BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Dec 26, 2025; 17(12): 114349
Published online Dec 26, 2025. doi: 10.4252/wjsc.v17.i12.114349
Hurdles to overcome for mesenchymal stem cell translation from bench to bedside
Naiyareen Fareeza Mayeen, Department of Biology, University of Rochester, Rochester, NY 14627, United States
Umme Salma, Regenerative Medicine, Dhaka 1209, Bangladesh
Noor Hayaty Abu Kasim, Department of Restorative Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia
Okba Mahmoud, Department of Clinical Science, Ajman University, Ajman 346, United Arab Emirates
Nazmul Haque, Department of Regenerative Medicine, Concord Stem Cell Limited, Dhaka 1209, Bangladesh
ORCID number: Naiyareen Fareeza Mayeen (0009-0007-2470-7149); Umme Salma (0009-0004-3836-4173); Noor Hayaty Abu Kasim (0000-0002-8889-842X); Okba Mahmoud (0000-0002-3355-1429); Nazmul Haque (0000-0002-8191-8993).
Co-first authors: Naiyareen Fareeza Mayeen and Umme Salma.
Author contributions: Mayeen NF and Salma U contributed equally to this manuscript and are co-first authors. Mayeen NF and Salma U drafted and wrote the article; Haque N contributed to the conception and design of the study; Abu Kasim NH, Mahmoud O, and Haque N contributed to the manuscript writing, and made critical revisions related to the relevant intellectual content of the manuscript; and all the authors have approved the final version of the article.
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: Nazmul Haque, PhD, Chief, Head, Department of Regenerative Medicine, Concord Stem Cell Limited, 3, Sima Blossom, Road 16, Dhanmondi, Dhaka 1209, Bangladesh. haquen1983@gmail.com
Received: September 17, 2025
Revised: October 20, 2025
Accepted: November 21, 2025
Published online: December 26, 2025
Processing time: 99 Days and 17.2 Hours

Abstract

Mesenchymal stem or stromal cells (MSCs) are among the most extensively studied cell populations in regenerative medicine due to their multipotent differentiation potential, secretion of trophic factors, and immunomodulatory effects. Over the past two decades, preclinical studies have demonstrated encouraging results across musculoskeletal, cardiovascular, neurological, and immune-mediated disorders. However, the translation of MSCs from the laboratory to routine clinical practice remains hindered by unresolved scientific, technical, and regulatory challenges. This review provides a critical appraisal of these hurdles, organized across three major stages of translation: In vitro research, in vivo animal studies, and clinical application. In vitro issues include the heterogeneity of isolation techniques, replicative senescence during expansion, genetic and epigenetic instability, and the need for xeno-free, standardized culture platforms. In vivo challenges arise from poor cell survival, low engraftment rates, off-target migration, and microenvironmental influences that shape therapeutic outcomes. Clinical translation introduces additional complexity, including inter-patient variability, large-scale manufacturing difficulties, stringent regulatory compliance, high production costs, and the absence of harmonized potency assays. Solutions under exploration include the use of automated bioreactors, biomimetic scaffolds, hypoxic preconditioning, extracellular vesicle-based therapies, and international standardization efforts. Addressing these hurdles through multidisciplinary collaboration is essential for MSC-based therapies to become reliable, safe, and accessible regenerative treatments.

Key Words: Regenerative medicine; Clinical translation; Stem cell expansion; Scaffolds; Immunomodulation; Good manufacturing practice

Core Tip: Regenerative medicine has been practiced in a variety of settings for decades. It is a multidisciplinary branch of medicine that aims at replacing degenerated cells or regenerating new human cells, tissues, or organs that have been lost or damaged due to the effects of aging, illness, or congenital defects. The main objective of regenerative medicines is to restore normal functions of the cells or tissues in patients affected by degenerative disorders. Mesenchymal stem cells (MSCs) have the potential to cure the most critical regions or take over the function of a wounded organ that is considered beyond repair using conventional therapies. In the last two decades, thousands of in vitro, in vivo, and clinical trials have been conducted using MSCs. Though the regenerative potential of MSCs has been extensively studied, there are still a lot of challenges that need to be addressed for their successful translation from bench to bedside.



INTRODUCTION

Regenerative medicine represents an interdisciplinary field aimed at restoring normal tissue and organ function that has been impaired by trauma, degenerative disease, or aging. At the center of this effort lies the therapeutic promise of stem cells, particularly mesenchymal stem or stromal cells (MSCs)[1,2]. MSCs are non-hematopoietic, multipotent cell capable of differentiating into osteoblasts, chondrocytes, adipocytes, and other lineages[3,4]. In addition to their differentiation capacity, MSCs exert powerful paracrine effects by secreting cytokines, chemokines, and extracellular vesicles (EVs) that modulate inflammation, enhance angiogenesis, and promote endogenous repair mechanisms[5,6]. These secretory functions allow MSCs to orchestrate a pro-regenerative microenvironment even in the absence of long-term engraftment or differentiation, broadening their therapeutic utility across a wide spectrum of diseases[7,8].

Indeed, MSCs have been explored not only in orthopedic and musculoskeletal disorders but also in cardiovascular, hepatic, neurological, and immune-mediated pathologies[9-11]. The preclinical literature documenting MSC efficacy is vast and compelling. In rodent and large-animal models, MSCs have been shown to improve fracture healing, regenerate articular cartilage, limit infarct size following myocardial ischemia, ameliorate liver fibrosis, and prevent severe immune complications such as graft-vs-host disease[12-16]. These findings have catalyzed rapid clinical translation: As of today, thousands of MSC-related clinical trials are registered worldwide, exploring indications as diverse as osteoarthritis, Crohn’s disease, spinal cord injury, acute respiratory distress syndrome, systemic lupus erythematosus and so on[17,18]. Despite this intense global activity, the clinical impact of MSC therapies has been modest relative to initial expectations. The vast majority of clinical trials remain in early phases, with variable and sometimes inconclusive outcomes. Only a few products have achieved regulatory approval, and these are restricted to narrow indications (Table 1)[19-21].

Table 1 Mesenchymal stem cell-based products that have received market approval from major regulatory agencies.
Product name
Developer company
Cell type
Indication
Regulatory body
Country
Year
Ref.
Ryoncil™ (remestemcel-L)MesoblastAllo-BM-MSCPediatric SR-aGvHDUnited States FDAUnited States2024[20]
Cupistem®AnterogenAuto-AD-MSCsPerianal Crohn’s fistulaMFDSSouth Korea2012[19]
Cartistem®MedipostUC-MSCKnee OAMFDSSouth Korea2012[19]
Temcell HS®JCR PharmaceuticalsAllo-BM-MSCSR-aGvHDPMDAJapan2015[19,20]
Amimestrocel (conditional approval)Platinum Life Excellence BiotechUC-MSCSR-aGvHD with gastrointestinal tract involvement in 14 years of age and olderNMPAChina2025[21]
Cellgram AMIPharmicellAuto-BM-MSCAMIMFDSSouth Korea2011[19]
NeuroNata-RCorestemAuto-BM-MSCALSMFDSSouth Korea2014[19]
StempeucelStempeutics ResearchAllo-BM-MSCCLIDCGIIndia2016[19]
MesestroCellCell Tech PharmedAuto-BM-MSCOAIFDAIran2018[19]
StemiracNipro CorpAuto-BM-MSCSCIPMDAJapan2018[19]

The reasons for this translational bottleneck are multifactorial. At the in vitro level, variability in MSC isolation techniques, donor heterogeneity, and culture practices influence cell quality and potency[22]. Expansion under suboptimal conditions can accelerate genetic drift and senescence, undermining reproducibility and safety[23-25]. At the in vivo level, challenges include poor survival and engraftment of transplanted cells, differences between controlled laboratory models and the complexity of human disease, and uncertainty regarding long-term safety[26,27]. Finally, at the clinical and commercial levels, large-scale manufacturing, quality control, regulatory approval, patient selection, and cost-effectiveness present formidable barriers that must be systematically addressed before MSC therapies can achieve widespread adoption[28].

The persistence of these hurdles has led to increasing recognition that a more critical and integrative perspective is necessary. Enthusiasm for MSCs has often outpaced rigorous mechanistic understanding, and inconsistent methodologies have clouded interpretation of trial results. To move the field forward, it is essential to delineate the full spectrum of translational barriers, identify the underlying causes, and highlight innovative strategies that are being developed to overcome them. Only through such a comprehensive assessment can the promise of MSC-based therapies be converted into reproducible clinical reality. Hence, the aim of this review is to provide a comprehensive analysis of the barriers facing MSC therapy, categorized into three domains: In vitro challenges (standardization of isolation and culture, genetic/epigenetic stability, and biomimetic systems), in vivo challenges (translation from controlled systems to complex organisms, tissue-specific differences, microenvironmental influences, and long-term safety), and clinical challenges (manufacturing, logistics, regulatory approval, patient heterogeneity, and commercialization). For each challenge, potential strategies to overcome limitations are critically evaluated, with a view toward guiding the future of MSC translation.

CHALLENGES IN VITRO STUDIES

The first stage of MSC translation involves their derivation, culture, and expansion in controlled laboratory conditions. While in vitro studies have provided essential knowledge regarding MSC biology, they also represent a critical bottleneck because the quality of cells generated at this stage profoundly affects downstream preclinical and clinical outcomes. Below, the major categories of in vitro hurdles are discussed in detail.

Isolation and expansion techniques

The process of isolating MSCs is foundational yet fraught with variability. Bone marrow aspiration remains the classical source, but yields decline with donor age and the procedure itself is invasive, often producing heterogeneous mixtures of stromal and hematopoietic cells[29,30]. Adipose tissue offers a minimally invasive alternative with abundant yields; however, enzymatic digestion protocols using collagenase raise concerns regarding residual enzyme contamination and non-good manufacturing practice (GMP) compliance[31,32]. Umbilical cord-derived MSCs, especially from Wharton’s jelly, have gained popularity due to their neonatal origin, high proliferative index, and immunoprivileged status. However, variability between cord processing methods limits reproducibility across laboratories[33,34].

Several techniques exist to enrich for MSCs during isolation. Density gradient centrifugation remains common to isolate MSCs from bone marrow aspirate, although it fails to guarantee purity[35,36]. Explant culture allows MSCs to migrate from tissue fragments, preserving their phenotype but yielding small numbers over long culture periods. While, enzymatic digestion increases yield but risks damaging surface epitopes[33,35]. More advanced methods include immunoselection using surface markers such as CD271, Stro-1, and CD146, which enrich for subpopulations with superior clonogenicity and differentiation potential[37,38]. Nevertheless, these approaches are expensive and technically demanding, complicating their integration into routine manufacturing.

Large-scale expansion presents further challenges. Clinical applications often require 107-109 cells per patient, necessitating extensive passaging[39,40]. Repeated culture drives replicative senescence characterized by flattened morphology, diminished clonogenic potential, reduced expression of immunomodulatory molecules, and secretion of pro-inflammatory cytokines as part of a senescence-associated secretory phenotype[41]. Such functional deterioration undermines therapeutic potency. Strategies under investigation include limiting passage numbers, employing three-dimensional (3D) bioreactor systems that expand cells more efficiently, and using pharmacological agents such as antioxidants to delay senescence[40,42-44].

Media composition, culture practices, and environment

Media formulation exerts profound effects on MSC quality. Fetal bovine serum has historically served as the gold standard supplement because of its nutrient richness, but its use is increasingly untenable for clinical applications. Risks include xenogeneic pathogen transmission, immunogenic reactions, and substantial batch-to-batch variability[45,46]. Alternatives such as human platelet lysate and human serum have shown strong potential, with clinical studies demonstrating comparable or superior proliferation rates compared to fetal bovine serum-cultured MSCs[26,45,47]. Chemically defined serum-free formulations eliminate biological variability altogether, although high costs and source-specific optimization remain obstacles[48,49].

Culture practices have also great impact on MSC fate. Seeding density plays a critical role in maintaining proliferation and differentiation potential. Studies reported that excessively low density leads to no proliferation, while high density induces premature senescence[50,51]. Passage number has also a strong correlation with functional decline of MSCs. Hence, early-passage cells (P1-P5) are generally recommended for therapeutic applications[52-54]. Media replacement schedules could further influence outcomes. While frequent complete changes replenish nutrients and reduce reactive oxygen species accumulation, they may inadvertently remove autocrine and paracrine factors important for MSC maintenance[55-57]. Gradual or partial media exchange could be proposed as a compromise, maintaining signaling cues while minimizing metabolic stress.

