Figure 2 - uploaded by Zongjin Li
Content may be subject to copyright.
RNA sequencing (RNA-seq) analysis revealed that nitric oxide (NO) improves the antioxidant capacity of engineered mesenchymal stem cells (eMSCs). (A) Gene Ontology (GO) category analysis of differentially expressed genes (DEGs) for biological processes. Only significantly enriched terms with corrected p< 0.05 are indicated. The top 20 enriched biological processes are ranked by the number of DEGs. (B) Gene set expression analysis (GSEA) revealed enrichment for the cell redox homeostasis pathway. The normalized enrichment score (NES), false discovery rate (FDR), and p value are indicated in the insert. (C, D) Heatmap of representative antioxidation (C) and proliferation/survival (D)-related genes. Fold change > 1.5, p < 0.05. Red indicates upregulation, and blue indicates downregulation. (E) GSEA revealed enrichment for apoptosis pathways. (F) Schematic representation of the prosurvival potential of the NO-eMSC system. eMSC+MGP, eMSCs with MGP administration. The online version of this article includes the following figure supplement(s) for figure 2:

RNA sequencing (RNA-seq) analysis revealed that nitric oxide (NO) improves the antioxidant capacity of engineered mesenchymal stem cells (eMSCs). (A) Gene Ontology (GO) category analysis of differentially expressed genes (DEGs) for biological processes. Only significantly enriched terms with corrected p< 0.05 are indicated. The top 20 enriched biological processes are ranked by the number of DEGs. (B) Gene set expression analysis (GSEA) revealed enrichment for the cell redox homeostasis pathway. The normalized enrichment score (NES), false discovery rate (FDR), and p value are indicated in the insert. (C, D) Heatmap of representative antioxidation (C) and proliferation/survival (D)-related genes. Fold change > 1.5, p < 0.05. Red indicates upregulation, and blue indicates downregulation. (E) GSEA revealed enrichment for apoptosis pathways. (F) Schematic representation of the prosurvival potential of the NO-eMSC system. eMSC+MGP, eMSCs with MGP administration. The online version of this article includes the following figure supplement(s) for figure 2:

Source publication
Article
Full-text available
ELife digest Animals are made up of cells of different types, with each type of cell specializing on a specific role. But for the body to work properly, the different types of cells must be able to coordinate with each other to respond to internal and external stimuli. This can be achieved through signaling molecules, that is, molecules released by...

Citations

... Mitochondria support multiple functions in cells, such as maintenance of redox balance, initiation of inflammation and regulation of cell death [30]. Increasing evidence has implicated mitochondrial oxidative stress, and cell death has been identified as the main cause of AKI, leading to subsequent extensive tissue damage [31,32]. We tested whether pEV treatment could reduce mitochondrial-related oxidative damage and cell death ( Figure 3A). ...
... Combining MSCs with NO-releasing biomaterials has been demonstrated to enhance their angiogenic, anti-apoptotic, and antioxidative stress capabilities, with great potential for treating peripheral ischemia, kidney diseases, and cardiovascular disease [18,31,47,48]. In this study, we pretreated MSCs with an NO-releasing polymer, which was linked to chondroitin sulfate and was capable of maintaining a stable low-concentration (nM-level) release for at least 48 h [18,20,21]. ...
... However, despite recent advances, the clinical application of NO donors remains limited by poor solubility, burst release effects, and off-target nitrosative stress [28]. Although our controlled-release NO system partially alleviates some of these issues, scale-up production still faces challenges, including batch variations in NO loading efficiency and the complexity of synthesizing biodegradable materials [31,66]. Moreover, standardizing the use of NO-primed MSCs remains a key challenge. ...
