Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Apr 26, 2025; 17(4): 102945
Published online Apr 26, 2025. doi: 10.4252/wjsc.v17.i4.102945
Stem cell therapy for intervertebral disc degeneration: Clinical progress with exosomes and gene vectors
Zhi-Peng Li, Yu-Hua Ruan, Peng Wang, Wei-Ping Fu, Rui-Bo Wang, Xiao-Dong Tang, Qi Zhang, Sen-Li Li, He Yin, Cheng-Jin Li, Yao-Bin Wang, Chang-Jiang Zhang, Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
Zhi-Peng Li, Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
Han Li, Department of Orthopedics, Affiliated Dongyang Hospital of Wenzhou Medical University, Jinhua 322100, Zhejiang Province, China
Meng-Ting Zhu, Department of Neurology, Union Medical College Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
Yi-Gong Tian, Rui-Ning Han, Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
Yao-Bin Wang, Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
ORCID number: Zhi-Peng Li (0000-0002-0355-7889); Han Li (0000-0001-7485-2642); Yu-Hua Ruan (0009-0001-7851-8155); Peng Wang (0009-0009-4211-5459); Meng-Ting Zhu (0009-0003-9429-1784); Wei-Ping Fu (0009-0000-6078-2671); Rui-Bo Wang (0009-0004-1280-9971); Xiao-Dong Tang (0000-0003-2366-6772); Cheng-Jin Li (0009-0000-5668-6839); Rui-Ning Han (0000-0001-8898-3627); Yao-Bin Wang (0000-0001-7004-5237); Chang-Jiang Zhang (0009-0006-2769-1413).
Co-first authors: Zhi-Peng Li and Han Li.
Co-corresponding authors: Yao-Bin Wang and Chang-Jiang Zhang.
Author contributions: Li ZP and Li H contributed equally to this work and are co-first authors of this manuscript; Li ZP and Li H conceptualized and designed the study, developed the research framework, and created the artwork; Ruan YH and Wang P were responsible for data curation and formal analysis, ensuring the accuracy and consistency of the results; Zhu MT developed the software essential for data processing; Fu WP supervised the project, providing critical oversight and guidance; Wang RB and Tang XD performed critical revisions of the manuscript, enhancing its clarity and scientific rigor; Zhang Q and Li SL conducted the validation and refined the methodology; Yin H and Li CJ managed resources and visualization, ensuring the effective presentation of findings, and contributed to the investigation and conducted key experimental work; Zhang CJ was responsible for funding acquisition and project administration, securing essential resources and ensuring efficient project coordination; Wang YB and Zhang CJ conducted the literature review, performed the data analysis and interpretation, and reviewed and edited the final manuscript; Wang YB and Zhang CJ contributed equally to this manuscript, and both served as co-corresponding authors because of their significant roles in resource management, data interpretation, and manuscript finalization; Each corresponding author made distinct and critical contributions to collaborative writing and the final review of the manuscript.
Supported by Henan Province Key Research and Development Program, No. 231111311000; Henan Provincial Science and Technology Research Project, No. 232102310411; Henan Province Medical Science and Technology Key Project, No. LHGJ20220566 and No. LHGJ20240365; Henan Province Medical Education Research Project, No. WJLX2023079; and Zhengzhou Medical and Health Technology Innovation Guidance Program, No. 2024YLZDJH022.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Chang-Jiang Zhang, MD, Chief Physician, Professor, Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfu Qianjie, Erqi District, Zhengzhou 450052, Henan Province, China. changjiangzhang1968@outlook.com
Received: November 5, 2024
Revised: January 25, 2025
Accepted: March 10, 2025
Published online: April 26, 2025
Processing time: 171 Days and 17.3 Hours

Abstract

Intervertebral disc degeneration is a leading cause of lower back pain and is characterized by pathological processes such as nucleus pulposus cell apoptosis, extracellular matrix imbalance, and annulus fibrosus rupture. These pathological changes result in disc height loss and functional decline, potentially leading to disc herniation. This comprehensive review aimed to address the current challenges in intervertebral disc degeneration treatment by evaluating the regenerative potential of stem cell-based therapies, with a particular focus on emerging technologies such as exosomes and gene vector systems. Through mechanisms such as differentiation, paracrine effects, and immunomodulation, stem cells facilitate extracellular matrix repair and reduce nucleus pulposus cell apoptosis. Despite recent advancements, clinical applications are hindered by challenges such as hypoxic disc environments and immune rejection. By analyzing recent preclinical and clinical findings, this review provided insights into optimizing stem cell therapy to overcome these obstacles and highlighted future directions in the field.

Key Words: Exosomes; Extracellular matrix repair; Gene vector system; Hypoxic environment; Intervertebral disc degeneration; Mesenchymal stem cells; Regenerative medicine; Stem cell therapy

Core Tip: The application of stem cell therapy in intervertebral disc degeneration has shown significant regenerative potential. Through differentiation, paracrine effects, and immunomodulation, stem cells can repair damaged extracellular matrix and slow the degenerative process. Emerging technologies such as exosomes and gene vector systems further enhance the therapeutic effects of stem cells, improving cell survival rates and regenerative capabilities. However, the long-term survival of stem cells in a hypoxic environment, the optimal injection dosage, and their long-term safety still requires further investigation. Future interdisciplinary collaboration will drive the clinical translation of stem cell therapy, making it a standard option for the treatment of intervertebral disc degeneration.



INTRODUCTION

Intervertebral disc degeneration (IVDD) is one of the primary causes of lower back pain, significantly contributing to disability and reducing the quality of life of millions of people worldwide. IVDD is characterized by the gradual loss of disc function with age and is driven primarily by the degeneration of nucleus pulposus cells (NPCs)[1,2]. NPCs play a crucial role in maintaining disc hydration and elasticity; however, over time, their number and functionality decline. Concurrently, the depletion of nucleus pulposus progenitor cells (ProNPs) exacerbates the reduction in extracellular matrix (ECM) synthesis, further diminishing disc hydration and elasticity[3-5]. Additionally, damage to the annulus fibrosus (AF) compromises the structural integrity of the disc, often leading to herniation[6,7]. This, in turn, can compress surrounding neural tissue, causing neuropathic pain and motor dysfunction[8]. These pathological changes result in significant impairment of daily activities, with severe cases progressing to disability.

Current treatment strategies for IVDD include conservative approaches such as medication and physical therapy as well as surgical interventions. Conservative treatments may provide symptomatic relief in the early stages but fail to halt disease progression. Surgical approaches, including discectomy and spinal fusion, address nerve compression but are invasive and unable to restore normal disc function[9]. Moreover, these methods only offer temporary relief and do not reverse the degenerative processes of the disc[10]. Given these limitations, there is a pressing need for innovative therapies capable of addressing the underlying mechanisms of IVDD, promoting tissue repair, and improving long-term outcomes.

Stem cell therapy has emerged as a promising regenerative approach for IVDD, offering the potential to repair damaged tissues, restore disc function, and slow disease progression. Stem cells can differentiate into NPCs and AF cells, and their paracrine effects promote ECM repair, inhibit inflammation, and reduce apoptosis[11-13]. This therapy provides a particularly viable option for patients who are unresponsive to traditional treatments or unable to tolerate surgery.

In recent years, advancements in technologies such as stem cell-derived exosomes and gene vector systems have further enhanced the potential of stem cell therapy. By introducing specific genes into stem cells, gene vector systems can increase cell survival in hypoxic environments, improve ECM repair, and increase anti-inflammatory capabilities[14-17]. Stem cell-derived exosomes, as nanoscale vesicles carrying therapeutic molecules, have demonstrated anti-inflammatory, antiapoptotic, and regenerative effects in IVDD models[12,16]. These developments collectively offer innovative strategies to optimize the therapeutic efficacy of stem cell-based treatments for IVDD.

This review aimed to synthesize the latest advancements in stem cell therapy for IVDD, focusing on the mechanisms underlying stem cell-mediated repair and the emerging roles of exosomes and gene vector systems. By addressing the challenges of clinical translation, including poor cell survival, limited ECM repair, and immune rejection, this work highlighted the potential of multidisciplinary approaches to bridge the gap between experimental findings and clinical applications.

MECHANISMS OF STEM CELL ACTION IN IVDD
Sources and types of stem cells

Stem cell therapy has promising applications in the treatment of IVDD. Stem cells from various sources offer distinct advantages in terms of their differentiation potential, anti-inflammatory properties, and clinical application prospects. Bone marrow-derived mesenchymal stem cells (BMSCs) are the most extensively studied stem cell type and possess significant self-renewal and multipotent differentiation abilities[18]. BMSCs regulate ECM production and reduce inflammation and apoptosis by secreting cytokines such as transforming growth factor beta (TGF-β), insulin-like growth factor 1 (IGF-1), and vascular endothelial growth factor (VEGF)[16,17]. In animal models, BMSC injections have demonstrated disc repair effects, with clinical studies also showing the potential of BMSCs to alleviate pain and promote NPC regeneration[3,16,19,20]. However, the hypoxic environment of the intervertebral disc presents a considerable challenge for BMSCs, and their long-term survival under hypoxic conditions requires further research[21,22].

Compared with BMSCs, adipose-derived mesenchymal stem cells (ADSCs) have similar multipotent differentiation potential but offer clinical advantages due to their wide availability and ease of acquisition[23,24]. ADSCs can promote NPC proliferation and enhance the production of essential ECM components, such as collagen and proteoglycans, thereby supporting disc structure and function[23,25,26]. Although ADSCs exhibit strong anti-inflammatory effects, their long-term survival in harsh, hypoxic disc environments remains challenging. Strategies to improve their therapeutic efficacy, such as genetic modification or combination with biomaterials such as hydrogels, are currently under exploration[26,27].

Owing to their low immunogenicity and high proliferative capacity, umbilical cord-derived mesenchymal stem cells (UC-MSCs) have become an attractive source for allogeneic transplantation[28]. By secreting anti-inflammatory and growth factors, including TGF-β, VEGF, and interleukin (IL)-10, UC-MSCs not only help mitigate inflammation but also promote ECM repair and regeneration[29-31]. While UC-MSCs have shown promising repair effects in vitro and in animal studies, their long-term efficacy and safety in clinical settings require further validation. Future research may focus on optimizing the application of UC-MSCs in disc repair through gene editing or exosome technology[32].

