stem cells ameliorate cerebral ischemic/reperfusion injury through modulation of UBIAD1 expression. Front Cell Neurosci. 14:5802062020. View Article : Google Scholar : PubMed/NCBI | |
Cao D, Qiao H, He D, Qin X, Zhang Q and Zhou Y: Mesenchymal stem cells inhibited the inflammation and oxidative stress in LPS-activated microglial cells through AMPK pathway. J Neural Transm (Vienna). 126:1589–1597. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang SS, Jia J and Wang Z: Mesenchymal stem cell-derived extracellular vesicles suppresses iNOS expression and ameliorates neural impairment in Alzheimer's disease mice. J Alzheimers Dis. 61:1005–1013. 2018. View Article : Google Scholar | |
Cai G, Cai G, Zhou H, Zhuang Z, Liu K, Pei S, Wang Y, Wang H, Wang X, Xu S, et al: Mesenchymal stem cell-derived exosome miR-542-3p suppresses inflammation and prevents cerebral infarction. Stem Cell Res Ther. 12:22021. View Article : Google Scholar : PubMed/NCBI | |
Fleischmann-Struzek C, Mellhammar L, Rose N, Cassini A, Rudd KE, Schlattmann P, Allegranzi B and Reinhart K: Incidence and mortality of hospital- and ICU-treated sepsis: Results from an updated and expanded systematic review and meta-analysis. Intensive Care Med. 46:1552–1562. 2020. View Article : Google Scholar : PubMed/NCBI | |
Aggarwal S and Pittenger MF: Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 105:1815–1822. 2005. View Article : Google Scholar | |
Li YP, Paczesny S, Lauret E, Poirault S, Bordigoni P, Mekhloufi F, Hequet O, Bertrand Y, Ou-Yang JP, Stoltz JF, et al: Human mesenchymal stem cells license adult CD34+ hemopoietic progenitor cells to differentiate into regulatory dendritic cells through activation of the Notch pathway. J Immunol. 180:1598–1608. 2008. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Zhang Y, Wang W, Liu Z, Meng J and Han Z: Mesenchymal stem cells modified with heme oxygenase-1 have enhanced paracrine function and attenuate lipopolysaccharide-induced inflammatory and oxidative damage in pulmonary microvascular endothelial cells. Cell Physiol Biochem. 49:101–122. 2018. View Article : Google Scholar : PubMed/NCBI | |
Koliaraki V, Pallangyo CK, Greten FR and Kollias G: Mesenchymal cells in colon cancer. Gastroenterology. 152:964–979. 2017. View Article : Google Scholar : PubMed/NCBI | |
Parikh SM, Yang Y, He L, Tang C, Zhan M and Dong Z: Mitochondrial function and disturbances in the septic kidney. Semin Nephrol. 35:108–119. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zheng K, Chen S and Hu X: Peroxisome proliferator-activated receptor gamma coactivator-1 alpha: A double-edged sword in prostate cancer. Curr Cancer Drug Targets. 22:541–559. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zheng D, Zhou H, Wang H, Zhu Y, Wu Y, Li Q, Li T and Liu L: Mesenchymal stem cell-derived microvesicles improve intestinal barrier function by restoring mitochondrial dynamic balance in sepsis rats. Stem Cell Res Ther. 12:2992021. View Article : Google Scholar : PubMed/NCBI | |
Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, Stolz DB, Watkins SC, Di YP, Leikauf GD, et al: Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun. 6:84722015. View Article : Google Scholar : PubMed/NCBI | |
Weber D, Hiergeist A, Weber M, Dettmer K, Wolff D, Hahn J, Herr W, Gessner A and Holler E: Detrimental effect of broad-spectrum antibiotics on intestinal microbiome diversity in patients after allogeneic stem cell transplantation: Lack of commensal sparing antibiotics. Clin Infect Dis. 68:1303–1310. 2019. View Article : Google Scholar | |
