Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Apr 26, 2024; 16(4): 324-333
Published online Apr 26, 2024. doi: 10.4252/wjsc.v16.i4.324
Adipose-derived regenerative therapies for the treatment of knee osteoarthritis
Ilias E Epanomeritakis, Division of Trauma and Orthopaedic Surgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
Wasim S Khan, Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
ORCID number: Ilias E Epanomeritakis (0000-0002-4810-4197); Wasim S Khan (0000-0003-3106-5424).
Author contributions: Epanomeritakis IE and Khan WS designed the overall concept and outline of the manuscript, contributed to the review of literature, and contributed to the writing and editing of the manuscript.
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: Wasim S Khan, FRCS, MBChB, MSc, PhD, Associate Professor, Surgeon, Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Hills Road, Cambridge CB2 0QQ, United Kingdom. wk280@cam.ac.uk
Received: January 17, 2024
Peer-review started: January 17, 2024
First decision: February 3, 2024
Revised: February 15, 2024
Accepted: March 1, 2024
Article in press: March 1, 2024
Published online: April 26, 2024

Abstract

Knee osteoarthritis is a degenerative condition with a significant disease burden and no disease-modifying therapy. Definitive treatment ultimately requires joint replacement. Therapies capable of regenerating cartilage could significantly reduce financial and clinical costs. The regenerative potential of mesenchymal stromal cells (MSCs) has been extensively studied in the context of knee osteoarthritis. This has yielded promising results in human studies, and is likely a product of immunomodulatory and chondroprotective biomolecules produced by MSCs in response to inflammation. Adipose-derived MSCs (ASCs) are becoming increasingly popular owing to their relative ease of isolation and high proliferative capacity. Stromal vascular fraction (SVF) and micro-fragmented adipose tissue (MFAT) are produced by the enzymatic and mechanical disruption of adipose tissue, respectively. This avoids expansion of isolated ASCs ex vivo and their composition of heterogeneous cell populations, including immune cells, may potentiate the reparative function of ASCs. In this editorial, we comment on a multicenter randomized trial regarding the efficacy of MFAT in treating knee osteoarthritis. We discuss the study’s findings in the context of emerging evidence regarding adipose-derived regenerative therapies. An underlying mechanism of action of ASCs is proposed while drawing important distinctions between the properties of isolated ASCs, SVF, and MFAT.

Key Words: Knee osteoarthritis, Mesenchymal stromal cells, Adipose tissue, Stromal vascular fraction, Micro-fragmented adipose tissue, Regeneration

Core Tip: Adipose tissue products are becoming increasingly popular regenerative therapies for treating knee osteoarthritis. Encouraging results have been demonstrated in numerous observational studies and randomized trials. However, it is important to distinguish among isolated adipose-derived mesenchymal stromal cells (ASCs), stromal vascular fraction (SVF), and micro-fragmented adipose tissue (MFAT) to avoid study heterogeneity and improve the quality of evidence regarding these therapies. Different modes of preparation, cell composition, and physical properties are likely to influence the regenerative function of ASCs. To elucidate which adipose-derived therapy is superior for cartilage regeneration, randomized trials are needed to compare ASCs, SVF, and MFAT as distinct therapies.



INTRODUCTION

Knee osteoarthritis is a degenerative joint disease that causes significant morbidity globally[1]. Currently, no disease-modifying therapies are available for this condition. Nonoperative measures, including exercise, analgesics, and intraarticular corticosteroids, provide temporary symptomatic relief[2,3]. Arthroscopic procedures such as debridement of torn menisci to alleviate mechanical symptoms provide little or no clinically important benefit[4]. The poor intrinsic healing ability of hyaline cartilage means that definitive treatment of the condition ultimately requires joint replacement[5].

Therapies capable of regenerating knee cartilage and avoiding knee replacement would lead to a significant reduction in financial and clinical costs[6]. Therapies aimed at cartilage regeneration include intraarticular platelet-rich plasma, mosaicplasty, and microfracture. These therapies may provide symptomatic relief and lead to radiographic improvements[3,7-9]. However, evidence for their use is inconsistent, and the lifespan of the repair is limited.

The search for regenerative therapies has led to the increasing use of tissue engineering techniques, including autologous chondrocyte implantation (ACI) and mesenchymal stromal cells (MSCs)[5]. Embedding cells in scaffolds allows targeted cell delivery to injury sites and improves cell retention and survival within the graft[10,11]. ACI and MSCs have proven promising in clinical trials for the treatment of knee osteoarthritis, as evidenced by patient-reported outcomes and radiographic evidence[12,13]. However, obstacles to the widespread adoption of cell therapies include the potential for donor site morbidity and the need to perform two separate procedures[14], which can reduce the acceptability of the therapy to patients. In addition, the isolation and expansion of cells in culture is expensive and can be met with regulatory restrictions on cell differentiation ex vivo[15].

MSCs have greater proliferative capacity than chondrocytes, are more widely available, and more often promote the formation of hyaline rather than fibrocartilage[10,15,16]. Their regenerative functions are likely due to the secretion of a combination of chondroprotective, pro-reparative, and immunomodulatory cytokines[15]. MSCs can be isolated from numerous sites, including the bone marrow, adipose tissue, umbilical cord, and synovium, but it remains unclear which MSC source is optimal for cartilage regeneration[17]. While bone marrow has traditionally been used as an abundant source of MSCs, adipose-derived MSCs (ASCs) have gained increasing popularity. Adipose tissue is easier to access and has less potential for donor site morbidity, and ASCs have a greater proliferation capacity than bone marrow-derived MSCs (BMSCs) in vitro[17]. Their ability to improve symptoms in patients with knee osteoarthritis has been proven superior to that of BMSCs[13].

Therapies that avoid the need for cell expansion ex vivo can facilitate a ‘one-step’ treatment and undergo less stringent regulatory processes[15]. In addition to isolated culture-expanded ASCs, preparations of adipose tissue that retain heterogeneous cell populations and do not involve cell culture have been tested for cartilage regeneration[18,19]. Stromal vascular fraction (SVF) is produced by the enzymatic digestion of fat tissue and contains a heterogeneous mix of ASCs, pericytes, immune cells, fibroblasts, erythrocytes, and endothelial cells[20]. Micro-fragmented adipose tissue (MFAT) is another relatively novel technique that has proven safe and effective in treating knee osteoarthritis[21]. MFAT involves mechanical, rather than enzymatic, disruption of adipose tissue and contains a similarly heterogeneous cell population[22,23]. Unlike SVF, the lack of enzymatic digestion maintains an intact extracellular matrix (ECM) that is more akin to that found in vivo.