Additionally, nuclear ProNPs, as precursor cells with multipotent differentiation capability, play a critical role in disc repair by differentiating into mature NPCs[33-35]. Studies indicate that ProNPs can promote NPC regeneration and slow disc degeneration under hypoxic conditions by activating signaling pathways such as the Wnt and TGF-β pathways[5,36,37]. Future research will focus on the potential clinical application of these progenitor cells, particularly in gene regulation and exosome therapy[37].

The choice between autologous and allogeneic stem cells in clinical applications should be based on specific case requirements. With a lower risk of immune rejection, autologous stem cells are suitable for personalized treatment but are limited by complex harvesting processes and limited cell numbers[38-40]. In contrast, allogeneic stem cells, especially UC-MSCs, are easier to obtain and are suitable for large-scale treatments, although they carry a certain risk of immune rejection[41]. Although allogeneic stem cells may trigger immune responses during transplantation, technologies such as gene editing and immune modulation can mitigate this risk[39]. Future studies will aim to optimize the application of both cell types to improve efficacy and minimize side effects (Tables 1 and 2).

Table 1 Comparison of stem cell types.
Stem cell source
Harvesting method
Differentiation potential
Anti-inflammatory action
Applicable scenarios
Prospects for IVDD repair application
BMSCsBone marrow extraction, complex procedureMultipotent differentiation potential, can differentiate into NPCs and AF cellsSecretes TGF-β, IGF-1, VEGF; regulates ECM metabolism, reduces NP cell apoptosis and inflammationSuitable for severe disc damageDemonstrates repair potential in in vivo and in vitro studies and early clinical research; long-term efficacy still requires validation
ADSCsFat extraction, relatively simpleMultipotent differentiation ability, promotes ECM productionSecretes anti-inflammatory factors, helps reduce NP cell apoptosis and inflammationMinimally invasive treatment, suitable for wide applicationShows promising results in preclinical studies; combining with materials like hydrogels can enhance therapeutic effects
UC-MSCsExtraction from umbilical cord tissue, easy to obtainMultipotent differentiation potential, high proliferative capacitySecretes TGF-β, VEGF, IL-10; exhibits significant anti-inflammatory and anti-apoptotic effectsSuitable for allogeneic transplantation and large-scale treatment, low immunogenicityShows good results in preclinical studies; can be further optimized with gene editing and exosome technologies
ProNPsExtraction from the disc, relatively complexDifferentiates into mature NP cellsPromotes NP tissue regeneration through paracrine effectsSuitable for NP regeneration, high potentialPromotes ECM production by activating specific signaling pathways, high future application potential
Table 2 Clinical and in vivo/in vitro experimental data.
Experiment type
Cell source
Main observed data
Comparative effects
Ref.
In vitro experimentBMSCs, NPCNP cell survival rate, ECM productionBMSCs and NPCs in coculture significantly increased NP cell survival and promoted the generation of type II collagen and proteoglycans. In a hypoxic environment, TGF-β and Notch pathways enhanced disc microenvironment repair[16,67]
In vivo experimentBMSCs, UC-MSCsDisc height, NP cell count, NP cell apoptosis rateIntradiscal injection of BMSCs and UC-MSCs significantly reduced NP cell apoptosis and restored disc height. Exosomes combined with hydrogels improved stem cell engraftment in the disc[32,55]
Clinical trialBMSCsPain relief, functional recovery, disc water contentEarly clinical trials of BMSC injections showed significant pain reduction in patients, with increased NP water content at 12-month follow-up, confirming BMSC repair potential. Several studies showed significant pain relief postinjection with no severe side effects[20]
In vitro testADSCsPain score, functional improvement, disc regenerationADSC injections demonstrated significant pain relief and functional improvement in patients with lower back pain, with substantial reductions in pain scores and no reported severe complications[25]
Animal experimentBMSC exosomesECM production, disc height, inflammationBMSC exosomes significantly enhanced NP cell antiapoptotic capacity, promoted ECM synthesis, and restored disc structure and elasticity. Exosomes reduced inflammation in disc degeneration models by activating the PI3K/AKT pathway[11]
In vitro testBMSC and ADSC exosomesDisc repair rate, inflammation modulation, cell survivalStem cell-derived exosomes promoted disc repair by reducing inflammation and enhancing NP cell survival, showing significant therapeutic potential when combined with gene editing technology and biomaterials[90,91]
Mechanisms of stem cell-mediated repair in IVDD

Differentiation capacity: Under suitable microenvironmental conditions, stem cells can differentiate into NPCs, AF cells, or chondrocyte-like cells, thus contributing to disc tissue regeneration[42,43]. Studies have shown that the activation of signaling pathways such as the TGF-β and Wnt/β-catenin pathways enable stem cells to effectively differentiate into the cell types required for the restoration of disc structure and function[44,45]. The TGF-β pathway facilitates ECM production and the secretion of collagen and proteoglycans, which help NP cells maintain disc elasticity and hydration[45]. The Wnt/β-catenin pathway plays a critical role in cell differentiation and regeneration by regulating β-catenin accumulation and nuclear translocation, initiating the expression of relevant genes[44].

Paracrine effects: Another therapeutic mechanism of stem cells is their potent paracrine effects. By secreting various cytokines, such as TGF-β, IGF-1, and VEGF, stem cells can regulate local inflammatory responses, reduce NP cell apoptosis, and promote ECM synthesis[35,45]. These paracrine factors modulate inflammatory mediators, improving the disc microenvironment to delay degeneration and facilitating tissue regeneration. Additionally, studies have shown that stem cell-derived exosomes, which carry microRNAs (miRNAs) and proteins, play a key role in disc regeneration[4].

ECM repair: Stem cells participate directly in disc repair by secreting essential ECM components such as proteoglycans and collagen[46-48]. These ECM elements are crucial for maintaining the structural integrity and elasticity of the disc[46]. As disc degeneration progresses, ECM synthesis decreases, and NP cells gradually undergo apoptosis, leading to a decrease in disc height and elasticity[49,50]. Stem cell therapy effectively enhances ECM production and improves ECM quality, making the repaired disc tissue closer to a healthy state[51,52]. Research indicates that in addition to directly secreting ECM components stem cells promote repair through paracrine effects, releasing cytokines such as TGF-β and IGF-1 to further stimulate ECM synthesis[53,54]. Together, these mechanisms help slow NP cell attrition, prevent disc height loss, and delay disc degeneration. Furthermore, recent studies have shown that combining stem cells with biomaterials, such as hydrogels, improves stem cell engraftment and ECM repair in the disc, enhancing their clinical application potential[51,55,56]. This combined approach offers new possibilities for optimizing stem cell therapy.

MOLECULAR MECHANISMS OF STEM CELL REPAIR IN IVDD

In recent years, the molecular mechanisms by which stem cells repair IVDD have been extensively studied and have gained significant attention. These studies highlighted the importance of hypoxic conditions in stem cell differentiation and the critical roles of various signaling pathways in regulating stem cell regeneration. Under the hypoxic environment of the disc, stem cells activate a series of molecular signaling pathways to promote ECM synthesis and repair damaged tissue. Additionally, stem cell-derived exosomes and the application of gene vector systems play essential roles in intercellular communication, further enhancing repair efficacy (Figure 1 and Table 3).

Figure 1
Figure 1 Main biological mechanisms of stem cell-mediated repair in damaged intervertebral discs. Growth factors such as insulin-like growth factor 1 and vascular endothelial growth factor activate the phosphatidylinositol 3-kinase/protein kinase B pathway, which enhances protein synthesis through mechanistic target of rapamycin and promotes cell cycle progression via cyclin D1 and CDK4, facilitating stem cell proliferation. Transforming growth factor beta signaling, which is mediated by Smad2/3/4, promotes the expression of Sry-related HMG box 9, which is critical for chondrogenic differentiation, and the synthesis of key extracellular matrix components (COL2A1 and ACAN), which contribute to tissue repair. Notch signaling, which is activated by ligand binding (Jagged, Delta) to Notch receptors, leads to the formation of the Notch intracellular domain. The Notch intracellular domain downregulates Hes and Hey transcription factors, influencing stem cell differentiation. The Wnt/β-catenin pathway is pivotal for cell survival, regulating the balance between antiapoptotic (Bcl-2, Bcl-xL) and proapoptotic (Bax, Bak) factors to control apoptosis, thereby supporting the survival of stem cells in the damaged disc environment. These pathways work together to enhance tissue regeneration and restore intervertebral disc function. IGF: Insulin-like growth factor; VEGF: Vascular endothelial growth factor; PI3K: Phosphatidylinositol 3-kinase; AKT: Protein kinase B; mTOR: Mechanistic target of rapamycin; SOX9: Sry-related HMG box 9; TGF: Transforming growth factor; NICD: Notch intracellular domain.
Table 3 Comparison of signaling pathways and effects.
Signaling pathway
Key factors
Regulatory mechanism
Functional effects
TGF-β signaling pathwayTGF-β, Smad2/3Promotes ECM production and reduces cell apoptosis by activating the Smad pathwayEnhances the survival of NPCs, promotes type II collagen and proteoglycan production, maintains disc elasticity
Wnt/β-catenin pathwayβ-cateninActivates the translocation of β-catenin into the nucleus, regulating the expression of cartilage-related genesPromotes differentiation of NPCs and chondrocyte-like cells, enhances ECM production, maintains disc structural integrity
PI3K/AKT pathwayPI3K, AKT, mTOR, Bcl-2Enhances stem cell survival and reduces apoptosis of nucleus pulposus cells by activating mTOR and Bcl-2Increases stem cell survival rate under hypoxic conditions, enhances regenerative ability, reduces inflammatory response
NF-κB signaling pathwayNF-κB, TNF-α, IL-6Inhibits the expression of inflammation-related molecules like TNF-α, IL-6, and IL-1β associated with NF-κB, delays tissue damage, and reduces apoptosis of NPCsImproves the survival rate of NPCs through anti-inflammatory and antiapoptotic effects, promoting tissue regeneration
Notch signaling pathwayNotch1, Notch2, CSL, NICDActivates the CSL transcription factor through ligand binding, promoting stem cell proliferation and differentiationEnhances the formation of NPCs and annulus fibrosus cells, increases the regenerative potential of disc tissue
HIF-1α signaling pathwayHIF-1α, IGF-1Regulates the metabolism of NPCs and ECM synthesis, enhancing cell survival in a hypoxic microenvironmentMaintains disc tissue elasticity, reduces cell apoptosis, delays disc degeneration
Regulation of NPC survival and function by hypoxia-inducible factor-1α in hypoxic environments

The intervertebral disc, especially the NP, is one of the few avascular tissues in the body and remains in a hypoxic environment over time. Hypoxia-inducible factor-1α (HIF-1α) plays a critical role in this environment, serving as an essential regulator of NP cell survival and function[57]. In response to local oxygen partial pressure changes, HIF-1α regulates cellular metabolism and matrix production, helping stem cells differentiate into NP-like cells and promoting the synthesis of ECM components, such as collagen and proteoglycans[58,59].

