International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Acute kidney injury (AKI) is a prevalent clinical condition characterized by a rapid elevation in serum creatinine levels and/or reduced urine output, resulting from various factors such as renal ischemia, sepsis, or drug toxicity (1,2). Global data suggests ~13.3 million individuals experience AKI annually, with a rising trend (3). In neonates, AKI is observed in one-third of infants and associated with increased risk of mortality (4). Besides its acute effects, AKI can advance to chronic kidney disease (CKD) and end-stage kidney disease, necessitating kidney transplantation or replacement therapy (5-9). These outcomes not only diminish patients' quality of life but also impose substantial financial strain on families and strain public healthcare systems. Currently, AKI management primarily involves supportive and preventive measures, lacking targeted pharmacological treatments (10,11). Moreover, due to AKI often co-occurring with conditions such as heart failure, hypoxic ischemic encephalopathy and sepsis, patients commonly receive multiple medications concurrently, heightening the risk of adverse drug interactions. Therefore, there is a critical demand for developing safer and more efficient approaches to manage and impede AKI progression.
Mesenchymal stem cells (MSCs) have garnered considerable attention for their potential in treating AKI due to their multipotency, robust self-renewal and ability to differentiate into various cell types (12,13). Derived from sources such as bone marrow, adipose tissue and umbilical cord blood, MSCs secrete a diverse array of bioactive molecules, including cytokines, chemokines, growth factors and microvesicles, which collectively promote cell proliferation, inhibit apoptosis, enhance angiogenesis, facilitate tissue repair, modulate immune responses and prevent fibrosis (14-18). In addition to these paracrine effects, MSC-derived extracellular vesicles serve as promising carriers for drug delivery, further expanding their therapeutic utility (19,20). Previous studies have also uncovered a novel mechanism whereby MSCs transfer healthy mitochondria to injured cells via tunneling nanotubes (21-23), thereby boosting cellular energy metabolism and contributing to tissue repair (24,25). The present review summarized the methods by which MSCs repair damaged cells in AKI (Fig. 1). Despite these promising attributes, clinical applications of MSCs in AKI are challenged by issues such as low cell retention, limited survival and suboptimal secretion of bioactive factors (26,27). Consequently, researchers are investigating innovative strategies, such as preconditioning techniques, optimized cell culture protocols, improved delivery methods, to overcome these limitations and enhance the efficacy of MSC-based therapies for AKI (28,29). The present review summarized these novel strategies and their underlying mechanisms in treating AKI.
Recent advances in MSCs biology and bioengineering have led to new strategies to overcome limitations of MSC-based therapies. As shown in Fig. 2, both extracellular and intracellular strategies have been employed to enhance the therapeutic potential of MSCs. Extracellular approaches include physical preconditioning (such as hypoxic culture and mechanical stimulation), chemical or drug-based preconditioning, biological stimulation with cytokines or hormones and cell-to-cell co-culture. Interestingly, 3D culture (hydrogels and spheroid formation) help recreate a more physiologically relevant microenvironment. On the intracellular level, genetic modification can further optimize MSCs functionality. Moreover, optimizing MSCs administration and selecting the suitable source of MSCs are also efficacies strategies.
Unlike the standard cell culture environment with regular oxygen levels (21%), transplanted MSCs face a hypoxic microenvironment in injured tissues. Culturing MSCs under hypoxic conditions enhance the function of MSCs by increasing proliferation, survival, homing, differentiation and paracrine activities (30-33), which may enhance their therapeutic potential in treatment of AKI.
Hypoxic preconditioning of MSCs has shown promising results in the treatment of AKI. Various oxygen concentrations have been tested for preconditioning MSCs, with different sources of MSCs and animal models. MSCs preconditioned under low oxygen conditions (1% O2, 5% O2 and CoCl2) contribute markedly to kidney function restoration; 1% O2-preconditioned human adipose-derived MSCs (hADMSCs) in rats with ischemia-reperfusion injury (IRI) showed improvements in apoptosis, enhanced anti-oxidative capacity and increased vascularization, leading to overall renal function improvement (34). Furthermore, 1% O2-preconditioned hADMSCs showed enhanced immunomodulatory effects, improved angiogenesis and reduced oxidative stress, leading to improved renal preservation (35). Additionally, 1% O2-preconditioned human bone marrow MSCs led to a marked renal function restoration in IRI rat models (36).
Notably, higher oxygen concentrations, such as 5% O2, also provided beneficial effects. A study reported that hypoxic preconditioning of human umbilical cord MSCs (hUCMSCs) with 5% O2 in a gentamicin-induced acute renal failure rat model led to increased expression of hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), integrins and stromal-derived factor-1, all of which ameliorated renal function (37). Moreover, a study showed that 5% O2-preconditioned AD-MSCs in mice with IRI demonstrated an increase in EVs particle number and protein concentrations, which reduced oxidative reactions and protected kidney function (38). Similarly, rat BMSCs exposed to CoCl2, which mimics the hypoxic environment, showed enhanced hypoxia-inducible factor 1 (HIF-1) and C-X-C chemokine receptor type 4 (CXCR4) expression, improved migration and prolonged retention, contributing to improved kidney preservation in IRI (39).
These findings highlight that hypoxic preconditioning is a potent strategy for optimizing MSCs-based therapies in AKI. The enhancement of key pathways such as angiogenesis, anti-oxidative mechanisms and immunomodulatory effects, along with improved cellular retention and migration, makes hypoxia preconditioned MSCs a promising candidate for clinical applications in AKI treatment. However, it is important to note that prolonged or excessive exposure to hypoxia may have a negative impact on cellular metabolism and proliferation, leading to unstable cell function or inducing apoptosis. The present review summarized the role of hypoxia pretreated MSCs in the preclinical model of AKI (Table I) (34-41).
Chemical and drugs preconditioningTo identify effective strategies for optimizing MSCs, researchers have explored chemical preconditioning approaches. These agents are primarily characterized by strong antioxidant, anti-inflammatory and tissue-repair properties. The aim is to protect the cellular membrane structure, improve the microenvironment of MSCs and enhance their stability within this environment. Recent studies which reported on drugs and chemical preconditioned MSCs in AKI models were summarized in Table II (42-46).
Chlorzoxane (CZ)CZ is a powerful muscle relaxant (47). As an FDA-approved drug, CZ has been shown to enhance the expression of anti-inflammatory cytokines, chemokines and critical immunoregulatory enzymes indoleamine 2,3-dioxygenase by promoting Forkhead box protein O3 phosphorylation. This process induces MSCs to adopt an anti-inflammatory phenotype, thereby strengthening their immunosuppressive capacity without increasing immunogenicity (44). In vitro, CZ markedly reduces T-cell activation and proliferation, substantially boosting the immunosuppressive function of hUCMSCs. In Thy1.1 antibody-induced AKI animal models, CZ-pretreated hUCMSCs effectively attenuated renal inflammation and decreased fibrinoid necrosis of the glomeruli, indicating a superior protective effect against kidney injury (44).
Atorvastatin (Ator)Ator has been shown to possess anti-apoptotic, antioxidant and anti-inflammatory properties (48). Ator improves the microenvironment not only by synergistically enhancing the inherent effects of BMSCs but also by introducing additional therapeutic benefits to BMSCs transplantation (49). Cai et al (45) proposed a combined therapeutic strategy for IRI in a rat model. Rats were administered Ator (lipitor; 5 mg/kg/day by gavage) starting 3 days before MSCs transplantation and continuing until sacrifice. Immediately following IRI, BMSCs were delivered via the left carotid artery. The results indicated that the Ator + MSCs combination therapy could synergistically promote post-ischemic renal functional and morphological recovery. These benefits may stem from the statin-mediated suppression of oxidative stress and inflammation in the injured kidney by inhibiting TLR4 signaling, thereby creating a more favorable environment for engrafted MSCs (45).
Darbepoetin (DPO)The erythropoietin receptor (EpoR) is not only expressed on hematopoietic cells but has also been reported in specific renal cell types, including tubular epithelial cells and mesangial cells (50). Growing evidence from animal studies indicates that erythropoietin (EPO) directly acts on these renal cells to mitigate AKI, a protective effect attributed to its antiapoptotic, antioxidative, anti-inflammatory properties and angiogenesis (51). DPO, a genetically modified EPO analog, exhibits a prolonged half-life and enhanced stability, enabling reduced dosing frequency while retaining biological activity comparable to native EPO (52). Altun et al (46), rats treated ischemia reperfusion injury (IRI)-induced AKI with either DPO, MSCs, or both (MSCs and DPO concomitantly). The results demonstrated that the combined administration of MSCs and DPO provided superior renoprotection and mitigated tissue damage, although the underlying mechanisms require further investigation.
Vitamin EVitamin E, a fat-soluble vitamin with antioxidant properties, can reduce reactive oxygen species (ROS) and prevent cell death, thus benefiting AKI therapy pathways (53-55). The therapeutic effects of combining BMSCs with Vitamin E in a gentamicin-induced AKI model have been shown to involve promoting tubular cell proliferation, inhibiting apoptosis and improving both renal function and pathological abnormalities (42). Another study (43) confirmed that co-administration of UCMSCs and Vitamin E markedly suppressed renal inflammation by modulating inflammatory cytokines [interleukin (IL)-lβ, tumor necrosis factor (TNF)-α and IL-10] in the AKI microenvironment, thereby reducing kidney injury. Moreover, this combined treatment achieved more favorable outcomes than using UCMSCs or Vitamin E alone.
OthersResearch has shown that ADMSCs from patients with stage 5 CKD, pretreated with metformin, can slow down the aging of CKD-MSCs (56). Subsequently, the study showed that metformin pretreated ADMSCs markedly attenuate renal inflammation and fibrosis in a CKD mouse model induced by unilateral ureteral obstruction (UUO). This finding suggests that even MSCs derived from CKD patients, can be functionally optimized through agents such as metformin and developed as patient-derived, autologous MSC-based therapeutic products for CKD, offering a novel approach to personalized stem cell therapy (56). Hu et al (57) demonstrated that CA-pretreated BMSCs from mice could alleviate renal fibrosis by inhibiting the TGF-β1/TNF-α/TNFR1 pathway. However, there have been no published studies combining these two drugs with MSCs for the treatment of AKI and further research is needed.
Overall, multiple studies have confirmed that chemical drug preconditioning effectively enhances the antioxidative and anti-inflammatory abilities of MSCs. However, these drugs may have certain effects on the kidneys or other organs. Therefore, the potential side effects and long-term impacts of drug preconditioning need to be further evaluated through additional research.