Valcz G, Krenács T, Sipos F, Leiszter K, Tóth K, Balogh Z, Csizmadia A, Muzes G, Molnár B and Tulassay Z: The role of the bone marrow derived mesenchymal stem cells in colonic epithelial regeneration. Pathol Oncol Res. 17:11–16. 2011. View Article : Google Scholar | |
Hayashi Y, Tsuji S, Tsujii M, Nishida T, Ishii S, Iijima H, Nakamura T, Eguchi H, Miyoshi E, Hayashi N and Kawano S: Topical implantation of mesenchymal stem cells has beneficial effects on healing of experimental colitis in rats. J Pharmacol Exp Ther. 326:523–531. 2008. View Article : Google Scholar : PubMed/NCBI | |
Nagashima K, Sawa S, Nitta T, Tsutsumi M, Okamura T, Penninger JM, Nakashima T and Takayanagi H: Identification of subepithelial mesenchymal cells that induce IgA and diversify gut microbiota. Nat Immunol. 18:675–682. 2017. View Article : Google Scholar : PubMed/NCBI | |
Levi M: Current understanding of disseminated intravascular coagulation. Br J Haematol. 124:567–576. 2004. View Article : Google Scholar : PubMed/NCBI | |
Levi M, de Jonge E and van der Poll T: Sepsis and disseminated intravascular coagulation. J Thromb Thrombolysis. 16:43–47. 2003. View Article : Google Scholar | |
Semeraro N, Ammollo CT, Semeraro F and Colucci M: Coagulopathy of acute sepsis. Semin Thromb Hemost. 41:650–658. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Wu S, Wang T, Ma Z and Liu K: Bone marrow-derived mesenchymal stem cells-mediated protection against organ dysfunction in disseminated intravascular coagulation is associated with peripheral immune responses. J Cell Biochem. 118:3184–3192. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Wu SM, Wang T, Liu K, Zhang G, Zhang XQ, Yu JH, Liu CZ and Fang CC: Pre-treatment with bone marrow-derived mesenchymal stem cells inhibits systemic intravascular coagulation and attenuates organ dysfunction in lipopolysaccharide-induced disseminated intravascular coagulation rat model. Chin Med J (Engl). 125:1753–1759. 2012.PubMed/NCBI | |
Baudry N, Starck J, Aussel C, Lund K, Aletti M, Duranteau J, Banzet S, Lataillade JJ, Vicaut E and Peltzer J: Effect of preconditioned mesenchymal stromal cells on early microvascular disturbance in a mouse sepsis model. Stem Cells Dev. 28:1595–1606. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ye Q, Qiu X, Wang J, Xu B, Su Y, Zheng C, Gui L, Yu L, Kuang H, Liu H, et al: MSCs-derived apoptotic extracellular vesicles promote muscle regeneration by inducing Pannexin 1 channel-dependent creatine release by myoblasts. Int J Oral Sci. 15:72023. View Article : Google Scholar : PubMed/NCBI | |
Sassoli C, Zecchi-Orlandini S and Formigli L: Trophic actions of bone marrow-derived mesenchymal stromal cells for muscle repair/regeneration. Cells. 1:832–850. 2012. View Article : Google Scholar : PubMed/NCBI | |
Gang EJ, Bosnakovski D, Simsek T, To K and Perlingeiro RCR: Pax3 activation promotes the differentiation of mesenchymal stem cells toward the myogenic lineage. Exp Cell Res. 314:1721–1733. 2008. View Article : Google Scholar : PubMed/NCBI | |
Dezawa M, Ishikawa H, Itokazu Y, Yoshihara T, Hoshino M, Takeda SI, Ide C and Nabeshima YI: Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science. 309:314–317. 2005. View Article : Google Scholar : PubMed/NCBI | |