During disc degeneration, HIF-1α not only supports NP cell survival but also promotes metabolic activity and inhibits apoptosis through downstream gene regulation[60]. The cooperative interaction between HIF-1α and the Notch signaling pathway helps slow the degenerative process by activating growth factors such as IGF-1 and TGF-β, thereby enhancing the regenerative capacity of NPCs[59]. Furthermore, studies have shown that activation of the HIF-1α pathway in stem cell therapy can significantly increase ECM production, improve disc elasticity and function, and delay tissue degeneration[4,60]. Future research may explore the application of HIF-1α as a potential therapeutic target, utilizing gene editing or pharmacological modulation to increase its activity in degenerated discs.

Signaling pathways involved in stem cell-mediated repair

Stem cells utilize multiple signaling pathways to exert reparative effects on disc degeneration. These pathways regulate stem cell survival, proliferation, and differentiation and influence the inflammatory response and ECM production. The Notch signaling pathway plays a crucial role in regulating stem cell self-renewal and differentiation[61]. When the Notch receptor binds with its corresponding ligand, it releases the intracellular domain, which enters the nucleus and initiates the transcription of genes that promote NPC generation[62,63]. Studies have shown that upon ligand binding, Notch releases Notch intracellular domain into the nucleus, activating the Sry-related HMG box 9 (Sox9) gene to stimulate the synthesis of type II collagen and proteoglycans, thereby restoring disc structure[64,65]. Proper activation of this pathway is essential for maintaining cellular balance, as overactivation may lead to fibrosis and other adverse effects[66,67].

The Wnt/β-catenin signaling pathway improves stem cell survival in adverse environments by enhancing antiapoptotic abilities and promoting NPC differentiation. When Wnt binds to the Frizzled receptor on the cell surface, β-catenin is released and enters the nucleus, activating antiapoptotic genes such as Bcl-2 and survivin[68-71]. This pathway also works in conjunction with HIF-1α to adapt to the hypoxic environment of the disc, supporting cellular energy metabolism[72,73]. The Wnt pathway is essential for stem cell differentiation into NPC and chondrocyte-like cells, promoting ECM synthesis and restoring disc elasticity.

The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway plays a central role in stem cell survival, proliferation, and differentiation[74]. PI3K activation generates PIP3, which activates downstream AKT, initiating signaling pathways related to antiapoptotic effects and cell proliferation[75,76]. AKT enhances cell survival in hypoxic environments by inhibiting proapoptotic factors such as Bad and Bax and activating antiapoptotic proteins such as Bcl-2 and Bcl-xL[77]. Additionally, the PI3K/AKT pathway regulates cell cycle-related proteins, such as cyclin D1 and CDK4, promoting cell proliferation to provide a sufficient cell source for disc repair[75,76]. This pathway also regulates the Sox9 gene, promoting the differentiation of stem cells into NP-like or chondrocyte-like cells, thus accelerating disc repair[66].

The TGF-β signaling pathway is a key regulator of ECM production and differentiation in stem cells[78,79]. Through both Smad-dependent and non-Smad-dependent pathways, TGF-β promotes the regeneration of NP and AF cells, particularly through regulation of the Sox9 gene, which governs the synthesis of type II collagen and proteoglycans, thereby restoring disc mechanical strength[80,81]. Moreover, TGF-β reduces inflammatory responses by inhibiting the nuclear factor-kappaB (NF-κB) pathway, improving the local microenvironment and further enhancing stem cell-mediated repair capacity[82].

The regulation of the mechanistic target of rapamycin (mTOR) and NF-κB signaling pathways is particularly crucial for stem cell survival in adverse environments[83,84]. mTOR promotes protein synthesis and cellular metabolism, enhances cell growth and antiapoptotic functions, while simultaneously suppressing the proinflammatory effects of NF-κB and reducing the release of inflammatory mediators[85-87]. By activating these pathways, stem cells can effectively respond to the hypoxic and inflammatory microenvironment of the disc, maintaining cellular function.

RESEARCH PROGRESS ON THE DEGENERATION OF STEM CELLS IN INTERVERTEBRAL DISC

The role of stem cells in the repair of IVDD has been extensively validated through in vitro experiments, animal models, and clinical studies. These studies revealed the regenerative potential of stem cells in disc tissues as well as the mechanisms of repair under various conditions and environments (Table 4).

Table 4 Comparison of stem cell therapy and traditional treatments.
Treatment method
Advantages
Indications
Limitations
Clinical application cases
Conservative treatmentHigh safety, suitable for early-stage patients, usually includes medication and physical therapy, low riskSuitable for patients with early mild symptomsCannot reverse disc degeneration, limited effectiveness, can only temporarily relieve symptomsCommonly used for early IVDD patients but cannot fundamentally stop disease progression
Minimally invasive surgeryMinimal surgical trauma, shorter recovery time, low riskApplicable to patients with moderate IVDDThe effect may not be as good as traditional surgery, and symptoms may recur in some patientsShows good short-term effects in some patients, suitable for those unwilling to undergo invasive surgery
Open surgeryRelieves nerve compression symptoms caused by disc herniation; spinal fusion can restore spinal stabilitySuitable for severe degenerative disc disease and neurological symptomsHigh trauma, long recovery time, high surgical risks, possible postoperative complications, and inability to restore normal disc functionSurgical treatment can effectively relieve pain and neurological symptoms, but recovery is slow, and there is a risk of recurrence
Stem cell therapyMinimally invasive, with regenerative potential, capable of repairing disc tissue through differentiation and paracrine mechanisms; has immunomodulatory effects, reduces inflammation, and inhibits apoptosisApplicable to early IVDD patients who do not respond to traditional treatmentsThe long-term survival rate and safety of stem cells in the disc require further research, with potential risks of immune rejection and tumor formationClinical trials indicate that BMSC injections can significantly reduce patient pain, with follow-up showing increased hydration of the nucleus pulposus
Stem cell + exosome therapyStem cell exosomes help enhance stem cell survival rate, and exosomes act as carriers of signaling molecules, promoting tissue repairSuitable for patients who are not candidates for surgeryLong-term efficacy needs further validation, and the isolation and preparation techniques for exosomes still need improvementCombining gene editing technology and biomaterials has enhanced its regenerative effects, with preclinical studies demonstrating significant regenerative potential
In vitro studies

In in vitro experiments, coculturing stem cells with NPCs has shown that stem cells significantly increase NPC survival and stimulate the secretion of more ECM components, such as collagen and proteoglycans[88]. Studies indicate that stem cells under hypoxic conditions in vitro contribute to the restoration of the disc microenvironment[36,57]. Stem cell-derived exosomes, which carry various bioactive molecules, further increase NPC survival and differentiation, improve the inflammatory environment, and increase the disc repair capacity[89-91].

Animal model studies

In animal models, stem cell injections have been shown to restore disc height, repair NP tissue, and significantly alleviate symptoms caused by degenerative changes. Research has demonstrated that injecting BMSCs or ADSCs into IVDD models effectively increases the number of NPCs, promotes ECM production, and thereby restores disc elasticity and function[92]. Additionally, the survival, migration, and differentiation of stem cells after injection into the disc are key indicators for evaluating therapeutic efficacy. In animal studies[93], stem cells significantly delay disc degeneration by activating hypoxic signaling pathways, increasing cell survival, and promoting the secretion of growth factors. Animal research has also explored the impact of various injection methods on stem cell survival and differentiation. For example, studies have shown that stem cells injected with exosomes or combined with biomaterials such as hydrogels are more likely to engraft in disc tissue, significantly enhancing their regenerative effects[93].

Clinical studies

In clinical studies, stem cell injections have shown efficacy in improving clinical symptoms and functional recovery in patients with IVDD. Multiple clinical trials have reported that intradiscal injection of BMSCs, ADSCs, and UC-MSCs can significantly reduce patient pain, restore disc function, and slow further disease progression[20]. Different sources of stem cells offer unique advantages. For example, BMSCs are widely studied and exhibit strong regenerative abilities, whereas UC-MSCs, owing to their low immunogenicity, have significant potential in clinical applications[92]. However, while clinical studies indicate the potential of stem cell therapy, optimizing injection methods, cell dosages, and long-term safety remains a research priority. Studies also suggest that future exploration of exosome and gene-editing technologies in conjunction with stem cell therapy may further enhance therapeutic effects[94]. In summary, experimental and clinical research on stem cells in IVDD has demonstrated their substantial promise in regenerative medicine. However, further large-scale clinical trials and long-term follow-up studies are essential to confirm the safety and efficacy of stem cell therapy.

APPLICATION OF STEM CELL EXOSOMES IN THE REPAIR OF IVDD

In recent years, stem cell-derived exosomes, as essential mediators of intercellular communication, have shown great potential in repairing IVDD. Exosomes are small membrane-bound vesicles with diameters ranging from 30 to 150 nm that carry various bioactive substances, including mRNAs, miRNAs, proteins, and lipids, that regulate target cell function and participate in multiple biological processes[95,96]. Owing to their low immunogenicity, ease of acquisition, and stability, stem cell-derived exosomes are gaining attention in regenerative medicine[97].