In a multicenter, randomized trial, Wu et al[24] investigated the efficacy of MFAT performed in conjunction with arthroscopic surgery in treating patients with Kellgren-Lawrence grade 2-3 osteoarthritis of the knee[25]. Compared with an intraarticular hyaluronic acid (HA) control, they demonstrated the efficacy of MFAT at 24 months of follow-up, as evidenced by patient-reported outcome measures (PROMs). This editorial reviews the trial by Wu et al[24] in the context of current evidence regarding adipose-derived regenerative therapies for knee osteoarthritis. Comparisons were made between ASC, SVF, and MFAT treatments to elucidate important distinctions and how the underlying mechanisms of action of ASCs may be affected by their differing compositions.

PATHOPHYSIOLOGY OF OSTEOARTHRITIS

Understanding the mechanisms governing osteoarthritis allows an understanding of the mechanisms of action of regenerative treatments. Osteoarthritis has a multifactorial etiology involving mechanical insults that drive inflammation and remodeling of the whole joint, including the cartilage, synovium, fat, and bone[26]. Under physiological conditions, chondrocytes generate a dense ECM composed of water, type II collagen, glycosaminoglycans (GAGs), and proteoglycans, which confer the shock-absorbing and gliding properties of hyaline cartilage[27]. The ECM plays a reciprocal role in maintaining chondrocyte homeostasis and phenotypic stability[28]. Therefore, loss of cartilage integrity has deleterious effects on chondrocytes, which in turn fail to preserve healthy ECM, leading to progressive degeneration of cartilage tissue.

Loss of ECM integrity following a mechanical insult leads to the production of pro-inflammatory cytokines by chondrocytes and the synovium, including interleukin (IL)-1β and tumor necrosis factor-alpha (TNF-α)[26], as well as the mechanosensitive induction of protease and chemokine expression[28,29]. Activation of nuclear factor-kappaB (NF-κB) signaling pathways by inflammatory cytokines upregulates the expression of cartilage-degrading enzymes by chondrocytes, including matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin-1 motifs[26].

Activation of NF-κB pathways by IL-1β has also been shown to increase the production of chemokines by chondrocytes, including chemokine (C-C motif) ligand (CCL)-2[30], which play an important role in osteoarthritis pathogenesis[31]. CCL-2 is considered a key factor in initiating and propagating chronic inflammation in osteoarthritis[32-34], and its deletion is considered chondroprotective. It is responsible for recruiting pro-inflammatory, M1, macrophages to the site of injury, and may act in an autocrine fashion leading to upregulated expression of CCL-2 itself, chondrocyte apoptosis, and greater MMP expression via the MAPK pathway[35], another principal pathway involved in osteoarthritis pathogenesis alongside NF-κB signaling[26].

Additional contributors to disease progression include the dysregulated expression of molecules involved in the homeostasis of healthy cartilage. These include transforming growth factor (TGF)-β and NOTCH[36,37]. Sustained high levels of TGF-β expression in osteoarthritis contribute to chondrocyte hypertrophy, bone remodeling, and osteophyte formation[36]. Pathological NOTCH signaling acts via the NF-κB and MAPK pathways to upregulate protease and chemokine production[37]. Both molecules are thought to activate pathways that converge on the upregulation of Runx2, a transcription factor that modulates MMP expression in chondrocytes[38]. Similarly, alterations in the physiological expression profiles of microRNAs (miRNAs), which are responsible for post-transcriptional alterations in gene expression, have been implicated in disease process[39,40]. The reduced expression of miRNAs responsible for promoting chondrocyte differentiation, inhibiting inflammation, and downregulating the expression of proteinases is coupled with an increase in the expression of pro-inflammatory and pro-apoptotic miRNAs.

The proximity of the articular cartilage to the underlying subchondral bone and their reciprocal interactions are important for understanding the pathogenesis of osteoarthritis[41]. The osteochondral unit is composed of articular hyaline cartilage and subchondral bone, separated by a zone of calcified cartilage, the tidemark[26]. Subchondral bone and cartilage interact both biomechanically and biochemically[42]. Altered transmission of loads between the cartilage and the underlying bone leads to pathological bone remodeling, which is associated with thickening of the cortical subchondral bone and thinning of the deeper cancellous bone, changes that precede cartilage degradation[26]. Increased RANKL expression in osteocytes in areas of reduced mechanical loading promotes osteoclastogenesis and bone resorption during early osteoarthritis[43]. Greater osteoclastic activity is associated with greater TGF-β1-induced bone formation by osteoblasts, which act reciprocally on osteoclasts to create a positive feedback loop that drives bone remodeling[43]. The increased stiffness of the subchondral bone in osteoarthritis exerts a greater load on the overlying cartilage, contributing to secondary cartilage damage[42].

As a result of remodeling, the tidemark is disrupted and calcification advances, contributing to cartilage thinning[26]. This effect is exacerbated by pathological angiogenesis[26,44]. Invasion of the overlying cartilage by new blood vessels is facilitated by the reduced integrity of the tidemark and cartilage ECM[45]. Biochemical mediators include vascular endothelial growth factor secreted by chondrocytes, in contrast to anti-angiogenic factors secreted under physiological conditions[45]. Osteoclast precursors within the osteoarthritic subchondral bone also drive this process by secreting excessive levels of platelet-derived growth factor-BB, which stimulates angiogenesis[44].

Although hyaline cartilage is aneural, pain is a hallmark of osteoarthritis. The bone is richly innervated by sensory nerves running alongside the vasculature[46]. This may explain why cystic bone lesions are independently associated with pain severity[47]. Furthermore, vessels extending into the overlying cartilage are accompanied by neuronal ingrowth, which contributes to pain associated with the disease[47].

Also relevant to the disease process is the biochemical crosstalk between cells within the subchondral bone and the overlying cartilage[41,42,45]. Although disruption of the tidemark weakens the barrier between the bone and cartilage, calcified cartilage is permeable to small molecules, even in healthy knee joints[42]. Therefore, it is likely that soluble factors crossing from the bone to articular cartilage and vice versa induce pathological changes in osteoarthritis. It has been shown that the co-culture of chondrocytes with osteoblasts from sclerotic bone induces a hypertrophic chondrocyte phenotype, expressing reduced levels of proteoglycans and elevated levels of MMPs[48]. TGF-β1 is one chemical mediator which has been implicated in the crosstalk between subchondral bone and cartilage, resulting in an increased chondrocyte apoptotic rate[43]. Similarly, the transmission of inflammatory cytokines produced by chondrocytes in osteoarthritis act to increases osteoclastogenesis and consequent bone resorption[43].