The culture environment extends beyond media composition. Oxygen tension plays a pivotal role during in vitro expansion of MSCs. In vivo, MSCs reside in hypoxic niches (2%-8% O2), but conventional culture exposes them to atmospheric levels (approximately 21% O2), accelerating oxidative stress and senescence[24,58]. Culturing under physiological hypoxia has been shown to extend proliferative lifespan, enhance immunomodulation, and preserve differentiation potential[59,60]. Mechanical cues such as substrate stiffness, shear stress, and cyclic stretch also modulate MSC behavior. For instance, rigid substrates promote osteogenesis, whereas softer matrices encourage neurogenic or adipogenic differentiation[61-64]. Advanced bioreactor platforms simulate these mechanical conditions, offering scalable and physiologically relevant environments.

The in vivo origin of MSCs and the perivascular niche

MSCs were first identified by Friedenstein et al[65] in the 1960s and, around 30 years later, renamed characterized by Caplan[66]. MSCs are multipotent cells capable of differentiating into osteoblasts, adipocytes, and chondrocytes[67-69]. They are found in nearly all tissues and can migrate to sites of injury when required[70,71]. However, it is unclear whether they are resident within tissues or replenished from the bone marrow[72]. Furthermore, the in vivo origin of MSCs remains uncertain as well. A landmark study by Crisan et al[73,74] revealed that many MSC-like cells originate from perivascular compartments, primarily pericytes and adventitial cells, found throughout human tissues. These perivascular cells, when cultured, show clonogenicity and multilineage differentiation, supporting the perivascular-origin hypothesis of MSCs[73].

Pericytes arise from multiple embryonic lineages that vary by tissue type. Their exact developmental origins remain incompletely defined, with evidence indicating diverse sources across organs[75]. In the cephalic region and thymus, pericytes originate from the neuroectoderm[75-78]. In the lung, heart, liver, and gut, they primarily derive from the mesothelium[75,77,79]. In most other organs, pericytes are of mesodermal origin[80,81]. This developmental diversity suggests that pericytes are a heterogeneous population whose origin reflects the embryological context of each tissue[77].

While it is traditionally considered that MSCs are mesoderm-derived, similar cells from ectodermal origins, such as dental pulp stem cells, also exhibit MSC-like features defined by Dominici et al[69]. Regardless of the origin and tissue source, MSCs show multi-differentiation, strong immunomodulatory and therapeutic potential[82]. Although their core properties are alike, their sensitivity and responsiveness to inflammation or regeneration may vary depending on their tissue of origin[72,83]. Source selection, isolation tactics, and efforts to replicate in vivo potency during therapeutic application after in vitro expansion would all benefit from a better mechanistic understanding of these native identities, including niche signals that maintain quiescence, mobilization cues following injury, and lineage-priming.

Genetic and epigenetic stability

Beyond extrinsic culture factors, intrinsic genetic and epigenetic mechanisms also determine MSC behavior. Prolonged culture induces telomere shortening and genomic instability, which may manifest as chromosomal aberrations or aneuploidy[84,85]. Though such alterations do not undergo malignant transformation, however, it raises concern regarding both safety and efficacy for clinical application[86]. At the transcriptional level, prolonged passaging downregulates stemness-associated genes including sex-determining region Y-box 2, octamer-binding protein 4, and NANOG, diminishing self-renewal and multipotency[87].

Epigenetic drift represents another layer of instability. DNA methylation changes can silence stemness genes or activate differentiation pathways prematurely[88]. Histone modifications, such as increased H3K9 methylation, promote transcriptional repression and senescence, while acetylation of H3K27 is associated with proliferation and renewal[89,90]. Non-coding RNAs, particularly microRNAs (miRNAs), are emerging as critical regulators: MiR-21 supports proliferation and immunomodulation, miR-1246 has roles in paracrine activity, and dysregulation of miR-92a impairs angiogenesis[91-93]. Disruption of these epigenetic regulators during in vitro expansion may therefore critically undermine therapeutic function of MSCs. To mitigate these risks, several approaches are under exploration. Hypoxic culture reduces oxidative DNA damage and preserves telomere length[94,95]. Genetic and epigenetic monitoring, including karyotyping, telomere length assessment, and methylation profiling, are increasingly recommended as part of GMP quality control frameworks[96-104] (Table 2).

Table 2 Methods to detect genetic stability of in vitro expanded mesenchymal stem cells.
Assay
What it detects
Strengths
Limitations
Suggested use/frequency
Ref.
Conventional karyotype (G-banding)Large chromosomal abnormalitiesLow cost; detects numerical and structural chromosomal abnormalitiesLow resolution (approximately 5-10 Mb), not sufficient to predict full genetic stability, laboriousAt master cell bank and before clinical release[96-98]
FISHLow mosaicism, minor structural abnormalitiesRapid, sensitive, easy data interpretationThe chromosomal aberration being searched for must be known beforehandNot a screening technique, complementary to karyotyping for further proof of genotypic stability[96-98]
M-FISHOverall view of all chromosomes in a single assayHigh resolution (approximately 1.5 Mb)Highly expensive, difficult to analyze and interpret dataNot to be used to replace karyotyping[96,99]
a-CGHCopy number variationsHigher resolution than karyotype (≤ 50 kb), medium cost, no need for metaphases, requires only the genomic DNACannot detect balanced translocations, inversion and intragenic rearrangement; need of experienced cytogenetic specialistAt defined passage thresholds and for new donor lines or when recurrent abnormalities are found[96-98]
WGSSingle-nucleotide changes and copy number alterationsHighest resolution; comprehensiveCostly; complex analysis; uncertain clinical significance for many variantsAs confirmatory test for master bank or if abnormalities suspected[100]
Telomere length (qPCR)Telomere attritionSimple surrogate for replicative senescenceNot definitive for malignancy riskPeriodic, after a certain number of passages[101]
DNA methylation profilingEpigenetic drift; aging signaturesUseful for senescence, potencyInterpretation still evolvingFor research and characterization; consider for potency correlation[102-104]
Biomimetic scaffold designs

Conventional two-dimensional plastic culture lacks the complexity of the native extracellular matrix, leading to altered cell morphology and loss of functional properties[105]. Biomimetic scaffolds provide a more physiologically relevant microenvironment, supporting cell adhesion, survival, and lineage-specific differentiation[106,107]. Natural scaffolds derived from collagen, fibrin, hyaluronic acid, and chitosan offer biocompatibility but often suffer from variability and limited mechanical strength[108]. Synthetic scaffolds such as polylactic acid, polycaprolactone, and polyethylene glycol allow precise control over stiffness and degradation kinetics but require biochemical modification to support cell adhesion[109,110].

Hybrid scaffolds combine the advantages of natural and synthetic materials, while advanced fabrication techniques such as electrospinning and 3D bioprinting enable precise control of architecture and spatial distribution of bioactive cues[110]. For example, scaffolds incorporating bone morphogenetic proteins and high stiffness are being tailored for bone regeneration, while soft, aligned hydrogel scaffolds are designed to support neural regeneration. Gradients in scaffold stiffness or biochemical signaling further enable engineering of complex, multi-lineage tissues[111,112].

The translational potential of scaffolds depends not only on their biological performance but also on their manufacturability and regulatory approval. GMP-compliant scaffold production requires reproducibility, sterility, and biocompatibility testing. Ultimately, biomimetic scaffolds not only serve as tools for MSC expansion but also as delivery vehicles in vivo, enhancing engraftment, survival, and paracrine activity in therapeutic contexts.

CHALLENGES IN VIVO STUDIES

While in vitro studies provide controlled insights into MSC biology, their translation into in vivo contexts introduces far greater complexity. Once transplanted into living organisms, MSCs encounter host immune responses, dynamic microenvironments, and mechanical challenges that can profoundly alter their fate. Understanding and overcoming these barriers are essential for bridging preclinical promise with clinical efficacy.

Translational gaps within in vitro and animal models

One of the most persistent hurdles in MSC research is the discrepancy between robust results observed in vitro and inconsistent outcomes in vivo. In vitro, MSCs demonstrate high proliferation, differentiation, and paracrine secretion. However, following transplantation, survival rates drop precipitously. Studies show that within hours to days of systemic infusion, a majority of cells are cleared from circulation or undergo apoptosis due to anoikis, hypoxia, and immune-mediated attack[113,114]. This low engraftment contrasts sharply with the large therapeutic effects sometimes observed, suggesting that paracrine signaling rather than direct tissue integration is the primary mechanism of action[114,115].

Animal models, while indispensable, do not always recapitulate human physiology. Rodent immune systems differ in sensitivity and responsiveness, often exaggerating or underestimating MSC effects. Moreover, scaling cell doses from rodents to humans is non-linear; a dose that is therapeutic in a 25-g mouse does not translate proportionally to a 70-kg human[116]. Large animal models, such as swine or nonhuman primates, provide closer approximations but are expensive and logistically challenging[117]. The reliance on reductionist models partly explains why many therapies succeed in rodents yet falter in clinical trials.

Efforts to bridge these gaps include the use of humanized mouse models that harbor human immune cells and more faithfully replicate host-donor interactions. In addition, advanced organ-on-chip systems are being developed to complement animal testing, offering platforms that integrate human cells, microvascular perfusion, and physiological mechanical stresses. These innovations hold promise for better predicting in vivo performance of MSCs prior to costly clinical translation.

Source-specific therapeutic efficacy

MSC origin exerts significant influence on in vivo outcomes. Bone marrow-derived MSCs remain the most studied due to their historical precedence and strong osteogenic and chondrogenic differentiation capacity[68,118]. They have demonstrated efficacy in models of bone defect repair and osteoarthritis, but their limited proliferative potential and age-dependent decline restrict scalability. Adipose-derived MSCs are abundant and proliferative, making them attractive for high-volume applications such as wound healing and ischemic injury[119,120]. However, they exhibit weaker osteogenic differentiation, which may limit utility in orthopedic applications[68,118].

Umbilical cord-derived MSCs, particularly from Wharton’s jelly, display high proliferative capacity, immune privilege, and a robust paracrine secretome[121]. These properties make them particularly suited for allogeneic use, where immune rejection is a concern[122,123]. Placenta-derived MSCs offer angiogenic and immunomodulatory benefits, showing promise in vascular and inflammatory conditions[124]. Dental pulp and synovium-derived MSCs provide lineage-specific advantages: Dental pulp MSCs excel in neural and vascular regeneration[125], while synovial MSCs are highly chondrogenic and effective for cartilage repair[126].

Tailoring MSC source selection to specific disease contexts has emerged as a critical determinant of therapeutic success. For example, adipose-derived MSCs may be ideal for ischemic heart disease where vascular regeneration is required, whereas bone marrow-derived MSCs remain superior for bone and cartilage repair. However, systematic head-to-head clinical comparisons remain rare, and most studies employ heterogeneous definitions and protocols, complicating direct comparison[68,118-120,127-134] (Table 3).

Table 3 Detailed comparison of the therapeutic potential of mesenchymal stem cells derived from various sources.
Source
Preferential lineage-specific differentiation
Typical best-fit indications
Pros
Cons
Ref.
Bone marrowStrong osteogenic and chondrogenicBone defects, osteoarthritis, non-union bonesExtensively studied; good for bone and cartilage repairLower yield; donor age effect; invasive harvest procedure[68,118,127]
Adipose tissueCardiomyocytes, skeletal muscle cells, neurons, hepatocytes, and tenocytesWound healing, ischemic injury, soft tissue repairHigh yield of cells from liposuction; abundant tissue source; proliferative; useful for soft tissue regenerationLower osteogenic and chondrogenic differentiation compared to bone marrow[119,120,127,128]
Umbilical cord/Wharton’s jellyOsteogenic, chondrogenic, adipogenic, vascular, neuronalImmune modulation, systemic inflammatory disorders, arthritis, cardio- and cerebrovascularNeonatal origin, high proliferation, immune-privileged, trophic activity, antifibrotic, good for allogeneic off-the-shelf useVariable processing methods; donor bank logistics[127,129]
Synovial fluidTissue-biased properties (e.g., chondrogenic, neurogenic)Cartilage repairUseful for lineage-specific repairLower availability; niche-specific handling[127,130,131]
Dental pulpOdontoblasts, osteoblasts, endothelial cells, and nerve cellsNeuronal, vascular and odontogenic disordersHighly proliferative; can be derived from deciduous teeth as wellTiny primary tissue that gives lower cell yield[127,132-134]
Microenvironmental influence and mode of delivery

Once introduced into host tissues, MSCs encounter diverse microenvironments that shape their behavior[135]. The inflammatory milieu is particularly influential. In conditions of acute inflammation, exposure to cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and interferon-γ can enhance MSC immunomodulatory functions by stimulating the secretion of anti-inflammatory mediators like prostaglandin E2, indoleamine 2,3-dioxygenase, and IL-10[135,136]. Conversely, chronic inflammation or high levels of oxidative stress may induce apoptosis or senescence, reducing efficacy[137,138].