Mechanisms of stem cell-derived exosomes in disc repair

Stem cell-derived exosomes have significant biological advantages in the inflammatory and degenerative environment of the intervertebral disc, particularly in suppressing inflammation and promoting cell survival[49]. As small vesicles secreted by cells, exosomes contain abundant proteins, nucleic acids, and lipids that enable signal transmission between cells and regulate key signaling pathways, such as the TGF-β, PI3K/AKT, and NF-κB pathways[98]. In the inflammatory microenvironment of IVDD, stem cell-derived exosomes modulate the release of the proinflammatory factors tumor necrosis factor-alpha and IL-6, effectively suppressing inflammation[98,99]. These proinflammatory factors not only accelerate apoptosis but also degrade the ECM. Stem cell-derived exosomes improve the disc microenvironment by carrying anti-inflammatory miRNAs and proteins, inhibiting NF-κB pathway activation and reducing proinflammatory factor secretion[100,101].

Additionally, stem cell-derived exosomes help NP cells resist the apoptosis induced by proinflammatory factors and oxidative stress by regulating antiapoptotic pathways such as the PI3K/AKT and mTOR pathways[4]. By activating AKT, stem cell-derived exosomes promote the expression of antiapoptotic proteins and inhibit proapoptotic factors, significantly reducing NP cell apoptosis[68,102]. Moreover, stem cell-derived exosomes carry TGF-β or regulate its signaling pathway, promoting ECM synthesis and suppressing local inflammation[84]. TGF-β signaling not only regulates cell differentiation and ECM production in NPCs but also modulates macrophage polarization, reducing inflammation intensity and creating a microenvironment conducive to disc repair[103].

By regulating the Wnt/β-catenin and TGF-β signaling pathways, stem cell-derived exosomes increase the differentiation efficiency of NP and chondrocyte-like cells, promote ECM synthesis, and restore disc elasticity and structural integrity[104]. Additionally, the immunomodulatory properties of stem cell-derived exosomes can reduce immune responses within the disc by regulating T cell and macrophage activity, further inhibiting disease progression and promoting regeneration[105,106].

Applications of exosomes in experimental studies

The effects of exosomes have been validated in various in vitro and in vivo studies. Research has shown that stem cell-derived exosomes significantly improve the NPC microenvironment, reduce inflammatory responses, and increase antiapoptotic capacity through the activation of survival pathways such as the PI3K/AKT pathway[11,107]. Furthermore, exosomes can facilitate stem cell differentiation into NP-like cells, accelerating the disc repair process[108]. Experimental studies have also revealed that exosomes can serve as efficient drug delivery systems for the targeted transport of anti-inflammatory drugs or growth factors to degenerated disc sites[109]. This delivery system enables drugs to act more directly on diseased tissues, enhancing therapeutic efficacy while reducing systemic side effects. For example, in animal models of disc degeneration, stem cell-derived exosomes significantly mitigate disc height loss and restore disc function by improving ECM synthesis[55,110]. Studies have also shown that combining exosomes with other biomaterials, such as hydrogels, further improves exosome engraftment and persistence within the disc, significantly delaying degenerative progression[55,56]. In summary, stem cell-derived exosomes have broad application potential in regulating NPC function and inhibiting disc degeneration. With further preclinical research, exosomes are expected to become novel biotherapies for early intervention and long-term management of IVDD.

APPLICATION OF GENE VECTOR SYSTEMS IN THE TREATMENT OF IVDD
Gene vector classification in IVDD therapy

Gene therapy holds significant promise for the treatment of IVDD because it allows precise modulation of gene expression in disc cells. A central element of this strategy involves selecting suitable vectors to deliver therapeutic genes, which can be broadly categorized as viral, nonviral, or nanomaterial-based[111]. Viral vectors, such as lentiviruses, adenoviruses, and adeno-associated viruses, provide robust transfection efficiencies but can pose immunogenicity and safety concerns. Nonviral vectors, including plasmid DNA, liposomes, and polyplex micelles, generally offer safer profiles owing to their lower immunogenicity, although at the expense of reduced transfection efficiency. Nanomaterial-based vectors, including nanoparticles, graphene oxide, and nanofibers, present advantages such as enhanced stability, targeted delivery, and improved cellular uptake[112]. Each vector type has distinct strengths and limitations, and the choice of vector depends on therapeutic objectives, target cell types, the desired duration of gene expression, and safety considerations. Current research efforts focus on optimizing these vectors to further improve both their efficacy and safety, thereby advancing the potential of gene therapy in IVDD treatment.

Enhancing stem cell efficacy in disc repair through gene vector systems

A gene vector system is a technique used to deliver specific genes to target cells or tissues. In stem cell therapy, gene vector systems can enhance stem cell repair abilities by introducing specific genes[14,15]. In the treatment of IVDD, gene vector systems help stem cells better adapt to the disc microenvironment, enhancing their differentiation, proliferation, and antiapoptotic capacities and thereby promoting disc regeneration (Figure 2 and Table 5).

Figure 2
Figure 2 Gene vector-based stem cell therapy for intervertebral disc degeneration. A gene vector containing a target gene is introduced into various types of stem cells, including bone marrow-derived mesenchymal stem cells, adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and progenitor nucleus pulposus cells. These genetically modified stem cells secrete exosomes, which are rich in biologically active contents such as RNA, proteins, and cytokines. These components contribute to the repair and regeneration of degenerated intervertebral discs by promoting cellular communication, reducing inflammation, and supporting tissue regeneration. Therapy aims to restore disc function and alleviate degenerative symptoms. BMSCs: Bone marrow-derived mesenchymal stem cells; ADSCs: Adipose-derived mesenchymal stem cells; US-MSCs: Umbilical cord-derived mesenchymal stem cells; ProNPs: Progenitor nucleus pulposus cells.
Table 5 Applications of combining gene editing with biomaterials in stem cell therapy.
Technology/material type
Target gene/material
Main mechanism of action
Application scenario
Experimental results
Ref.
CRISPR-Cas9ParkinCRISPR-dCas9-KRAB system used to silence the expression of ParkinTargeting Parkin provides a new approach for IVDD repairInhibition of Parkin significantly reduces mitophagy and accelerates apoptosis of NPCs[128]
siRNABcl-2Reduces apoptosis by inhibiting the expression of apoptosis-related gene Bcl-2Inhibition of NPC apoptosisDecreases the apoptosis rate of NPCs, promoting disc tissue repair[113,114]
Gene editing + hydrogelHIF-1αEnhances HIF-1α expression in hypoxic environments, improving stem cell survivalDisc regeneration and cell protection under hypoxic conditionsSignificantly improves stem cell colonization and survival, promoting regeneration of nucleus pulposus tissue[26,27]
Gene editing + chitosanIGF-1Enhances IGF-1 expression, promoting cell proliferation and differentiationNP tissue repair and regenerationIncreases the proliferation rate of NPCs, enhances type II collagen and proteoglycan production[123]
Nanofiber scaffoldNo gene editingProvides 3D structural support, enhancing stem cell colonization efficiencyTissue regeneration and structural repairNanofiber scaffold provides ideal mechanical support, significantly improving tissue structure restoration[112]
CRISPR + exosomemiRNAIncreases miRNA content in exosomes, regulates inflammation, and promotes tissue repairInflammation control and disc regenerationSignificantly reduces inflammation, promotes ECM production, and enhances cell repair capacity[30]
Gene editing + nanoparticlesVEGFOverexpression of the VEGF gene increases angiogenesis, improving blood supply to the discDisc vascularization and regenerationPromotes angiogenesis in disc tissue, enhancing regeneration capacity[30,128]
Application of gene vector systems in disc degeneration

In stem cell therapy for disc degeneration, gene vector systems improve stem cell survival rates in the hypoxic, nutrient-poor disc environment by introducing specific genes. For example, antiapoptotic genes (e.g., Bcl-2) or prodifferentiation genes (e.g., TGF-β, IGF-1) can be delivered into stem cells via viral or nonviral vectors, promoting their directed differentiation into NP or AF cells within the disc[16]. This genetic modification not only enhances cell survival but also increases ECM production, thereby improving disc elasticity and function. Additionally, gene vector systems can suppress inflammation associated with disc degeneration. Through the introduction of anti-inflammatory genes (e.g., IL-10 and tumor necrosis factor-alpha inhibitors) or the use of gene silencing techniques (e.g., small interfering RNA), inflammation within the disc can be significantly reduced, slowing disease progression[4,113,114]. This combination of gene editing with stem cell therapy addresses limitations in traditional stem cell therapies related to inflammation control, thereby increasing overall treatment efficacy.

Combination of stem cell-derived exosomes and gene vector systems

In recent years, researchers have explored the use of gene vector systems to modulate exosomes secreted by stem cells to enhance therapeutic effects. Stem cell-derived exosomes, as extracellular vesicles, carry repair-related genes, proteins, and miRNAs and facilitate intercellular communication[115,116]. By introducing specific genes into stem cells via gene vectors, the content of regenerative factors in their exosomes, such as antiapoptotic miRNAs or growth-promoting factors, can be increased, thereby improving the efficiency of disc regeneration[117-119].

ADVANTAGES AND LIMITATIONS OF STEM CELL THERAPY

Stem cell therapy has several advantages in the treatment of IVDD, particularly in terms of tissue regeneration, minimally invasive treatment, and immune modulation[120]. By differentiating into NP and AF cells, stem cells can directly repair disc tissue and secrete growth factors and anti-inflammatory factors, promoting ECM production, suppressing inflammation, and slowing the progression of disc degeneration[4,10]. Compared with traditional surgical treatments, stem cell therapy is less invasive, offers faster recovery, and presents a lower risk of complications. Clinical studies have shown that stem cell therapy can effectively relieve pain and promote disc regeneration.

However, stem cell therapy also faces certain limitations, especially concerning cell survival rates in hypoxic environments and uncertain long-term efficacy[57,121]. The complex structure of the AF limits the repair capacity of stem cells, particularly in cases of extensive damage[122,123]. Additionally, allogeneic stem cells may carry the risk of immune rejection, and their long-term safety remains to be further verified, especially regarding potential risks of tumor formation and ectopic differentiation, which requires more clinical data support. Therefore, although stem cell therapy shows great promise, future research must focus on optimizing its efficacy and safety[124,125].

CURRENT RESEARCH FOCUS AND FUTURE OUTLOOK

In recent years, research on stem cell therapy for IVDD has focused primarily on combining stem cells with biomaterials, gene editing technology, and optimized injection methods[57,126,127]. Combining stem cells with biomaterials such as hydrogels provides mechanical support, enhances cell engraftment and differentiation within the disc, and mimics the physical properties of the NP to promote tissue regeneration. Gene editing techniques (e.g., CRISPR-Cas9) further increase the reparative potential of stem cells by regulating specific genes to improve their differentiation and antiapoptotic abilities, while hypoxic preconditioning aids stem cells in adapting to the hypoxic environment of the disc[57,127,128]. Additionally, optimizing the injection dosage and methods is crucial; studies indicate that multiple small-dose injections are more effective in enhancing cell survival and efficacy than single large-dose injections[129].