Biological compounds preconditioningMSCs typically exhibit low immunogenicity, thereby rarely triggering strong rejection responses in allogeneic transplantation (58,59). Moreover, research has demonstrated that the immunomodulatory and immunosuppressive capacities of MSCs can be further activated when exposed to inflammatory stimuli or residing within an inflammatory microenvironment (60-62). Numerous studies have shown that both exogenous and endogenous pro-inflammatory stimuli, such as interferon gamma (IFN-γ) (63), TNF-α (64,65), IL-17 (66), IL-1β (67), Toll-like receptor (TLR) (68) and TLR ligands (69), can activate or precondition MSCs, enhancing their secretion of immunoregulatory factors and their therapeutic potential. Current preconditioning studies targeting AKI have primarily focused on IFN-γ, IL-17 and IL-1β. While several reports indicate that TNF-α and TLR4 ligand-preconditioned MSCs can boost therapeutic efficacy in other disease models (65,70-72), there is relatively little published literature regarding the efficacy of TNF-α or TLR4-induced MSCs preconditioning in improving AKI outcomes. Additionally, the combined administration of MSCs with low doses of hormones has demonstrated significant renoprotective effects in various animal models (73,74). Biological compound preconditioning MSCs may offer novel insights into enhancing their therapeutic applications in AKI via hormones and cytokines. The recent findings were summarized in Table III (75-81).
IL-17A modulates MSCs immunoregulatory functions without affecting MHC expression (66,82,83), has been shown to enhance the therapeutic potential of BMSCs in mouse models of AKI (76). Mechanistically, IL-17A induces the expansion of regulatory T cells (Tregs), thereby reinforcing immunosuppression and repairing renal function. Conversely, the absence of Tregs diminishes T-cell suppression and negates the beneficial effects of IL-17A preconditioning (76).
IFN-γIFN-γ has been employed to pretreat MSCs (63,84). IFN-γ-preconditioned BMSCs administered via the abdominal aorta in rat models of IRI and UUO exhibited marked reductions in immune cell infiltration and renal fibrosis (78). In mechanism, IFN-γ-preconditioned BMSCs upregulated PGE2 related responses, which induced the polarization of immunosuppressive CD163+CD206+ macrophages in the local renal microenvironment. This immunomodulatory shift markedly attenuated inflammatory cell infiltration, restored immune homeostasis and ultimately protected against IRI-induced AKI (78).
IL-1βIL-1β has been used to pretreat MSCs and has been shown to enhance MSCs immune regulation functions. Intravenous injection of IL-1β pretreated MSCs in hemorrhagic shock rats resulted in decreased plasma creatinine, urea nitrogen, cystatin C and kidney injury molecule kidney injury molecule 1 levels on renal tissue sections. In vitro experiments demonstrated that IL-1β-pretreated MSCs reduced systemic cytokines (IL-1α, IL-6 and IL-10) of monocytes and granulocytes, as well as CD80 and CD86, inhibiting activation of PD-1/PDL-1 axis. This suggests that IL-1β enhances MSCs efficiency by promoting its immunomodulatory activity (75).
Melatonin, a neurohormone primarily secreted by the pineal gland, plays a crucial role in regulating circadian rhythms (85-89). It is recognized as an effective free radical scavenger with protective effects on various dysfunctional organs, including the kidney (73,74). Given that inflammation and oxidative stress are key contributors to the impaired function of transplanted MSCs in disease environments, preconditioning MSCs with melatonin may prove beneficial (73,90,91). Animal studies in models of lung injury and cerebral and limb ischemia have demonstrated that melatonin preconditioning enhances the therapeutic outcomes of MSCs (92-96). Notably, melatonin not only exerts potent antioxidant effects by reducing free radical production, but also its receptors, MT1 and MT2, widely expressed on MSCs, may also modulate cell fate via receptor-dependent mechanisms (97-99). AKI caused by sepsis is a particularly severe subtype with high morbidity and mortality. Chen et al (77). compared melatonin-preconditioned ADMSCs with ADMSCs alone in a cecal ligation and puncture model, finding that the combined therapy produced superior anti-inflammatory, antioxidant, anti-apoptotic and anti-fibrotic responses, as well as lower serum creatinine levels and reduced renal damage on histological examination. In vitro, melatonin preconditioning markedly enhances ADMSCs proliferation and upregulates key survival and antioxidant proteins such as phosphorylated Erk1/2, Akt, SOD-1 and heme oxygenase-1 (HO-1) (77). The conditioned medium from these cells further promoted proliferation, migration and anti-apoptotic effects in cisplatin-exposed human renal epithelial cells (80). Moreover, Saberi et al (100) demonstrated that pretreatment of BMSCs with melatonin enhanced their homing to the injured kidney, reduced renal cell apoptosis, decreased the expression of TNF-α, α-SMA and TGF-β1 and increased E-cadherin levels in UUO. Ultimately, this approach improved the integrity of the tubular basement membrane, ameliorated tissue architecture and effectively mitigated renal fibrosis.
Erythropoietin (EPO)EPO is an endogenous hormone primarily secreted by the kidney that regulates red blood cell production (101). It stimulates the proliferation and differentiation of hematopoietic stem cells (HSCs) in the bone marrow through signaling via the EpoR (102). Notably, both BMSCs and HSCs express homologous EpoR. EPO has shown therapeutic potential in various conditions such as myocardial infarction (103), cerebral ischemia (104), chronic heart failure (105) and kidney injury (101,106) through its anti-inflammatory and antioxidant effects. Zhou et al (107). demonstrated that 48-h EPO pretreatment markedly enhances the proliferation rate, cytoskeletal reorganization, migration capacity and expression of the chemokine receptor CXCR4 in BMSCs, thereby improving their homing ability and cellular functionality in vitro. Subsequently, compared with untreated BMSCs, EPO-preconditioned BMSCs not only more effectively restored renal function but also markedly reduced interstitial lymphocyte infiltration, tubular swelling, necrosis and interstitial fibrosis in a cyclosporine A-induced nephrotoxicity rat model (107). Zhou et al (81) further reported that EPO pretreatment upregulated the expression of SIRT1 and the anti-apoptotic protein Bcl-2 while downregulating the pro-apoptotic protein p53 in BMSCs, thereby enhancing their survival rate and anti-apoptotic capacity. The authors observed that in a rat AKI model, GFP-labeled BMSCs infused after 24 h primarily accumulated in the lungs, whereas EPO pretreatment reduced pulmonary retention and increased their distribution to the injured kidneys. Moreover, AKI rats treated with EPO-preconditioned BMSCs exhibited markedly lower serum levels of IL-1β and TNF-α, along with elevated IL-10 levels and demonstrated more pronounced improvements in renal function (81). These findings indicate that EPO pretreatment not only enhances the survival and anti-apoptotic capabilities of BMSCs but also improves their homing and distribution in vivo, thereby markedly augmenting their therapeutic efficacy in AKI.
RelaxinRelaxin is primarily secreted by the corpus luteum during pregnancy, with minor expression in the brain, heart and kidneys (108). It plays a key role in mediating adaptive hemodynamic changes, including increased cardiac output, enhanced renal blood flow and improved arterial compliance (109-113). Several investigations have explored relaxin preconditioning of MSCs (114,115). Although direct evidence of relaxin preconditioned MSCs protecting against AKI is currently lacking, combined therapy has been shown to mitigate fibrosis progression in a UUO model in normotensive mice (116,117). Badawi et al (117) developed a novel combinatorial approach for CKD patients using synthetic relaxin (Serelaxin) with BMSCs to restore and enhance endothelial progenitor cell populations. While BMSCs alone have demonstrated blood pressure reduction and anti-fibrotic effects comparable to perindopril, serelaxin monotherapy markedly decreases tubular injury and moderately reduces renal fibrosis (114). These findings suggest that further research is needed to fully elucidate the renoprotective potential of relaxin preconditioned MSCs in the setting of AKI.
In summarily, biological preconditioning strategies enhance the therapeutic effects in AKI by activating the immunomodulatory function of MSCs through cytokines or hormones. However, its main drawback is the potential risk of overstimulation, leading to unwanted side effects, such as excessive inflammation or immune modulation, which could complicate therapeutic use.
Three-dimensional (3D) cultureConventionally, cell cultures are primarily performed on flat, two-dimensional substrates. Nevertheless, in a two-dimensional setup, the interplay among cells and between cells and the extracellular matrix (ECM) is not entirely consistent with that in vivo. In order to more closely mimic the in vivo microenvironment and improve cellular function as well as the physiological relevance of experimental data, MSCs 3D cell culture methods have been developed and applied in the study of AKI (Table IV) (118-125).
Hydrogel-focusedHydrogels are regarded as one of the most promising options for MSCs 3D culture and delivery (126,127). Hydrogels, formed through chemical or physical crosslinking, are 3D porous polymer networks with high water content and tunable chemical and physical attributes (126,128). Owing to their substantial water content and porous structure, hydrogels facilitate the diffusion of nutrients and metabolites within the network (129). In this way, hydrogels can serve as an artificial ECM around the cells, providing the necessary conditions for cell-to-matrix and cell-to-cell interactions, thus influencing MSCs behavior and function (130). At present, hydrogels have been explored to replicate the native microenvironment of cells in vivo and they are categorized by source or fabrication approach into natural hydrogels, synthetic hydrogels and hybrid hydrogels (130). Notably, the effectiveness of hydrogels as scaffolds to support cellular growth and function has been confirmed in various preclinical AKI models (119,121,131).
Natural hydrogelsHydrogels made from natural materials have gained significant interest in recent years because of their excellent biocompatibility, biodegradability and eco-friendly nature (132). Common natural polymers utilized are agarose, alginate, chitosan, collagen, hyaluronic acid, gelatin and fibrin (133-136). These materials can be categorized as either polysaccharide-based or protein-based hydrogels (137).
AlginateAlginate, a linear hydrophilic polysaccharide from brown algae and specific bacteria, comprises β-d-mannuronic acid and α-l-guluronic acid (135). This biopolymer, soluble and cost-effective, with high biocompatibility and appropriate rheological properties, is widely used in 3D bioprinting (138). Before its bioink application, alginate was extensively researched in regenerative medicine and tissue engineering for MSCs culture and delivery (139-141). It further facilitates the regulated secretion of paracrine factors derived from MSCs (142,143). Nonetheless, the limited biodegradability and cell-adhesive characteristics of alginate restrict its potential uses (138,139). Efforts are ongoing to address these constraints. A recent study proposed that an alginate-hyaluronic acid hydrogel blend could be a promising option for AKI treatment (121).
ChitosanChitosan is a naturally occurring linear polysaccharide, is derived from crustacean exoskeletons and fungal cell walls (134,139). Beyond its structural resemblance to glycosaminoglycans in the ECM, it exhibits biocompatibility, biodegradability, antimicrobial properties, non-toxicity and affordability (144). These hydrogels demonstrate notable responsiveness to changes in pH and temperature (145), enhancing their adaptability for diverse applications. In vivo research has shown that chitosan hydrogels improve MSCs retention, viability and therapeutic efficacy in AKI (118,146). However, their weak mechanical properties and limited solubility in physiological environments restrict biomedical utility. Combining chitosan with peptides or alternative hydrogels offers a potential solution to these limitations (137,145).