Article
Full-text available
Background: The disruption of mitochondrial homeostasis in acute kidney injury (AKI) is an important factor that drives persistent renal dysfunction. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown great therapeutic potential in AKI, but insufficient specificity of targeting the impaired mitochondrial function. Herein, we developed an engineered nitric oxide (NO)-primed MSC-EVs (pEVs) to restore mitochondrial homeostasis for AKI therapy. Methods: A cisplatin-induced AKI model was established to investigate the therapeutic effects of MSC-EVs. Proteomic and Western blot analyses compared mitochondrial cargos and functional assays included mitochondrial complex I activity and Adenosine triphosphate (ATP) quantification. Mitochondrial transfer was tracked using flow cytometry and confocal imaging. Mitochondrial dynamics, oxidative stress, and apoptosis were evaluated through ATP measurement, western blotting and rotenone-mediated respiratory chain inhibition. Results: Our data indicated that pEVs outperformed cEVs in restoring renal function and histopathology. Additionally, a reduction in mitochondria-associated oxidative stress and cell death was observed. Proteomic profiling revealed that NO priming enriched pEVs with mitochondrial complex I components, which directly enhanced their bioenergetic capacity, as evidenced by higher mitochondrial complex I activity and elevated ATP production compared to cEVs. In vivo tracking confirmed targeted delivery of pEV-derived mitochondrial contents to renal tubular cells, reducing mitochondrial reactive oxygen species (ROS) and restoring mitochondrial mass. Crucially, mitochondria-depleted pEVs abolished these therapeutic effects, establishing mitochondrial cargos as the primary therapeutic driver. Furthermore, pEVs activated a pro-survival cascade in recipient cells, showing superior efficacy in promoting mitochondrial biogenesis, dynamics, and mitophagy, thereby restoring renal mitochondrial homeostasis. Conclusion: Our study elucidated a mitochondria-targeted therapeutic strategy enabled by engineered EVs that deliver functional cargo to restore mitochondrial homeostasis. These advances provide transformative potential for AKI and other mitochondrial disorders.
... Human placenta-derived mesenchymal stem cells (hP-MSCs), purchased from AmCellGene Co. Ltd. (Tianjin, China), were cultured in line with previous reports [30][31][32]. Briefly, hP-MSCs were maintained in Dulbecco's modified Eagle medium/nutrient mixture F-12 (DMEM/F-12; Gibco, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS; Gibco), 100 U/mL penicillin and 100 µg/mL streptomycin (Gibco) at 37 °C in 5% CO 2 . ...
Article
Full-text available
Background Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD) globally, presenting a significant therapeutic challenge. Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs) have emerged as promising therapeutic agents. This study explored the therapeutic effects and mechanisms of EVs derived from human placental mesenchymal stem cells (hP-MSCs) on DKD. Methods EVs were isolated from cultured hP-MSCs and administered to streptozotocin (STZ)-induced diabetic mice and high glucose–treated glomerular mesangial cells. The therapeutic impact of EVs was assessed through histological analysis and biochemical assays. miR-99b-5p expression in EVs and its role in modulating the mechanistic target of rapamycin (mTOR)/autophagy pathway were examined via western blotting and RT‒qPCR. Results Treatment with hP-MSC-derived EVs significantly alleviated renal fibrosis and improved renal function in DKD models. These EVs were enriched with miR-99b-5p, which targeted and inhibited mTOR signaling, thereby increasing autophagic activity and reducing cellular proliferation and extracellular matrix accumulation in renal tissues. Conclusions hP-MSC-derived EVs can mitigate renal injury in DKD by modulating the miR-99b-5p/mTOR/autophagy pathway. These findings suggest a potential cell-free therapeutic strategy for managing DKD.
... Electrical field stimulation enhances the localization of MSCs, thereby improving their therapeutic potential against acute cisplatin nephrotoxicity [37]. Furthermore, the development of an MSC system for nitric oxide delivery facilitates AKI therapy by promoting angiogenesis, stimulating cell proliferation, and exerting anti-inflammatory effects through macrophage polarization [38]. ...