Mechanical properties of the host tissue also regulate MSC fate through mechanotransduction pathways. Rigid microenvironments encourage osteogenesis, whereas softer, elastic tissues promote adipogenesis or neurogenesis. This context dependence necessitates careful consideration of delivery site and mode[139-141]. Delivery strategies strongly influence engraftment and efficacy. Intravenous administration, while minimally invasive, results in the majority of cells being trapped in the pulmonary capillary bed, limiting systemic biodistribution[142]. This pulmonary first-pass effect has been harnessed in some contexts, such as acute lung injury, but represents a barrier in others[142]. Local injection into target tissues enhances retention and site-specific activity but is invasive and often associated with poor cell survival due to limited oxygen and nutrient supply[143]. Scaffold- or hydrogel-based delivery systems represent a promising compromise, providing structural support, mimicking extracellular matrix cues, and sustaining local paracrine signaling[144-146].

Long-term fate and safety

Long-term outcomes remain a major uncertainty in MSC transplantation. The majority of infused MSCs disappear within weeks, raising questions about the durability of their therapeutic effects[113,114]. While paracrine signaling may initiate repair cascades that persist beyond MSC survival, the lack of long-term engraftment limits their potential for direct tissue replacement[114].

Safety concerns are paramount. Over two decades of clinical use demonstrate that MSC therapy has a favorable short- to mid-term safety profile across various diseases and delivery routes (intravenous, intra-arterial, intra-articular, intrathecal, and intramuscular)[147,148]. Comprehensive systematic reviews and meta-analyses involving thousands of patients show consistent safety, with no increase in overall or serious adverse events, or in mortality, compared with controls, indicating a low short-term risk[148]. Studies revealed that only transient fever was consistently linked to MSC treatment, while arrhythmia and injection-site reactions showed minor, non-significant trends. All other adverse events displayed no meaningful association with MSC administration[148].

Although MSCs are generally considered safe and non-tumorigenic, rare reports have raised concerns about ectopic tissue formation or malignant transformation, particularly following extensive in vitro manipulation. The immunoprivileged nature of MSCs also raises the possibility of immune escape in the event of malignant transformation[149]. These risks underscore the importance of stringent genomic monitoring during culture and careful long-term surveillance following transplantation.

Advanced tracking technologies are being developed to study MSC fate in vivo. Imaging modalities such as magnetic resonance imaging, positron emission tomography, and bioluminescence allow non-invasive monitoring of cell survival and migration[150-152]. Genetic labeling with reporter constructs enables precise lineage tracing, although safety concerns limit their use in clinical contexts[153]. The integration of such tools into preclinical and clinical studies will be essential for clarifying the long-term behavior of transplanted MSCs.

Strategies to overcome in vivo challenges

Several approaches have been developed to improve MSC survival and functionality in vivo. Preconditioning, or “licensing”, of MSCs with hypoxia, cytokines, or pharmacological agents primes cells for hostile environments by upregulating survival and immunomodulatory pathways[113,154,155]. Genetic engineering or culture condition could help overexpression of C-X-C chemokine receptor type 4 that could enhances homing to injured tissues, while anti-apoptotic genes such as Bcl-2 improve survival[156-158]. Additionally, MSCs can be engineered to secrete therapeutic proteins, extending their repertoire beyond native capabilities[159].

Biomaterial-based delivery platforms, including hydrogels and scaffolds, improve engraftment by providing mechanical support and localized trophic signaling[106,109,112]. Encapsulation strategies can shield MSCs from immune attack while enabling controlled release of paracrine factors[160,161]. Co-delivery with supportive cells, such as endothelial progenitor cells, further enhances survival and functional integration[162,163].

An alternative approach gaining momentum is the use of EVs derived from MSCs. These vesicles, carrying proteins, lipids, and nucleic acids, mediate many of the paracrine effects attributed to MSCs[164,165]. EV-based therapies bypass the risks associated with live-cell transplantation, such as immune rejection and tumorigenicity, while offering scalability and stability[166]. Early preclinical data support their efficacy in cardiovascular, neurological, and musculoskeletal models, positioning EVs as a promising next-generation therapy[167-169].

CHALLENGES IN CLINICAL TRANSLATION

The leap from preclinical promise to clinical application represents the most formidable stage in the translational pathway of MSCs. Despite extensive investigation in vitro and in vivo, the transition into human patients exposes a new array of challenges that span biological variability, manufacturing scalability, regulatory complexity, and economic sustainability. This section critically examines the hurdles faced during clinical translation and outlines emerging strategies to overcome them.

Bridging preclinical and clinical studies

One of the first barriers lies in the translation of findings from animal models to human patients. While small animal models such as mice and rats provide mechanistic insights, their immune systems, metabolic rates, and tissue repair capacities differ substantially from those of humans. As a result, MSCs that demonstrate efficacy in rodents often fail to reproduce similar outcomes in clinical trials[116]. Large animal models such as pigs, dogs, and nonhuman primates offer closer physiological relevance but are costly, ethically challenging, and logistically complex[117,170]. Consequently, preclinical evidence often overestimates the magnitude and durability of clinical benefits.

Furthermore, dosing regimens established in animal studies rarely translate directly to human contexts. Whereas rodent experiments may use cell doses equivalent to millions of MSCs per kilogram of body weight, such quantities are neither practical nor safe in human applications[171]. Extrapolating an optimal therapeutic dose for humans remains uncertain, as there is no consensus on whether efficacy depends primarily on absolute cell number, total secretory output, or duration of exposure[172]. The absence of standardized potency assays compounds this uncertainty, hindering rational trial design.

Variability in clinical trial outcomes

Clinical trials investigating MSC therapy have produced mixed results. While some studies report significant improvements in conditions such as graft-vs-host disease, osteoarthritis, and Crohn’s disease, others demonstrate only marginal or no benefit compared to standard care[82,173,174]. This variability reflects a confluence of factors. Patient characteristics, including age, comorbidities, immune status, and disease stage, significantly influence responsiveness to MSCs[82]. Older patients often exhibit reduced MSC efficacy, likely due to systemic inflammatory environments and impaired endogenous repair mechanisms[175,176].

Trial heterogeneity further complicates interpretation. MSCs are derived from diverse sources, including bone marrow, adipose tissue, and umbilical cord, each have unique properties[68,119,124]. Differences in isolation techniques, expansion protocols, passage numbers, and storage conditions introduce additional variability[177,178]. Delivery routes, ranging from intravenous infusion to local injection, further diversify outcomes[142,143]. Without harmonized standards for cell characterization and potency testing, direct comparisons between trials are nearly impossible.

An additional concern is the placebo effect, which is particularly strong in regenerative medicine and musculoskeletal disorders. Patients often report subjective improvements in pain and function, complicating the interpretation of trial endpoints[179]. Objective biomarkers of MSC activity are lacking, making it difficult to distinguish true biological effects from placebo responses. Hence, there is an urgent need for reliable biomarkers to guide patient stratification and early response assessment in MSC therapy. Promising candidates include circulating inflammatory cytokines (e.g., IL-6, TNF-α), chemokines, exosomal miRNA signatures, and functional immune readouts such as T-cell activity. Preliminary data from rheumatic diseases suggest that elevated IL-6 and TNF-α may predict poor therapeutic response or help identify patients most likely to benefit from MSC-based immunomodulation[180,181]. However, these findings need prospective validation in randomized clinical trials. Future studies should incorporate biomarker endpoints particularly cytokine and EV profiles to enable adaptive patient selection and establish surrogate markers for MSC efficacy[72].

Manufacturing and supply chain challenges

A critical step in clinical translation is the production of MSCs at a scale and quality suitable for therapeutic use. Manufacturing must adhere to GMP standards, which mandate stringent control over every step of the process including tissue procurement, isolation, expansion, cryopreservation, and delivery. These requirements ensure safety and reproducibility but substantially increase cost and complexity[182,183].

The expansion of MSCs to clinically relevant doses is resource-intensive. Conventional two-dimensional culture flasks are laborious, while large-scale bioreactor systems provide greater efficiency but require significant capital investment and technical expertise[184,185]. Cryopreservation introduces further challenges: While it enables batch production and long-term storage, the freeze-thaw process can impair cell viability, alter surface marker expression, and reduce immunomodulatory potency[186]. Optimizing cryoprotectants and thawing protocols remains an area of active research.

Supply chain logistics represent another bottleneck. MSCs must be transported under tightly controlled temperature and sterility conditions, often across long distances from centralized production facilities to hospitals[187]. Any deviation in cold chain management risks compromising cell viability. Decentralized manufacturing at the point of care has been proposed, but standardizing quality control across multiple sites presents its own challenges[188]. A hybrid model involving regional hubs with robust distribution networks may balance scalability and quality assurance.

Regulatory and ethical complexities

Regulatory agencies such as the United States Food and Drug Administration, the European Medicines Agency, and Japan’s Pharmaceuticals and Medical Devices Agency classify MSCs as advanced therapy medicinal products, subjecting them to rigorous evaluation for safety, efficacy, and manufacturing consistency[189]. However, regulatory frameworks vary across regions, creating inconsistencies that hinder global collaboration and commercialization[190]. Harmonizing international standards is therefore a priority to facilitate multicenter trials and global approval pathways.

A persistent regulatory challenge lies in defining potency assays for MSCs. Unlike small molecules or monoclonal antibodies, MSCs exert therapeutic effects through multifaceted mechanisms, including immunomodulation, angiogenesis, and trophic support. Capturing this complexity in a single assay is not feasible. Instead, regulators encourage a panel of functional tests such as T-cell suppression assays, cytokine secretion profiles, and differentiation capacity to serve as surrogates[191-193]. Yet, these remain poorly standardized, leading to delays in approval.

Ethical considerations also come to the forefront in MSC therapy. Donor tissue sourcing, particularly from perinatal tissues such as umbilical cord and placenta, requires informed consent and careful ethical oversight. Autologous MSC therapies raise fewer ethical concerns but are limited by donor variability and the time required for expansion. Allogeneic therapies promise off-the-shelf availability but necessitate stringent donor screening and raise concerns about long-term immunogenicity[194,195]. Public perception and trust are fragile; cases of unregulated “stem cell clinics” offering unproven treatments have damaged credibility, emphasizing the need for robust ethical and regulatory oversight[196].

Economic and commercialization barriers

Even if MSC therapies clear scientific and regulatory hurdles, economic realities may limit widespread adoption. Manufacturing costs are high, with estimates ranging from $10000 to $30000 per patient dose depending on cell source and expansion method[19]. Without reimbursement strategies, access will be restricted to affluent healthcare systems and patients, exacerbating global health disparities. Demonstrating cost-effectiveness is therefore critical. Long-term follow-up studies that capture reductions in hospitalizations, surgeries, or disability are essential to justify reimbursement by payers.

Commercialization also requires addressing intellectual property and market competition. Patent protections around MSC sources, manufacturing methods, and delivery systems are often fragmented, creating barriers for new entrants. Moreover, the rise of alternative regenerative modalities, including induced pluripotent stem cells and EV-based therapies, threatens to outpace MSC commercialization unless scalability and affordability are improved. Partnerships between academia, industry, and healthcare systems are likely essential to create sustainable business models.

Future strategies for clinical translation

To overcome these hurdles, several innovative strategies are being pursued. Standardization of isolation, expansion, and potency assays across laboratories would reduce variability and enhance reproducibility. Automation and closed-system bioreactors promise to reduce contamination risks, labor costs, and variability, thereby improving scalability. Cryopreservation protocols are being optimized with novel cryoprotectants and controlled thawing systems to preserve potency.