Although stem cell therapy shows significant promise, gene vector systems are also receiving attention for their potential to increase therapeutic efficacy. However, challenges remain in the application of gene vectors. Viral vectors offer high gene delivery efficiency but may pose risks of immune rejection and tumor formation, whereas nonviral vectors such as plasmids and liposomes are safer but have lower gene delivery efficiency, limiting their clinical application. Future research should aim to optimize gene vector design to minimize side effects and improve transfection efficiency while promoting personalized gene therapies tailored to individual patient needs.

Future research directions should continue to explore the molecular mechanisms of stem cell regeneration, accumulate more clinical data, and refine personalized treatment strategies. Combining new technologies, such as exosome delivery and gene vectors, can further improve therapeutic precision and efficiency, expanding the clinical potential of stem cell therapy in the treatment of IVDD[130].

CONCLUSION

In recent years, stem cell therapy for IVDD has shown immense promise because of its inherent regenerative capacity. Stem cells can differentiate into key disc cell types, stimulating ECM production. Moreover, they exert paracrine effects, modulating inflammation and reducing cell death. Stem cell-derived exosomes amplify this regenerative response by delivering signaling molecules to target cells within the degenerated disc. However, challenges exist. The harsh disc environment hinders stem cell survival. Regenerating the complex AF structure presents difficulties. Additionally, the use of allogeneic stem cells necessitates addressing the risk of immune rejection.

To realize the clinical potential of stem cell therapy, future research must prioritize enhancing stem cell survival through strategies such as biomaterial support and gene editing. Addressing AF regeneration will require innovative approaches, including bioengineered scaffolds and manipulation of stem cell differentiation. Minimizing immune rejection will necessitate strategies such as immunosuppression, immunomodulation, and the use of patient-specific stem cells. Finally, rigorous clinical trials are crucial for evaluating safety and efficacy and refining treatment protocols. Through continued research, stem cell therapy has the potential to revolutionize IVDD treatment, offering patients a more effective and durable solution.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade A, Grade B, Grade C

Novelty: Grade A, Grade B, Grade C

Creativity or Innovation: Grade A, Grade B, Grade C

Scientific Significance: Grade A, Grade A, Grade B

P-Reviewer: Mengistu BA; Shi Q; Ventura C S-Editor: Wang JJ L-Editor: Filipodia P-Editor: Zheng XM