Article
Full-text available
Renal diseases, particularly acute kidney injury (AKI) and chronic kidney disease (CKD), are significant global health challenges. These conditions impair kidney function and can lead to serious complications, including cardiovascular diseases, which further exacerbate the public health burden. Currently, the global AKI mortality rate is alarmingly high (20%–50%); CKD is projected to emerge as a major global health burden by 2040. Existing treatments such as hemodialysis and kidney transplantation have limited effectiveness and are often associated with adverse effects. Mesenchymal stem cells (MSCs) offer considerable potential for treating renal diseases owing to their regenerative and immunomodulatory properties. Thus, this review focuses on the application of MSCs in renal disease, discusses fundamental research findings, and evaluates their application in clinical trials. Moreover, we discuss the impact and safety of MSCs as a therapeutic option and highlight challenges and potential directions for their clinical application. We selected research articles from PubMed published within the last 5 years (from 2019), focusing on high-impact journals and clinical trial data, and included a few key studies predating 2019. Considerations included the novelty of the research, sample size, experimental design, and data reliability. With advancements in single-cell sequencing, CRISPR/Cas9 gene editing, and other cutting-edge technologies, future MSC research will explore combination therapies and personalized treatments to provide more promising, safer treatments with reduced adverse reactions and enhanced therapeutic outcomes. These advances will improve kidney disease treatment methods, enhance patient quality of life, and maximize the benefits of MSC therapies.
... S-nitrosylated l-serine-modified dendrimer as NO donor targeting the kidney for IRI prevention [279]. In addition, genetically engineered mesenchymal stem cells as nitric oxide reservoirs and can be used to treat AKI [280]. Targeting the Keap1-Nrf2 system with drugs such as entacapone prevents kidney disease progression [281,282] (Table 3). ...
Article
Full-text available
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI. Graphical abstract
... Strategy through regulation of an instructive microenvironment is needed to recruit and stimulate resident renal progenitor cells or stem cells in vivo, to provide signals for native healing cascades, and to promote cell proliferation and differentiation of vulnerable proximal tubules for kidney repair after AKI. 4,5 Recent studies have shown that injury-induced PGE 2 secretion plays an important role in the regulation of endogenous stem cells for myocardial regeneration, intestinal regeneration and liver regeneration after injury. [6][7][8][9] Furthermore, Sox9 is upregulated in the early developing nephron and is located at the tips of the branching ureteral network where progenitors reside for the entire ureteral network. ...
Article
Full-text available
Uncovering mechanisms of endogenous regeneration and repair through resident stem cell activation will allow us to develop specific therapies for injuries and diseases by targeting resident stem cell lineages. Sox9⁺ stem cells have been reported to play an essential role in acute kidney injury (AKI). However, a complete view of the Sox9⁺ lineage was not well investigated to accurately elucidate the functional end state and the choice of cell fate during tissue repair after AKI. To identify the mechanisms of fate determination of Sox9⁺ stem cells, we set up an AKI model with prostaglandin E2 (PGE2) treatment in a Sox9 lineage tracing mouse model. Single‐cell RNA sequencing (scRNA‐seq) was performed to analyse the transcriptomic profile of the Sox9⁺ lineage. Our results revealed that PGE2 could activate renal Sox9⁺ cells and promote the differentiation of Sox9⁺ cells into renal proximal tubular epithelial cells and inhibit the development of fibrosis. Furthermore, single‐cell transcriptome analysis demonstrated that PGE2 could regulate the restoration of lipid metabolism homeostasis in proximal tubular epithelial cells by participating in communication with different cell types. Our results highlight the prospects for the activation of endogenous renal Sox9⁺ stem cells with PGE2 for the regenerative therapy of AKI.