ASCS: MECHANISM OF ACTION

The International Society for Cellular Therapy outlines a three-part definition of MSC characterization[49]. MSCs display plastic adherence and chondrogenic, osteogenic, and adipogenic differentiation in vitro and are characterized by the expression of specific cluster of differentiation (CD) markers, including CD73, CD90, and CD105, as well as the absence of CD14, CD19, CD45, and HLA-DR. We identified an additional panel of cell surface markers that should be tested for accurate characterization of ASCs, allowing for consistent identification between studies[50].

Although initially termed mesenchymal “stem” cells, this is now recognized as a misnomer[51]. Despite the multipotency of MSCs in vitro, the concept that their regenerative function of MSCs is reliant on their differentiation into resident tissues in vivo has been disregarded. MSCs are present in all vascularized tissues and arise from perivascular pericytes[51]. Rather than regenerating tissues by direct differentiation, the regenerative capacity of MSCs in vivo is thought to rely on their ability to migrate to sites of injury and secrete bioactive molecules with immunomodulatory and regenerative functions in response to local cues. One such molecule is prostaglandin-E2 (PGE2), which is produced by MSCs in response to inflammatory cytokines, such as TNF-α[52]. PGE2 converts pro-inflammatory M1 macrophages into anti-inflammatory M2 macrophages. MSCs also induce a transition from TH1 to TH2 lymphocytes, increase IL-10 and IL-4 levels, and decrease interferon gamma production, which have been implicated in their ability to promote cartilage repair[52].

The low reported survival rate of MSCs implanted into knee joints[17] and their tendency to migrate to the synovium and menisci, not solely to the damaged cartilage itself[53], further strengthens the doubt over their role in direct differentiation into neocartilage. Instead, regeneration is likely promoted by ASCs via immunomodulatory mechanisms and secretion of chondroprotective biomolecules[52,54,55].

We previously investigated the effect of co-culturing infrapatellar fat pad-derived ASCs and chondrocytes in vitro to investigate changes in chondrogenic differentiation in comparison with ASC monocultures[56]. Although there are doubts about the ability of MSCs to differentiate into resident tissues in vivo[51], our findings suggest that chondrocyte-ASC crosstalk may be involved in the pro-reparative function of ASCs. We found that a high relative ratio of chondrocytes to ASC resulted in greater expression levels of chondrogenic genes, as detected using polymerase chain reaction, including COL2A1, L-SOX5, SOX6, SOX9, ACAN, and COMP. Our findings suggest that chondrocyte-ASC crosstalk is relevant to the upregulation of chondrogenic genes. The underlying mechanisms may include bidirectional paracrine signaling and cell-to-cell contact (juxtacrine). Although we suspect that the upregulated expression in this case represents chondrogenic differentiation of ASCs in vitro, it is possible that this reflects the reciprocal activity of ASCs in maintaining chondrocyte differentiation. Cell sorting techniques to separate the two cell types following co-culture would have allowed the analysis of the gene expression of each cell type.

Other studies investigating co-cultures of ASCs and chondrocytes have analyzed the expression profiles of these two cell populations individually[54,57]. These results provide further evidence that ASCs secrete chondroprotective molecules that promote cartilage repair and reduce the expression of proinflammatory cytokines and chemokines in chondrocytes. Hepatocyte growth factor produced by ASCs downregulates TGF-β expression and fibrotic markers (collagen I and III) in chondrocytes, whereas PGE2 decreases the expression of pro-inflammatory molecules, including IL-1β, TNF-α, and CCL-2.

The immunomodulatory function of MSCs has been widely reported[52] and is likely to play a key role in the treatment of osteoarthritis. Aggarwal and Pittenger[58] co-cultured human MSCs with subpopulations of immune cells to investigate alterations in their cytokine expression profiles. They found that MSCs reduced the expression of proinflammatory cytokines and induced anti-inflammatory phenotypic changes in various immune cells. MSCs themselves were found to display increased expression of PGE2 in co-culture, implying that PGE2 was responsible for promoting anti-inflammatory changes and that its expression was mediated by crosstalk between immune cells and MSCs.

The concept of reciprocal interactions between MSCs and immune cells has been demonstrated repeatedly[59], including the induction of PGE2 expression using TNF-α, which in turn leads to an anti-inflammatory switch by immune cells. Pro-inflammatory M1 macrophages that produce IL-1β, TGF-β, and TNF-α drive osteoarthritis, whereas the anti-inflammatory, pro-reparative (M2) phenotype that produces IL-10 is considered chondroprotective[17,53].

MiRNAs are additional bioactive molecules implicated in the immunomodulatory functions of ASCs[60]. In response to the synovial fluid obtained from human patients with osteoarthritis, ASCs demonstrate altered expression of miRNAs embedded in extracellular vesicles, which act on chondrocytes and macrophages to increase the expression of chondrogenic genes and promote the M2 phenotype, respectively. This provides additional evidence that MSCs secrete bioactive molecules in response to local injurious stimuli.

Although studies on the effectiveness of ASCs in treating osteoarthritis primarily address their ability to regenerate cartilage, there is emerging evidence of their ability to promote bone repair[61]. A study investigating the effect of ASCs on osteoclastogenesis, both in vitro and in a mouse model, demonstrated that RANKL-induced osteoclastogenesis was suppressed following ASC treatment[62]. As osteoclasts are derived from the myeloid lineage, the anti-inflammatory function of MSCs may also be applicable to the modulation of osteoclast activity[61]. The mechanisms involved may include cell-cell contact or paracrine effects. For example, secretion of osteoprotegerin by MSCs, a decoy receptor that competitively binds to RANKL, reduces osteoclastogenesis[61]. Therefore, as proposed for cartilage repair, evidence for the potential reparative role of ASCs in damaged bone suggests that the tissue may be regenerated through a combination of anti-inflammatory mechanisms and protection from further tissue destruction.

In addition to their local action, ASCs injected into human osteoarthritic knee joints mediated a systemic immune regulatory function[63]. Three months after the intraarticular injection of ASCs, increased peripheral regulatory T cells and decreased circulating classical monocytes were observed in fresh peripheral blood. Systemic effects of MSCs on immune cells have also been demonstrated in other diseases. In addition to interactions in colocalized areas, there is evidence for the role of MSCs in inducing the migration of macrophages to sites of inflammation, including atherosclerotic plaques and infections[64,65]. Interestingly, CCL-2 expression in MSCs was responsible for this effect. Therefore, although elevated CCL-2 levels are considered proinflammatory, its production by MSCs in the context of osteoarthritis could be explained by CCL-2’s beneficial role in the initial recruitment of M1 macrophages to the site of injury. Through reciprocal interactions with MSCs, they may induce a switch to M2 pro-reparative macrophages.