References
1.  Tu J, Li W, Yang S, Yang P, Yan Q, Wang S, Lai K, Bai X, Wu C, Ding W, Cooper-White J, Diwan A, Yang C, Yang H, Zou J. Single-Cell Transcriptome Profiling Reveals Multicellular Ecosystem of Nucleus Pulposus during Degeneration Progression. Adv Sci (Weinh). 2022;9:e2103631.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
2.  Wang F, Cai F, Shi R, Wang XH, Wu XT. Aging and age related stresses: a senescence mechanism of intervertebral disc degeneration. Osteoarthritis Cartilage. 2016;24:398-408.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in RCA: 350]  [Article Influence: 38.9]  [Reference Citation Analysis (0)]
3.  Zhang W, Sun T, Li Y, Yang M, Zhao Y, Liu J, Li Z. Application of stem cells in the repair of intervertebral disc degeneration. Stem Cell Res Ther. 2022;13:70.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
4.  Krut Z, Pelled G, Gazit D, Gazit Z. Stem Cells and Exosomes: New Therapies for Intervertebral Disc Degeneration. Cells. 2021;10:2241.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 97]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
5.  Gao B, Jiang B, Xing W, Xie Z, Luo Z, Zou W. Discovery and Application of Postnatal Nucleus Pulposus Progenitors Essential for Intervertebral Disc Homeostasis and Degeneration. Adv Sci (Weinh). 2022;9:e2104888.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 36]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
6.  Wang Z, Perez-Terzic CM, Smith J, Mauck WD, Shelerud RA, Maus TP, Yang TH, Murad MH, Gou S, Terry MJ, Dauffenbach JP, Pingree MJ, Eldrige JS, Mohammed K, Benkhadra K, van Wijnen AJ, Qu W. Efficacy of intervertebral disc regeneration with stem cells - a systematic review and meta-analysis of animal controlled trials. Gene. 2015;564:1-8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
7.  Hunt CL, Shen S, Nassr A, van Wijnen AJ, Larson AN, Eldrige JS, Mauck WD, Pingree MJ, Erwin PJ, Bydon M, Qu W. Current understanding of safety and efficacy of stem cell therapy for discogenic pain—A systematic review of human studies. Tech Reg Anesth Pain Manag. 2015;19:32-37.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
8.  Li W, Gong Y, Liu J, Guo Y, Tang H, Qin S, Zhao Y, Wang S, Xu Z, Chen B. Peripheral and Central Pathological Mechanisms of Chronic Low Back Pain: A Narrative Review. J Pain Res. 2021;14:1483-1494.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
9.  Kamatani T, Hagizawa H, Yarimitsu S, Morioka M, Koyamatsu S, Sugimoto M, Kodama J, Yamane J, Ishiguro H, Shichino S, Abe K, Fujibuchi W, Fujie H, Kaito T, Tsumaki N. Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials. 2022;284:121491.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
10.  Soufi KH, Castillo JA, Rogdriguez FY, DeMesa CJ, Ebinu JO. Potential Role for Stem Cell Regenerative Therapy as a Treatment for Degenerative Disc Disease and Low Back Pain: A Systematic Review. Int J Mol Sci. 2023;24:8893.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
11.  Luo L, Jian X, Sun H, Qin J, Wang Y, Zhang J, Shen Z, Yang D, Li C, Zhao P, Liu M, Tian Z, Zhou Y. Cartilage endplate stem cells inhibit intervertebral disc degeneration by releasing exosomes to nucleus pulposus cells to activate Akt/autophagy. Stem Cells. 2021;39:467-481.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in RCA: 87]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
12.  Xia Y, Yang R, Hou Y, Wang H, Li Y, Zhu J, Fu C. Application of mesenchymal stem cell-derived exosomes from different sources in intervertebral disc degeneration. Front Bioeng Biotechnol. 2022;10:1019437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 33]  [Reference Citation Analysis (0)]
13.  Hajiesmailpoor A, Mohamadi O, Farzanegan G, Emami P, Ghorbani M. Overview of Stem Cell Therapy in Intervertebral Disc Disease: Clinical Perspective. Curr Stem Cell Res Ther. 2023;18:595-607.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
14.  Huining L, Yi Z, Dihong T, Yifeng P, Man X, Ting Y, Jingting C. Inhibition of choriocarcinoma by Fe3O4-dextran-anti-β-human chorionic gonadotropin nanoparticles containing antisense oligodeoxynucleotide of heparanase. Int J Nanomedicine. 2013;8:4371-4378.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
15.  Yu KR, Natanson H, Dunbar CE. Gene Editing of Human Hematopoietic Stem and Progenitor Cells: Promise and Potential Hurdles. Hum Gene Ther. 2016;27:729-740.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
16.  Wen T, Wang H, Li Y, Lin Y, Zhao S, Liu J, Chen B. Bone mesenchymal stem cell-derived extracellular vesicles promote the repair of intervertebral disc degeneration by transferring microRNA-199a. Cell Cycle. 2021;20:256-270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
17.  Wangler S, Kamali A, Wapp C, Wuertz-Kozak K, Häckel S, Fortes C, Benneker LM, Haglund L, Richards RG, Alini M, Peroglio M, Grad S. Uncovering the secretome of mesenchymal stromal cells exposed to healthy, traumatic, and degenerative intervertebral discs: a proteomic analysis. Stem Cell Res Ther. 2021;12:11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
18.  Hu J, Li C, Jin S, Ye Y, Fang Y, Xu P, Zhang C. Salvianolic acid B combined with bone marrow mesenchymal stem cells piggybacked on HAMA hydrogel re-transplantation improves intervertebral disc degeneration. Front Bioeng Biotechnol. 2022;10:950625.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
19.  Tang W, Zhang H, Liu D, Jiao F. Icariin accelerates cartilage defect repair by promoting chondrogenic differentiation of BMSCs under conditions of oxygen-glucose deprivation. J Cell Mol Med. 2022;26:202-215.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
20.  Gornet MF, Beall DP, Davis TT, Coric D, LaBagnara M, Krull A, DePalma MJ, Hsieh PC, Mallempati S, Schranck FW, Kelly C, Foley KT. Allogeneic Disc Progenitor Cells Safely Increase Disc Volume and Improve Pain, Disability, and Quality of Life in Patients With Lumbar Disc Degeneration-Results of an FDA-Approved Biologic Therapy Randomized Clinical Trial. Int J Spine Surg. 2024;18:237-248.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
21.  Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B. Mesenchymal stem cells and hypoxia: where are we? Mitochondrion. 2014;19 Pt A:105-112.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in RCA: 95]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
22.  Khasawneh RR, Abu-El-Rub E. Hypoxia disturbs the Migration and Adhesion characteristics of Mesenchymal Stem Cells. Cell Mol Biol (Noisy-le-grand). 2022;68:28-32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
23.  Yuan C, Song W, Jiang X, Wang Y, Li C, Yu W, He Y. Adipose-derived stem cell-based optimization strategies for musculoskeletal regeneration: recent advances and perspectives. Stem Cell Res Ther. 2024;15:91.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
24.  Khazaei S, Keshavarz G, Bozorgi A, Nazari H, Khazaei M. Adipose tissue-derived stem cells: a comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank. 2022;23:1-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
25.  Yang L, Li Z, Zhang C, Li S, Chen L, Yang S, Guo Y. Psoralen synergizes with exosome-loaded SPC25 to alleviate senescence of nucleus pulposus cells in intervertebral disc degeneration. J Orthop Surg Res. 2023;18:622.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
26.  Kim S, Kwon OJ, Lee J, Kim J, Kim T, Kim K. A Brief Overview of Recent Engineering Approaches for Intervertebral Disc Regeneration Using Adipose Derived Mesenchymal Stem Cell Administration. Biotechnol Bioproc E. 2021;26:335-347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Farzanehpour M, Miri A, Alvanegh AG, Gouvarchinghaleh HE. Viral Vectors, Exosomes, and Vexosomes: Potential armamentarium for delivering CRISPR/Cas to cancer cells. Biochem Pharmacol. 2023;212:115555.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
28.  Huang H, Liu X, Wang J, Suo M, Zhang J, Sun T, Zhang W, Li Z. Umbilical cord mesenchymal stem cells for regenerative treatment of intervertebral disc degeneration. Front Cell Dev Biol. 2023;11:1215698.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
29.  Salehpour A, Karimi Z, Ghasemi Zadeh M, Afshar M, Kameli A, Mooseli F, Zare M, Afshar A. Therapeutic potential of mesenchymal stem cell-derived exosomes and miRNAs in neuronal regeneration and rejuvenation in neurological disorders: a mini review. Front Cell Neurosci. 2024;18:1427525.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
30.  Deng Y, Li Y, Chu Z, Dai C, Ge J. Exosomes from umbilical cord-derived mesenchymal stem cells combined with gelatin methacryloyl inhibit vein graft restenosis by enhancing endothelial functions. J Nanobiotechnology. 2023;21:380.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
31.  Chen R, Jiang Y, Lu L, Wang P, Huang D, Wang J, Liu Z, Qin S, Yin F. Bibliometric analysis of research trends in stem cell therapy for knee osteoarthritis over the period 2001-2021. Front Cell Dev Biol. 2022;10:996273.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
32.  Ekram S, Khalid S, Bashir I, Salim A, Khan I. Human umbilical cord-derived mesenchymal stem cells and their chondroprogenitor derivatives reduced pain and inflammation signaling and promote regeneration in a rat intervertebral disc degeneration model. Mol Cell Biochem. 2021;476:3191-3205.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
33.  Hu B, Wang P, Zhang S, Liu W, Lv X, Shi D, Zhao L, Liu H, Wang B, Chen S, Shao Z. HSP70 attenuates compression-induced apoptosis of nucleus pulposus cells by suppressing mitochondrial fission via upregulating the expression of SIRT3. Exp Mol Med. 2022;54:309-323.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 25]  [Reference Citation Analysis (0)]
34.  Zheng L, Cao Y, Ni S, Qi H, Ling Z, Xu X, Zou X, Wu T, Deng R, Hu B, Gao B, Chen H, Li Y, Zhu J, Tintani F, Demehri S, Jain A, Kebaish KM, Liao S, Séguin CA, Crane JL, Wan M, Lu H, Sponseller PD, Riley LH 3rd, Zhou X, Hu J, Cao X. Ciliary parathyroid hormone signaling activates transforming growth factor-β to maintain intervertebral disc homeostasis during aging. Bone Res. 2018;6:21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
35.  Zhang YY, Hu ZL, Qi YH, Li HY, Chang X, Gao XX, Liu CH, Li YY, Lou JH, Zhai Y, Li CQ. Pretreatment of nucleus pulposus mesenchymal stem cells with appropriate concentration of H(2)O(2) enhances their ability to treat intervertebral disc degeneration. Stem Cell Res Ther. 2022;13:340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
36.  Shen J, Chen A, Cai Z, Chen Z, Cao R, Liu Z, Li Y, Hao J. Exhausted local lactate accumulation via injectable nanozyme-functionalized hydrogel microsphere for inflammation relief and tissue regeneration. Bioact Mater. 2022;12:153-168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 46]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
37.  Wang H, Li F, Ban W, Zhang J, Zhang G. Human Bone Marrow Mesenchymal Stromal Cell-Derived Extracellular Vesicles Promote Proliferation of Degenerated Nucleus Pulposus Cells and the Synthesis of Extracellular Matrix Through the SOX4/Wnt/β-Catenin Axis. Front Physiol. 2021;12:723220.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
38.  Zhu Y, You T, Ai S, Jin M, Cong M, Wu D, Huang H. A Retrospective Comparison of Allogenic and Autologous Hematopoietic Stem Cell Transplantation in Patients with T-Cell Lymphoblastic Lymphoma. Blood. 2023;142:7112.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
39.  Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci. 2021;11:187.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
40.  Fodor Duric L, Basic Jukic N, Vujicic B. Comparison of Autologous and Allogeneic Adipose-Derived Stem Cells in Kidney Transplantation: Immunological Considerations and Therapeutic Efficacy. J Clin Med. 2024;13:5763.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
41.  Wang Z, Wei Q, Wang H, Han L, Dai H, Qian X, Yu H, Yin M, Shi F, Qi N. Mesenchymal Stem Cell Therapy Using Human Umbilical Cord in a Rat Model of Autoimmune-Induced Premature Ovarian Failure. Stem Cells Int. 2020;2020:3249495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
42.  Yan L, Wu X. Exosomes produced from 3D cultures of umbilical cord mesenchymal stem cells in a hollow-fiber bioreactor show improved osteochondral regeneration activity. Cell Biol Toxicol. 2020;36:165-178.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in RCA: 148]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