... The therapeutic effectiveness of MSCs was enhanced by priming them with biofactors and chemical factors, which controlled their secretion [10,12] (Table 2). Previous works have investigated the potential roles of MSC priming by a variety of biofactors, such as IFN-γ [13,31,32], TNF-α [14,33], IL-1α-β [15,34], FGF-2 [16], LPS [35], IL-17A [17], TLR3 [36], IGF-1 [37,38], IL-6 [39], IL-8 [40,41], IL-3 [42,43], IL-25 [44], even gaseous signal molecule nitric oxide [45][46][47]. Their findings showed promise in enhancing MSC treatment profiles for various diseases. For example, priming MSCs with TNF-and IFN-γ aided bone regeneration and immune modulation and induced unique microRNA expression in MSCs and their exosomes [18,20]. ...
Article
Full-text available
Mesenchymal stem cells (MSCs) have gained substantial attention in regenerative medicine due to their multi-lineage differentiation potential and immunomodulatory capabilities. MSCs have demonstrated therapeutic promise in numerous preclinical and clinical studies across a variety of diseases, including neurodegenerative disorders, cardiovascular diseases, and autoimmune conditions. Recently, priming MSCs has emerged as a novel strategy to enhance their therapeutic efficacy by preconditioning them for optimal survival and function in challenging in vivo environments. This study presented a comprehensive bibliometric analysis of research activity in the field of priming mesenchymal stem cells (MSCs) from 2003 to 2023. Utilizing a dataset of 585 documents, we explored research trends, leading authors and countries, productive journals, and frequently used keywords. We also explored priming strategies to augment the therapeutic efficacy of MSCs. Our findings show increasing research productivity with a peak in 2019, identified the United States as the leading contributor, and highlighted WANG JA as the most prolific author. The most published journal was Stem Cell Research & Therapy. Keyword analysis revealed core research areas emerging hotspots, while coword and cited sources visualizations elucidated the conceptual framework and key information sources. Further studies are crucial to advance the translation of primed MSCs from bench to bedside, potentially revolutionizing the landscape of regenerative medicine.
Article
Full-text available
This review summarizes recent advances in understanding the role of the nitric oxide (NO) and cyclic GMP (cGMP) pathway in stem cells. The levels of expression of various components of the pathway are changed during the differentiation of pluripotent embryonic stem cells. In undifferentiated stem cells, NO regulates self-renewal and survival predominantly through cGMP-independent mechanisms. Natriuretic peptides influence the growth of undifferentiated stem cells by activating particulate isoforms of guanylyl cyclases in a cGMP-mediated manner. The differentiation, recruitment, survival, migration, and homing of partially differentiated precursor cells of various types are sensitive to regulation by endogenous levels of NO and natriuretic peptides produced by stem cells, within surrounding tissues, and by the application of various pharmacological agents known to influence the cGMP pathway. Numerous drugs and formulations target various components of the cGMP pathway to influence the therapeutic efficacy of stem cell-based therapies. Thus, pharmacological manipulation of the cGMP pathway in stem cells can be potentially used to develop novel strategies in regenerative medicine.
Article
Interleukin-4 (IL-4), which is traditionally associated with inflammation, has emerged as a key player in tissue regeneration. Produced primarily by T-helper 2 (Th2) and other immune cells, IL-4 activates endogenous lymphocytes and promotes M2 macrophage polarization, both of which are crucial for tissue repair. Moreover, IL-4 stimulates the proliferation and differentiation of various cell types, contributing to efficient tissue regeneration, and shows promise for promoting tissue regeneration after injury. This review explores the multifaceted roles of IL-4 in tissue repair, summarizing its mechanisms and potential for clinical application. This review delves into the multifaceted functions of IL-4, including its immunomodulatory effects, its involvement in tissue regeneration, and its potential therapeutic applications. We discuss the mechanisms underlying IL-4-induced M2 macrophage polarization, a crucial process for tissue repair. Additionally, we explore innovative strategies for delivering IL-4, including gene therapy, protein-based therapies, and cell-based therapies. By leveraging the regenerative properties of IL-4, we can potentially develop novel therapies for various diseases, including chronic inflammatory disorders, autoimmune diseases, and organ injuries. While early research has shown promise for the application of IL-4 in regenerative medicine, further studies are needed to fully elucidate its therapeutic potential and optimize its use.