On the regulatory front, international collaborations such as the International Society for Cell & Gene Therapy are working to harmonize definitions and guidelines, enabling smoother global approval pathways. Adaptive trial designs, incorporating biomarkers of response, may accelerate evaluation and regulatory approval. From a therapeutic perspective, MSC-derived EVs represent a cell-free alternative that circumvents many manufacturing and safety concerns[197]. Early studies suggest that EVs may replicate the paracrine effects of MSCs with greater scalability and stability[198,199]. Ultimately, the successful clinical translation of MSCs will depend on a combination of scientific innovation, regulatory clarity, ethical integrity, and economic feasibility. With coordinated efforts across stakeholders, MSC-based therapies have the potential to move beyond experimental promise and become an integral component of mainstream regenerative medicine.

FUTURE PERSPECTIVES

The field of MSC-based therapy is poised at a transformative juncture. Although significant hurdles remain, emerging technologies and evolving paradigms suggest new strategies to overcome current barriers. One promising avenue is the development of MSC-derived EVs and exosomes as cell-free therapies[198,199]. These nanoscale vesicles recapitulate many of the paracrine benefits of MSCs, including immunomodulation, angiogenesis, and tissue repair, while avoiding the risks associated with live cell transplantation such as immune rejection or tumorigenicity[166]. Early preclinical data support their efficacy across cardiovascular, neurological, and musculoskeletal disorders, and clinical translation efforts are already underway[200-202].

Another frontier is genetic and metabolic engineering of MSCs to enhance therapeutic potency. CRISPR-Cas9 and other editing platforms allow precise modification of genes related to survival, homing, or secretion of therapeutic factors. Engineering MSCs to overexpress chemokine receptors like C-X-C chemokine receptor type 4, or anti-apoptotic proteins such as Bcl-2, could demonstrate improved engraftment and functional outcomes[156,203]. Metabolic modulation, such as optimizing mitochondrial function or glycolytic pathways, may further refine their resilience in hostile microenvironments[55,204].

Biomaterials and bioengineering innovations offer complementary opportunities. Advanced hydrogels, 3D bioprinted scaffolds, and smart biomaterials capable of releasing growth factors or providing mechanical support are being integrated with MSC therapy to enhance survival and site-specific effects[109,110]. These platforms may also allow controlled delivery, reducing the need for repeated administrations.

On the translational side, standardization and harmonization of protocols remain urgent. The adoption of universal potency assays, shared biobanks, and international trial frameworks will improve reproducibility and accelerate approval pathways. In parallel, automation and closed-system bioreactors will reduce costs and variability, making MSC therapies more economically viable. Future clinical trial designs should incorporate precision medicine principles, stratifying patients by age, immune status, or disease subtype to maximize response rates.

Finally, ethical and societal considerations must remain central. Ensuring equitable access, preventing exploitation by unregulated stem cell clinics, and fostering public trust through transparency will be critical to the responsible development of MSC therapies. Engagement with policymakers, patient advocacy groups, and healthcare providers will help ensure that MSC-based therapies are not only effective but also accessible and ethically sound.

CONCLUSION

MSCs represent one of the most intensively studied platforms in regenerative medicine, offering multipotent differentiation capacity and robust paracrine activity. Despite decades of research, their translation from bench to bedside remains challenged by biological variability, limited engraftment, manufacturing bottlenecks, and regulatory complexity. However, advances in genetic engineering, biomaterials, and cell-free strategies such as EV therapies are reshaping the landscape. With concerted efforts toward standardization, scalability, and ethical oversight, MSC-based interventions may soon progress from experimental promise to reliable clinical reality (Figure 1). The future of regenerative medicine will likely see MSCs integrated into multifaceted therapeutic strategies, serving as both direct cellular agents and as sources of novel biologics, ultimately broadening the horizons of human healing and repair.

Figure 1
Figure 1 Challenges encountered during the translation of mesenchymal stem cell therapy from bench to bedside, and suggested solutions to overcome them. MSC: Mesenchymal stem cell; GMP: Good manufacturing practice.
ACKNOWLEDGEMENTS

The authors would like to thank Mahfuzul Islam and Ariful Islam for their support and reviewing this manuscript.

Footnotes

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

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: Bangladesh

Peer-review report’s classification

Scientific Quality: Grade B, Grade B

Novelty: Grade B, Grade C

Creativity or Innovation: Grade B, Grade C

Scientific Significance: Grade B, Grade B

P-Reviewer: Dai XL, Assistant Professor, Chief Physician, China; Ding LJ, PhD, Professor, China S-Editor: Wang JJ L-Editor: A P-Editor: Wang CH