43.  Yang X, Chen Z, Chen C, Han C, Zhou Y, Li X, Tian H, Cheng X, Zhang K, Qin A, Zhou T, Zhao J. Bleomycin induces fibrotic transformation of bone marrow stromal cells to treat height loss of intervertebral disc through the TGFβR1/Smad2/3 pathway. Stem Cell Res Ther. 2021;12:34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
44.  Gumede DB, Abrahamse H, Houreld NN. Targeting Wnt/β-catenin signaling and its interplay with TGF-β and Notch signaling pathways for the treatment of chronic wounds. Cell Commun Signal. 2024;22:244.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Du X, Liang K, Ding S, Shi H. Signaling Mechanisms of Stem Cell Therapy for Intervertebral Disc Degeneration. Biomedicines. 2023;11:2467.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
46.  Cheng F, Wang C, Ji Y, Yang B, Shu J, Shi K, Wang L, Wang S, Zhang Y, Huang X, Zhou X, Xia K, Liang C, Chen Q, Li F. Partial reprogramming strategy for intervertebral disc rejuvenation by activating energy switch. Aging Cell. 2022;21:e13577.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 19]  [Reference Citation Analysis (0)]
47.  Farooq M, Hameed H, Dimanche-Boitrel MT, Piquet-Pellorce C, Samson M, Le Seyec J. Switching to Regular Diet Partially Resolves Liver Fibrosis Induced by High-Fat, High-Cholesterol Diet in Mice. Nutrients. 2022;14:386.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
48.  Jiang Y, Tuan RS. Bioactivity of human adult stem cells and functional relevance of stem cell-derived extracellular matrix in chondrogenesis. Stem Cell Res Ther. 2023;14:160.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
49.  Guo Z, Su W, Zhou R, Zhang G, Yang S, Wu X, Qiu C, Cong W, Shen N, Guo J, Liu C, Yang SY, Xing D, Wang Y, Chen B, Xiang H. Exosomal MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc Degeneration by Antisenescence Effects and Promotes NPC Proliferation and ECM Synthesis by Activating TGF-β. Oxid Med Cell Longev. 2021;2021:5542241.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
50.  Wang D, Qu H, Kang H, Xu F, Huang W, Cai X. Kukoamine A attenuates lipopolysaccharide-induced apoptosis, extracellular matrix degradation, and inflammation in nucleus pulposus cells by activating the P13K/Akt pathway. Bioengineered. 2022;13:8772-8784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
51.  Ma B, Wang T, Li J, Wang Q. Extracellular matrix derived from Wharton's Jelly-derived mesenchymal stem cells promotes angiogenesis via integrin αVβ3/c-Myc/P300/VEGF. Stem Cell Res Ther. 2022;13:327.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
52.  Novoseletskaya ES, Grigorieva OA, Efimenko AY, Kalinina NI. Extracellular Matrix in the Regulation of Stem Cell Differentiation. Biochemistry (Mosc). 2019;84:232-240.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
53.  Leung VY, Aladin DM, Lv F, Tam V, Sun Y, Lau RY, Hung SC, Ngan AH, Tang B, Lim CT, Wu EX, Luk KD, Lu WW, Masuda K, Chan D, Cheung KM. Mesenchymal stem cells reduce intervertebral disc fibrosis and facilitate repair. Stem Cells. 2014;32:2164-2177.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in RCA: 74]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
54.  Hou J, Peng X, Wang J, Zhang H, Xia J, Ge Q, Wang X, Chen X, Wu X. Mesenchymal stem cells promote endothelial progenitor cell proliferation by secreting insulinlike growth factor1. Mol Med Rep. 2017;16:1502-1508.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
55.  Xing H, Zhang Z, Mao Q, Wang C, Zhou Y, Zhou X, Ying L, Xu H, Hu S, Zhang N. Injectable exosome-functionalized extracellular matrix hydrogel for metabolism balance and pyroptosis regulation in intervertebral disc degeneration. J Nanobiotechnology. 2021;19:264.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 156]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
56.  Peng B, Du L, Zhang T, Chen J, Xu B. Research progress in decellularized extracellular matrix hydrogels for intervertebral disc degeneration. Biomater Sci. 2023;11:1981-1993.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
57.  Li H, Wu QY, Teng XH, Li ZP, Zhu MT, Gu CJ, Chen BJ, Xie QQ, LuO XJ. The pathogenesis and regulatory role of HIF-1 in rheumatoid arthritis. Cent Eur J Immunol. 2023;48:338-345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Kim JW, An HJ, Yeo H, Jeong Y, Lee H, Lee J, Nam K, Lee J, Shin DE, Lee S. Activation of Hypoxia-Inducible Factor-1α Signaling Pathway Has the Protective Effect of Intervertebral Disc Degeneration. Int J Mol Sci. 2021;22:11355.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
59.  Xiong Z, Ding J, Zhou J, Yao S, Zheng J, Guo X. Correlation between the HIF-1α/Notch signaling pathway and Modic changes in nucleus pulposus cells isolated from patients with low back pain. BMC Musculoskelet Disord. 2020;21:500.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
60.  Kim JW, Jeon N, Shin DE, Lee SY, Kim M, Han DH, Shin JY, Lee S. Regeneration in Spinal Disease: Therapeutic Role of Hypoxia-Inducible Factor-1 Alpha in Regeneration of Degenerative Intervertebral Disc. Int J Mol Sci. 2021;22:5281.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
61.  Guo Y, Tan J, Xiong W, Chen S, Fan L, Li Y. Notch3 promotes 3T3-L1 pre-adipocytes differentiation by up-regulating the expression of LARS to activate the mTOR pathway. J Cell Mol Med. 2020;24:1116-1127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
62.  Gagliani EK, Gutzwiller LM, Kuang Y, Odaka Y, Hoffmeister P, Hauff S, Turkiewicz A, Harding-Theobald E, Dolph PJ, Borggrefe T, Oswald F, Gebelein B, Kovall RA. A Drosophila Su(H) model of Adams-Oliver Syndrome reveals cofactor titration as a mechanism underlying developmental defects. PLoS Genet. 2022;18:e1010335.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
63.  Li Z, Xin S, Yu S, Liang J, Zhang X. Prognostic Signatures and Therapeutic Value Based on the Notch Pathway in Renal Clear Cell Carcinoma. Oxid Med Cell Longev. 2022;2022:1669664.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
64.  Chen H, Tan XN, Hu S, Liu RQ, Peng LH, Li YM, Wu P. Molecular Mechanisms of Chondrocyte Proliferation and Differentiation. Front Cell Dev Biol. 2021;9:664168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 66]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
65.  Chen S, Tao J, Bae Y, Jiang MM, Bertin T, Chen Y, Yang T, Lee B. Notch gain of function inhibits chondrocyte differentiation via Rbpj-dependent suppression of Sox9. J Bone Miner Res. 2013;28:649-659.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in RCA: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
66.  Kubo Y, Beckmann R, Fragoulis A, Conrads C, Pavanram P, Nebelung S, Wolf M, Wruck CJ, Jahr H, Pufe T. Nrf2/ARE Signaling Directly Regulates SOX9 to Potentially Alter Age-Dependent Cartilage Degeneration. Antioxidants (Basel). 2022;11:263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
67.  Bakalenko N, Kuznetsova E, Malashicheva A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int J Mol Sci. 2024;25:10803.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Nežić L, Amidžić L, Škrbić R, Gajanin R, Mandić D, Dumanović J, Milovanović Z, Jaćević V. Amelioration of Endotoxin-Induced Acute Lung Injury and Alveolar Epithelial Cells Apoptosis by Simvastatin Is Associated with Up-Regulation of Survivin/NF-kB/p65 Pathway. Int J Mol Sci. 2022;23:2596.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
69.  Yu H, Gao R, Chen S, Liu X, Wang Q, Cai W, Vemula S, Fahey AC, Henley D, Kobayashi M, Liu SZ, Qian Z, Kapur R, Broxmeyer HE, Gao Z, Xi R, Liu Y. Bmi1 Regulates Wnt Signaling in Hematopoietic Stem and Progenitor Cells. Stem Cell Rev Rep. 2021;17:2304-2313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
70.  Zhang L, Yan R, Zhang Q, Wang H, Kang X, Li J, Yang S, Zhang J, Liu Z, Yang X. Survivin, a key component of the Wnt/β-catenin signaling pathway, contributes to traumatic brain injury-induced adult neurogenesis in the mouse dentate gyrus. Int J Mol Med. 2013;32:867-875.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
71.  Trejo-Solis C, Escamilla-Ramirez A, Jimenez-Farfan D, Castillo-Rodriguez RA, Flores-Najera A, Cruz-Salgado A. Crosstalk of the Wnt/β-Catenin Signaling Pathway in the Induction of Apoptosis on Cancer Cells. Pharmaceuticals (Basel). 2021;14:871.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
72.  Meng F, Luo X, Li C, Wang G. LncRNA LINC00525 activates HIF-1α through miR-338-3p / UBE2Q1 / β-catenin axis to regulate the Warburg effect in colorectal cancer. Bioengineered. 2022;13:2554-2567.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
73.  Jridi I, Canté-Barrett K, Pike-Overzet K, Staal FJT. Inflammation and Wnt Signaling: Target for Immunomodulatory Therapy? Front Cell Dev Biol. 2020;8:615131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 66]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
74.  Drastichova Z, Trubacova R, Novotny J. β-Arrestin2 Is Critically Involved in the Differential Regulation of Phosphosignaling Pathways by Thyrotropin-Releasing Hormone and Taltirelin. Cells. 2022;11:1473.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
75.  Sun J, Zhao H, Shen C, Li S, Zhang W, Ma J, Li Z, Zhang M, Yang J. Tideglusib promotes wound healing in aged skin by activating PI3K/Akt pathway. Stem Cell Res Ther. 2022;13:269.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
76.  Zhang Y, Lv P, Ma J, Chen N, Guo H, Chen Y, Gan X, Wang R, Liu X, Fan S, Cong B, Kang W. Antrodia cinnamomea exerts an anti-hepatoma effect by targeting PI3K/AKT-mediated cell cycle progression in vitro and in vivo. Acta Pharm Sin B. 2022;12:890-906.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
77.  Zhang ZD, Yang YJ, Qin Z, Liu XW, Li SH, Bai LX, Li JY. Protective Activity of Aspirin Eugenol Ester on Paraquat-Induced Cell Damage in SH-SY5Y Cells. Oxid Med Cell Longev. 2021;2021:6697872.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
78.  Liu M, Han X, Liu H, Chen D, Li Y, Hu W. The effects of CRISPR-Cas9 knockout of the TGF-β1 gene on antler cartilage cells in vitro. Cell Mol Biol Lett. 2019;24:44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
79.  Khalid S, Ekram S, Salim A, Chaudhry GR, Khan I. Transcription regulators differentiate mesenchymal stem cells into chondroprogenitors, and their in vivo implantation regenerated the intervertebral disc degeneration. World J Stem Cells. 2022;14:163-182.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
80.  Wang J, Wan X, Le Q. Cross-regulation between SOX9 and the canonical Wnt signalling pathway in stem cells. Front Mol Biosci. 2023;10:1250530.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
81.  Cui P, Pan P, Qin L, Wang X, Chen X, Deng Y, Zhang X. Nanoengineered hydrogels as 3D biomimetic extracellular matrix with injectable and sustained delivery capability for cartilage regeneration. Bioact Mater. 2023;19:487-498.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
82.  Luo K. Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol. 2017;9:a022137.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in RCA: 416]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
83.  Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1:a001651.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2497]  [Cited by in RCA: 3495]  [Article Influence: 218.4]  [Reference Citation Analysis (0)]
84.  Kracht M, Müller-Ladner U, Schmitz ML. Mutual regulation of metabolic processes and proinflammatory NF-κB signaling. J Allergy Clin Immunol. 2020;146:694-705.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in RCA: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
85.  Pei X, Zheng F, Li Y, Lin Z, Han X, Feng Y, Tian Z, Ren D, Cao K, Li C. Niclosamide Ethanolamine Salt Alleviates Idiopathic Pulmonary Fibrosis by Modulating the PI3K-mTORC1 Pathway. Cells. 2022;11:346.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
86.  Cui D, Qu R, Liu D, Xiong X, Liang T, Zhao Y. The Cross Talk Between p53 and mTOR Pathways in Response to Physiological and Genotoxic Stresses. Front Cell Dev Biol. 2021;9:775507.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
87.  Surti M, Patel M, Redhwan A, Al-Keridis LA, Adnan M, Alshammari N, Reddy MN. Ilimaquinone (Marine Sponge Metabolite) Induces Apoptosis in HCT-116 Human Colorectal Carcinoma Cells via Mitochondrial-Mediated Apoptosis Pathway. Mar Drugs. 2022;20:582.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