CollagenCollagen is a fibrous structure, constitutes the predominant structural component in mammalian extracellular matrices (147). Owing to properties such as biodegradability, biocompatibility, elasticity and resemblance to natural tissue structures, collagen-based hydrogels are widely utilized as biomimetic scaffolds for 3D culture applications (139). Evidence indicates that scaffolds constructed from collagen effectively promote MSCs retention, proliferation, functional performance and phenotypic stability, consequently augmenting their therapeutic potential in treating AKI (122,139,148). A decellularized vascular matrix combined with collagen (co-gel) and MSCs were jointly transplanted into the kidneys of rats with IRI-induced AKI, the survival rate of MSCs and their paracrine effects in the injured kidneys were improved (122). More importantly, the co-gel markedly augmented the therapeutic efficacy of MSCs in AKI, including reducing cell death and tissue damage and improving angiogenesis and renal function (149). Duragen is an absorbable collagen-based artificial matrix that was used as a biological membrane to encapsulate MSCs transplanted into AKI mouse kidneys, preventing MSCs escape, prolonging their survival in vivo, improving renal function, ameliorating tubular lesions and reducing apoptosis. The authors also demonstrated in cell-based experiments that duragen-encapsulated MSCs protected renal cells from myoglobin-induced apoptosis. Thus, this confirms the potential of duragen-encapsulated MSCs as a new therapeutic strategy for AKI (150).
GelatinGelatin, a biocompatible polypeptide obtained from bovine or porcine collagen, comprises 18 distinct amino acids (139,151) and possesses lower antigenicity relative to collagen (151). Due to its advantageous properties, such as affordability, commercial accessibility, water solubility, good adhesion and straightforward processing, gelatin has become widely adopted in biomedical applications (152). Research involving AKI models demonstrates that gelatin-based hydrogels combined with MSCs effectively promote MSC survival, thus facilitating tissue repair (119). However, challenges of gelatin hydrogels include low mechanical strength, quick enzymatic breakdown and poor thermal stability (153). Pure gelatin undergoes a sol-gel shift close to body temperature, allowing it to be injected as a thin liquid at 37°C, but it does not solidify into a stable hydrogel in vivo (154). Modifications are necessary to enhance the overall properties of natural gelatin.
ECMKidney ECM hydrogels have attracted significant interest as biomaterials for regenerative medicine (155). By selectively removing cellular components while preserving the native proteins, glycosaminoglycans and growth factors, these hydrogels maintain essential bioactive properties, biocompatibility and minimal immunogenic responses (156,157). Compared with hydrogels derived from individual ECM components, kidney ECM hydrogels uniquely preserve the complete biochemical profile of renal tissue and do not include proteins obtained from tumorigenic sources as does Matrigel (158). Kidney ECM hydrogels are currently being explored as injectable scaffolds to facilitate renal regeneration and tissue repair, showing promising outcomes (159). In a related investigation, embedding ADMSCs within kidney ECM hydrogels markedly enhanced cell retention and survival after transplantation into ischemic rat kidneys. Moreover, ECM hydrogels promoted growth factor secretion while reducing oxidative damage and apoptosis, suggesting therapeutic promise for AKI treatment (160). However, due to inherent variability associated with natural biomaterials, the characteristics of kidney ECM hydrogels and their specific influences on cellular behavior require further elucidation (158). Additional studies should further clarify these issues to provide deeper insights into AKI management.
Hyaluronic acid (HA)HA is extensively found within mammalian tissues, including connective tissues, synovial fluid and vitreous humor (61,78). HA exhibits distinctive properties, including inherent bioactivity, excellent hydrophilicity, biodegradability and low cellular adhesiveness, making it a promising biomaterial candidate (145). These advantages make HA-based hydrogels increasingly suitable for numerous biomedical applications. As a crucial ECM constituent, HA markedly contributes to various biological processes, such as cellular proliferation, angiogenesis, embryogenesis, wound repair, stromal structuring and morphogenetic events (126). Implanting stem cells in HA hydrogels influences the release of cytokines/chemokines, counteracts pro-inflammatory mediators secreted by immune cells, thereby modulating immune responses and improving AKI (161). A previous investigation demonstrated that injecting MSCs encapsulated within HA hydrogels directly into kidneys subjected to IRI effectively attenuated ECM remodeling at one month post injury in mice (162). Notably, the highly thermoreversible nature of HA hydrogels provides favorable conditions for their use as injectable scaffolds for MSCs culture and delivery, or as implantable materials for soft and hard tissue repair and reconstruction (163). However, due to insufficient stability at body temperature and limited controlled-release capacity for bioactive molecules, chemical modifications and covalent crosslinking are required to improve the performance of HA hydrogels (164). Anyway, the aforementioned research proved that HA hydrogels have the potential to alleviate AKI.
OthersAgarose (129), Matrigel (165) and fibrin (166)-based hydrogels have also been employed to culture and deliver MSCs and serve as viable scaffolds for MSCs cultivation, but research on their application in AKI remains limited and warrants further investigation.
Synthetic hydrogelsSynthetic hydrogels are artificially engineered, 3D polymer networks that can absorb and retain large amounts of water, mimicking the properties of the natural ECM. They are widely used in biomedical applications such as tissue engineering, drug delivery and wound healing. Fu et al (119) discovered a novel therapeutic strategy for rhabdomyolysis. The authors used 3D modeling and printing to create an elastic sac that mimicked the kidney's dimensions and shape. This sac was applied as an outer coating for the kidney along with MSC-laden hydrogel (119). This innovative technology not only offers a versatile solution for organ restoration but also presents a promising option for AKI treatment. In another study, a new β-sheet supramolecular self-assembling peptide hydrogel was developed by combining insulin-like growth factor-1C (IGF-1C) with a D-assembling motif Nap-DFDFG, along with human placenta-derived mesenchymal stem cells for AKI. The results demonstrated that this hydrogel enhanced cell engraftment, ameliorated renal function, stimulated angiogenesis and reduced renal fibrosis in a mouse model of IRI induced AKI (125). Furthermore, an injectable nitric oxide (NO)-releasing hydrogel was created through physical crosslinking using short aromatic dipeptides. This hydrogel (Fmoc-diphenylalanine S-nitroso-N-acetylpenicillamine; Fmoc-FFSNAP) enabled localized and sustained NO release, leading to enhanced regeneration post-IRI and improved renal function (167). A recent study compared the therapeutic efficacy of Fmoc-FF + Fmoc-RGD-MSCs with free MSCs and Fmoc-FF + Fmoc-RGD-SNAP-MSCs in a renal IRI injury model. The hydrogel was combined with Wharton's jelly-derived MSCs and administered via intrarenal injection at three sites in IRI mice. Results demonstrated that the hydrogel treatment markedly enhanced renal function biomarkers, decreased reactive oxygen species production, improved histopathological changes and promoted the recovery and endothelial regeneration of the injured kidney (123).
Hybrid hydrogelsFeng et al (118) demonstrated that a bioactive hydrogel, formed by immobilizing the C domain peptide of IGF-1C on chitosan, provided an artificial microenvironment for adipose-derived ADMSCs, which enhanced cell viability through paracrine effects and protected against IRI.
Spheroid cultureAn alternative approach to 3D cell culture involves spheroid formation. This scaffold-free method enables MSCs and their ECM to self-assemble into tissue-like structures. Various techniques facilitate spheroid production, including liquid overlay, rotary culture, magnetic levitation, microparticle culture and the hanging drop method (168,169). Research suggests therapeutic applications for MSCs spheroids in kidney disorders. For example, Xu et al (124) demonstrated the enhanced therapeutic effects of human ADMSCs spheroids in AKI models. Compared with traditional monolayer cultures, these 3D aggregates showed increased secretion of ECM components (collagen I, fibronectin and laminin) along with superior antioxidative and anti-apoptotic capabilities.
Additionally, MSCs cultured with 3D spheroids exhibit enhanced secretion of paracrine cytokines such as VEGF and bFGF (angiogenic factors), EGF and HGF (anti-apoptotic factors), IGF (antioxidant) and TSG-6 (anti-inflammatory). These spheroids display greater in vivo survival and sustained paracrine effects, aligning with in vitro observations. In rat models of IRI-induced AKI, intrarenal injection of 3D spheroids provides superior renal protection compared to 2D-cultured cells, mitigating apoptosis, tissue injury and enhancing angiogenesis and functional recovery (124). Hybrid 3D spheroids composed of MSCs, vascular endothelial cells and podocytes can mimic glomerular microenvironments. Transplantation of these constructs into the renal cortex of hypertensive nephropathy mice improves kidney function, suggesting that engineered hybrid spheroids could markedly enhance podocyte replacement strategies and therapeutic outcomes (170).
3D culture of MSCs offers significant advantages in enhancing cell function and therapeutic effects, but it may also pose certain risks. First, the technical complexity and controllability of 3D culture systems are relatively low, which may lead to abnormalities in cell growth and differentiation. Second, hypoxia may occur in the center of the cells, affecting their survival and function. In addition, the therapeutic effects of 3D culture may vary due to inconsistencies in culture conditions and it may trigger immune rejection responses. Finally, the long-term effects and safety are not yet clear, particularly in clinical applications and further optimization and evaluation are needed.
Intracellular strategies (genetic modification)To enhance MSCs therapeutic potential in AKI, intracellular engineering methods such as genetic and epigenetic modifications have been extensively studied. Genetic modification involves changing MSCs' DNA sequence to regulate the expression of important genes. For example, boosting HO-1 and VEGF, genes crucial for antioxidant defense and angiogenesis, has been found to notably enhance MSCs migration, vascular targeting, anti-inflammatory abilities and viability, ultimately improving renal healing (171). These protective impacts are mainly mediated through activating signaling pathways such as PI3K/Akt and MEK/ERK (172,173).
Gene modification currently used include viral vector-based systems such as lentiviruses, retroviruses, adenoviruses and adeno-associated viruses, as well as non-viral systems such as plasmid DNA, RNA and protein-RNA complexes (171,174). In AKI preclinical models, these approaches have been used to boost the regenerative and immunomodulatory capabilities of MSCs, enhancing their proliferation, influencing differentiation potential and enhancing their anti-inflammatory and antioxidative functions (Table V) (175-182). Despite encouraging outcomes, the clinical application of these methods is restricted. Genetic modification might unintentionally affect genomic regions outside the target, leading to potential risks of genotoxicity, cytotoxicity, or tumorigenesis (183). Furthermore, ethical and safety issues associated with gene editing technologies are impeding their broad clinical implementation. Therefore, although genetic manipulation is a potent tool for refining MSC-based therapies for AKI, further optimization and thorough safety assessment are crucial before clinical adoption.
The mechanisms by which preconditioning strategies enhance MSCs function can be briefly summarized as follows: Physical preconditioning (such as hypoxic culture) enhances MSCs function by promoting proliferation, survival, homing, differentiation and paracrine activity, thereby improving renal function recovery. Chemical preconditioning primarily enhances MSCs stability by protecting cell membrane structures and improving the MSCs microenvironment through antioxidant, anti-inflammatory and tissue repair properties. Biological preconditioning activates MSC's immunomodulatory functions through cytokines or hormones, further enhancing their therapeutic effects. In addition, genetic modification optimizes MSC therapy for AKI by altering gene expression to enhance MSCs migration, vascular targeting and anti-inflammatory abilities. Further mechanisms specific pathways are still being explored.