References
1.  Wang S, Qu X, Zhao RC. Mesenchymal stem cells hold promise for regenerative medicine. Front Med. 2011;5:372-378.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 52]  [Cited by in RCA: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
2.  Patel DM, Shah J, Srivastava AS. Therapeutic potential of mesenchymal stem cells in regenerative medicine. Stem Cells Int. 2013;2013:496218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 132]  [Cited by in RCA: 157]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
3.  Rastegar F, Shenaq D, Huang J, Zhang W, Zhang BQ, He BC, Chen L, Zuo GW, Luo Q, Shi Q, Wagner ER, Huang E, Gao Y, Gao JL, Kim SH, Zhou JZ, Bi Y, Su Y, Zhu G, Luo J, Luo X, Qin J, Reid RR, Luu HH, Haydon RC, Deng ZL, He TC. Mesenchymal stem cells: Molecular characteristics and clinical applications. World J Stem Cells. 2010;2:67-80.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 145]  [Cited by in RCA: 165]  [Article Influence: 10.3]  [Reference Citation Analysis (2)]
4.  Soliman H, Theret M, Scott W, Hill L, Underhill TM, Hinz B, Rossi FMV. Multipotent stromal cells: One name, multiple identities. Cell Stem Cell. 2021;28:1690-1707.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 106]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
5.  Shammaa R, El-Kadiry AE, Abusarah J, Rafei M. Mesenchymal Stem Cells Beyond Regenerative Medicine. Front Cell Dev Biol. 2020;8:72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
6.  Dabrowska S, Andrzejewska A, Janowski M, Lukomska B. Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cells and Extracellular Vesicles: Therapeutic Outlook for Inflammatory and Degenerative Diseases. Front Immunol. 2020;11:591065.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 42]  [Cited by in RCA: 156]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
7.  Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016;7:125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 438]  [Cited by in RCA: 632]  [Article Influence: 63.2]  [Reference Citation Analysis (0)]
8.  Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC. Paracrine Mechanisms of Mesenchymal Stem Cells in Tissue Repair. Methods Mol Biol. 2016;1416:123-146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 220]  [Cited by in RCA: 320]  [Article Influence: 35.6]  [Reference Citation Analysis (0)]
9.  Zhidu S, Ying T, Rui J, Chao Z. Translational potential of mesenchymal stem cells in regenerative therapies for human diseases: challenges and opportunities. Stem Cell Res Ther. 2024;15:266.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 68]  [Reference Citation Analysis (0)]
10.  Zeng L, Liu C, Wu Y, Liu S, Zheng Y, Hao W, Wang D, Sun L. Efficacy and safety of mesenchymal stromal cell transplantation in the treatment of autoimmune and rheumatic immune diseases: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res Ther. 2025;16:65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
11.  Trounson A, McDonald C. Stem Cell Therapies in Clinical Trials: Progress and Challenges. Cell Stem Cell. 2015;17:11-22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 846]  [Cited by in RCA: 1034]  [Article Influence: 103.4]  [Reference Citation Analysis (0)]
12.  Kim MS, Chung HJ, Kim KI. Optimal concentration of mesenchymal stem cells for fracture healing in a rat model with long bone fracture. World J Stem Cells. 2022;14:839-850.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
13.  Charles CJ, Li RR, Yeung T, Mazlan SMI, Lai RC, de Kleijn DPV, Lim SK, Richards AM. Systemic Mesenchymal Stem Cell-Derived Exosomes Reduce Myocardial Infarct Size: Characterization With MRI in a Porcine Model. Front Cardiovasc Med. 2020;7:601990.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
14.  Huang B, Cheng X, Wang H, Huang W, la Ga Hu Z, Wang D, Zhang K, Zhang H, Xue Z, Da Y, Zhang N, Hu Y, Yao Z, Qiao L, Gao F, Zhang R. Mesenchymal stem cells and their secreted molecules predominantly ameliorate fulminant hepatic failure and chronic liver fibrosis in mice respectively. J Transl Med. 2016;14:45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 133]  [Cited by in RCA: 141]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
15.  Feng X, Feng B, Zhou J, Yang J, Pan Q, Yu J, Shang D, Li L, Cao H. Mesenchymal stem cells alleviate mouse liver fibrosis by inhibiting pathogenic function of intrahepatic B cells. Hepatology. 2025;81:1211-1227.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 28]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
16.  Wang L, Zhang H, Guan L, Zhao S, Gu Z, Wei H, Gao Z, Wang F, Yang N, Luo L, Li Y, Wang L, Liu D, Gao C. Mesenchymal stem cells provide prophylaxis against acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation: A meta-analysis of animal models. Oncotarget. 2016;7:61764-61774.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
17.  Jovic D, Yu Y, Wang D, Wang K, Li H, Xu F, Liu C, Liu J, Luo Y. A Brief Overview of Global Trends in MSC-Based Cell Therapy. Stem Cell Rev Rep. 2022;18:1525-1545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 157]  [Article Influence: 39.3]  [Reference Citation Analysis (0)]
18.  Chen Y, Li X, Zhang J, Peng J, Huang F, Bao J, Fan Y, Huang S. Global clinical trials on stem cell therapy for autoimmune diseases: trends and future directions. Front Immunol. 2025;16:1616231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
19.  Fernández-Garza LE, Barrera-Barrera SA, Barrera-Saldaña HA. Mesenchymal Stem Cell Therapies Approved by Regulatory Agencies around the World. Pharmaceuticals (Basel). 2023;16:1334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 28]  [Reference Citation Analysis (0)]
20.  Giebel B. A milestone for the therapeutic EV field: FDA approves Ryoncil, an allogeneic bone marrow-derived mesenchymal stromal cell therapy. Extracell Vesicles Circ Nucl Acids. 2025;6:183-190.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
21.  Liu Y, Ge C, Chen X, Lu S, Wang T. Trends in the development of cellular and gene therapy in China. Nat Rev Drug Discov. 2025;24:656-657.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
22.  Calcat-I-Cervera S, Rendra E, Scaccia E, Amadeo F, Hanson V, Wilm B, Murray P, O'Brien T, Taylor A, Bieback K. Harmonised culture procedures minimise but do not eliminate mesenchymal stromal cell donor and tissue variability in a decentralised multicentre manufacturing approach. Stem Cell Res Ther. 2023;14:120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
23.  Neri S. Genetic Stability of Mesenchymal Stromal Cells for Regenerative Medicine Applications: A Fundamental Biosafety Aspect. Int J Mol Sci. 2019;20:2406.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 73]  [Cited by in RCA: 126]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
24.  Haque N, Rahman MT, Abu Kasim NH, Alabsi AM. Hypoxic culture conditions as a solution for mesenchymal stem cell based regenerative therapy. ScientificWorldJournal. 2013;2013:632972.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 125]  [Cited by in RCA: 160]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
25.  Wang J, Zhang M, Wang H. Emerging Landscape of Mesenchymal Stem Cell Senescence Mechanisms and Implications on Therapeutic Strategies. ACS Pharmacol Transl Sci. 2024;7:2306-2325.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
26.  Haque N, Khan IM, Abu Kasim NH. Survival and immunomodulation of stem cells from human extracted deciduous teeth expanded in pooled human and foetal bovine sera. Cytokine. 2019;120:144-154.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Mastrolia I, Foppiani EM, Murgia A, Candini O, Samarelli AV, Grisendi G, Veronesi E, Horwitz EM, Dominici M. Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review. Stem Cells Transl Med. 2019;8:1135-1148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 112]  [Cited by in RCA: 237]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
28.  Williams ME, Banche Niclot F, Rota S, Lim J, Machado J, de Azevedo R, Castillo K, Adebiyi S, Sreenivasan R, Kota D, Mcculloch PC, Taraballi F. Optimizing mesenchymal stem cell therapy: from isolation to GMP-compliant expansion for clinical application. BMC Mol Cell Biol. 2025;26:15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
29.  Wu LW, Wang YL, Christensen JM, Khalifian S, Schneeberger S, Raimondi G, Cooney DS, Lee WP, Brandacher G. Donor age negatively affects the immunoregulatory properties of both adipose and bone marrow derived mesenchymal stem cells. Transpl Immunol. 2014;30:122-127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 74]  [Cited by in RCA: 69]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
30.  Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS One. 2014;9:e115963.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 210]  [Cited by in RCA: 250]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
31.  Guiotto M, Palombella S, Brambilla S, Applegate LA, Riehle M, Hart A, Raffoul W, di Summa PG. Isolation and Culture of Human Adipose-Derived Stem Cells With an Innovative Xenogeneic-Free Method for Human Therapy. J Vis Exp.  2023.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
32.  Athanasiadis A, Ambele MA, Pepper MS. Isolation of Adipose-Derived Mesenchymal Stromal/Stem Cells. Methods Mol Biol. 2025;2938:125-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
33.  Paladino FV, Peixoto-Cruz JS, Santacruz-Perez C, Goldberg AC. Comparison between isolation protocols highlights intrinsic variability of human umbilical cord mesenchymal cells. Cell Tissue Bank. 2016;17:123-136.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
34.  Vieira Paladino F, de Moraes Rodrigues J, da Silva A, Goldberg AC. The Immunomodulatory Potential of Wharton's Jelly Mesenchymal Stem/Stromal Cells. Stem Cells Int. 2019;2019:3548917.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
35.  García-Muñoz E, Vives J. Towards the standardization of methods of tissue processing for the isolation of mesenchymal stromal cells for clinical use. Cytotechnology. 2021;73:513-522.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
36.  Gudleviciene Z, Kundrotas G, Liudkeviciene R, Rascon J, Jurga M. Quick and effective method of bone marrow mesenchymal stem cell extraction. Open Med (Wars). 2015;10:44-49.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
37.  Mo M, Wang S, Zhou Y, Li H, Wu Y. Mesenchymal stem cell subpopulations: phenotype, property and therapeutic potential. Cell Mol Life Sci. 2016;73:3311-3321.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 95]  [Cited by in RCA: 98]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
38.  Chen S, Liang B, Xu J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med. 2024;22:459.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 25]  [Reference Citation Analysis (0)]
39.  Hassan MNFB, Yazid MD, Yunus MHM, Chowdhury SR, Lokanathan Y, Idrus RBH, Ng AMH, Law JX. Large-Scale Expansion of Human Mesenchymal Stem Cells. Stem Cells Int. 2020;2020:9529465.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
40.  Soder RP, Dudley NR, Dawn B. Microcarrier-based clinical-grade manufacturing of therapeutic Wharton's jelly mesenchymal stromal cells. Cytotherapy. 2024;26:1556-1565.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
41.  Weng Z, Wang Y, Ouchi T, Liu H, Qiao X, Wu C, Zhao Z, Li L, Li B. Mesenchymal Stem/Stromal Cell Senescence: Hallmarks, Mechanisms, and Combating Strategies. Stem Cells Transl Med. 2022;11:356-371.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 168]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
42.  Zheng Z, Wang X, Ouyang L, Chen W, Zhang L, Cao Y. Antioxidants Improve the Proliferation and Efficacy of hUC-MSCs against H(2)O(2)-Induced Senescence. Antioxidants (Basel). 2023;12:1334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
43.  Li X, Zhan J, Hou Y, Hou Y, Chen S, Luo D, Luan J, Wang L, Lin D. Coenzyme Q10 Regulation of Apoptosis and Oxidative Stress in H(2)O(2) Induced BMSC Death by Modulating the Nrf-2/NQO-1 Signaling Pathway and Its Application in a Model of Spinal Cord Injury. Oxid Med Cell Longev. 2019;2019:6493081.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 51]  [Cited by in RCA: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
44.  Wang X, Ouyang L, Chen W, Cao Y, Zhang L. Efficient expansion and delayed senescence of hUC-MSCs by microcarrier-bioreactor system. Stem Cell Res Ther. 2023;14:284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
45.  Witzeneder K, Lindenmair A, Gabriel C, Höller K, Theiß D, Redl H, Hennerbichler S. Human-derived alternatives to fetal bovine serum in cell culture. Transfus Med Hemother. 2013;40:417-423.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
46.  Pilgrim CR, McCahill KA, Rops JG, Dufour JM, Russell KA, Koch TG. A Review of Fetal Bovine Serum in the Culture of Mesenchymal Stromal Cells and Potential Alternatives for Veterinary Medicine. Front Vet Sci. 2022;9:859025.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 44]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
47.  Haque N, Abu Kasim NH. Pooled Human Serum Increases Regenerative Potential of In Vitro Expanded Stem Cells from Human Extracted Deciduous Teeth. Adv Exp Med Biol. 2018;1083:29-44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
48.  Tekkatte C, Gunasingh GP, Cherian KM, Sankaranarayanan K. "Humanized" stem cell culture techniques: the animal serum controversy. Stem Cells Int. 2011;2011:504723.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 116]  [Cited by in RCA: 145]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
49.  Gottipamula S, Muttigi MS, Kolkundkar U, Seetharam RN. Serum-free media for the production of human mesenchymal stromal cells: a review. Cell Prolif. 2013;46:608-627.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 62]  [Cited by in RCA: 82]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
50.  Kim SN, Choi B, Lee CJ, Moon JH, Kim MK, Chung E, Song SU. Culturing at Low Cell Density Delays Cellular Senescence of Human Bone Marrow-Derived Mesenchymal Stem Cells in Long-Term Cultures. Int J Stem Cells. 2021;14:103-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 13]  [Reference Citation Analysis (0)]
51.  Mareschi K, Rustichelli D, Calabrese R, Gunetti M, Sanavio F, Castiglia S, Risso A, Ferrero I, Tarella C, Fagioli F. Multipotent mesenchymal stromal stem cell expansion by plating whole bone marrow at a low cellular density: a more advantageous method for clinical use. Stem Cells Int. 2012;2012:920581.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 47]  [Cited by in RCA: 51]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
52.  Drela K, Stanaszek L, Nowakowski A, Kuczynska Z, Lukomska B. Experimental Strategies of Mesenchymal Stem Cell Propagation: Adverse Events and Potential Risk of Functional Changes. Stem Cells Int. 2019;2019:7012692.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 54]  [Cited by in RCA: 90]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
53.  Tian L, Wang W, Li X, Chen Y, Song Q, Yuan L, Hao T, Gu J, Dong J. Whole transcriptome scanning and validation of negatively related genes in UC-MSCs. Heliyon. 2024;10:e27996.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