ADIPOSE-DERIVED REGENERATIVE THERAPIES FOR KNEE OSTEOARTHRITIS: IMPORTANT DISTINCTIONS

An array of heterogeneous clinical studies using different permutations of adipose-derived therapies[66] has demonstrated growing interest in harnessing the pro-reparative properties of adipose tissue for cartilage regeneration. Given the differences in the preparation, composition, and physical properties of different adipose-derived therapies, it is pertinent to avoid conflating these and make an early distinction between their properties and, therefore, their therapeutic potential.

ASCs have gained popularity over traditionally studied BMSCs because of their ease of isolation and high proliferative ability[17]. In a recent systematic review, we found that isolated ASCs are a successful and safe therapy for the treatment of focal cartilage defects of the knee that risk progression to osteoarthritis[67]. Similarly, ASCs have shown promise for the treatment of established osteoarthritis[68] and are superior to BMSCs in this field[13].

SVF is a related therapy produced by the enzymatic digestion of adipose tissue. The composition of various cell types, including ASCs, immune cells, pericytes, fibroblasts, and endothelial cells, is thought to offer a higher therapeutic potential because of the complementary action of different cell populations[69]. SVF has demonstrated therapeutic benefits in patients with established osteoarthritis[19,69]. However, SVF is not strictly an ASC treatment. Although meta-analyses have demonstrated the promise of both ASCs and SVF as adipose-derived therapies[67,68], it is more appropriate to consider that these distinct treatments should be investigated separately or in comparison to one another.

MFAT was developed to disrupt the adipose tissue while maintaining its in vivo composition[22]. Unlike SVF, this technique involves mechanical, rather than enzymatic, disruption. This has the advantage of maintaining an adipose ECM composed of collagen, laminin, fibronectin, and GAGs, which can improve graft fixation and cell retention within the site of delivery[70]. Again, although MFAT contains a fraction of ASCs, it is not strictly an ASC therapy because of the lack of cell isolation and culture expansion, preserved ECM, and additional immune cells[71].

Numerous studies have demonstrated the efficacy of MFAT in alleviating osteoarthritis symptoms in recent years, but few have evaluated its treatment in randomized controlled trials (RCTs)[72-74]. More RCTs are needed to provide more evidence regarding MFAT[15]. Wu et al[24] expanded on emerging evidence by reporting the outcomes of their prospective, multicenter, randomized trial, recruiting a larger, more heterogeneous population than those in previous studies. Intraarticular delivery of MFAT, performed alongside various other arthroscopic treatments for osteoarthritis, was compared with that of an intraarticular HA control. Patients with Kellgren-Lawrence grade 2-3 osteoarthritis of the knee (i.e., moderate disease) that had been symptomatic for over six months and failed to respond to conservative measures were included. Follow-up was conducted over 24 months. Outcome measures included changes in various PROMs, occurrence of adverse events, and magnetic resonance imaging (MRI) evidence of osteoarthritis resolution.

No serious adverse events were observed in either group. While there was no significant difference in the PROMs at baseline, the MFAT performed significantly better at all follow-up time points. Both groups demonstrated improvement relative to baseline, which was possibly related to the arthroscopic intervention. Notably, in the treatment group, the total Western Ontario and McMaster Universities Osteoarthritis index declined between 12 and 24 months. This might indicate a limited duration of treatment efficacy; however, a longer follow-up period is necessary to draw any conclusions. While the MFAT group demonstrated MRI improvements relative to baseline (P < 0.05), this was not significant compared to the control group at 24 months. Discrepancies between clinical and imaging outcomes following adipose-derived therapies have been noted in other studies and may require more than 24 months to correlate[15]. Again, a longer follow-up period or more sensitive imaging techniques may be needed to determine the true extent of morphological improvements.

The strength of this study lies in its generalizability. The multi-centre, adequately powered design, and use of intention-to-treat analysis confer external validity. This is likely reflected by the fact that the improvements observed were more modest than those in other trials. Although unavoidable, both the patients and surgeons were not blinded, possibly introducing a degree of bias. In addition, the repeated use of PROMs tools at various follow-up points may have introduced a response bias.

In conclusion, Wu et al[24] demonstrated the efficacy of MFAT in treating moderately advanced knee osteoarthritis, in accordance with the existing literature. The ability to offer a minimally invasive therapy that provides sustained functional improvements at two years follow-up is encouraging. Further follow-up data will be important in determining the longevity of cartilage repair by MFAT and whether this correlates with the evidence of healing seen on imaging.

PROPERTIES OF DIFFERENT ADIPOSE-DERIVED THERAPIES

It is important to consider how SVF and MFAT function as therapies that are distinct from isolated ASCs. Unpassaged (p0) stromal cells comprise a more heterogeneous cell population, whereas culture passages select for plastic-adherent ASCs[75]. ASCs in culture may also undergo genetic alterations, depending on the culture conditions[76] which may alter their behavior in vivo. Furthermore, given that MSC function relies on immunomodulatory mechanisms and bidirectional signaling with native cells, it is expected that their delivery within an adipose niche composed of various cell types, including immune cells, will have an impact on their therapeutic function.

Desando et al[53] investigated how cell migration patterns differ between ASCs, SVF, and MFAT in a rabbit model of knee osteoarthritis. Isolated ASCs and those in the SVF showed higher tropism for the synovium and menisci than for the cartilage. Conversely, MFAT showed the highest tropism for the cartilage on day seven, followed by the synovium on day 30. The authors suggested that the collagen network in MFAT permitted greater survival of cells embedded within the hypoxic environment of the osteoarthritic cartilage and protection from lytic enzymes. The migration pattern may also be explained by the structure of MFAT, which acts as a reservoir of cells until the collagen degrades, a concept that has been supported in vitro and is associated with a prolonged release of anti-inflammatory cytokines[77]. Furthermore, while all three therapies showed strong positivity for MSC markers, MFAT contained a greater abundance of blood vessels, preadipocytes, and monocytes, as well as more CD163+ (M2) macrophages, than SVF. MSCs from MFAT also demonstrated greater co-localization with CD163+ cells within the knee joint.

Other authors have commented on how the preparation of MFAT may improve its efficiency compared to other adipose-derived therapies. Vezzani et al[78] showed that mechanical disruption of adipose tissue maintains ASCs within the perivascular environment observed in vivo, unlike the enzymatic digestion process involved in SVF. This was associated with significantly higher levels of secreted growth factors and cytokines that promote tissue regeneration, which may be explained by the improved function of ASCs when retained within their usual niche and the altered gene expression induced by enzymatic tissue digestion.