Although these strategies have their advantages, they also present some challenges. Physical preconditioning and 3D culture methods are relatively simple, but there may be variability in cell growth and differentiation consistency. Chemical and biological preconditioning methods are comparatively simple and have high reproducibility, but more research may be needed to evaluate their long-term effects and potential side effects. Genetic modification strategies can markedly enhance MSCs functionality, but their safety and ethical concerns still need further evaluation. A combination of these strategies may be the best approach for treating AKI in the future, but further research and optimization are still required.
Improvement in administration routesTo fully leverage the diverse therapeutic capabilities of MSCs, it is crucial to ensure the adequate delivery of cells to the site of injury, which is a fundamental requirement for the efficacy of MSC-based therapy. The selection of the administration route markedly influences therapeutic results, with no consensus on the most effective approach to date (148). Systemic and local injections are currently the main methods used in preclinical and clinical studies, while innovative strategies such as encapsulated delivery of biomaterials and pulsed focused ultrasound (pFUS) guidance are gaining attention (184) (Fig. 3).
Systemic administration included intransient (IV), intra-arterial (IA), intraperitoneal (IP), accounting for 74% of MSCs injected. IV is commonly used for systemic delivery of bone marrow MSCs because it is simple and minimally invasive, with tail vein injection being the most dominant mode. However, an important drawback is the 'lung first-pass effect', which causes MSCs to become trapped in the lungs (185,186). In addition, MSCs tend to accumulate in the liver after intravenous injection, thereby reducing their concentration to reach the kidneys (187). Although administering higher numbers of MSCs could mitigate this problem, it potentially elevates the likelihood of complications, including thrombotic events and pulmonary embolism (188-190). By contrast, IA has shown higher efficacy by bypassing lung filters, improving MSCs homing and increasing cell delivery to target tissues, despite the risk of cell embolism and higher invasiveness (191,192). Whether intraperitoneally administered MSCs can enter the systemic circulation has not been reported, although they have the ability to reach the renal parenchyma.
Topical administration accounts for ~40% of MSCs injection, including intra-renal, subcapsular, renal-arterial. The MSC sheet transplantation has emerged in recent years as a novel localized delivery strategy for MSCs. Like above topical administration, this approach enables direct and precise renal implantation, enhancing cellular engraftment and therapeutic efficacy while circumventing pulmonary entrapment and reducing pulmonary complication risks (193). Notably, MSC sheet transplantation uniquely maintains high local cell concentrations at target sites, thereby promoting renal tissue regeneration (193). Various topical administration methods have been evaluated in AKI models showing good results, with intra-renal injections being extensively studied for their ease of use. Other methods, such as hydrogel, microgel and biomaterials aforementioned, simulate the interaction between cells in vivo and improve the viability of MSCs. In addition, pFUS-guided delivery of MSCs was reported to improve microenvironment, up-regulate adhesion factors and guide MSCs homing and viability (184). Due to the small number of published reports, more studies are needed to confirm its safety and effectiveness.
Although preclinical studies have demonstrated the therapeutic potential of MSCs and preconditioned MSCs in AKI, their clinical translation remains a crucial step in validating efficacy and safety in human patients. A review of current clinical trials (Table VI; https://clinicaltrials.gov/) reveals multiple studies exploring MSC-based therapies for AKI, including autologous or allogeneic MSCs derived from sources such as umbilical cord (UCMSCs), bone marrow (BMSCs) and adipose tissue (ADMSCs). These trials aim to evaluate parameters such as renal function recovery, inflammatory marker modulation and long-term safety, with results confirming the safety of MSC infusion for AKI treatment. Preconditioning strategies (such as hypoxia, cytokine priming, or pharmacological agents such as melatonin) have been employed to enhance MSCs survival and reparative function. However, challenges such as optimizing dosage, administration routes and quality control of MSCs products must still be addressed to ensure reproducible therapeutic outcomes. Future research should focus on biomarker-guided patient stratification and combination therapies (such as MSC-derived exosomes) to maximize treatment efficacy.
In summary, MSCs show promise for treating AKI. However, clinical outcomes have not matched preclinical findings, indicating challenges for translation to clinical practice. Researchers have investigated strategies including pretreatment, changing cell culture environment, alternative delivery routes to address these hurdles. Moreover, due to patient heterogeneity, treatment approaches should be personalized based on factors such as age, genetics and overall health. Further research is warranted to enhance MSC-based therapies for effective clinical application in AKI treatment.
Not applicable.
ND was responsible for writing, reviewing and editing, supervision and project administration. LW was responsible for conceptualization, writing, reviewing and editing, supervision and funding acquisition. JK was responsible for supervision and project administration. YZ, JD and JF were responsible for writing, reviewing and editing, writing the original draft, visualization, software, methodology, investigation, formal analysis, data curation and conceptualization. MY and HW were responsible for writing, reviewing and editing, supervision and funding acquisition. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Professor Li Wang ORCID:0000000338813149 Professor Nathupakorn Dechsupa ORCID: 0000-0002-0412-9659.
|
AKI |
acute kidney injury |
|
CKD |
chronic kidney disease |
|
MSCs |
mesenchymal stem cells |
|
hAD-MSCs |
human adipose-derived MSCs |
|
hUC-MSCs |
human umbilical cord MSCs |
|
EVs |
extracellular vesicles |
|
CZ |
Chlorzoxane |
|
Ator |
Atorvastatin |
|
DPO |
darbepoetin |
|
EpoR |
erythropoietin receptor |
|
EPO |
erythropoietin |
|
ROS |
reactive oxygen species |
|
UUO |
unilateral ureteral obstruction |
|
Tregs |
regulatory T cells |
|
IRI |
ischemia reperfusion injury |
|
ADMSCs |
adipose-derived MSCs |
|
HSCs |
hematopoietic stem cells |
|
3D |
three-dimensional |
|
ECM |
extracellular matrix |
|
HA |
hyaluronic acid |
|
GAG |
glycosaminoglycan |
|
IGF-1C |
insulin-like growth factor-1C |
|
WJ-MSCs |
Wharton's jelly-derived MSCs |
|
pFUS |
pulsed focused ultrasound |
|
IV |
intransient |
|
IA |
intra-arterial |
|
IP |
intraperitoneal |
|
HO-1 |
heme oxygenase-1 |
|
VEGF |
vascular endothelial growth factor |
The authors would like to thank BioRender.com (https://app.biorender.com/) for providing the illustration platform used to create the figures (Figs. 1, 2 and 3) in the present review.
The present review was supported by National Natural Science Foundation (grant no. 82200830); Sichuan Science and Technology Program (grant no. 2025ZNSFSC0617); The Project of Southwest Medical University Affiliated Traditional Medicine Hospital (grant no. 2022-CXTD-03) and Southwest Medical University Technology Program (grant no. 2023ZYYQ01).
|
Ronco C, Bellomo R and Kellum JS: Acute kidney injury. Lancet. 394:1949–1964. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Y and Dong Z: Immune response in COVID-19-associated acute kidney injury and maladaptive kidney repair. Integrative Medicine in Nephrology and Andrology. 10:e000222023. View Article : Google Scholar | |
|
Hoste EAJ, Kellum JA, Selby NM, Zarbock A, Palevsky PM, Bagshaw SM, Goldstein SL, Cerdá J and Chawla LS: Global epidemiology and outcomes of acute kidney injury. Nat Rev Nephrol. 14:607–625. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Meena J, Kumar J, Kocharlakota JP, Gupta H, Mittal P, Kumar A, Sinha A, Hari P and Bagga A: Acute kidney injury in neonates: A meta-analysis. Pediatrics. 154:e20230651822024. View Article : Google Scholar : PubMed/NCBI | |
|
Gautam SC, Lim J and Jaar BG: Complications associated with continuous RRT. Kidney360. 3:1980–1990. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang R, Widdop RE and Ricardo SD: Transition from acute kidney injury to chronic kidney disease: Mechanisms, models, and biomarkers. Am J Physiol Renal Physiol. 327:F788–F805. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Duan J, Hua C, Pan S, Li G, Feng Q, Liu D and Liu Z: Nanomedicine embraces the treatment and prevention of acute kidney injury to chronic kidney disease transition: Evidence, challenges, and opportunities. Burns Trauma. 12:tkae0442024. View Article : Google Scholar : PubMed/NCBI | |
|
Koh ES and Chung S: Recent update on acute kidney injury-to-chronic kidney disease transition. Yonsei Med J. 65:247–256. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Song Z and Gong X: Research progress on the potential mechanisms of acute kidney injury and chronic kidney disease induced by proton pump inhibitors. Integrative Medicine in Nephrology and Andrology. 10:2023. View Article : Google Scholar | |
|
Stanski NL, Rodrigues CE, Strader M, Murray PT, Endre ZH and Bagshaw SM: Precision management of acute kidney injury in the intensive care unit: Current state of the art. Intensive Care Med. 49:1049–1061. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Krishnasamy S, Sinha A and Bagga A: Management of acute kidney injury in critically Ill children. Indian J Pediatr. 90:481–491. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kou M, Huang L, Yang J, Chiang Z, Chen S, Liu J, Guo L, Zhang X, Zhou X, Xu X, et al: Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: A next generation therapeutic tool? Cell Death Dis. 13:5802022. View Article : Google Scholar : PubMed/NCBI | |
|
Allinson CS, Pollock CA and Chen X: Mesenchymal stem cells in the treatment of acute kidney injury (AKI), chronic kidney disease (CKD) and the AKI-to-CKD transition. Integrative Medicine in Nephrology and Andrology. 10:e000142023. View Article : Google Scholar | |
|
Sávio-Silva C, Soinski-Sousa PE, Balby-Rocha MTA, Lira ÁO and Rangel ÉB: Mesenchymal stem cell therapy in acute kidney injury (AKI): Review and perspectives. Rev Assoc Med Bras (1992). 13(66 Suppl 1): S45–S54. 2020. View Article : Google Scholar | |
|
Lee PW, Wu BS, Yang CY and Lee OK: Molecular mechanisms of mesenchymal stem cell-based therapy in acute kidney injury. Int J Mol Sci. 22:114062021. View Article : Google Scholar : PubMed/NCBI | |