54.  Patntirapong S, Khankhow J, Julamorn S. Long-term passage impacts human dental pulp stem cell activities and cell response to drug addition in vitro. PeerJ. 2024;12:e17913.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
55.  Cenini G, Lloret A, Cascella R. Oxidative Stress in Neurodegenerative Diseases: From a Mitochondrial Point of View. Oxid Med Cell Longev. 2019;2019:2105607.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 221]  [Cited by in RCA: 345]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
56.  Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther. 2023;8:282.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 249]  [Reference Citation Analysis (0)]
57.  Mahrouf-Yorgov M, Augeul L, Da Silva CC, Jourdan M, Rigolet M, Manin S, Ferrera R, Ovize M, Henry A, Guguin A, Meningaud JP, Dubois-Randé JL, Motterlini R, Foresti R, Rodriguez AM. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017;24:1224-1238.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 131]  [Cited by in RCA: 219]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
58.  Di Mattia M, Mauro A, Citeroni MR, Dufrusine B, Peserico A, Russo V, Berardinelli P, Dainese E, Cimini A, Barboni B. Insight into Hypoxia Stemness Control. Cells. 2021;10:2161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
59.  Jaraba-Álvarez WV, Uscanga-Palomeque AC, Sanchez-Giraldo V, Madrid C, Ortega-Arellano H, Halpert K, Quintero-Gil C. Hypoxia-induced metabolic reprogramming in mesenchymal stem cells: unlocking the regenerative potential of secreted factors. Front Cell Dev Biol. 2025;13:1609082.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
60.  Kwon SY, Chun SY, Ha YS, Kim DH, Kim J, Song PH, Kim HT, Yoo ES, Kim BS, Kwon TG. Hypoxia Enhances Cell Properties of Human Mesenchymal Stem Cells. Tissue Eng Regen Med. 2017;14:595-604.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 58]  [Cited by in RCA: 94]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
61.  Olivares-Navarrete R, Lee EM, Smith K, Hyzy SL, Doroudi M, Williams JK, Gall K, Boyan BD, Schwartz Z. Substrate Stiffness Controls Osteoblastic and Chondrocytic Differentiation of Mesenchymal Stem Cells without Exogenous Stimuli. PLoS One. 2017;12:e0170312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 111]  [Cited by in RCA: 150]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
62.  Kozaniti FK, Deligianni DD, Georgiou MD, Portan DV. The Role of Substrate Topography and Stiffness on MSC Cells Functions: Key Material Properties for Biomimetic Bone Tissue Engineering. Biomimetics (Basel). 2021;7:7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
63.  Li J, Liu Y, Zhang Y, Yao B, Enhejirigala, Li Z, Song W, Wang Y, Duan X, Yuan X, Fu X, Huang S. Biophysical and Biochemical Cues of Biomaterials Guide Mesenchymal Stem Cell Behaviors. Front Cell Dev Biol. 2021;9:640388.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 79]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
64.  Nellinger S, Kluger PJ. How Mechanical and Physicochemical Material Characteristics Influence Adipose-Derived Stem Cell Fate. Int J Mol Sci. 2023;24:3551.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
65.  Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393-403.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 408]  [Cited by in RCA: 962]  [Article Influence: 17.2]  [Reference Citation Analysis (1)]
66.  Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641-650.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3539]  [Cited by in RCA: 3327]  [Article Influence: 95.1]  [Reference Citation Analysis (0)]
67.  Orbay H, Tobita M, Mizuno H. Mesenchymal stem cells isolated from adipose and other tissues: basic biological properties and clinical applications. Stem Cells Int. 2012;2012:461718.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 142]  [Cited by in RCA: 163]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
68.  Via AG, Frizziero A, Oliva F. Biological properties of mesenchymal Stem Cells from different sources. Muscles Ligaments Tendons J. 2012;2:154-162.  [PubMed]  [DOI]
69.  Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315-317.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11055]  [Cited by in RCA: 12943]  [Article Influence: 681.2]  [Reference Citation Analysis (10)]
70.  Eggenhofer E, Luk F, Dahlke MH, Hoogduijn MJ. The life and fate of mesenchymal stem cells. Front Immunol. 2014;5:148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 270]  [Cited by in RCA: 349]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
71.  Wu X, Jiang J, Gu Z, Zhang J, Chen Y, Liu X. Mesenchymal stromal cell therapies: immunomodulatory properties and clinical progress. Stem Cell Res Ther. 2020;11:345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 213]  [Cited by in RCA: 216]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
72.  Zhou J, Shi Y. Mesenchymal stem/stromal cells (MSCs): origin, immune regulation, and clinical applications. Cell Mol Immunol. 2023;20:555-557.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 67]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
73.  Crisan M, Corselli M, Chen WC, Péault B. Perivascular cells for regenerative medicine. J Cell Mol Med. 2012;16:2851-2860.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 211]  [Cited by in RCA: 225]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
74.  Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, Norotte C, Teng PN, Traas J, Schugar R, Deasy BM, Badylak S, Buhring HJ, Giacobino JP, Lazzari L, Huard J, Péault B. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301-313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2900]  [Cited by in RCA: 2902]  [Article Influence: 161.2]  [Reference Citation Analysis (0)]
75.  Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21:193-215.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1689]  [Cited by in RCA: 2027]  [Article Influence: 135.1]  [Reference Citation Analysis (0)]
76.  Müller SM, Stolt CC, Terszowski G, Blum C, Amagai T, Kessaris N, Iannarelli P, Richardson WD, Wegner M, Rodewald HR. Neural crest origin of perivascular mesenchyme in the adult thymus. J Immunol. 2008;180:5344-5351.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 111]  [Cited by in RCA: 104]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
77.  Dias Moura Prazeres PH, Sena IFG, Borges IDT, de Azevedo PO, Andreotti JP, de Paiva AE, de Almeida VM, de Paula Guerra DA, Pinheiro Dos Santos GS, Mintz A, Delbono O, Birbrair A. Pericytes are heterogeneous in their origin within the same tissue. Dev Biol. 2017;427:6-11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 112]  [Cited by in RCA: 111]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
78.  Zachariah MA, Cyster JG. Neural crest-derived pericytes promote egress of mature thymocytes at the corticomedullary junction. Science. 2010;328:1129-1135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 170]  [Cited by in RCA: 162]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
79.  Que J, Wilm B, Hasegawa H, Wang F, Bader D, Hogan BL. Mesothelium contributes to vascular smooth muscle and mesenchyme during lung development. Proc Natl Acad Sci U S A. 2008;105:16626-16630.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 197]  [Cited by in RCA: 212]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
80.  Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond). 2015;128:81-93.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 200]  [Cited by in RCA: 181]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
81.  Harrell CR, Simovic Markovic B, Fellabaum C, Arsenijevic A, Djonov V, Volarevic V. Molecular mechanisms underlying therapeutic potential of pericytes. J Biomed Sci. 2018;25:21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 83]  [Cited by in RCA: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
82.  Han X, Liao R, Li X, Zhang C, Huo S, Qin L, Xiong Y, He T, Xiao G, Zhang T. Mesenchymal stem cells in treating human diseases: molecular mechanisms and clinical studies. Signal Transduct Target Ther. 2025;10:262.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 21]  [Article Influence: 21.0]  [Reference Citation Analysis (2)]
83.  Li J, Wu Z, Zhao L, Liu Y, Su Y, Gong X, Liu F, Zhang L. The heterogeneity of mesenchymal stem cells: an important issue to be addressed in cell therapy. Stem Cell Res Ther. 2023;14:381.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 89]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
84.  Estrada JC, Torres Y, Benguría A, Dopazo A, Roche E, Carrera-Quintanar L, Pérez RA, Enríquez JA, Torres R, Ramírez JC, Samper E, Bernad A. Human mesenchymal stem cell-replicative senescence and oxidative stress are closely linked to aneuploidy. Cell Death Dis. 2013;4:e691.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 158]  [Cited by in RCA: 181]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
85.  Nikitina V, Astrelina T, Nugis V, Ostashkin A, Karaseva T, Dobrovolskaya E, Usupzhanova D, Suchkova Y, Lomonosova E, Rodin S, Brunchukov V, Lauk-Dubitskiy S, Brumberg V, Machova A, Kobzeva I, Bushmanov A, Samoilov A. Clonal chromosomal and genomic instability during human multipotent mesenchymal stromal cells long-term culture. PLoS One. 2018;13:e0192445.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
86.  Wang Y, Zhang Z, Chi Y, Zhang Q, Xu F, Yang Z, Meng L, Yang S, Yan S, Mao A, Zhang J, Yang Y, Wang S, Cui J, Liang L, Ji Y, Han ZB, Fang X, Han ZC. Long-term cultured mesenchymal stem cells frequently develop genomic mutations but do not undergo malignant transformation. Cell Death Dis. 2013;4:e950.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 107]  [Cited by in RCA: 128]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
87.  Tsai CC, Su PF, Huang YF, Yew TL, Hung SC. Oct4 and Nanog directly regulate Dnmt1 to maintain self-renewal and undifferentiated state in mesenchymal stem cells. Mol Cell. 2012;47:169-182.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 246]  [Cited by in RCA: 289]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
88.  Wang R, Wang Y, Zhu L, Liu Y, Li W. Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int. 2020;2020:8836258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
89.  Fernández AF, Bayón GF, Urdinguio RG, Toraño EG, García MG, Carella A, Petrus-Reurer S, Ferrero C, Martinez-Camblor P, Cubillo I, García-Castro J, Delgado-Calle J, Pérez-Campo FM, Riancho JA, Bueno C, Menéndez P, Mentink A, Mareschi K, Claire F, Fagnani C, Medda E, Toccaceli V, Brescianini S, Moran S, Esteller M, Stolzing A, de Boer J, Nisticò L, Stazi MA, Fraga MF. H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Res. 2015;25:27-40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 114]  [Cited by in RCA: 100]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
90.  Schellenberg A, Lin Q, Schüler H, Koch CM, Joussen S, Denecke B, Walenda G, Pallua N, Suschek CV, Zenke M, Wagner W. Replicative senescence of mesenchymal stem cells causes DNA-methylation changes which correlate with repressive histone marks. Aging (Albany NY). 2011;3:873-888.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 129]  [Cited by in RCA: 143]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
91.  Efimenko A, Sagaradze G, Akopyan Z, Lopatina T, Kalinina N. Data supporting that miR-92a suppresses angiogenic activity of adipose-derived mesenchymal stromal cells by down-regulating hepatocyte growth factor. Data Brief. 2016;6:295-310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
92.  Morente-López M, Mato-Basalo R, Lucio-Gallego S, Gil C, Carrera M, Fafián-Labora JA, Mateos J, Arufe MC. Effect of miR-21 in mesenchymal stem cells-derived extracellular vesicles behavior. Stem Cell Res Ther. 2023;14:383.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
93.  Ramadan F, Saab R, Ghamloush F, Khoueiry R, Herceg Z, Gomez L, Badran B, Clezardin P, Hussein N, Cohen PA, Ghayad SE. Exosome-Mediated Paracrine Signaling Unveils miR-1246 as a Driver of Aggressiveness in Fusion-Negative Rhabdomyosarcoma. Cancers (Basel). 2024;16:1652.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
94.  Bigot N, Mouche A, Preti M, Loisel S, Renoud ML, Le Guével R, Sensebé L, Tarte K, Pedeux R. Hypoxia Differentially Modulates the Genomic Stability of Clinical-Grade ADSCs and BM-MSCs in Long-Term Culture. Stem Cells. 2015;33:3608-3620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 35]  [Cited by in RCA: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
95.  Estrada JC, Albo C, Benguría A, Dopazo A, López-Romero P, Carrera-Quintanar L, Roche E, Clemente EP, Enríquez JA, Bernad A, Samper E. Culture of human mesenchymal stem cells at low oxygen tension improves growth and genetic stability by activating glycolysis. Cell Death Differ. 2012;19:743-755.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 187]  [Cited by in RCA: 216]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
96.  Catalina P, Cobo F, Cortés JL, Nieto AI, Cabrera C, Montes R, Concha A, Menendez P. Conventional and molecular cytogenetic diagnostic methods in stem cell research: a concise review. Cell Biol Int. 2007;31:861-869.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 41]  [Cited by in RCA: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
97.  Barkholt L, Flory E, Jekerle V, Lucas-Samuel S, Ahnert P, Bisset L, Büscher D, Fibbe W, Foussat A, Kwa M, Lantz O, Mačiulaitis R, Palomäki T, Schneider CK, Sensebé L, Tachdjian G, Tarte K, Tosca L, Salmikangas P. Risk of tumorigenicity in mesenchymal stromal cell-based therapies--bridging scientific observations and regulatory viewpoints. Cytotherapy. 2013;15:753-759.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 216]  [Cited by in RCA: 303]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
98.  Afonso Cornélio D, Batistuzzo de Medeiros SR. Genetic evaluation of mesenchymal stem cells. Rev Bras Hematol Hemoter. 2014;36:238-240.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
99.  Meisner LF, Johnson JA. Protocols for cytogenetic studies of human embryonic stem cells. Methods. 2008;45:133-141.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 70]  [Cited by in RCA: 70]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
100.  Cai J, Miao X, Li Y, Smith C, Tsang K, Cheng L, Wang QF. Whole-genome sequencing identifies genetic variances in culture-expanded human mesenchymal stem cells. Stem Cell Reports. 2014;3:227-233.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
101.  Samsonraj RM, Raghunath M, Hui JH, Ling L, Nurcombe V, Cool SM. Telomere length analysis of human mesenchymal stem cells by quantitative PCR. Gene. 2013;519:348-355.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 34]  [Cited by in RCA: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
102.  Bentivegna A, Miloso M, Riva G, Foudah D, Butta V, Dalprà L, Tredici G. DNA Methylation Changes during In Vitro Propagation of Human Mesenchymal Stem Cells: Implications for Their Genomic Stability? Stem Cells Int. 2013;2013:192425.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 39]  [Cited by in RCA: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
103.  Choi MR, In YH, Park J, Park T, Jung KH, Chai JC, Chung MK, Lee YS, Chai YG. Genome-scale DNA methylation pattern profiling of human bone marrow mesenchymal stem cells in long-term culture. Exp Mol Med. 2012;44:503-512.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