Haghighipour N, Heidarian S, Shokrgozar MA and Amirizadeh N: Differential effects of cyclic uniaxial stretch on human mesenchymal stem cell into skeletal muscle cell. Cell Biol Int. 36:669–675. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ninagawa NT, Isobe E, Hirayama Y, Murakami R, Komatsu K, Nagai M, Kobayashi M, Kawabata Y and Torihashi S: Transplantated mesenchymal stem cells derived from embryonic stem cells promote muscle regeneration and accelerate functional recovery of injured skeletal muscle. Biores Open Access. 2:295–306. 2013. View Article : Google Scholar : PubMed/NCBI | |
Natsu K, Ochi M, Mochizuki Y, Hachisuka H, Yanada S and Yasunaga Y: Allogeneic bone marrow-derived mesenchymal stromal cells promote the regeneration of injured skeletal muscle without differentiation into myofibers. Tissue Eng. 10:1093–1112. 2004. View Article : Google Scholar : PubMed/NCBI | |
Winkler T, von Roth P, Matziolis G, Mehta M, Perka C and Duda GN: Dose-response relationship of mesenchymal stem cell transplantation and functional regeneration after severe skeletal muscle injury in rats. Tissue Eng Part A. 15:487–492. 2009. View Article : Google Scholar | |
Winkler T, von Roth P, Radojewski P, Urbanski A, Hahn S, Preininger B, Duda GN and Perka C: Immediate and delayed transplantation of mesenchymal stem cells improve muscle force after skeletal muscle injury in rats. J Tissue Eng Regen Med. 6(Suppl 3): s60–s67. 2012. View Article : Google Scholar : PubMed/NCBI | |
von Roth P, Duda GN, Radojewski P, Preininger B, Perka C and Winkler T: Mesenchymal stem cell therapy following muscle trauma leads to improved muscular regeneration in both male and female rats. Gend Med. 9:129–136. 2012. View Article : Google Scholar : PubMed/NCBI | |
Meregalli M, Farini A, Belicchi M, Parolini D, Cassinelli L, Razini P, Sitzia C and Torrente Y: Perspectives of stem cell therapy in Duchenne muscular dystrophy. FEBS J. 280:4251–4262. 2013. View Article : Google Scholar | |
Jiang J, Yao P, Gu Y, Xu L, Xu J and Tan H: Adult rat mesenchymal stem cells delay denervated muscle atrophy. Cell Mol Neurobiol. 32:1287–1298. 2012. View Article : Google Scholar : PubMed/NCBI | |
Merritt EK, Cannon MV, Hammers DW, Le LN, Gokhale R, Sarathy A, Song TJ, Tierney MT, Suggs LJ, Walters TJ and Farrar RP: Repair of traumatic skeletal muscle injury with bone-marrow-derived mesenchymal stem cells seeded on extracellular matrix. Tissue Eng Part A. 16:2871–2881. 2010. View Article : Google Scholar : PubMed/NCBI | |
Stana F, Vujovic M, Mayaki D, Leduc-Gaudet JP, Leblanc P, Huck L and Hussain SNA: Differential regulation of the autophagy and proteasome pathways in skeletal muscles in sepsis. Crit Care Med. 45:e971–e979. 2017. View Article : Google Scholar : PubMed/NCBI | |
Khalil R: Ubiquitin-proteasome pathway and muscle atrophy. Adv Exp Med Biol. 1088:235–248. 2018. View Article : Google Scholar : PubMed/NCBI | |
Peruchi BB, Petronilho F, Rojas HA, Constantino L, Mina F, Vuolo F, Cardoso MR, Gonçalves CL, Rezin GT, Streck EL and Dal-Pizzol F: Skeletal muscle electron transport chain dysfunction after sepsis in rats. J Surg Res. 167:e333–e338. 2011. View Article : Google Scholar : PubMed/NCBI | |
Thoma A and Lightfoot AP: NF-kB and inflammatory cytokine signalling: Role in skeletal muscle atrophy. Adv Exp Med Biol. 1088:267–279. 2018. View Article : Google Scholar : PubMed/NCBI | |