|
Khubutiya MS, Vagabov AV, Temnov AA and Sklifas AN: Paracrine mechanisms of proliferative, anti-apoptotic and anti-inflammatory effects of mesenchymal stromal cells in models of acute organ injury. Cytotherapy. 16:579–585. 2014. View Article : Google Scholar | |
|
Han Q, Wang X, Ding X, He J, Cai G and Zhu H: Immunomodulatory effects of mesenchymal stem cells on drug-induced acute kidney injury. Front Immunol. 12:6830032021. View Article : Google Scholar : PubMed/NCBI | |
|
Fazekas B and Griffin MD: Mesenchymal stromal cell-based therapies for acute kidney injury: Progress in the last decade. Kidney Int. 97:1130–1140. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Hu C, Han F, Wang J and Chen J: Regenerative abilities of mesenchymal stem cells via acting as an ideal vehicle for subcellular component delivery in acute kidney injury. J Cell Mol Med. 24:4882–4891. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Birtwistle L, Chen XM and Pollock C: Mesenchymal stem cell-derived extracellular vesicles to the rescue of renal injury. Int J Mol Sci. 22:65962021. View Article : Google Scholar : PubMed/NCBI | |
|
Mukkala AN, Jerkic M, Khan Z, Szaszi K, Kapus A and Rotstein O: Therapeutic effects of mesenchymal stromal cells require mitochondrial transfer and quality control. Int J Mol Sci. 24:157882023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang D, Chen FX, Zhou H, Lu YY, Tan H, Yu SJ, Yuan J, Liu H, Meng W and Jin ZB: Bioenergetic Crosstalk between mesenchymal stem cells and various ocular cells through the intercellular trafficking of mitochondria. Theranostics. 10:7260–7272. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Paliwal S, Chaudhuri R, Agrawal A and Mohanty S: Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci. 25:312018. View Article : Google Scholar : PubMed/NCBI | |
|
Michaeloudes C, Li X, Mak JCW and Bhavsar PK: Study of mesenchymal stem cell-mediated mitochondrial transfer in in vitro models of oxidant-mediated airway epithelial and smooth muscle cell injury. Methods Mol Biol. 2269:93–105. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Iorio R, Petricca S, Mattei V and Monache SD: Horizontal mitochondrial transfer as a novel bioenergetic tool for mesenchymal stromal/stem cells: Molecular mechanisms and therapeutic potential in a variety of diseases. J Transl Med. 22:4912024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Hu C, Zhang P, Jiang H and Chen J: Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med. 23:720–730. 2019. View Article : Google Scholar : | |
|
Zhao L, Hu C, Zhang P, Jiang H and Chen J: Novel preconditioning strategies for enhancing the migratory ability of mesenchymal stem cells in acute kidney injury. Stem Cell Res Ther. 9:2252018. View Article : Google Scholar : PubMed/NCBI | |
|
Makkar D, Gakhar D, Mishra V and Rakha A: Fine tuning mesenchymal stromal cells-code for mitigating kidney diseases. Stem Cell Rev Rep. 20:738–754. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Z, Zhang Y, Geng X, Chi K, Liu C, Song C, Cai G, Chen X and Hong Q: Optimization strategies of mesenchymal stem cell-based therapy for acute kidney injury. Stem Cell Res Ther. 14:1162023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Lee EH and Yang Z: Hypoxia-conditioned mesenchymal stem cells in tissue regeneration application. Tissue Eng Part B Rev. 28:966–977. 2022. View Article : Google Scholar | |
|
Yasan GT and Gunel-Ozcan A: Hypoxia and hypoxia mimetic agents as potential priming approaches to empower mesenchymal stem cells. Curr Stem Cell Res Ther. 19:33–54. 2024. View Article : Google Scholar | |
|
Li H, Ji XQ, Zhang SM and Bi RH: Hypoxia and inflammatory factor preconditioning enhances the immunosuppressive properties of human umbilical cord mesenchymal stem cells. World J Stem Cells. 15:999–1016. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Muscari C, Giordano E, Bonafè F, Govoni M, Pasini A and Guarnieri C: Priming adult stem cells by hypoxic pretreatments for applications in regenerative medicine. J Biomed Sci. 20:632013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Liu L, Huo Y, Yang Y and Wang Y: Hypoxia-pretreated human MSCs attenuate acute kidney injury through enhanced angiogenic and antioxidative capacities. Biomed Res Int. 2014:4624722014.PubMed/NCBI | |
|
Collino F, Lopes JA, Corrêa S, Abdelhay E, Takiya CM, Wendt CHC, de Miranda KR, Vieyra A and Lindoso R: Adipose-derived mesenchymal stromal cells under hypoxia: Changes in extracellular vesicles secretion and improvement of renal recovery after ischemic injury. Cell Physiol Biochem. 52:1463–1483. 2019.PubMed/NCBI | |
|
Jang MJ, You D, Park JY, Kim K, Aum J, Lee C, Song G, Shin HC, Suh N, Kim YM and Kim CS: Hypoxic preconditioned mesenchymal stromal cell therapy in a rat model of renal ischemia-reperfusion injury: development of optimal protocol to potentiate therapeutic efficacy. Int J Stem Cells. 11:157–167. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Putra A, Pertiwi D, Milla MN, Indrayani UD, Jannah D, Sahariyani M, Trisnadi S and Wibowo JW: Hypoxia-preconditioned MSCs have superior effect in ameliorating renal function on acute renal failure animal model. Open Access Maced J Med Sci. 7:305–310. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan F, Liu J, Zhong L, Liu P, Li T, Yang K, Gao W, Zhang G, Sun J and Zou X: Enhanced therapeutic effects of hypoxia-preconditioned mesenchymal stromal cell-derived extracellular vesicles in renal ischemic injury. Stem Cell Res Ther. 16:392025. View Article : Google Scholar : PubMed/NCBI | |
|
Overath JM, Gauer S, Obermüller N, Schubert R, Schäfer R, Geiger H and Baer PC: Short-term preconditioning enhances the therapeutic potential of adipose-derived stromal/stem cell-conditioned medium in cisplatin-induced acute kidney injury. Exp Cell Res. 342:175–183. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Lu C, Liu H, Rao S, Cai J, Liu S, Kriegel AJ, Greene AS, Liang M and Ding X: Hypoxic preconditioning with cobalt of bone marrow mesenchymal stem cells improves cell migration and enhances therapy for treatment of ischemic acute kidney injury. PLoS One. 8:e627032013. View Article : Google Scholar : PubMed/NCBI | |
|
Tseng WC, Lee PY, Tsai MT, Chang FP, Chen NJ, Chien CT, Hung SC and Tarng DC: Hypoxic mesenchymal stem cells ameliorate acute kidney ischemia-reperfusion injury via enhancing renal tubular autophagy. Stem Cell Res Ther. 12:3672021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu P, Feng Y, Dong C, Liu D, Wu X, Wu H, Lv P and Zhou Y: Study on therapeutic action of bone marrow derived mesenchymal stem cell combined with vitamin E against acute kidney injury in rats. Life Sci. 92:829–837. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Q and Wang J: Effect of combination of vitamin E and umbilical cord-derived mesenchymal stem cells on inflammation in mice with acute kidney injury. Immunopharmacol Immunotoxicol. 40:168–172. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Deng L, Li H, Su X, Zhang Y, Xu H, Fan L, Fan J, Han Q, Bai X and Zhao RC: Chlorzoxazone, a small molecule drug, augments immunosuppressive capacity of mesenchymal stem cells via modulation of FOXO3 phosphorylation. Cell Death Dis. 11:1582020. View Article : Google Scholar : PubMed/NCBI | |
|
Cai J, Yu X, Zhang B, Zhang H, Fang Y, Liu S, Liu T and Ding X: Atorvastatin improves survival of implanted stem cells in a rat model of renal ischemia-reperfusion injury. Am J Nephrol. 39:466–475. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Altun B, Yilmaz R, Aki T, Akoglu H, Zeybek D, Piskinpasa S, Uckan D, Purali N, Korkusuz P and Turgan C: Use of mesenchymal stem cells and darbepoetin improve ischemia-induced acute kidney injury outcomes. Am J Nephrol. 35:531–539. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
LiverTox: Clinical and Research Information on Drug-Induced Liver Injury (Internet). National Institute of Diabetes and Digestive and Kidney Diseases; Bethesda, MD: 2012, https://www.ncbi.nlm.nih.gov/books/NBK548137/. | |
|
Dh HS, Sultana R, Prabhu A, Pavan SR, Mohanto S and Subramaniyan V: Biomedicine and pharmacotherapeutic effectiveness of combinatorial atorvastatin and quercetin on diabetic nephropathy: An in vitro study. Biomed Pharmacother. 174:1165332024. View Article : Google Scholar : PubMed/NCBI | |
|
Qu H, Xu H, Tian Y and Jiang X: Atorvastatin improves microenvironment to enhance the beneficial effects of BMSCs therapy in a rabbit model of acute myocardial infarction. Cell Physiol Biochem. 32:380–389. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y and Yang B: Erythropoietin Receptor/β common receptor: A shining light on acute kidney injury induced by ischemia-reperfusion. Front Immunol. 12:6977962021. View Article : Google Scholar | |
|
Ghassemi-Barghi N, Ehsanfar Z, Mohammadrezakhani O, Ashari S, Ghiabi S and Bayrami Z: Mechanistic approach for protective effect of ARA290, a specific ligand for the erythropoietin/CD131 heteroreceptor, against cisplatin-induced nephrotoxicity, the involvement of apoptosis and inflammation pathways. Inflammation. 46:342–358. 2023. View Article : Google Scholar | |
|
Johnson DW, Pat B, Vesey DA, Guan Z, Endre Z and Gobe GC: Delayed administration of darbepoetin or erythropoietin protects against ischemic acute renal injury and failure. Kidney Int. 69:1806–1813. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Hakiminia B, Goudarzi A and Moghaddas A: Has vitamin E any shreds of evidence in cisplatin-induced toxicity. J Biochem Mol Toxicol. 33:e223492019. View Article : Google Scholar : PubMed/NCBI | |
|
Darwish MA, Abo-Youssef AM, Khalaf MM, Abo-Saif AA, Saleh IG and Abdelghany TM: Vitamin E mitigates cisplatin-induced nephrotoxicity due to reversal of oxidative/nitrosative stress, suppression of inflammation and reduction of total renal platinum accumulation. J Biochem Mol Toxicol. 31:1–9. 2017. View Article : Google Scholar | |
|
Abo-Elmaaty AMA, Behairy A, El-Naseery NI and Abdel-Daim MM: The protective efficacy of vitamin E and cod liver oil against cisplatin-induced acute kidney injury in rats. Environ Sci Pollut Res Int. 27:44412–44426. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kim H, Yu MR, Lee H, Kwon SH, Jeon JS, Han DC and Noh H: Metformin inhibits chronic kidney disease-induced DNA damage and senescence of mesenchymal stem cells. Aging Cell. 20:e133172021. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Q, Zhu B, Yang G, Jia J, Wang H, Tan R, Zhang Q, Wang L and Kantawong F: Calycosin pretreatment enhanced the therapeutic efficacy of mesenchymal stem cells to alleviate unilateral ureteral obstruction-induced renal fibrosis by inhibiting necroptosis. J Pharmacol Sci. 151:72–83. 2023. 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 : | |
|