104.  Gómez R, Barter MJ, Alonso-Pérez A, Skelton AJ, Proctor C, Herrero-Beaumont G, Young DA. DNA methylation analysis identifies key transcription factors involved in mesenchymal stem cell osteogenic differentiation. Biol Res. 2023;56:9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
105.  Kapałczyńska M, Kolenda T, Przybyła W, Zajączkowska M, Teresiak A, Filas V, Ibbs M, Bliźniak R, Łuczewski Ł, Lamperska K. 2D and 3D cell cultures - a comparison of different types of cancer cell cultures. Arch Med Sci. 2018;14:910-919.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 263]  [Cited by in RCA: 635]  [Article Influence: 79.4]  [Reference Citation Analysis (0)]
106.  Ye B, Wu B, Su Y, Sun T, Guo X. Recent Advances in the Application of Natural and Synthetic Polymer-Based Scaffolds in Musculoskeletal Regeneration. Polymers (Basel). 2022;14:4566.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 28]  [Reference Citation Analysis (0)]
107.  Nourany M, Makaremy A, Bazrpash S, Hosseini S. Bimodal macroporous 3D scaffolds based on compatibilized PCL and PLA blend using PCL-PEG-PCL block copolymers and cellulose nanocrystals for osteogenic differentiation of hMSCs. Int J Biol Macromol. 2025;299:140149.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
108.  Zhao L, Zhou Y, Zhang J, Liang H, Chen X, Tan H. Natural Polymer-Based Hydrogels: From Polymer to Biomedical Applications. Pharmaceutics. 2023;15:2514.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 136]  [Reference Citation Analysis (0)]
109.  Ferrari M, Taboni S, Chan HHL, Townson J, Gualtieri T, Franz L, Ruaro A, Mathews S, Daly MJ, Douglas CM, Eu D, Sahovaler A, Muhanna N, Ventura M, Dey K, Pandini S, Pasini C, Re F, Bernardi S, Bosio K, Mattavelli D, Doglietto F, Joshi S, Gilbert RW, Nicolai P, Viswanathan S, Sartore L, Russo D, Irish JC. Hydrogel-chitosan and polylactic acid-polycaprolactone bioengineered scaffolds for reconstruction of mandibular defects: a preclinical in vivo study with assessment of translationally relevant aspects. Front Bioeng Biotechnol. 2024;12:1353523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
110.  Samadian H, Farzamfar S, Vaez A, Ehterami A, Bit A, Alam M, Goodarzi A, Darya G, Salehi M. A tailored polylactic acid/polycaprolactone biodegradable and bioactive 3D porous scaffold containing gelatin nanofibers and Taurine for bone regeneration. Sci Rep. 2020;10:13366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 59]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
111.  Sun Y, Wu Q, Zhang Y, Dai K, Wei Y. 3D-bioprinted gradient-structured scaffold generates anisotropic cartilage with vascularization by pore-size-dependent activation of HIF1α/FAK signaling axis. Nanomedicine. 2021;37:102426.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
112.  Lee SS, Santschi M, Ferguson SJ. A Biomimetic Macroporous Hybrid Scaffold with Sustained Drug Delivery for Enhanced Bone Regeneration. Biomacromolecules. 2021;22:2460-2471.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
113.  Haque N, Kasim NH, Rahman MT. Optimization of pre-transplantation conditions to enhance the efficacy of mesenchymal stem cells. Int J Biol Sci. 2015;11:324-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 47]  [Cited by in RCA: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
114.  Laurila JP, Laatikainen L, Castellone MD, Trivedi P, Heikkila J, Hinkkanen A, Hematti P, Laukkanen MO. Human embryonic stem cell-derived mesenchymal stromal cell transplantation in a rat hind limb injury model. Cytotherapy. 2009;11:726-737.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 53]  [Cited by in RCA: 56]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
115.  Tang YL, Zhao Q, Qin X, Shen L, Cheng L, Ge J, Phillips MI. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann Thorac Surg. 2005;80:229-36; discussion 236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 320]  [Cited by in RCA: 315]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
116.  Meyerholz DK, Burrough ER, Kirchhof N, Anderson DJ, Helke KL. Swine models in translational research and medicine. Vet Pathol. 2024;61:512-523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
117.  Sridharan D, Pracha N, Rana SJ, Ahmed S, Dewani AJ, Alvi SB, Mergaye M, Ahmed U, Khan M. Preclinical Large Animal Porcine Models for Cardiac Regeneration and Its Clinical Translation: Role of hiPSC-Derived Cardiomyocytes. Cells. 2023;12:1090.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
118.  Shafiee A, Seyedjafari E, Soleimani M, Ahmadbeigi N, Dinarvand P, Ghaemi N. A comparison between osteogenic differentiation of human unrestricted somatic stem cells and mesenchymal stem cells from bone marrow and adipose tissue. Biotechnol Lett. 2011;33:1257-1264.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 116]  [Cited by in RCA: 129]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
119.  Kotani T, Saito T, Suzuka T, Matsuda S. Adipose-derived mesenchymal stem cell therapy for connective tissue diseases and complications. Inflamm Regen. 2024;44:35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
120.  Kim WS, Park BS, Sung JH. The wound-healing and antioxidant effects of adipose-derived stem cells. Expert Opin Biol Ther. 2009;9:879-887.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 186]  [Cited by in RCA: 222]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
121.  Drobiova H, Sindhu S, Ahmad R, Haddad D, Al-Mulla F, Al Madhoun A. Wharton's jelly mesenchymal stem cells: a concise review of their secretome and prospective clinical applications. Front Cell Dev Biol. 2023;11:1211217.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
122.  Kamal MM, Kassem DH. Therapeutic Potential of Wharton's Jelly Mesenchymal Stem Cells for Diabetes: Achievements and Challenges. Front Cell Dev Biol. 2020;8:16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 55]  [Cited by in RCA: 53]  [Article Influence: 8.8]  [Reference Citation Analysis (1)]
123.  Mebarki M, Moine-Picard C, Enjaume-Rauch R, Laurent-Puig A, Suissa A, Feyants V, Larghero J, Cras A. Pooling umbilical cord-mesenchymal stromal cells derived from selected multiple donors reduces donor-dependent variability and improves their immunomodulatory properties. Stem Cell Res Ther. 2025;16:252.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
124.  Xie N, Li Z, Adesanya TM, Guo W, Liu Y, Fu M, Kilic A, Tan T, Zhu H, Xie X. Transplantation of placenta-derived mesenchymal stem cells enhances angiogenesis after ischemic limb injury in mice. J Cell Mol Med. 2016;20:29-37.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 39]  [Cited by in RCA: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
125.  Luo L, He Y, Wang X, Key B, Lee BH, Li H, Ye Q. Potential Roles of Dental Pulp Stem Cells in Neural Regeneration and Repair. Stem Cells Int. 2018;2018:1731289.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 65]  [Cited by in RCA: 104]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
126.  Jeyaraman M, Muthu S, Jeyaraman N, Ranjan R, Jha SK, Mishra P. Synovium Derived Mesenchymal Stromal Cells (Sy-MSCs): A Promising Therapeutic Paradigm in the Management of Knee Osteoarthritis. Indian J Orthop. 2022;56:1-15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
127.  Rehman A, Nigam A, Laino L, Russo D, Todisco C, Esposito G, Svolacchia F, Giuzio F, Desiderio V, Ferraro G. Mesenchymal Stem Cells in Soft Tissue Regenerative Medicine: A Comprehensive Review. Medicina (Kaunas). 2023;59:1449.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
128.  Suchanecka M, Grzelak J, Farzaneh M, Azizidoost S, Dari MAG, Józkowiak M, Data K, Domagała D, Niebora J, Kotrych K, Czerny B, Kamiński A, Torlińska-Walkowiak N, Bieniek A, Szepietowski J, Piotrowska-Kempisty H, Dzięgiel P, Mozdziak P, Kempisty B. Adipose derived stem cells - Sources, differentiation capacity and a new target for reconstructive and regenerative medicine. Biomed Pharmacother. 2025;186:118036.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
129.  Xie Q, Liu R, Jiang J, Peng J, Yang C, Zhang W, Wang S, Song J. What is the impact of human umbilical cord mesenchymal stem cell transplantation on clinical treatment? Stem Cell Res Ther. 2020;11:519.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 177]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
130.  Cao M, Ou Z, Sheng R, Wang Q, Chen X, Zhang C, Dai G, Wang H, Li J, Zhang X, Gao Y, Shi L, Rui Y. Efficacy and safety of mesenchymal stem cells in knee osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res Ther. 2025;16:122.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
131.  To K, Zhang B, Romain K, Mak C, Khan W. Synovium-Derived Mesenchymal Stem Cell Transplantation in Cartilage Regeneration: A PRISMA Review of in vivo Studies. Front Bioeng Biotechnol. 2019;7:314.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 44]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
132.  Govindasamy V, Abdullah AN, Ronald VS, Musa S, Ab Aziz ZA, Zain RB, Totey S, Bhonde RR, Abu Kasim NH. Inherent differential propensity of dental pulp stem cells derived from human deciduous and permanent teeth. J Endod. 2010;36:1504-1515.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 108]  [Cited by in RCA: 117]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
133.  Liu Y, Xiong W, Li J, Feng H, Jing S, Liu Y, Zhou H, Li D, Fu D, Xu C, He Y, Ye Q. Application of dental pulp stem cells for bone regeneration. Front Med (Lausanne). 2024;11:1339573.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
134.  Liu Y, Liu Y, Hu J, Han J, Song L, Liu X, Han N, Xia X, He J, Meng H, Wan M, Wang H, Liu X, Gao Z, Wang X, Wu C, Wang S. Impact of allogeneic dental pulp stem cell injection on tissue regeneration in periodontitis: a multicenter randomized clinical trial. Signal Transduct Target Ther. 2025;10:239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
135.  Lee HJ, Lee WJ, Hwang SC, Choe Y, Kim S, Bok E, Lee S, Kim SJ, Kim HO, Ock SA, Noh HS, Rho GJ, Lee SI, Lee SL. Chronic inflammation-induced senescence impairs immunomodulatory properties of synovial fluid mesenchymal stem cells in rheumatoid arthritis. Stem Cell Res Ther. 2021;12:502.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
136.  Kulesza A, Paczek L, Burdzinska A. The Role of COX-2 and PGE2 in the Regulation of Immunomodulation and Other Functions of Mesenchymal Stromal Cells. Biomedicines. 2023;11:445.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 78]  [Reference Citation Analysis (0)]
137.  Lee SS, Vũ TT, Weiss AS, Yeo GC. Stress-induced senescence in mesenchymal stem cells: Triggers, hallmarks, and current rejuvenation approaches. Eur J Cell Biol. 2023;102:151331.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
138.  Vono R, Jover Garcia E, Spinetti G, Madeddu P. Oxidative Stress in Mesenchymal Stem Cell Senescence: Regulation by Coding and Noncoding RNAs. Antioxid Redox Signal. 2018;29:864-879.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 57]  [Cited by in RCA: 81]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
139.  Zhao W, Li X, Liu X, Zhang N, Wen X. Effects of substrate stiffness on adipogenic and osteogenic differentiation of human mesenchymal stem cells. Mater Sci Eng C Mater Biol Appl. 2014;40:316-323.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 83]  [Cited by in RCA: 87]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
140.  Xie J, Zhang D, Zhou C, Yuan Q, Ye L, Zhou X. Substrate elasticity regulates adipose-derived stromal cell differentiation towards osteogenesis and adipogenesis through β-catenin transduction. Acta Biomater. 2018;79:83-95.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 61]  [Cited by in RCA: 81]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
141.  Raman N, Imran SAM, Ahmad Amin Noordin KB, Zaman WSWK, Nordin F. Mechanotransduction in Mesenchymal Stem Cells (MSCs) Differentiation: A Review. Int J Mol Sci. 2022;23:4580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 31]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
142.  Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, Laine GA, Cox CS Jr. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev. 2009;18:683-692.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 794]  [Cited by in RCA: 953]  [Article Influence: 56.1]  [Reference Citation Analysis (0)]
143.  Zhao Y, Wang M, Liang F, Li J. Recent strategies for enhancing the therapeutic efficacy of stem cells in wound healing. Stem Cell Res Ther. 2021;12:588.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
144.  Rustad KC, Wong VW, Sorkin M, Glotzbach JP, Major MR, Rajadas J, Longaker MT, Gurtner GC. Enhancement of mesenchymal stem cell angiogenic capacity and stemness by a biomimetic hydrogel scaffold. Biomaterials. 2012;33:80-90.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 271]  [Cited by in RCA: 292]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
145.  Wong VW, Rustad KC, Glotzbach JP, Sorkin M, Inayathullah M, Major MR, Longaker MT, Rajadas J, Gurtner GC. Pullulan hydrogels improve mesenchymal stem cell delivery into high-oxidative-stress wounds. Macromol Biosci. 2011;11:1458-1466.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 43]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
146.  Marquardt LM, Heilshorn SC. Design of Injectable Materials to Improve Stem Cell Transplantation. Curr Stem Cell Rep. 2016;2:207-220.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 129]  [Cited by in RCA: 130]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
147.  Mesa Bedoya LE, Camacho Barbosa JC, López Quiceno L, Barrios Arroyave F, Halpert K, España Peña JA, Salazar Uribe JC. The safety profile of mesenchymal stem cell therapy administered through intrathecal injections for treating neurological disorders: a systematic review and meta-analysis of randomised controlled trials. Stem Cell Res Ther. 2024;15:146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
148.  Wang Y, Yi H, Song Y. The safety of MSC therapy over the past 15 years: a meta-analysis. Stem Cell Res Ther. 2021;12:545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 38]  [Cited by in RCA: 130]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
149.  Zhang Y, Wang C, Li JJ. Revisiting the role of mesenchymal stromal cells in cancer initiation, metastasis and immunosuppression. Exp Hematol Oncol. 2024;13:64.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
150.  Parashurama N, Ahn BC, Ziv K, Ito K, Paulmurugan R, Willmann JK, Chung J, Ikeno F, Swanson JC, Merk DR, Lyons JK, Yerushalmi D, Teramoto T, Kosuge H, Dao CN, Ray P, Patel M, Chang YF, Mahmoudi M, Cohen JE, Goldstone AB, Habte F, Bhaumik S, Yaghoubi S, Robbins RC, Dash R, Yang PC, Brinton TJ, Yock PG, McConnell MV, Gambhir SS. Multimodality Molecular Imaging of Cardiac Cell Transplantation: Part II. In Vivo Imaging of Bone Marrow Stromal Cells in Swine with PET/CT and MR Imaging. Radiology. 2016;280:826-836.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
151.  Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107:2290-2293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 569]  [Cited by in RCA: 509]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