Pelosi M, De Rossi M, Barberi L and Musarò A: IL-6 impairs myogenic differentiation by downmodulation of p90RSK/eEF2 and mTOR/p70S6K axes, without affecting AKT activity. Biomed Res Int. 2014:2060262014. View Article : Google Scholar : PubMed/NCBI | |
Zhu S, Nagashima M, Khan MAS, Yasuhara S, Kaneki M and Martyn JAJ: Lack of caspase-3 attenuates immobilization-induced muscle atrophy and loss of tension generation along with mitigation of apoptosis and inflammation. Muscle Nerve. 47:711–721. 2013. View Article : Google Scholar : PubMed/NCBI | |
Okamura LH, Cordero P, Palomino J, Parraguez VH, Torres CG and Peralta OA: Myogenic differentiation potential of mesenchymal stem cells derived from fetal bovine bone marrow. Anim Biotechnol. 29:1–11. 2018. View Article : Google Scholar | |
Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, et al: Bone marrow cells regenerate infarcted myocardium. Nature. 410:701–705. 2001. View Article : Google Scholar : PubMed/NCBI | |
Egusa H, Kobayashi M, Matsumoto T, Sasaki J, Uraguchi S and Yatani H: Application of cyclic strain for accelerated skeletal myogenic differentiation of mouse bone marrow-derived mesenchymal stromal cells with cell alignment. Tissue Eng Part A. 19:770–782. 2013. View Article : Google Scholar | |
Drost AC, Weng S, Feil G, Schäfer J, Baumann S, Kanz L, Sievert KD, Stenzl A and Möhle R: In vitro myogenic differentiation of human bone marrow-derived mesenchymal stem cells as a potential treatment for urethral sphincter muscle repair. Ann N Y Acad Sci. 1176:135–143. 2009. View Article : Google Scholar : PubMed/NCBI | |
Maeda Y, Yonemochi Y, Nakajyo Y, Hidaka H, Ikeda T and Ando Y: CXCL12 and osteopontin from bone marrow-derived mesenchymal stromal cells improve muscle regeneration. Sci Rep. 7:33052017. View Article : Google Scholar : PubMed/NCBI | |
Wakitani S, Saito T and Caplan AI: Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve. 18:1417–1426. 1995. View Article : Google Scholar : PubMed/NCBI | |
Meligy FY, Shigemura K, Behnsawy HM, Fujisawa M, Kawabata M and Shirakawa T: The efficiency of in vitro isolation and myogenic differentiation of MSCs derived from adipose connective tissue, bone marrow, and skeletal muscle tissue. In Vitro Cell Dev Biol Anim. 48:203–215. 2012. View Article : Google Scholar : PubMed/NCBI | |
Asakura A: Stem cells in adult skeletal muscle. rends Cardiovasc Med. 13:123–128. 2003. View Article : Google Scholar | |
Dumont NA, Wang YX and Rudnicki MA: Intrinsic and extrinsic mechanisms regulating satellite cell function. Development. 142:1572–1581. 2015. View Article : Google Scholar : PubMed/NCBI | |
Thomas K, Engler AJ and Meyer GA: Extracellular matrix regulation in the muscle satellite cell niche. Connect Tissue Res. 56:1–8. 2015. View Article : Google Scholar : | |
Tonkin J, Temmerman L, Sampson RD, Gallego-Colon E, Barberi L, Bilbao D, Schneider MD, Musarò A and Rosenthal N: Monocyte/macrophage-derived IGF-1 orchestrates murine skeletal muscle regeneration and modulates autocrine polarization. Mol Ther. 23:1189–1200. 2015. View Article : Google Scholar : PubMed/NCBI | |
Karp JM and Teo GSL: Mesenchymal stem cell homing: The devil is in the details. Cell Stem Cell. 4:206–216. 2009. View Article : Google Scholar : PubMed/NCBI | |
Zhou Q, Yang C and Yang P: The promotional effect of mesenchymal stem cell homing on bone tissue regeneration. Curr Stem Cell Res Ther. 12:365–376. 2017. View Article : Google Scholar | |