Huang Y, Wu Q and Tam PKH: Immunomodulatory mechanisms of mesenchymal stem cells and their potential clinical applications. Int J Mol Sci. 23:100232022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou J and Shi Y: Mesenchymal stem/stromal cells (MSCs): Origin, immune regulation, and clinical applications. Cell Mol Immunol. 20:555–557. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Fang J, Liu B, Shao C and Shi Y: Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell. 29:1515–1530. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li P, Ou Q, Shi S and Shao C: Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications. Cell Mol Immunol. 20:558–569. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Umrath F, Cen W, Salgado AJ, Reinert S and Alexander D: Pre-conditioning with IFN-γ and hypoxia enhances the angiogenic potential of iPSC-Derived MSC secretome. Cells. 11:9882022. View Article : Google Scholar | |
|
Wu J, Wu J, Xiang W, Gong Y, Feng D, Fang S, Wu Y, Liu Z, Li Y, Chen R, et al: Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis. J Nanobiotechnology. 22:5552024. View Article : Google Scholar | |
|
Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D, Watanabe Y, Hayashi C, et al: Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 122:306–324. 2021. View Article : Google Scholar | |
|
Ma T, Wang X, Jiao Y, Wang H, Qi Y, Gong H, Zhang L and Jiang D: Interleukin 17 (IL-17)-induced mesenchymal stem cells prolong the survival of allogeneic skin grafts. Ann Transplant. 23:615–621. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao C, Li J, Cai H, Wu D, Tao S, Pi C, Zhu L, Xu N and Zhang T: An injectable hydrogel scaffold with IL-1β-activated MSC-derived exosomes for the treatment of endometritis. Biomater Sci. 11:1422–1436. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pezzanite LM, Chow L, Phillips J, Griffenhagen GM, Moore AR, Schaer TP, Engiles JB, Werpy N, Gilbertie J, Schnabel LV, et al: TLR-activated mesenchymal stromal cell therapy and antibiotics to treat multi-drug resistant Staphylococcal septic arthritis in an equine model. Ann Transl Med. 10:11572022. View Article : Google Scholar : PubMed/NCBI | |
|
Giuliani M, Bennaceur-Griscelli A, Nanbakhsh A, Oudrhiri N, Chouaib S, Azzarone B, Durrbach A and Lataillade JJ: TLR ligands stimulation protects MSC from NK killing. Stem Cells. 32:290–300. 2014. View Article : Google Scholar | |
|
Kwon YW, Heo SC, Jeong GO, Yoon JW, Mo WM, Lee MJ, Jang IH, Kwon SM, Lee JS and Kim JH: Tumor necrosis factor-α-activated mesenchymal stem cells promote endothelial progenitor cell homing and angiogenesis. Biochim Biophys Acta. 1832:2136–2144. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Heo SC, Jeon ES, Lee IH, Kim HS, Kim MB and Kim JH: Tumor necrosis factor-α-activated human adipose tissue-derived mesenchymal stem cells accelerate cutaneous wound healing through paracrine mechanisms. J Invest Dermatol. 131:1559–1567. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Bai X, Xi J, Bi Y, Zhao X, Bing W, Meng X, Liu Y, Zhu Z and Song G: TNF-alpha promotes survival and migration of MSCs under oxidative stress via NF-κB pathway to attenuate intimal hyperplasia in vein grafts. J Cell Mol Med. 21:2077–2091. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Hu C, Zhang P, Jiang H and Chen J: Melatonin preconditioning is an effective strategy for mesenchymal stem cell-based therapy for kidney disease. J Cell Mol Med. 24:25–33. 2020. View Article : Google Scholar | |
|
Alzahrani FA: Melatonin improves therapeutic potential of mesenchymal stem cells-derived exosomes against renal ischemia-reperfusion injury in rats. Am J Transl Res. 11:2887–2907. 2019.PubMed/NCBI | |
|
Aussel C, Baudry N, Grosbot M, Caron C, Vicaut E, Banzet S and Peltzer JS: IL-1β primed mesenchymal stromal cells moderate hemorrhagic shock-induced organ injuries. Stem Cell Res Ther. 12:4382021. View Article : Google Scholar | |
|
Bai M, Zhang L, Fu B, Bai J, Zhang Y, Cai G, Bai X, Feng Z, Sun S and Chen X: IL-17A improves the efficacy of mesenchymal stem cells in ischemic-reperfusion renal injury by increasing Treg percentages by the COX-2/PGE2 pathway. Kidney Int. 93:814–825. 2018. View Article : Google Scholar | |
|
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 | |
|
Kanai R, Nakashima A, Doi S, Kimura T, Yoshida K, Maeda S, Ishiuchi N, Yamada Y, Ike T, Doi T, et al: Interferon-gamma enhances the therapeutic effect of mesenchymal stem cells on experimental renal fibrosis. Sci Rep. 11:8502021. View Article : Google Scholar | |
|
Mias C, Trouche E, Seguelas MH, Calcagno F, Dignat-George F, Sabatier F, Piercecchi-Marti MD, Daniel L, Bianchi P, Calise D, et al: Ex vivo pretreatment with melatonin improves survival, proangiogenic/mitogenic activity, and efficiency of mesenchymal stem cells injected into ischemic kidney. Stem Cells. 26:1749–1757. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Young YK, Fradette J and Eliopoulos N: Melatonin pretreatment of human adipose tissue-derived mesenchymal stromal cells enhances their prosurvival and protective effects on human kidney cells. Am J Physiol Renal Physiol. 308:F1474–F1483. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou S, Qiao YM, Liu YG, Liu D, Hu JM, Liao J, Li M, Guo Y, Fan LP, Li LY and Zhao M: Bone marrow derived mesenchymal stem cells pretreated with erythropoietin accelerate the repair of acute kidney injury. Cell Biosci. 10:1302020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Wang G, Zhang L, Li F, Liu K, Wang Y, Shi Y and Cao K: Interleukin-17 promotes nitric oxide-dependent expression of PD-L1 in mesenchymal stem cells. Cell Biosci. 10:732020. View Article : Google Scholar : PubMed/NCBI | |
|
Han X, Yang Q, Lin L, Xu C, Zheng C, Chen X, Han Y, Li M, Cao W, Cao K, et al: Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death Differ. 21:1758–1768. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Jiang C, Yang Y, Xi C, Yin Y, Wu H and Qian C: Therapeutic potential of HUC-MSC-exos primed with IFN-γ against LPS-induced acute lung injury. Iran J Basic Med Sci. 27:375–382. 2024. | |
|
Stazi M, Negro S, Megighian A, D'Este G, Solimena M, Jockers R, Lista F, Montecucco C and Rigoni M: Melatonin promotes regeneration of injured motor axons via MT1 receptors. J Pineal Res. 70:e126952021. View Article : Google Scholar | |
|
Romero A, Morales-Garcia JA and Ramos E: Melatonin: A multitasking indoleamine to modulate hippocampal neurogenesis. Neural Regen Res. 18:503–505. 2023. View Article : Google Scholar | |
|
Reiter RJ, Tan DX, Poeggeler B, Menendez-Pelaez A, Chen LD and Saarela S: Melatonin as a free radical scavenger: Implications for aging and age-related diseases. Ann N Y Acad Sci. 719:1–12. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Reiter RJ, Tan DX and Fuentes-Broto L: Melatonin: A multitasking molecule. Prog Brain Res. 181:127–151. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Reiter RJ: Melatonin: The chemical expression of darkness. Mol Cell Endocrinol. 79:C153–C158. 1991. View Article : Google Scholar : PubMed/NCBI | |
|
Feng ZY, Yang SD, Wang T and Guo S: Effect of melatonin for regulating mesenchymal stromal cells and derived extracellular vesicles. Front Cell Dev Biol. 9:7179132021. View Article : Google Scholar : PubMed/NCBI | |
|
Elzainy A, El Sadik A and Altowayan WM: Comparison between the regenerative and therapeutic impacts of bone marrow mesenchymal stem cells and adipose mesenchymal stem cells pre-treated with melatonin on liver fibrosis. Biomolecules. 14:2972024. View Article : Google Scholar : PubMed/NCBI | |
|
Zou D, Liao J, Xiao M, Liu L and Xu M: Melatonin alleviates hyperoxia-induced lung injury through elevating MSC exosomal miR-18a-5p expression to repress PUM2 signaling. FASEB J. 38:e700122024. View Article : Google Scholar : PubMed/NCBI | |
|
Rafiq H, Ayaz M, Khan HA, Iqbal M, Quraish S, Afridi SG, Khan A, Khan B, Sher A, Siraj F and Shams S: Therapeutic potential of stem cell and melatonin on the reduction of CCl4-induced liver fibrosis in experimental mice model. Braz J Biol. 84:e2530612022. View Article : Google Scholar : PubMed/NCBI | |
|
Mintoft A, Vallatos A and Robertson NJ: Mesenchymal stromal cell therapy for hypoxic ischemic encephalopathy: Future directions for combination therapy with hypothermia and/or melatonin. Semin Perinatol. 48:1519292024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee JH, Han YS and Lee SH: Melatonin-induced PGC-1α improves angiogenic potential of mesenchymal stem cells in hindlimb ischemia. Biomol Ther (Seoul). 28:240–249. 2020. View Article : Google Scholar | |
|
Hanna M, Elnassag SS, Mohamed DH, Elbaset MA, Shaker O, Khowailed EA and Gouda SAA: Melatonin and mesenchymal stem cells co-administration alleviates chronic obstructive pulmonary disease via modulation of angiogenesis at the vascular-alveolar unit. Pflugers Arch. 476:1155–1168. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tripathy S and Bhattamisra SK: Cellular signalling of melatonin and its role in metabolic disorders. Mol Biol Rep. 52:1932025. View Article : Google Scholar : PubMed/NCBI | |
|
Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS and Slominski AT: Melatonin membrane receptors in peripheral tissues: Distribution and functions. Mol Cell Endocrinol. 351:152–166. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Charif SE and Dorfman VB: Melatonin, modulation of hypothalamic activity, and reproduction. Vitam Horm. 127:283–303. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Saberi K, Pasbakhsh P, Omidi A, Borhani-Haghighi M, Nekoonam S, Omidi N, Ghasemi S and Kashani IR: Melatonin preconditioning of bone marrow-derived mesenchymal stem cells promotes their engraftment and improves renal regeneration in a rat model of chronic kidney disease. J Mol Histol. 50:129–140. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yasuoka Y, Izumi Y, Fukuyama T, Oshima T, Yamazaki T, Uematsu T, Kobayashi N, Nanami M, Shimada Y, Nagaba Y, et al: Tubular endogenous erythropoietin protects renal function against ischemic reperfusion injury. Int J Mol Sci. 25:12232024. View Article : Google Scholar : PubMed/NCBI | |
|
Bi B, Guo J, Marlier A, Lin SR and Cantley LG: Erythropoietin expands a stromal cell population that can mediate renoprotection. Am J Physiol Renal Physiol. 295:F1017–F1022. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zuo L, Li DD, Ma XX, Shi SH, Lyu DC, Shen J, Zhang WF, Gao EH and Cao JM: Erythropoietin promotes myocardial infarction repair in mice by improving the function of Sca-1(+) stem cells. Sheng Li Xue Bao. 75:36–48. 2023.PubMed/NCBI | |
|
Wu YW, Comstock BA, Gonzalez FF, Mayock DE, Goodman AM, Maitre NL, Chang T, Van Meurs KP, Lampland AL, Bendel-Stenzel E, et al: Trial of erythropoietin for hypoxic-ischemic encephalopathy in newborns. N Engl J Med. 387:148–159. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bianchi VE and von Haehling S: The treatment of chronic anemia in heart failure: A global approach. Clin Res Cardiol. 113:1117–1136. 2024. View Article : Google Scholar | |