152.  Fu Y, Mease R, Chen Y, Wang G, Kedziorek D, Solaiyappan M, Kraitchman D. PET-MRI tracking of imaging-visible microencapsulated stem cells in immunocompetent rabbits. J Cardiovasc Magn Reson. 2013;15:M1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
153.  Serganova I, Blasberg RG. Molecular Imaging with Reporter Genes: Has Its Promise Been Delivered? J Nucl Med. 2019;60:1665-1681.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 47]  [Cited by in RCA: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
154.  Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther. 2022;13:146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
155.  Hertel FC, Silva ASD, Sabino AP, Valente FL, Reis ECC. Preconditioning Methods to Improve Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Bone Regeneration-A Systematic Review. Biology (Basel). 2022;11:733.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
156.  Damasceno PKF, de Santana TA, Santos GC, Orge ID, Silva DN, Albuquerque JF, Golinelli G, Grisendi G, Pinelli M, Ribeiro Dos Santos R, Dominici M, Soares MBP. Genetic Engineering as a Strategy to Improve the Therapeutic Efficacy of Mesenchymal Stem/Stromal Cells in Regenerative Medicine. Front Cell Dev Biol. 2020;8:737.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 81]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
157.  Haque N, Kasim NHA, Kassim NLA, Rahman MT. Autologous serum supplement favours in vitro regenerative paracrine factors synthesis. Cell Prolif. 2017;50:e12354.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
158.  Haque N, Fareez IM, Fong LF, Mandal C, Abu Kasim NH, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells. 2020;12:938-951.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (2)]
159.  Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res. 2025;70:103-124.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 61]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
160.  Cheng HY, Anggelia MR, Liu SC, Lin CF, Lin CH. Enhancing Immunomodulatory Function of Mesenchymal Stromal Cells by Hydrogel Encapsulation. Cells. 2024;13:210.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
161.  Farasati Far B, Safaei M, Nahavandi R, Gholami A, Naimi-Jamal MR, Tamang S, Ahn JE, Ramezani Farani M, Huh YS. Hydrogel Encapsulation Techniques and Its Clinical Applications in Drug Delivery and Regenerative Medicine: A Systematic Review. ACS Omega. 2024;9:29139-29158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 30]  [Reference Citation Analysis (0)]
162.  Li Q, Wang Z. Influence of mesenchymal stem cells with endothelial progenitor cells in co-culture on osteogenesis and angiogenesis: an in vitro study. Arch Med Res. 2013;44:504-513.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 43]  [Cited by in RCA: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
163.  Xu C, Liu H, He Y, Li Y, He X. Endothelial progenitor cells promote osteogenic differentiation in co-cultured with mesenchymal stem cells via the MAPK-dependent pathway. Stem Cell Res Ther. 2020;11:537.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 36]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
164.  Williams T, Salmanian G, Burns M, Maldonado V, Smith E, Porter RM, Song YH, Samsonraj RM. Versatility of mesenchymal stem cell-derived extracellular vesicles in tissue repair and regenerative applications. Biochimie. 2023;207:33-48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 33]  [Cited by in RCA: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
165.  Manzoor T, Saleem A, Farooq N, Dar LA, Nazir J, Saleem S, Ismail S, Gugjoo MB, Shiekh PA, Ahmad SM. Extracellular vesicles derived from mesenchymal stem cells - a novel therapeutic tool in infectious diseases. Inflamm Regen. 2023;43:17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 44]  [Reference Citation Analysis (0)]
166.  Haque N, Widera D, Govindasamy V, Soesilawati P, Abu Kasim NH. Extracellular Vesicles from Stem and Progenitor Cells for Cell-Free Regenerative Therapy. Curr Mol Med. 2022;22:120-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
167.  Haque N, Abu Kasim NH. Specific Bioactive Factors and Application of Bioinformatics Tools for the Successful Use of Secretome and Extracellular Vesicles as Cell-Free Therapeutics. Curr Mol Med. 2022;22:83-84.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
168.  Rather HA, Almousa S, Craft S, Deep G. Therapeutic efficacy and promise of stem cell-derived extracellular vesicles in Alzheimer's disease and other aging-related disorders. Ageing Res Rev. 2023;92:102088.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
169.  Laura Francés J, Pagiatakis C, Di Mauro V, Climent M. Therapeutic Potential of EVs: Targeting Cardiovascular Diseases. Biomedicines. 2023;11:1907.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
170.  Neziri S, Köseoğlu AE, Deniz Köseoğlu G, Özgültekin B, Özgentürk NÖ. Animal models in neuroscience with alternative approaches: Evolutionary, biomedical, and ethical perspectives. Animal Model Exp Med. 2024;7:868-880.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
171.  Marshall LJ, Bailey J, Cassotta M, Herrmann K, Pistollato F. Poor Translatability of Biomedical Research Using Animals - A Narrative Review. Altern Lab Anim. 2023;51:102-135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 77]  [Reference Citation Analysis (0)]
172.  Wysoczynski M, Khan A, Bolli R. New Paradigms in Cell Therapy: Repeated Dosing, Intravenous Delivery, Immunomodulatory Actions, and New Cell Types. Circ Res. 2018;123:138-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 108]  [Cited by in RCA: 129]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
173.  Hetta HF, Elsaghir A, Sijercic VC, Ahmed AK, Gad SA, Zeleke MS, Alanazi FE, Ramadan YN. Clinical Progress in Mesenchymal Stem Cell Therapy: A Focus on Rheumatic Diseases. Immun Inflamm Dis. 2025;13:e70189.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
174.  Kim N, Cho SG. New strategies for overcoming limitations of mesenchymal stem cell-based immune modulation. Int J Stem Cells. 2015;8:54-68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 91]  [Cited by in RCA: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
175.  Kizilay Mancini Ö, Lora M, Shum-Tim D, Nadeau S, Rodier F, Colmegna I. A Proinflammatory Secretome Mediates the Impaired Immunopotency of Human Mesenchymal Stromal Cells in Elderly Patients with Atherosclerosis. Stem Cells Transl Med. 2017;6:1132-1140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 30]  [Cited by in RCA: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
176.  Zhang Y, Ravikumar M, Ling L, Nurcombe V, Cool SM. Age-Related Changes in the Inflammatory Status of Human Mesenchymal Stem Cells: Implications for Cell Therapy. Stem Cell Reports. 2021;16:694-707.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
177.  Todtenhaupt P, Franken LA, Groene SG, van Hoolwerff M, van der Meeren LE, van Klink JMM, Roest AAW, de Bruin C, Ramos YFM, Haak MC, Lopriore E, Heijmans BT, van Pel M. A robust and standardized method to isolate and expand mesenchymal stromal cells from human umbilical cord. Cytotherapy. 2023;25:1057-1068.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
178.  Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products-a review. Front Cell Dev Biol. 2024;12:1400347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 57]  [Reference Citation Analysis (0)]
179.  Neogi T, Colloca L. Placebo effects in osteoarthritis: implications for treatment and drug development. Nat Rev Rheumatol. 2023;19:613-626.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
180.  Wei ST, Sun YH, Zong SH, Xiang YB. Serum Levels of IL-6 and TNF-α May Correlate with Activity and Severity of Rheumatoid Arthritis. Med Sci Monit. 2015;21:4030-4038.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 56]  [Cited by in RCA: 87]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
181.  El-Jawhari JJ, El-Sherbiny Y, McGonagle D, Jones E. Multipotent Mesenchymal Stromal Cells in Rheumatoid Arthritis and Systemic Lupus Erythematosus; From a Leading Role in Pathogenesis to Potential Therapeutic Saviors? Front Immunol. 2021;12:643170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
182.  Galipeau J, Sensébé L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell. 2018;22:824-833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1009]  [Cited by in RCA: 1269]  [Article Influence: 158.6]  [Reference Citation Analysis (0)]
183.  Sensebé L, Bourin P, Tarte K. Good manufacturing practices production of mesenchymal stem/stromal cells. Hum Gene Ther. 2011;22:19-26.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 171]  [Cited by in RCA: 160]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
184.  Heathman TR, Nienow AW, McCall MJ, Coopman K, Kara B, Hewitt CJ. The translation of cell-based therapies: clinical landscape and manufacturing challenges. Regen Med. 2015;10:49-64.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 207]  [Cited by in RCA: 212]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
185.  Jossen V, van den Bos C, Eibl R, Eibl D. Manufacturing human mesenchymal stem cells at clinical scale: process and regulatory challenges. Appl Microbiol Biotechnol. 2018;102:3981-3994.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 104]  [Cited by in RCA: 144]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
186.  Cottle C, Porter AP, Lipat A, Turner-Lyles C, Nguyen J, Moll G, Chinnadurai R. Impact of Cryopreservation and Freeze-Thawing on Therapeutic Properties of Mesenchymal Stromal/Stem Cells and Other Common Cellular Therapeutics. Curr Stem Cell Rep. 2022;8:72-92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 89]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
187.  Fernández-Santos ME, Garcia-Arranz M, Andreu EJ, García-Hernández AM, López-Parra M, Villarón E, Sepúlveda P, Fernández-Avilés F, García-Olmo D, Prosper F, Sánchez-Guijo F, Moraleda JM, Zapata AG. Optimization of Mesenchymal Stromal Cell (MSC) Manufacturing Processes for a Better Therapeutic Outcome. Front Immunol. 2022;13:918565.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 83]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
188.  Harrison RP, Ruck S, Medcalf N, Rafiq QA. Decentralized manufacturing of cell and gene therapies: Overcoming challenges and identifying opportunities. Cytotherapy. 2017;19:1140-1151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
189.  Shin H, Kim E. Analysis of GMP for marketing authorization of ATMPs: comparison in the US, the EU, Japan and South Korea. Regen Med. 2022;17:283-297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
190.  Dahiya A, Singh K, Ashish A, Nipun, Bhadyaria A, Thakur S, Kumar M, Das Gupta G, Das Kurmi B, Pal RR. Global harmonization in advanced therapeutics: balancing innovation, safety, and access. Per Med. 2025;22:181-191.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
191.  Galipeau J, Krampera M, Barrett J, Dazzi F, Deans RJ, DeBruijn J, Dominici M, Fibbe WE, Gee AP, Gimble JM, Hematti P, Koh MB, LeBlanc K, Martin I, McNiece IK, Mendicino M, Oh S, Ortiz L, Phinney DG, Planat V, Shi Y, Stroncek DF, Viswanathan S, Weiss DJ, Sensebe L. International Society for Cellular Therapy perspective on immune functional assays for mesenchymal stromal cells as potency release criterion for advanced phase clinical trials. Cytotherapy. 2016;18:151-159.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 303]  [Cited by in RCA: 384]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
192.  Sensebé L, Gadelorge M, Fleury-Cappellesso S. Production of mesenchymal stromal/stem cells according to good manufacturing practices: a review. Stem Cell Res Ther. 2013;4:66.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 115]  [Cited by in RCA: 125]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
193.  Phinney DG, Galipeau J; MSC Committee of the International Society of Cell and Gene Therapy. Manufacturing mesenchymal stromal cells for clinical applications: A survey of Good Manufacturing Practices at U.S. academic centers. Cytotherapy. 2019;21:782-792.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 41]  [Cited by in RCA: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
194.  Turner L, Knoepfler P. Selling Stem Cells in the USA: Assessing the Direct-to-Consumer Industry. Cell Stem Cell. 2016;19:154-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 230]  [Cited by in RCA: 250]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
195.  Lukomska B, Stanaszek L, Zuba-Surma E, Legosz P, Sarzynska S, Drela K. Challenges and Controversies in Human Mesenchymal Stem Cell Therapy. Stem Cells Int. 2019;2019:9628536.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 220]  [Cited by in RCA: 379]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
196.  Sipp D, Robey PG, Turner L. Clear up this stem-cell mess. Nature. 2018;561:455-457.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 163]  [Cited by in RCA: 201]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
197.  Mendt M, Rezvani K, Shpall E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant. 2019;54:789-792.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 193]  [Cited by in RCA: 356]  [Article Influence: 59.3]  [Reference Citation Analysis (0)]
198.  Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9:63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 315]  [Cited by in RCA: 657]  [Article Influence: 82.1]  [Reference Citation Analysis (0)]
199.  Phinney DG, Pittenger MF. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells. 2017;35:851-858.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 831]  [Cited by in RCA: 1294]  [Article Influence: 143.8]  [Reference Citation Analysis (0)]
200.  Kou M, Huang L, Yang J, Chiang Z, Chen S, Liu J, Guo L, Zhang X, Zhou X, Xu X, Yan X, Wang Y, Zhang J, Xu A, Tse HF, Lian Q. Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: a next generation therapeutic tool? Cell Death Dis. 2022;13:580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 296]  [Cited by in RCA: 360]  [Article Influence: 90.0]  [Reference Citation Analysis (6)]
201.  Tsiapalis D, O'Driscoll L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells. 2020;9:991.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 177]  [Cited by in RCA: 229]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
202.  Park KS, Bandeira E, Shelke GV, Lässer C, Lötvall J. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther. 2019;10:288.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 116]  [Cited by in RCA: 191]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
203.  Mooney R, Majid AA, Mota D, He A, Aramburo S, Flores L, Covello-Batalla J, Machado D, Gonzaga J, Aboody KS. Bcl-2 Overexpression Improves Survival and Efficacy of Neural Stem Cell-Mediated Enzyme Prodrug Therapy. Stem Cells Int. 2018;2018:7047496.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
204.  Federico A, Cardaioli E, Da Pozzo P, Formichi P, Gallus GN, Radi E. Mitochondria, oxidative stress and neurodegeneration. J Neurol Sci. 2012;322:254-262.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 505]  [Cited by in RCA: 572]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]