88.  Yoon SH, Kim DH, Cho S, Kim KJ. Evaluation of Bone Marrow-derived Stem Cells and Adipose-derived Stem Cells Co-cultured on Human Nucleus Pulposus Cells: A Pilot Study. Korean J Neurotrauma. 2020;16:138-146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
89.  Liao Z, Luo R, Li G, Song Y, Zhan S, Zhao K, Hua W, Zhang Y, Wu X, Yang C. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics. 2019;9:4084-4100.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in RCA: 283]  [Article Influence: 47.2]  [Reference Citation Analysis (0)]
90.  Harrell CR, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells. 2019;8:1605.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in RCA: 486]  [Article Influence: 81.0]  [Reference Citation Analysis (0)]
91.  Jin Y, Xu M, Zhu H, Dong C, Ji J, Liu Y, Deng A, Gu Z. Therapeutic effects of bone marrow mesenchymal stem cells-derived exosomes on osteoarthritis. J Cell Mol Med. 2021;25:9281-9294.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 83]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
92.  Zhang XB, Chen XY, Qi J, Zhou HY, Zhao XB, Hu YC, Zhang RH, Yu DC, Gao XD, Wang KP, Ma L. New Hope for Intervertebral Disc Degeneration: Bone Marrow Mesenchymal Stem Cells and Exosomes Derived from Bone Marrow Mesenchymal Stem Cell Transplantation. Curr Gene Ther. 2022;22:291-302.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
93.  Luo L, Gong J, Wang Z, Liu Y, Cao J, Qin J, Zuo R, Zhang H, Wang S, Zhao P, Yang D, Zhang M, Wang Y, Zhang J, Zhou Y, Li C, Ni B, Tian Z, Liu M. Injectable cartilage matrix hydrogel loaded with cartilage endplate stem cells engineered to release exosomes for non-invasive treatment of intervertebral disc degeneration. Bioact Mater. 2022;15:29-43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
94.  Kostyushev D, Kostyusheva A, Brezgin S, Smirnov V, Volchkova E, Lukashev A, Chulanov V. Gene Editing by Extracellular Vesicles. Int J Mol Sci. 2020;21:7362.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
95.  Wei J, Qi T, Hao C, Zong S, Wang Z, Cui Y. Optical microscopic and spectroscopic detection of exosomes. TrAC Trends Anal Chem. 2023;163:117077.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
96.  Chen H, Zeng B, Li X, Zhao Q, Liu D, Chen Y, Zhang Y, Wang J, Xing HR. High-Metastatic Melanoma Cells Promote the Metastatic Capability of Low-Metastatic Melanoma Cells via Exosomal Transfer of miR-411-5p. Front Oncol. 2022;12:895164.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
97.  Shyu KG, Wang BW, Fang WJ, Pan CM, Lin CM. Hyperbaric oxygen-induced long non-coding RNA MALAT1 exosomes suppress MicroRNA-92a expression in a rat model of acute myocardial infarction. J Cell Mol Med. 2020;24:12945-12954.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
98.  Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2822]  [Cited by in RCA: 6013]  [Article Influence: 1202.6]  [Reference Citation Analysis (0)]
99.  Bhujel B, Shin HE, Choi DJ, Han I. Mesenchymal Stem Cell-Derived Exosomes and Intervertebral Disc Regeneration: Review. Int J Mol Sci. 2022;23:7306.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
100.  Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, Gao L, Xie J, Xu B. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res. 2019;115:1205-1216.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in RCA: 519]  [Article Influence: 103.8]  [Reference Citation Analysis (0)]
101.  Mitchell R, Mellows B, Sheard J, Antonioli M, Kretz O, Chambers D, Zeuner MT, Tomkins JE, Denecke B, Musante L, Joch B, Debacq-Chainiaux F, Holthofer H, Ray S, Huber TB, Dengjel J, De Coppi P, Widera D, Patel K. Secretome of adipose-derived mesenchymal stem cells promotes skeletal muscle regeneration through synergistic action of extracellular vesicle cargo and soluble proteins. Stem Cell Res Ther. 2019;10:116.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 135]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
102.  Zhang Z, Shang J, Yang Q, Dai Z, Liang Y, Lai C, Feng T, Zhong D, Zou H, Sun L, Su Y, Yan S, Chen J, Yao Y, Shi Y, Huang X. Exosomes derived from human adipose mesenchymal stem cells ameliorate hepatic fibrosis by inhibiting PI3K/Akt/mTOR pathway and remodeling choline metabolism. J Nanobiotechnology. 2023;21:29.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 51]  [Reference Citation Analysis (0)]
103.  Gao T, Huang F, Wang W, Xie Y, Wang B. Interleukin-10 genetically modified clinical-grade mesenchymal stromal cells markedly reinforced functional recovery after spinal cord injury via directing alternative activation of macrophages. Cell Mol Biol Lett. 2022;27:27.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
104.  Chen S, Zhao L, Deng X, Shi D, Wu F, Liang H, Huang D, Shao Z. Mesenchymal Stem Cells Protect Nucleus Pulposus Cells from Compression-Induced Apoptosis by Inhibiting the Mitochondrial Pathway. Stem Cells Int. 2017;2017:9843120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
105.  Lee DS, Yi TG, Lee HJ, Kim SN, Park S, Jeon MS, Song SU. Mesenchymal stem cells infected with Mycoplasma arginini secrete complement C3 to regulate immunoglobulin production in B lymphocytes. Cell Death Dis. 2014;5:e1192.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
106.  Yang S, Liang X, Song J, Li C, Liu A, Luo Y, Ma H, Tan Y, Zhang X. A novel therapeutic approach for inflammatory bowel disease by exosomes derived from human umbilical cord mesenchymal stem cells to repair intestinal barrier via TSG-6. Stem Cell Res Ther. 2021;12:315.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 98]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
107.  Xia C, Zeng Z, Fang B, Tao M, Gu C, Zheng L, Wang Y, Shi Y, Fang C, Mei S, Chen Q, Zhao J, Lin X, Fan S, Jin Y, Chen P. Mesenchymal stem cell-derived exosomes ameliorate intervertebral disc degeneration via anti-oxidant and anti-inflammatory effects. Free Radic Biol Med. 2019;143:1-15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 210]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
108.  Zhuang Y, Song S, Xiao D, Liu X, Han X, Du S, Li Y, He Y, Zhang S. Exosomes Secreted by Nucleus Pulposus Stem Cells Derived From Degenerative Intervertebral Disc Exacerbate Annulus Fibrosus Cell Degradation via Let-7b-5p. Front Mol Biosci. 2021;8:766115.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
109.  Wang Y, Guo M, Lin D, Liang D, Zhao L, Zhao R, Wang Y. Docetaxel-loaded exosomes for targeting non-small cell lung cancer: preparation and evaluation in vitro and in vivo. Drug Deliv. 2021;28:1510-1523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 42]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
110.  Ambrosio L, Schol J, Ruiz-Fernandez C, Tamagawa S, Soma H, Tilotta V, Di Giacomo G, Cicione C, Nakayama S, Kamiya K, Papalia R, Sato M, Vadalà G, Watanabe M, Denaro V, Sakai D. ISSLS PRIZE in Basic Science 2024: superiority of nucleus pulposus cell- versus mesenchymal stromal cell-derived extracellular vesicles in attenuating disc degeneration and alleviating pain. Eur Spine J. 2024;33:1713-1727.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
111.  Takeoka Y, Yurube T, Nishida K. Gene Therapy Approach for Intervertebral Disc Degeneration: An Update. Neurospine. 2020;17:3-14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 37]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
112.  Ruan WJ, Xu SS, Xu DH, Li ZP. Orthopedic revolution: The emerging role of nanotechnology. World J Orthop. 2024;15:932-938.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
113.  Shahabipour F, Banach M, Sahebkar A. Exosomes as nanocarriers for siRNA delivery: paradigms and challenges. Arch Med Sci. 2016;12:1324-1326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
114.  Wu X, Song Y, Liu W, Wang K, Gao Y, Li S, Duan Z, Shao Z, Yang S, Yang C. IAPP modulates cellular autophagy, apoptosis, and extracellular matrix metabolism in human intervertebral disc cells. Cell Death Discov. 2017;3:16107.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
115.  Zou X, Zou D, Li L, Yu R, Li X, Du X, Guo J, Wang K, Liu W. Multi-omics analysis of an in vitro photoaging model and protective effect of umbilical cord mesenchymal stem cell-conditioned medium. Stem Cell Res Ther. 2022;13:435.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
116.  Han Q, Xie QR, Li F, Cheng Y, Wu T, Zhang Y, Lu X, Wong AST, Sha J, Xia W. Targeted inhibition of SIRT6 via engineered exosomes impairs tumorigenesis and metastasis in prostate cancer. Theranostics. 2021;11:6526-6541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in RCA: 80]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
117.  Fahs A, Hussein N, Zalzali H, Ramadan F, Ghamloush F, Tamim H, El Homsi M, Badran B, Boulos F, Tawil A, Ghayad SE, Saab R. CD147 Promotes Tumorigenesis via Exosome-Mediated Signaling in Rhabdomyosarcoma. Cells. 2022;11:2267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
118.  Hade MD, Suire CN, Suo Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells. 2021;10:1959.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 257]  [Article Influence: 64.3]  [Reference Citation Analysis (0)]
119.  Lin Z, Xu G, Lu X, Liu S, Zou F, Ma X, Jiang J, Wang H, Song J. Chondrocyte-targeted exosome-mediated delivery of Nrf2 alleviates cartilaginous endplate degeneration by modulating mitochondrial fission. J Nanobiotechnology. 2024;22:281.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
120.  He F, Umrath F, Reinert S, Alexander D. Jaw Periosteum-Derived Mesenchymal Stem Cells Regulate THP-1-Derived Macrophage Polarization. Int J Mol Sci. 2021;22:4310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
121.  Li XC, Wang MS, Liu W, Zhong CF, Deng GB, Luo SJ, Huang CM. Co-culturing nucleus pulposus mesenchymal stem cells with notochordal cell-rich nucleus pulposus explants attenuates tumor necrosis factor-α-induced senescence. Stem Cell Res Ther. 2018;9:171.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
122.  Sloan SR Jr, Wipplinger C, Kirnaz S, Navarro-Ramirez R, Schmidt F, McCloskey D, Pannellini T, Schiavinato A, Härtl R, Bonassar LJ. Combined nucleus pulposus augmentation and annulus fibrosus repair prevents acute intervertebral disc degeneration after discectomy. Sci Transl Med. 2020;12:eaay2380.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in RCA: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
123.  Liu C, Ge X, Li Y. Repair of annulus fibrosus defects using decellularized annulus fibrosus matrix/chitosan hybrid hydrogels. J Orthop Surg Res. 2024;19:535.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
124.  Peng YQ, Wu ZC, Xu ZB, Fang SB, Chen DH, Zhang HY, Liu XQ, He BX, Chen D, Akdis CA, Fu QL. Mesenchymal stromal cells-derived small extracellular vesicles modulate DC function to suppress Th2 responses via IL-10 in patients with allergic rhinitis. Eur J Immunol. 2022;52:1129-1140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
125.  Yang Y, Ma B, Chen J, Liu D, Ma J, Li B, Hao J, Zhou X. Epigenetic regulation and factors that influence the effect of iPSCs-derived neural stem/progenitor cells (NS/PCs) in the treatment of spinal cord injury. Clin Epigenetics. 2024;16:30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
126.  Patmanathan SN, Gnanasegaran N, Lim MN, Husaini R, Fakiruddin KS, Zakaria Z. CRISPR/Cas9 in Stem Cell Research: Current Application and Future Perspective. Curr Stem Cell Res Ther. 2018;13:632-644.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
127.  Liu J, He J, Ge L, Xiao H, Huang Y, Zeng L, Jiang Z, Lu M, Hu Z. Hypoxic preconditioning rejuvenates mesenchymal stem cells and enhances neuroprotection following intracerebral hemorrhage via the miR-326-mediated autophagy. Stem Cell Res Ther. 2021;12:413.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
128.  Lan T, Zheng YC, Li ND, Chen XS, Shen Z, Yan B. CRISPR/dCas9-Mediated Parkin Inhibition Impairs Mitophagy and Aggravates Apoptosis of Rat Nucleus Pulposus Cells Under Oxidative Stress. Front Mol Biosci. 2021;8:674632.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
129.  Shin JH, Ryu CM, Ju H, Yu HY, Song S, Shin DM, Choo MS. Synergistic Effects of N-Acetylcysteine and Mesenchymal Stem Cell in a Lipopolysaccharide-Induced Interstitial Cystitis Rat Model. Cells. 2019;9:86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
130.  Liu W, Ma Z, Wang Y, Yang J. Multiple nano-drug delivery systems for intervertebral disc degeneration: Current status and future perspectives. Bioact Mater. 2023;23:274-299.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]