Ringe J, Strassburg S, Neumann K, Endres M, Notter M, Burmester GR, Kaps C and Sittinger M: Towards in situ tissue repair: Human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2. J Cell Biochem. 101:135–146. 2007. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Li Y, Chen W, Han N, Li K, Guo R, Liu Z and Xiao Y: Enhanced recruitment and hematopoietic reconstitution of bone marrow-derived mesenchymal stem cells in bone marrow failure by the SDF-1/CXCR4. J Tissue Eng Regen Med. 14:1250–1260. 2020.PubMed/NCBI | |
Moll NM and Ransohoff RM: CXCL12 and CXCR4 in bone marrow physiology. Expert Rev Hematol. 3:315–322. 2010. View Article : Google Scholar : PubMed/NCBI | |
Pozzobon T, Goldoni G, Viola A and Molon B: CXCR4 signaling in health and disease. Immunol Lett. 177:6–15. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mousavi A: CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett. 217:91–115. 2020. View Article : Google Scholar | |
Guo J, Zhang H, Xiao J, Wu J, Ye Y, Li Z, Zou Y and Li X: Monocyte chemotactic protein-1 promotes the myocardial homing of mesenchymal stem cells in dilated cardiomyopathy. Int J Mol Sci. 14:8164–8178. 2013. View Article : Google Scholar : PubMed/NCBI | |
Schenk S, Mal N, Finan A, Zhang M, Kiedrowski M, Popovic Z, McCarthy PM and Penn MS: Monocyte chemotactic protein-3 is a myocardial mesenchymal stem cell homing factor. Stem Cells. 25:245–251. 2007. View Article : Google Scholar | |
Ahn SY, Park WS, Kim YE, Sung DK, Sung SI, Ahn JY and Chang YS: Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Exp Mol Med. 50:1–12. 2018. | |
Shams S, Mohsin S, Nasir GA, Khan M and Khan SN: Mesenchymal stem cells pretreated with HGF and FGF4 can reduce liver fibrosis in mice. Stem Cells Int. 2015:7472452015. View Article : Google Scholar : PubMed/NCBI | |
Kumar S and Ponnazhagan S: Bone homing of mesenchymal stem cells by ectopic alpha 4 integrin expression. FASEB J. 21:3917–3927. 2007. View Article : Google Scholar : PubMed/NCBI | |
Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G and Mavilio F: Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 279:1528–1530. 1998. View Article : Google Scholar : PubMed/NCBI | |
Winkler T, von Roth P, Schuman MR, Sieland K, Stoltenburg-Didinger G, Taupitz M, Perka C, Duda GN and Matziolis G: In vivo visualization of locally transplanted mesenchymal stem cells in the severely injured muscle in rats. Tissue Eng Part A. 14:1149–1160. 2008. View Article : Google Scholar : PubMed/NCBI | |
Le Blanc K and Mougiakakos D: Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol. 12:383–396. 2012. View Article : Google Scholar : PubMed/NCBI | |
da Justa Pinheiro CH, de Queiroz JC, Guimarães-Ferreira L, Vitzel KF, Nachbar RT, de Sousa LGO, de Souza AL Jr, Nunes MT and Curi R: Local injections of adipose-derived mesenchymal stem cells modulate inflammation and increase angiogenesis ameliorating the dystrophic phenotype in dystrophin-deficient skeletal muscle. Stem Cell Rev Rep. 8:363–374. 2012. View Article : Google Scholar | |
Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, Robey PG, Leelahavanichkul K, Koller BH, Brown JM, et al: Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 15:42–49. 2009. View Article : Google Scholar | |
Chen L, Tredget EE, Wu PY and Wu Y: Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 3:e18862008. View Article : Google Scholar : PubMed/NCBI | |