FUTURE CHALLENGES

As previously mentioned, ASCs have gained popularity owing to their ease of isolation, abundance in adipose tissue, anti-inflammatory properties, and proliferative potential[17]. Furthermore, the intraarticular delivery of adipose-derived therapies, including SVF and MFAT, is conducive to relatively noninvasive treatment techniques[79]. However, various challenges exist in translating adipose-derived regenerative therapies from trial settings to clinical practice.

One challenge is our incomplete understanding of how to stratify patients to predict successful treatment outcomes. For example, ASCs obtained from morbidly obese patients show reduced proliferation rates and chondrogenic differentiation capacity[80,81]. Furthermore, studies have often been conducted using cells obtained from healthy donors. The proliferative capacity of MSCs decreases with age, which may affect the applicability of the experimental evidence in elderly patients with advanced osteoarthritis[80,81]. Tailoring MSC therapy to particular patient groups is also complicated because the phenotype of osteoarthritis differs depending on its etiology[82].

Safety concerns raised regarding ASC therapies include the potential for immunoreactivity with animal-derived products used for culture expansion and the theoretical possibility of tumorigenesis, as the genetic instability of ASCs increases following long-term culture[83]. However, a meta-analysis of 11 trials using MSC therapies demonstrated no increase in the incidence of complications compared to control therapies[84]. Finally, regulatory restrictions necessitating minimal manipulation of the transplanted tissue may act as a barrier to the clinical implementation of adipose-derived therapies, including SVF, which requires the enzymatic digestion of adipose tissue[83]. The preparation of MFAT, which involves the mechanical disruption of tissue and no cell culture[24], may overcome several safety and regulatory concerns.

CONCLUSION

Osteoarthritis is a progressive disease that results from the degeneration of hyaline cartilage. The lack of available disease-modifying therapies has led to ample research on therapies with the potential to regenerate articular cartilage. ASCs are a promising cell therapy that have demonstrated efficacy in human trials and are likely to function through the production of immunomodulatory and chondroprotective biomolecules in response to local inflammatory stimuli.

Adipose tissue is gaining popularity as a source of ASCs because of the ease of harvesting and high proliferative capacity of ASCs compared to other MSC sources. SVF and MFAT are other adipose-derived regenerative therapies which retain a heterogenous population of cell types including, but not limited to, ASCs. Both have successfully alleviated osteoarthritis symptoms in humans. The lack of enzymatic processing of adipose tissue required to produce MFAT, unlike SVF, is likely to confer additional beneficial properties that might potentiate the mechanism of action of MSCs. This may explain the encouraging results of MFAT treatment observed across multiple trials in recent years. The success of MFAT in treating osteoarthritis, as evidenced by both symptomatic and radiological improvements, is certainly encouraging and warrants continued investigation to determine the optimal treatment approach and longevity of cartilage repair.

Footnotes

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

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country/Territory of origin: United Kingdom

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Xu T, China S-Editor: Wang JJ L-Editor: A P-Editor: Zhao YQ