|
Jun JH, Jun NH, Shim JK, Shin EJ and Kwak YL: Erythropoietin protects myocardium against ischemia-reperfusion injury under moderate hyperglycemia. Eur J Pharmacol. 745:1–9. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou S, Liu YG, Zhang Y, Hu JM, Liu D, Chen H, Li M, Guo Y, Fan LP, Li LY and Zhao M: Bone mesenchymal stem cells pretreated with erythropoietin enhance the effect to ameliorate cyclosporine A-induced nephrotoxicity in rats. J Cell Biochem. 119:8220–8232. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Verdino P, Lee SL, Cooper FN, Cottle SR, Grealish PF, Hu CC, Meyer CM, Lin J, Copeland V, Porter G, et al: Development of a long-acting relaxin analogue, LY3540378, for treatment of chronic heart failure. Br J Pharmacol. 180:1965–1980. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Samuel CS and Hewitson TD: Relaxin in cardiovascular and renal disease. Kidney Int. 69:1498–1502. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Martins RC, Pintalhao M, Leite-Moreira A and Castro-Chaves P: Relaxin and the cardiovascular system: From basic science to clinical practice. Curr Mol Med. 20:167–184. 2020. View Article : Google Scholar | |
|
Feiteng C, Lei C, Deng L, Chaoliang X, Zijie X, Yi S and Minglei S: Relaxin inhibits renal fibrosis and the epithelial-to-mesenchymal transition via the Wnt/beta-catenin signaling pathway. Ren Fail. 44:513–524. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Conrad KP, von Versen-Hoynck F and Baker VL: Potential role of the corpus luteum in maternal cardiovascular adaptation to pregnancy and preeclampsia risk. Am J Obstet Gynecol. 226:683–699. 2022. View Article : Google Scholar | |
|
Bathgate RA, Ivell R, Sanborn BM, Sherwood OD and Summers RJ: International union of pharmacology LVII: Recommendations for the nomenclature of receptors for relaxin family peptides. Pharmacol Rev. 58:7–31. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Shen M, Ferens D, Broughton BRS, Murthi P, Saini S, Widdop RE, Ricardo SD, Pinar AA and Samuel CS: Combining mesenchymal stem cells with serelaxin provides enhanced renoprotection against 1K/DOCA/salt-induced hypertension. Br J Pharmacol. 178:1164–1181. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Lu W, Wang A, Jiang H and Lyu J: Changing epidemiology of chronic kidney disease as a result of type 2 diabetes mellitus from 1990 to 2017: Estimates from global burden of disease 2017. J Diabetes Investig. 12:346–356. 2021. View Article : Google Scholar | |
|
Huuskes BM, Wise AF, Cox AJ, Lim EX, Payne NL, Kelly DJ, Samuel CS and Ricardo SD: Combination therapy of mesenchymal stem cells and serelaxin effectively attenuates renal fibrosis in obstructive nephropathy. FASEB J. 29:540–553. 2015. View Article : Google Scholar | |
|
Badawi A, Jefferson OC, Huuskes BM, Ricardo SD, Kerr PG, Samuel CS and Murthi P: A novel approach to enhance the regenerative potential of circulating endothelial progenitor cells in patients with end-stage kidney disease. Biomedicines. 10:8832022. View Article : Google Scholar : PubMed/NCBI | |
|
Feng G, Zhang J, Li Y, Nie Y, Zhu D, Wang R, Liu J, Gao J, Liu N, He N, et al: IGF-1 C domain-modified hydrogel enhances cell therapy for AKI. J Am Soc Nephrol. 27:2357–2369. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Z, Chu Y, Geng X, Ma Y, Chi K, Song C, Liao S, Hong Q, Wu D and Wang Y: Artificial kidney capsule packed with mesenchymal stem cell-laden hydrogel for the treatment of rhabdomyolysis-induced acute kidney injury. ACS Biomater Sci Eng. 8:1726–1734. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Geng X, Hong Q, Wang W, Zheng W, Li O, Cai G, Chen X and Wu D: Biological membrane-packed mesenchymal stem cells treat acute kidney disease by ameliorating mitochondrial-related apoptosis. Sci Rep. 7:411362017. View Article : Google Scholar : PubMed/NCBI | |
|
Han DS, Erickson C, Hansen KC, Kirkbride-Romeo L, He Z, Rodell CB and Soranno DE: Mesenchymal stem cells delivered locally to ischemia-reperfused kidneys via injectable hyaluronic acid hydrogels decrease extracellular matrix remodeling 1 month after injury in male mice. Cells. 12:17712023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang S, Li Y, Wang X, Ma X and Zhang X: Injectable co-gels of collagen and decellularized vascular matrix improve MSC-based therapy for acute kidney injury. J Biomater Sci Polym Ed. 28:2186–2195. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Najafi H, Abolmaali SS, Heidari R, Valizadeh H, Tamaddon AM and Azarpira N: Integrin receptor-binding nanofibrous peptide hydrogel for combined mesenchymal stem cell therapy and nitric oxide delivery in renal ischemia/reperfusion injury. Stem Cell Res Ther. 13:3442022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Shi T, Xu A and Zhang L: 3D spheroid culture enhances survival and therapeutic capacities of MSCs injected into ischemic kidney. J Cell Mol Med. 20:1203–1213. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Shang Y, Chen X, Wang Z, Zhu D, Liu Y, Zhang C, Chen P, Wu J, Wu L, et al: Delivery of MSCs with a hybrid beta-sheet peptide hydrogel consisting IGF-1C domain and D-Form peptide for acute kidney injury therapy. Int J Nanomedicine. 15:4311–4324. 2020. View Article : Google Scholar : | |
|
Ho TC, Chang CC, Chan HP, Chung TW, Shu CW, Chuang KP, Duh TH, Yang MH and Tyan YC: Hydrogels: Properties and applications in biomedicine. Molecules. 27:29022022. View Article : Google Scholar : PubMed/NCBI | |
|
Hameed H, Faheem S, Paiva-Santos AC, Sarwar HS and Jamshaid M: A Comprehensive review of hydrogel-based drug delivery systems: classification, properties, recent trends, and applications. AAPS PharmSciTech. 25:642024. View Article : Google Scholar : PubMed/NCBI | |
|
Seliktar D: Designing cell-compatible hydrogels for biomedical applications. Science. 336:1124–1128. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Yap JX, Leo CP, Yasin NH, Show PL, Chu DT, Singh V and Derek CJC: Recent advances of natural biopolymeric culture scaffold: Synthesis and modification. Bioengineered. 13:2226–2247. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wechsler ME, Rao VV, Borelli AN and Anseth KS: Engineering the MSC Secretome: A hydrogel focused approach. Adv Healthc Mater. 10:e20019482021. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Yang T, Chen S, Deng N, Luo X, Liao R and Su B: Utilization of hydrogels in mesenchymal stem cell-based therapy for kidney diseases. Tissue Eng Part B Rev. 30:315–326. 2024. View Article : Google Scholar | |
|
Zhao L, Zhou Y, Zhang J, Liang H, Chen X and Tan H: Natural polymer-based hydrogels: From polymer to biomedical applications. Pharmaceutics. 15:25142023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M and Zhao X: Alginate hydrogel dressings for advanced wound management. Int J Biol Macromol. 162:1414–1428. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kou SG, Peters LM and Mucalo MR: Chitosan: A review of sources and preparation methods. Int J Biol Macromol. 169:85–94. 2021. View Article : Google Scholar | |
|
Yazdi MK, Taghizadeh A, Taghizadeh M, Stadler FJ, Farokhi M, Mottaghitalab F, Zarrintaj P, Ramsey JD, Seidi F, Saeb MR and Mozafari M: Agarose-based biomaterials for advanced drug delivery. J Control Release. 326:523–543. 2020. View Article : Google Scholar | |
|
Burdick JA and Prestwich GD: Hyaluronic acid hydrogels for biomedical applications. Adv Mater. 23:H41–H56. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Hu X, Xia Z and Cai K: Recent advances in 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B. 10:1486–1507. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Axpe E and Oyen ML: Applications of alginate-based bioinks in 3D Bioprinting. Int J Mol Sci. 17:19762016. View Article : Google Scholar : PubMed/NCBI | |
|
Troy E, Tilbury MA, Power AM and Wall JG: Nature-based biomaterials and their application in biomedicine. Polymers (Basel). 13:33212021. View Article : Google Scholar : PubMed/NCBI | |
|
Lee KY and Mooney DJ: Alginate: Properties and biomedical applications. Prog Polym Sci. 37:106–126. 2012. View Article : Google Scholar | |
|
Khatab S, Leijs MJ, van Buul G, Haeck J, Kops N, Nieboer M, Bos PK, Verhaar JAN, Bernsen M and van Osch GJVM: MSC encapsulation in alginate microcapsules prolongs survival after intra-articular injection, a longitudinal in vivo cell and bead integrity tracking study. Cell Biol Toxicol. 36:553–570. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Mohammadi M, Luong JC, Rodriguez SM, Cao R, Wheeler AE, Lau H, Li S, Shabestari SK, Chadarevian JP, Alexander M, et al: Controlled release of stem cell secretome attenuates inflammatory response against implanted biomaterials. Adv Healthc Mater. 9:e19018742020. View Article : Google Scholar : PubMed/NCBI | |
|
Bari E, Scocozza F, Perteghella S, Sorlini M, Auricchio F, Torre ML and Conti M: 3D bioprinted scaffolds containing mesenchymal stem/stromal lyosecretome: Next generation controlled release device for bone regenerative medicine. Pharmaceutics. 13:5152021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Onyeri S, Siewe M, Moshfeghian A and Madihally SV: In vitro characterization of chitosan-gelatin scaffolds for tissue engineering. Biomaterials. 26:7616–7627. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Wagenbrenner M, Mayer-Wagner S, Rudert M, Holzapfel BM and Weissenberger M: Combinations of hydrogels and mesenchymal stromal cells (MSCs) for cartilage tissue engineering-A review of the literature. Gels. 7:2172021. View Article : Google Scholar : PubMed/NCBI | |
|
Gao J, Liu R, Wu J, Liu Z, Li J, Zhou J, Hao T, Wang Y, Du Z, Duan C and Wang C: The use of chitosan based hydrogel for enhancing the therapeutic benefits of adipose-derived MSCs for acute kidney injury. Biomaterials. 33:3673–3681. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Lee CH, Singla A and Lee Y: Biomedical applications of collagen. Int J Pharm. 221:1–22. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Huang M, Li D, Chen J, Ji Y, Su T, Chen Y, Zhang Y, Wang Y, Li F, Chen S, et al: Comparison of the treatment efficacy of umbilical mesenchymal stem cell transplantation via renal subcapsular and parenchymal routes in AKI-CKD mice. Stem Cell Res Ther. 13:1282022. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Q, Zou Y, Arno MC, Chen S, Wang T, Gao J, Dove AP and Du J: Hydrogel scaffolds for differentiation of adipose-derived stem cells. Chem Soc Rev. 46:6255–6275. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
La Manna G, Bianchi F, Cappuccilli M, Cenacchi G, Tarantino L, Pasquinelli G, Valente S, Della Bella E, Cantoni S, Claudia C, et al: Mesenchymal stem cells in renal function recovery after acute kidney injury: Use of a differentiating agent in a rat model. Cell Transplant. 20:1193–1208. 2011. View Article : Google Scholar | |