Bencze M, Negroni E, Vallese D, Yacoub-Youssef H, Chaouch S, Wolff A, Aamiri A, Di Santo JP, Chazaud B, Butler-Browne G, et al: Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation. Mol Ther. 20:2168–2179. 2012. View Article : Google Scholar : PubMed/NCBI | |
Waterman RS, Tomchuck SL, Henkle SL and Betancourt AM: A new mesenchymal stem cell (MSC) paradigm: Polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS One. 5:e100882010. View Article : Google Scholar : PubMed/NCBI | |
de Couto G, Gallet R, Cambier L, Jaghatspanyan E, Makkar N, Dawkins JF, Berman BP and Marbán E: Exosomal MicroRNA transfer into macrophages mediates cellular postconditioning. Circulation. 136:200–214. 2017. View Article : Google Scholar : PubMed/NCBI | |
Xu R, Zhang F, Chai R, Zhou W, Hu M, Liu B, Chen X, Liu M, Xu Q, Liu N and Liu S: Exosomes derived from pro-inflammatory bone marrow-derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization. J Cell Mol Med. 23:7617–7631. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, Gao L, Xie J and Xu B: Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res. 115:1205–1216. 2019. View Article : Google Scholar : PubMed/NCBI | |
Simovic Markovic B, Gazdic M, Arsenijevic A, Jovicic N, Jeremic J, Djonov V, Arsenijevic N, Lukic ML and Volarevic V: Mesenchymal stem cells attenuate cisplatin-induced nephrotoxicity in iNOS-dependent manner. Stem Cells Int. 2017:13153782017. View Article : Google Scholar : PubMed/NCBI | |
Sassoli C, Pini A, Chellini F, Mazzanti B, Nistri S, Nosi D, Saccardi R, Quercioli F, Zecchi-Orlandini S and Formigli L: Bone marrow mesenchymal stromal cells stimulate skeletal myoblast proliferation through the paracrine release of VEGF. PLoS One. 7:e375122012. View Article : Google Scholar : PubMed/NCBI | |
Murphy MB, Moncivais K and Caplan AI: Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 45:e542013. View Article : Google Scholar : PubMed/NCBI | |
Sassoli C, Frati A, Tani A, Anderloni G, Pierucci F, Matteini F, Chellini F, Zecchi Orlandini S, Formigli L and Meacci E: Mesenchymal stromal cell secreted sphingosine 1-phosphate (S1P) exerts a stimulatory effect on skeletal myoblast proliferation. PLoS One. 9:e1086622014. View Article : Google Scholar : PubMed/NCBI | |
Nakamura Y, Miyaki S, Ishitobi H, Matsuyama S, Nakasa T, Kamei N, Akimoto T, Higashi Y and Ochi M: Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration. FEBS Lett. 589:1257–1265. 2015. View Article : Google Scholar : PubMed/NCBI | |
Leoni G, Neumann PA, Kamaly N, Quiros M, Nishio H, Jones HR, Sumagin R, Hilgarth RS, Alam A, Fredman G, et al: Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J Clin Invest. 125:1215–1227. 2015. View Article : Google Scholar : PubMed/NCBI | |
Bizzarro V, Petrella A and Parente L: Annexin A1: Novel roles in skeletal muscle biology. J Cell Physiol. 227:3007–3015. 2012. View Article : Google Scholar : PubMed/NCBI | |
Kim S, Lee MJ, Choi JY, Park DH, Kwak HB, Moon S, Koh JW, Shin HK, Ryu JK, Park CS, et al: Roles of exosome-like vesicles released from inflammatory C2C12 myotubes: Regulation of myocyte differentiation and myokine expression. Cell Physiol Biochem. 48:1829–1842. 2018. View Article : Google Scholar : PubMed/NCBI | |
Choi JS, Yoon HI, Lee KS, Choi YC, Yang SH, Kim IS and Cho YW: Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J Control Release. 222:107–115. 2016. View Article : Google Scholar | |