References
1.  Cross M, Smith E, Hoy D, Nolte S, Ackerman I, Fransen M, Bridgett L, Williams S, Guillemin F, Hill CL, Laslett LL, Jones G, Cicuttini F, Osborne R, Vos T, Buchbinder R, Woolf A, March L. The global burden of hip and knee osteoarthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. 2014;73:1323-1330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1801]  [Cited by in F6Publishing: 2050]  [Article Influence: 205.0]  [Reference Citation Analysis (0)]
2.  Fransen M, McConnell S, Harmer AR, Van der Esch M, Simic M, Bennell KL. Exercise for osteoarthritis of the knee: a Cochrane systematic review. Br J Sports Med. 2015;49:1554-1557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 392]  [Article Influence: 43.6]  [Reference Citation Analysis (0)]
3.  Richards MM, Maxwell JS, Weng L, Angelos MG, Golzarian J. Intra-articular treatment of knee osteoarthritis: from anti-inflammatories to products of regenerative medicine. Phys Sportsmed. 2016;44:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 98]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
4.  Felson DT. Arthroscopy as a treatment for knee osteoarthritis. Best Pract Res Clin Rheumatol. 2010;24:47-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
5.  Żylińska B, Silmanowicz P, Sobczyńska-Rak A, Jarosz Ł, Szponder T. Treatment of Articular Cartilage Defects: Focus on Tissue Engineering. In Vivo. 2018;32:1289-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
6.  Bhandari M, Smith J, Miller LE, Block JE. Clinical and economic burden of revision knee arthroplasty. Clin Med Insights Arthritis Musculoskelet Disord. 2012;5:89-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 121]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
7.  Lim HC, Bae JH, Song SH, Park YE, Kim SJ. Current treatments of isolated articular cartilage lesions of the knee achieve similar outcomes. Clin Orthop Relat Res. 2012;470:2261-2267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 91]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
8.  Gracitelli GC, Moraes VY, Franciozi CE, Luzo MV, Belloti JC. Surgical interventions (microfracture, drilling, mosaicplasty, and allograft transplantation) for treating isolated cartilage defects of the knee in adults. Cochrane Database Syst Rev. 2016;9:CD010675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
9.  Devitt BM, Bell SW, Webster KE, Feller JA, Whitehead TS. Surgical treatments of cartilage defects of the knee: Systematic review of randomised controlled trials. Knee. 2017;24:508-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 88]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
10.  Oldershaw RA. Cell sources for the regeneration of articular cartilage: the past, the horizon and the future. Int J Exp Pathol. 2012;93:389-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 65]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
11.  Huang BJ, Hu JC, Athanasiou KA. Cell-based tissue engineering strategies used in the clinical repair of articular cartilage. Biomaterials. 2016;98:1-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 258]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
12.  Epanomeritakis IE, Lee E, Lu V, Khan W. The Use of Autologous Chondrocyte and Mesenchymal Stem Cell Implants for the Treatment of Focal Chondral Defects in Human Knee Joints-A Systematic Review and Meta-Analysis. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
13.  Jeyaraman M, Muthu S, Ganie PA. Does the Source of Mesenchymal Stem Cell Have an Effect in the Management of Osteoarthritis of the Knee? Meta-Analysis of Randomized Controlled Trials. Cartilage. 2021;13:1532S-1547S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
14.  Shetty AA, Kim SJ, Shanmugasundaram S, Shetty N, Stelzeneder D, Kim CS. Injectable cultured bone marrow derived mesenchymal cells vs chondrocytes in the treatment of chondral defects of the knee - RCT with 6 years follow-up. J Clin Orthop Trauma. 2022;28:101845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
15.  Ranmuthu CDS, Ranmuthu CKI, Khan WS. Evaluating the Current Literature on Treatments Containing Adipose-Derived Stem Cells for Osteoarthritis: a Progress Update. Curr Rheumatol Rep. 2018;20:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
16.  Le H, Xu W, Zhuang X, Chang F, Wang Y, Ding J. Mesenchymal stem cells for cartilage regeneration. J Tissue Eng. 2020;11:2041731420943839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 110]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
17.  Lv Z, Cai X, Bian Y, Wei Z, Zhu W, Zhao X, Weng X. Advances in Mesenchymal Stem Cell Therapy for Osteoarthritis: From Preclinical and Clinical Perspectives. Bioengineering (Basel). 2023;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
18.  Yokota N, Hattori M, Ohtsuru T, Otsuji M, Lyman S, Shimomura K, Nakamura N. Comparative Clinical Outcomes After Intra-articular Injection With Adipose-Derived Cultured Stem Cells or Noncultured Stromal Vascular Fraction for the Treatment of Knee Osteoarthritis. Am J Sports Med. 2019;47:2577-2583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
19.  Goncharov EN, Koval OA, Nikolaevich Bezuglov E, Encarnacion Ramirez MJ, Engelgard M, Igorevich EI, Saporiti A, Valentinovich Kotenko K, Montemurro N. Stromal Vascular Fraction Therapy for Knee Osteoarthritis: A Systematic Review. Medicina (Kaunas). 2023;59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
20.  Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, Redl H, Rubin JP, Yoshimura K, Gimble JM. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy. 2013;15:641-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1199]  [Cited by in F6Publishing: 1245]  [Article Influence: 113.2]  [Reference Citation Analysis (2)]
21.  Vinet-Jones H, F Darr K. Clinical use of autologous micro-fragmented fat progressively restores pain and function in shoulder osteoarthritis. Regen Med. 2020;15:2153-2161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
22.  Bianchi F, Maioli M, Leonardi E, Olivi E, Pasquinelli G, Valente S, Mendez AJ, Ricordi C, Raffaini M, Tremolada C, Ventura C. A new nonenzymatic method and device to obtain a fat tissue derivative highly enriched in pericyte-like elements by mild mechanical forces from human lipoaspirates. Cell Transplant. 2013;22:2063-2077.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 193]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
23.  Carelli S, Messaggio F, Canazza A, Hebda DM, Caremoli F, Latorre E, Grimoldi MG, Colli M, Bulfamante G, Tremolada C, Di Giulio AM, Gorio A. Characteristics and Properties of Mesenchymal Stem Cells Derived From Microfragmented Adipose Tissue. Cell Transplant. 2015;24:1233-1252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
24.  Wu CZ, Shi ZY, Wu Z, Lin WJ, Chen WB, Jia XW, Xiang SC, Xu HH, Ge QW, Zou KA, Wang X, Chen JL, Wang PE, Yuan WH, Jin HT, Tong PJ. Mid-term outcomes of microfragmented adipose tissue plus arthroscopic surgery for knee osteoarthritis: A randomized, active-control, multicenter clinical trial. World J Stem Cells. 2023;15:1063-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
25.  Kohn MD, Sassoon AA, Fernando ND. Classifications in Brief: Kellgren-Lawrence Classification of Osteoarthritis. Clin Orthop Relat Res. 2016;474:1886-1893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 680]  [Cited by in F6Publishing: 599]  [Article Influence: 74.9]  [Reference Citation Analysis (0)]
26.  Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum. 2012;64:1697-1707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1550]  [Cited by in F6Publishing: 1806]  [Article Influence: 150.5]  [Reference Citation Analysis (1)]
27.  Sophia Fox AJ, Bedi A, Rodeo SA. The basic science of articular cartilage: structure, composition, and function. Sports Health. 2009;1:461-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1254]  [Cited by in F6Publishing: 1399]  [Article Influence: 116.6]  [Reference Citation Analysis (0)]