|
Rohanizadeh R, Swain MV and Mason RS: Gelatin sponges (Gelfoam) as a scaffold for osteoblasts. J Mater Sci Mater Med. 19:1173–1182. 2008. View Article : Google Scholar | |
|
Bello AB, Kim D, Kim D, Park H and Lee SH: Engineering and functionalization of gelatin biomaterials: From cell culture to medical applications. Tissue Eng Part B Rev. 26:164–180. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Helminger M, Wu B, Kollmann T, Benke D, Schwahn D, Pipich V, Faivre D, Zahn D and Cölfen H: Synthesis and characterization of gelatin-based magnetic hydrogels. Adv Funct Mater. 24:3187–3196. 2014. View Article : Google Scholar | |
|
Mushtaq F, Raza ZA, Batool SR, Zahid M, Onder OC, Rafique A and Nazeer MA: Preparation, properties, and applications of gelatin-based hydrogels (GHs) in the environmental, technological, and biomedical sectors. Int J Biol Macromol. 218:601–633. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sobreiro-Almeida R, Quinteira R and Neves NM: Renal regeneration: The role of extracellular matrix and current ECM-based tissue engineered strategies. Adv Healthc Mater. 10:e21001602021. View Article : Google Scholar : PubMed/NCBI | |
|
Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP and Ott HC: Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med. 19:646–651. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JW, Nam SA, Yi J, Kim JY, Lee JY, Park SY, Sen T, Choi YM, Lee JY, Kim HL, et al: Kidney decellularized extracellular matrix enhanced the vascularization and maturation of human kidney organoids. Adv Sci (Weinh). 9:e21035262022. View Article : Google Scholar : PubMed/NCBI | |
|
Saldin LT, Cramer MC, Velankar SS, White LJ and Badylak SF: Extracellular matrix hydrogels from decellularized tissues: Structure and function. Acta Biomater. 49:1–15. 2017. View Article : Google Scholar : | |
|
Zhou C, Zhou L, Liu J, Xu L, Xu Z, Chen Z, Ge Y, Zhao F, Wu R, Wang X, et al: Kidney extracellular matrix hydrogel enhances therapeutic potential of adipose-derived mesenchymal stem cells for renal ischemia reperfusion injury. Acta Biomater. 115:250–263. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Chee HL, Chong YT, Chan BQY, Xue K, Lim PC, Loh XJ and Wang F: Hofmeister effect mediated strong PHEMA-Gelatin hydrogel actuator. ACS Appl Mater Interfaces. 14:23826–23838. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zullo JA, Nadel EP, Rabadi MM, Baskind MJ, Rajdev MA, Demaree CM, Vasko R, Chugh SS, Lamba R, Goligorsky MS and Ratliff BB: The secretome of hydrogel-coembedded endothelial progenitor cells and mesenchymal stem cells instructs macrophage polarization in endotoxemia. Stem Cells Transl Med. 4:852–861. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Tietze S, Kräter M, Jacobi A, Taubenberger A, Herbig M, Wehner R, Schmitz M, Otto O, List C, Kaya B, et al: Spheroid culture of mesenchymal stromal cells results in morphorheological properties appropriate for improved microcirculation. Adv Sci (Weinh). 6:18021042019. View Article : Google Scholar : PubMed/NCBI | |
|
Shoma Suresh K, Bhat S, Guru BR, Muttigi MS and Seetharam RN: A nanocomposite hydrogel delivery system for mesenchymal stromal cell secretome. Stem Cell Res Ther. 11:2052020. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Jha AK, Harrington DA, Farach-Carson MC and Jia X: Hyaluronic acid-based hydrogels: From a natural polysaccharide to complex networks. Soft Matter. 8:3280–3294. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kleinman HK and Martin GR: Matrigel: Basement membrane matrix with biological activity. Semin Cancer Biol. 15:378–386. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Goetzke R, Franzen J, Ostrowska A, Vogt M, Blaeser A, Klein G, Rath B, Fischer H, Zenke M and Wagner W: Does soft really matter? Differentiation of induced pluripotent stem cells into mesenchymal stromal cells is not influenced by soft hydrogels. Biomaterials. 156:147–158. 2018. View Article : Google Scholar | |
|
Najafi H, Abolmaali SS, Heidari R, Valizadeh H, Jafari M, Tamaddon AM and Azarpira N: Nitric oxide releasing nanofibrous Fmoc-dipeptide hydrogels for amelioration of renal ischemia/reperfusion injury. J Control Release. 337:1–13. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lin RZ and Chang HY: Chang, recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol J. 3:1172–1184. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ryu NE, Lee SH and Park H: Spheroid culture system methods and applications for mesenchymal stem cells. Cells. 8:16202019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WY, Chen LC, Jhuang YT, Lin YJ, Hung PY, Ko YC, Tsai MY, Lee YW, Hsu LW, Yeh CK, et al: Injection of hybrid 3D spheroids composed of podocytes, mesenchymal stem cells, and vascular endothelial cells into the renal cortex improves kidney function and replenishes glomerular podocytes. Bioeng Transl Med. 6:e102122021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Chen C, Hu L and Hou J: Gene-modified mesenchymal stem cell-based therapy in renal ischemia-reperfusion injury. Curr Gene Ther. 17:453–460. 2017. View Article : Google Scholar | |
|
Roudkenar MH, Halabian R, Tehrani HA, Amiri F, Jahanian-Najafabadi A, Roushandeh AM, Abbasi-Malati Z and Kuwahara Y: Lipocalin 2 enhances mesenchymal stem cell-based cell therapy in acute kidney injury rat model. Cytotechnology. 70:103–117. 2018. View Article : Google Scholar : | |
|
Ni W, Zhang Y and Yin Z: The protective mechanism of Klotho gene-modified bone marrow mesenchymal stem cells on acute kidney injury induced by rhabdomyolysis. Regen Ther. 18:255–267. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Yuan Z, Zhong W and Wei Y: Stem cell as vehicles of antibody in treatment of lymphoma: A novel and potential targeted therapy. Stem Cell Rev Rep. 17:829–841. 2021. View Article : Google Scholar | |
|
Chen Y, Qian H, Zhu W, Zhang X, Yan Y, Ye S, Peng X, Li W and Xu W: Hepatocyte growth factor modification promotes the amelioration effects of human umbilical cord mesenchymal stem cells on rat acute kidney injury. Stem Cells Dev. 20:103–113. 2011. View Article : Google Scholar | |
|
Hagiwara M, Shen B, Chao L and Chao J: Kallikrein-modified mesenchymal stem cell implantation provides enhanced protection against acute ischemic kidney injury by inhibiting apoptosivgs and inflammation. Hum Gene Ther. 19:807–819. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Patzak A and Zhang J: CXCR4-overexpressing bone marrow-derived mesenchymal stem cells improve repair of acute kidney injury. Am J Physiol Renal Physiol. 305:F1064–F1073. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Tian J, Cheng J and Zhang J: Migration of CXCR4 gene-modified bone marrow-derived mesenchymal stem cells to the acute injured kidney. J Cell Biochem. 114:2677–2689. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Wang H, Han G, Cheng J, Hu W and Zhang J: Enhanced proliferation and differentiation of HO-1 gene-modified bone marrow-derived mesenchymal stem cells in the acute injured kidney. Int J Mol Med. 42:946–956. 2018.PubMed/NCBI | |
|
Liu N, Wang H, Han G, Tian J, Hu W and Zhang J: Alleviation of apoptosis of bone marrow-derived mesenchymal stem cells in the acute injured kidney by heme oxygenase-1 gene modification. Int J Biochem Cell Biol. 69:85–94. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
da Cunha MGM, Zia S, Beckmann DV, Carlon MS, Arcolino FO, Albersen M, Pippi NL, Graça DL, Gysemans C, Carmeliet P, et al: Vascular endothelial growth factor up-regulation in human amniotic fluid stem cell enhances nephroprotection after ischemia-reperfusion injury in the rat. Crit Care Med. 45:e86–e96. 2017. View Article : Google Scholar | |
|
Yuzeng Q, Weiyang H, Xin G, Qingson Z, Youlin K and Ke R: Effects of transplantation with marrow-derived mesenchymal stem cells modified with survivin on renal ischemia-reperfusion injury in mice. Yonsei Med J. 55:1130–1137. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Pawitan JA, Bui TA, Mubarok W, Antarianto RD, Nurhayati RW, Dilogo IH and Oceandy D: Enhancement of the therapeutic capacity of mesenchymal stem cells by genetic modification: A systematic review. Front Cell Dev Biol. 8:5877762020. View Article : Google Scholar : PubMed/NCBI | |
|
Kresse JC, Gregersen E, Atay JCL, Eijken M and Nørregaard R: Does the route matter? A preclinical review of mesenchymal stromal cell delivery to the kidney. APMIS. 131:687–697. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Schrepfer S, Deuse T, Reichenspurner H, Fischbein MP, Robbins RC and Pelletier MP: Stem cell transplantation: The lung barrier. Transplant Proc. 39:573–576. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, Laine GA and Cox CS Jr: Pulmonary passage is a major obstacle for intravenous stem cell delivery: The pulmonary first-pass effect. Stem Cells Dev. 18:683–692. 2009. View Article : Google Scholar | |
|
Schmuck EG, Koch JM, Centanni JM, Hacker TA, Braun RK, Eldridge M, Hei DJ, Hematti P and Raval AN: Biodistribution and clearance of human mesenchymal stem cells by quantitative three-dimensional cryo-imaging after intravenous infusion in a rat lung injury model. Stem Cells Transl Med. 5:1668–1675. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, Granton J and Stewart DJ; Canadian Critical Care Trials Group: Safety of cell therapy with mesenchymal stromal cells (SafeCell): A systematic review and meta-analysis of clinical trials. PLoS One. 7:e475592012. View Article : Google Scholar : PubMed/NCBI | |
|
Kansu E: Thrombosis in stem cell transplantation. Hematology. 17(Suppl 1): S159–S162. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Cui LL, Kerkelä E, Bakreen A, Nitzsche F, Andrzejewska A, Nowakowski A, Janowski M, Walczak P, Boltze J, Lukomska B and Jolkkonen J: The cerebral embolism evoked by intra-arterial delivery of allogeneic bone marrow mesenchymal stem cells in rats is related to cell dose and infusion velocity. Stem Cell Res Ther. 6:112015. View Article : Google Scholar : PubMed/NCBI | |
|
Sanchez-Diaz M, Quiñones-Vico MI, de la Torre RS, Montero-Vílchez T, Sierra-Sánchez A, Molina-Leyva A and Arias-Santiago S: Biodistribution of mesenchymal stromal cells after administration in animal models and humans: A systematic review. J Clin Med. 10:29252021. View Article : Google Scholar : PubMed/NCBI | |
|
Bagno LL, Salerno AG, Balkan W and Hare JM: Mechanism of Action of Mesenchymal Stem Cells (MSCs): Impact of delivery method. Expert Opin Biol Ther. 22:449–463. 2022. View Article : Google Scholar : | |
|
Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW and Okano T: Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. J Control Release. 330:696–704. 2021. View Article : Google Scholar |