Forterre A, Jalabert A, Berger E, Baudet M, Chikh K, Errazuriz E, De Larichaudy J, Chanon S, Weiss-Gayet M, Hesse AM, et al: Proteomic analysis of C2C12 myoblast and myotube exosome-like vesicles: A new paradigm for myoblast-myotube cross talk? PLoS One. 9:e841532014. View Article : Google Scholar : PubMed/NCBI | |
Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, Bornhäuser M and Werner C: Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells. 22:377–384. 2004. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Li Y, Yang M, Zou Y, Liu H, Liang Z, Yin Y, Niu G, Yan Z and Zhang B: Efficient differentiation of bone marrow mesenchymal stem cells into endothelial cells in vitro. Eur J Vasc Endovasc Surg. 55:257–265. 2018. View Article : Google Scholar | |
Lu W, Xiu X, Zhao Y and Gui M: Improved proliferation and differentiation of bone marrow mesenchymal stem cells into vascular endothelial cells with sphingosine 1-phosphate. Transplant Proc. 47:2035–2040. 2015. View Article : Google Scholar : PubMed/NCBI | |
Grote K, Petri M, Liu C, Jehn P, Spalthoff S, Kokemüller H, Luchtefeld M, Tschernig T, Krettek C, Haasper C and Jagodzinski M: Toll-like receptor 2/6-dependent stimulation of mesenchymal stem cells promotes angiogenesis by paracrine factors. Eur Cell Mater. 26:66–79. 2013. View Article : Google Scholar : PubMed/NCBI | |
Leroux L, Descamps B, Tojais NF, Séguy B, Oses P, Moreau C, Daret D, Ivanovic Z, Boiron JM, Lamazière JM, et al: Hypoxia preconditioned mesenchymal stem cells improve vascular and skeletal muscle fiber regeneration after ischemia through a Wnt4-dependent pathway. Mol Ther. 18:1545–1552. 2010. View Article : Google Scholar : PubMed/NCBI | |
Berebichez-Fridman R and Montero-Olvera PR: Sources and clinical applications of mesenchymal stem cells: State-of-the-art review. Sultan Qaboos Univ Med J. 18:e264–e277. 2018. View Article : Google Scholar | |
Mushahary D, Spittler A, Kasper C, Weber V and Charwat V: Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A. 93:19–31. 2018. View Article : Google Scholar | |
Spath L, Rotilio V, Alessandrini M, Gambara G, De Angelis L, Mancini M, Mitsiadis TA, Vivarelli E, Naro F, Filippini A and Papaccio G: Explant-derived human dental pulp stem cells enhance differentiation and proliferation potentials. J Cell Mol Med. 14:1635–1644. 2010. View Article : Google Scholar | |
Ueda N, Atsuta I, Ayukawa Y, Yamaza T, Furuhashi A, Narimatsu I, Matsuura Y, Kondo R, Watanabe Y, Zhang X and Koyano K: Novel application method for mesenchymal stem cell therapy utilizing its attractant-responsive accumulation property. Appl Sci. 9:49082019. View Article : Google Scholar | |
Castelo-Branco MTL, Soares IDO, Lopes DV, Buongusto F, Martinusso CA, do Rosario A Jr, Souza SA, Gutfilen B, Fonseca LM, Elia C, et al: Intraperitoneal but not intravenous cryopreserved mesenchymal stromal cells home to the inflamed colon and ameliorate experimental colitis. PLoS One. 7:e333602012. View Article : Google Scholar : PubMed/NCBI | |
Gonçalves Fda C, Schneider N, Pinto FO, Meyer FS, Visioli F, Pfaffenseller B, Lopez PL, Passos EP, Cirne-Lima EO, Meurer L and Paz AH: Intravenous vs intraperitoneal mesenchymal stem cells administration: What is the best route for treating experimental colitis? World J Gastroenterol. 20:18228–18239. 2014. View Article : Google Scholar | |
Roux C, Saviane G, Pini J, Belaïd N, Dhib G, Voha C, Ibáñez L, Boutin A, Mazure NM, Wakkach A, et al: Immunosuppressive mesenchymal stromal cells derived from human-induced pluripotent stem cells induce human regulatory T cells in vitro and in vivo. Front Immunol. 8:19912018. View Article : Google Scholar : PubMed/NCBI | |