28.  Sherwood J, Bertrand J, Nalesso G, Poulet B, Pitsillides A, Brandolini L, Karystinou A, De Bari C, Luyten FP, Pitzalis C, Pap T, Dell'Accio F. A homeostatic function of CXCR2 signalling in articular cartilage. Ann Rheum Dis. 2015;74:2207-2215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
29.  Burleigh A, Chanalaris A, Gardiner MD, Driscoll C, Boruc O, Saklatvala J, Vincent TL. Joint immobilization prevents murine osteoarthritis and reveals the highly mechanosensitive nature of protease expression in vivo. Arthritis Rheum. 2012;64:2278-2288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
30.  Miotla Zarebska J, Chanalaris A, Driscoll C, Burleigh A, Miller RE, Malfait AM, Stott B, Vincent TL. CCL2 and CCR2 regulate pain-related behaviour and early gene expression in post-traumatic murine osteoarthritis but contribute little to chondropathy. Osteoarthritis Cartilage. 2017;25:406-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 88]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
31.  Sandell LJ, Xing X, Franz C, Davies S, Chang LW, Patra D. Exuberant expression of chemokine genes by adult human articular chondrocytes in response to IL-1beta. Osteoarthritis Cartilage. 2008;16:1560-1571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 113]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
32.  Zhang Y, Liu D, Vithran DTA, Kwabena BR, Xiao W, Li Y. CC chemokines and receptors in osteoarthritis: new insights and potential targets. Arthritis Res Ther. 2023;25:113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
33.  Xu YK, Ke Y, Wang B, Lin JH. The role of MCP-1-CCR2 ligand-receptor axis in chondrocyte degradation and disease progress in knee osteoarthritis. Biol Res. 2015;48:64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
34.  Raghu H, Lepus CM, Wang Q, Wong HH, Lingampalli N, Oliviero F, Punzi L, Giori NJ, Goodman SB, Chu CR, Sokolove JB, Robinson WH. CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment, inflammation and cartilage destruction in osteoarthritis. Ann Rheum Dis. 2017;76:914-922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 260]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
35.  Wilcockson HH, Esterellas A, Jowdy C, Ozkan H, Loeser RF, Longobardi L. Identification of signaling pathways mediating human chondrocyte degeneration induced by the chemokine CCL-2. Osteoarthritis and Cartilage. 2019;27:S41-S42.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  van der Kraan PM. The changing role of TGFβ in healthy, ageing and osteoarthritic joints. Nat Rev Rheumatol. 2017;13:155-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 122]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
37.  Liu Z, Chen J, Mirando AJ, Wang C, Zuscik MJ, O'Keefe RJ, Hilton MJ. A dual role for NOTCH signaling in joint cartilage maintenance and osteoarthritis. Sci Signal. 2015;8:ra71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
38.  Chen D, Shen J, Zhao W, Wang T, Han L, Hamilton JL, Im HJ. Osteoarthritis: toward a comprehensive understanding of pathological mechanism. Bone Res. 2017;5:16044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 635]  [Cited by in F6Publishing: 628]  [Article Influence: 89.7]  [Reference Citation Analysis (0)]
39.  Iulian Stanciugelu S, Homorogan C, Selaru C, Patrascu JM, Patrascu JM Jr, Stoica R, Nitusca D, Marian C. Osteoarthritis and microRNAs: Do They Provide Novel Insights into the Pathophysiology of This Degenerative Disorder? Life (Basel). 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
40.  Panagopoulos PK, Lambrou GI. The Involvement of MicroRNAs in Osteoarthritis and Recent Developments: A Narrative Review. Mediterr J Rheumatol. 2018;29:67-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
41.  Goldring MB, Goldring SR. Articular cartilage and subchondral bone in the pathogenesis of osteoarthritis. Ann N Y Acad Sci. 2010;1192:230-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 527]  [Cited by in F6Publishing: 531]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
42.  Li G, Yin J, Gao J, Cheng TS, Pavlos NJ, Zhang C, Zheng MH. Subchondral bone in osteoarthritis: insight into risk factors and microstructural changes. Arthritis Res Ther. 2013;15:223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 464]  [Article Influence: 46.4]  [Reference Citation Analysis (0)]
43.  Hu W, Chen Y, Dou C, Dong S. Microenvironment in subchondral bone: predominant regulator for the treatment of osteoarthritis. Ann Rheum Dis. 2021;80:413-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 161]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
44.  Su W, Liu G, Liu X, Zhou Y, Sun Q, Zhen G, Wang X, Hu Y, Gao P, Demehri S, Cao X, Wan M. Angiogenesis stimulated by elevated PDGF-BB in subchondral bone contributes to osteoarthritis development. JCI Insight. 2020;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 91]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
45.  Suri S, Walsh DA. Osteochondral alterations in osteoarthritis. Bone. 2012;51:204-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 167]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
46.  Steverink JG, Oostinga D, van Tol FR, van Rijen MHP, Mackaaij C, Verlinde-Schellekens SAMW, Oosterman BJ, Van Wijck AJM, Roeling TAP, Verlaan JJ. Sensory Innervation of Human Bone: An Immunohistochemical Study to Further Understand Bone Pain. J Pain. 2021;22:1385-1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
47.  Morgan M, Nazemian V, Harrington K, Ivanusic JJ. Mini review: The role of sensory innervation to subchondral bone in osteoarthritis pain. Front Endocrinol (Lausanne). 2022;13:1047943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
48.  Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY, Henrotin YE. Subchondral bone osteoblasts induce phenotypic changes in human osteoarthritic chondrocytes. Osteoarthritis Cartilage. 2005;13:988-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 131]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
49.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11055]  [Cited by in F6Publishing: 11799]  [Article Influence: 694.1]  [Reference Citation Analysis (1)]
50.  Mildmay-White A, Khan W. Cell Surface Markers on Adipose-Derived Stem Cells: A Systematic Review. Curr Stem Cell Res Ther. 2017;12:484-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 93]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
51.  Caplan AI. Mesenchymal Stem Cells: Time to Change the Name! Stem Cells Transl Med. 2017;6:1445-1451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 558]  [Cited by in F6Publishing: 621]  [Article Influence: 88.7]  [Reference Citation Analysis (0)]
52.  Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 786]  [Cited by in F6Publishing: 811]  [Article Influence: 73.7]  [Reference Citation Analysis (0)]
53.  Desando G, Bartolotti I, Martini L, Giavaresi G, Nicoli Aldini N, Fini M, Roffi A, Perdisa F, Filardo G, Kon E, Grigolo B. Regenerative Features of Adipose Tissue for Osteoarthritis Treatment in a Rabbit Model: Enzymatic Digestion Versus Mechanical Disruption. Int J Mol Sci. 2019;20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
54.  Maumus M, Manferdini C, Toupet K, Peyrafitte JA, Ferreira R, Facchini A, Gabusi E, Bourin P, Jorgensen C, Lisignoli G, Noël D. Adipose mesenchymal stem cells protect chondrocytes from degeneration associated with osteoarthritis. Stem Cell Res. 2013;11:834-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
55.  Ohashi H, Nishida K, Yoshida A, Nasu Y, Nakahara R, Matsumoto Y, Takeshita A, Kaneda D, Saeki M, Ozaki T. Adipose-Derived Extract Suppresses IL-1β-Induced Inflammatory Signaling Pathways in Human Chondrocytes and Ameliorates the Cartilage Destruction of Experimental Osteoarthritis in Rats. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
56.  Mak CCH, To K, Fekir K, Brooks RA, Khan WS. Infrapatellar fat pad adipose-derived stem cells co-cultured with articular chondrocytes from osteoarthritis patients exhibit increased chondrogenic gene expression. Cell Commun Signal. 2022;20:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