Braid LR, Wood CA, Wiese DM and Ford BN: Intramuscular administration potentiates extended dwell time of mesenchymal stromal cells compared to other routes. Cytotherapy. 20:232–244. 2018. View Article : Google Scholar | |
Rangarajan A, Hong SJ, Gifford A and Weinberg RA: Erratum to species- and cell type-specific requirements for cellular transformation [Cancer Cell 6, (2004) 171-183]. Cancer Cell. 24:394–398. 2013. View Article : Google Scholar | |
Wang Y, Han ZB, Song YP and Han ZC: Safety of mesenchymal stem cells for clinical application. Stem Cells Int. 2012:6520342012. View Article : Google Scholar : PubMed/NCBI | |
Golpanian S, DiFede DL, Khan A, Schulman IH, Landin AM, Tompkins BA, Heldman AW, Miki R, Goldstein BJ, Mushtaq M, et al: Allogeneic human mesenchymal stem cell infusions for aging frailty. J Gerontol A Biol Sci Med Sci. 72:1505–1512. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tompkins BA, DiFede DL, Khan A, Landin AM, Schulman IH, Pujol MV, Heldman AW, Miki R, Goldschmidt-Clermont PJ, Goldstein BJ, et al: Allogeneic mesenchymal stem cells ameliorate aging frailty: A phase II randomized, double-blind, placebo-controlled clinical trial. J Gerontol A Biol Sci Med Sci. 72:1513–1522. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kabat M, Bobkov I, Kumar S and Grumet M: Trends in mesenchymal stem cell clinical trials 2004-2018: Is efficacy optimal in a narrow dose range? Stem Cells Transl Med. 9:17–27. 2020. View Article : Google Scholar | |
Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al: The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 315:801–810. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mankowski RT, Laitano O, Darden D, Kelly L, Munley J, Loftus TJ, Mohr AM, Efron PA and Thomas RM: Sepsis-Induced myopathy and gut microbiome dysbiosis: Mechanistic links and therapeutic targets. Shock. 57:15–23. 2022. View Article : Google Scholar : | |
Liu Y, Wang D, Li T, Yang F, Li Z, Bai X and Wang Y: The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol. 13:10357092022. View Article : Google Scholar : PubMed/NCBI | |
Liu D, Huang SY, Sun JH, Zhang HC, Cai QL, Gao C, Li L, Cao J, Xu F, Zhou Y, et al: Sepsis-induced immunosuppression: Mechanisms, diagnosis and current treatment options. Mil Med Res. 9:562022.PubMed/NCBI | |
Kramer CL: Intensive care unit-acquired weakness. Neurol Clin. 35:723–736. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fu X, Liu G, Halim A, Ju Y, Luo Q and Song AG: Mesenchymal stem cell migration and tissue repair. Cells. 8:7842019. View Article : Google Scholar : PubMed/NCBI | |
Song J, Liu J, Cui C, Hu H, Zang N, Yang M, Yang J, Zou Y, Li J, Wang L, et al: Mesenchymal stromal cells ameliorate diabetes-induced muscle atrophy through exosomes by enhancing AMPK/ULK1-mediated autophagy. J Cachexia Sarcopenia Muscle. 14:915–929. 2023. View Article : Google Scholar : PubMed/NCBI | |
Abrigo J, Rivera JC, Aravena J, Cabrera D, Simon F, Ezquer F, Ezquer M and Cabello-Verrugio C: High fat diet-induced skeletal muscle wasting is decreased by mesenchymal stem cells administration: Implications on oxidative stress, ubiquitin proteasome pathway activation, and myonuclear apoptosis. Oxid Med Cell Longev. 2016:90478212016. View Article : Google Scholar : PubMed/NCBI |