57.  Manferdini C, Maumus M, Gabusi E, Piacentini A, Filardo G, Peyrafitte JA, Jorgensen C, Bourin P, Fleury-Cappellesso S, Facchini A, Noël D, Lisignoli G. Adipose-derived mesenchymal stem cells exert antiinflammatory effects on chondrocytes and synoviocytes from osteoarthritis patients through prostaglandin E2. Arthritis Rheum. 2013;65:1271-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
58.  Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815-1822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3271]  [Cited by in F6Publishing: 3170]  [Article Influence: 158.5]  [Reference Citation Analysis (0)]
59.  Ghannam S, Bouffi C, Djouad F, Jorgensen C, Noël D. Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications. Stem Cell Res Ther. 2010;1:2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 358]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
60.  Ragni E, Colombini A, Viganò M, Libonati F, Perucca Orfei C, Zagra L, de Girolamo L. Cartilage Protective and Immunomodulatory Features of Osteoarthritis Synovial Fluid-Treated Adipose-Derived Mesenchymal Stem Cells Secreted Factors and Extracellular Vesicles-Embedded miRNAs. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
61.  Ibáñez L, Guillem-Llobat P, Marín M, Guillén MI. Connection between Mesenchymal Stem Cells Therapy and Osteoclasts in Osteoarthritis. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (1)]
62.  Chang Q, Li C, Lu Y, Geng R, Wei JN, Hu JZ. Adipose-derived mesenchymal stromal cells suppress osteoclastogenesis and bone erosion in collagen-induced arthritis. Scand J Immunol. 2020;92:e12877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
63.  Pers YM, Quentin J, Feirreira R, Espinoza F, Abdellaoui N, Erkilic N, Cren M, Dufourcq-Lopez E, Pullig O, Nöth U, Jorgensen C, Louis-Plence P. Injection of Adipose-Derived Stromal Cells in the Knee of Patients with Severe Osteoarthritis has a Systemic Effect and Promotes an Anti-Inflammatory Phenotype of Circulating Immune Cells. Theranostics. 2018;8:5519-5528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 42]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
64.  Shi C, Jia T, Mendez-Ferrer S, Hohl TM, Serbina NV, Lipuma L, Leiner I, Li MO, Frenette PS, Pamer EG. Bone marrow mesenchymal stem and progenitor cells induce monocyte emigration in response to circulating toll-like receptor ligands. Immunity. 2011;34:590-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 382]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
65.  Del Toro R, Chèvre R, Rodríguez C, Ordóñez A, Martínez-González J, Andrés V, Méndez-Ferrer S. Nestin(+) cells direct inflammatory cell migration in atherosclerosis. Nat Commun. 2016;7:12706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
66.  Frazier T, March K, Garza JR, Bunnell BA, Darr KF, Rogers E, Hamel K, Gimble JM. Non-homologous use of adipose-derived cell and tissue therapies: Osteoarthritis as a case study. Bone Rep. 2022;17:101601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
67.  Meng HY, Lu V, Khan W. Adipose Tissue-Derived Mesenchymal Stem Cells as a Potential Restorative Treatment for Cartilage Defects: A PRISMA Review and Meta-Analysis. Pharmaceuticals (Basel). 2021;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
68.  Kim KI, Kim MS, Kim JH. Intra-articular Injection of Autologous Adipose-Derived Stem Cells or Stromal Vascular Fractions: Are They Effective for Patients With Knee Osteoarthritis? A Systematic Review With Meta-analysis of Randomized Controlled Trials. Am J Sports Med. 2023;51:837-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
69.  Boada-Pladellorens A, Avellanet M, Pages-Bolibar E, Veiga A. Stromal vascular fraction therapy for knee osteoarthritis: a systematic review. Ther Adv Musculoskelet Dis. 2022;14:1759720X221117879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
70.  Greenwood V, Clausen P, Matuska AM. Micro-fragmented adipose tissue cellular composition varies by processing device and analytical method. Sci Rep. 2022;12:16107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
71.  Mautner K, Bowers R, Easley K, Fausel Z, Robinson R. Functional Outcomes Following Microfragmented Adipose Tissue Versus Bone Marrow Aspirate Concentrate Injections for Symptomatic Knee Osteoarthritis. Stem Cells Transl Med. 2019;8:1149-1156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
72.  Zaffagnini S, Andriolo L, Boffa A, Poggi A, Cenacchi A, Busacca M, Kon E, Filardo G, Di Martino A. Microfragmented Adipose Tissue Versus Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: A Prospective Randomized Controlled Trial at 2-Year Follow-up. Am J Sports Med. 2022;50:2881-2892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
73.  Ulivi M, Meroni V, Viganò M, Colombini A, Lombardo MDM, Rossi N, Orlandini L, Messina C, Sconfienza LM, Peretti GM, Mangiavini L, de Girolamo L. Micro-fragmented adipose tissue (mFAT) associated with arthroscopic debridement provides functional improvement in knee osteoarthritis: a randomized controlled trial. Knee Surg Sports Traumatol Arthrosc. 2023;31:3079-3090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
74.  Baria M, Pedroza A, Kaeding C, Durgam S, Duerr R, Flanigan D, Borchers J, Magnussen R. Platelet-Rich Plasma Versus Microfragmented Adipose Tissue for Knee Osteoarthritis: A Randomized Controlled Trial. Orthop J Sports Med. 2022;10:23259671221120678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
75.  Locke M, Windsor J, Dunbar PR. Human adipose-derived stem cells: isolation, characterization and applications in surgery. ANZ J Surg. 2009;79:235-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 179]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
76.  Dahl JA, Duggal S, Coulston N, Millar D, Melki J, Shahdadfar A, Brinchmann JE, Collas P. Genetic and epigenetic instability of human bone marrow mesenchymal stem cells expanded in autologous serum or fetal bovine serum. Int J Dev Biol. 2008;52:1033-1042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 131]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
77.  Nava S, Sordi V, Pascucci L, Tremolada C, Ciusani E, Zeira O, Cadei M, Soldati G, Pessina A, Parati E, Slevin M, Alessandri G. Long-Lasting Anti-Inflammatory Activity of Human Microfragmented Adipose Tissue. Stem Cells Int. 2019;2019:5901479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
78.  Vezzani B, Shaw I, Lesme H, Yong L, Khan N, Tremolada C, Péault B. Higher Pericyte Content and Secretory Activity of Microfragmented Human Adipose Tissue Compared to Enzymatically Derived Stromal Vascular Fraction. Stem Cells Transl Med. 2018;7:876-886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
79.  Pasculli RM, Kenyon CD, Berrigan WA, Mautner K, Hammond K, Jayaram P. Mesenchymal stem cells for subchondral bone marrow lesions: From bench to bedside. Bone Rep. 2022;17:101630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
80.  Diekman BO, Guilak F. Stem cell-based therapies for osteoarthritis: challenges and opportunities. Curr Opin Rheumatol. 2013;25:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 98]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
81.  Loo SJQ, Wong NK. Advantages and challenges of stem cell therapy for osteoarthritis (Review). Biomed Rep. 2021;15:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
82.  Sun AR, Udduttula A, Li J, Liu Y, Ren PG, Zhang P. Cartilage tissue engineering for obesity-induced osteoarthritis: Physiology, challenges, and future prospects. J Orthop Translat. 2021;26:3-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
83.  Zhang J, Liu Y, Chen Y, Yuan L, Liu H, Wang J, Liu Q, Zhang Y. Adipose-Derived Stem Cells: Current Applications and Future Directions in the Regeneration of Multiple Tissues. Stem Cells Int. 2020;2020:8810813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 61]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
84.  Yubo M, Yanyan L, Li L, Tao S, Bo L, Lin C. Clinical efficacy and safety of mesenchymal stem cell transplantation for osteoarthritis treatment: A meta-analysis. PLoS One. 2017;12:e0175449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 111]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]