Open Access

Advantages and challenges of stem cell therapy for osteoarthritis (Review)

  • Authors:
    • Stephanie Jyet Quan Loo
    • Nyet Kui Wong
  • View Affiliations

  • Published online on: June 10, 2021     https://doi.org/10.3892/br.2021.1443
  • Article Number: 67
  • Copyright: © Loo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteoarthritis (OA) is a degenerative disorder of the cartilage and is one of the leading causes of disability, particularly amongst the elderly, wherein patients with advanced‑stage OA experience chronic pain and functional impairment of the limbs, thus resulting in a significantly reduced quality of life. The currently available treatments primarily revolve around symptom management, and is thus palliative rather than curative. The aim of the present review is to briefly discuss the limitations of some of the currently available treatments for patients with OA, and highlight the value of the potential use of stem cells in cellular therapy, which is widely regarded as the breakthrough that can address the present unmet medical needs for treatment of degenerative diseases, such as OA. The advantages of stem cell therapy, particularly mesenchymal stem cells, and the challenges involved are also discussed in this review.

1. Introduction

Arthritis is an umbrella term used to refer to diseases that cause pain and inflammation of the joints, and it characterized by painful inflammation and stiffness of the joints (1). Osteoarthritis (OA) is one of the most commonly diagnosed types of arthritis, and it is considered a chronic, debilitating and prevalent joint disease, accounting for ~23% of all cases of musculoskeletal disorders, according to the Global Burden Disease study 2017(2). In brief, OA occurs due to the loss of articular cartilage within the synovial joints with the natural propensity to occur in elderly individuals (3,4). The high count of years lived with disability of patients with OA makes it one of the leading causes of disability, where patients with advanced-stage OA tend to experience chronic pain and functional impairment of the limbs, thus resulting in a poor quality of life (1).

Generally, a healthy joint possesses a layer of slippery tissue known as the articular cartilage, which is comprised primarily of chondrocytes and extracellular matrix (ECM). The ECM is predominantly made up of proteoglycan and type II collagen fiber (5). With several roles in the musculoskeletal system, the articular cartilage lubricates the bones during angular movements and absorbs shock to prevent the bones from impacting one another. This involves the spreading of the load evenly across the joints during weight-bearing activities (such as walking and weight-lifting), as well as high-intensity activities (such as running and jumping). Additionally, the articular cartilage also acts as a reservoir that stores synovial fluid; a fluid that transports nutrients to the joints (1).

Cartilage can be damaged through several factors, such as injuries as well as autoimmune diseases including rheumatoid arthritis (6). However, it can also be damaged from wear-and-tear over time. Typically, the occurrence of wear-and-tear are often counteracted by the repair and renewal of articular cartilage. However, the regenerative capacity is dependent on several aspects, including genetic background, age, sex, body weight and the level of physical activity an individual partakes in (7). When the cartilage damage outweighs the regenerative capacity of the body, thinning of the articular cartilage occurs followed by a progressive loss of articular cartilage. Under such circumstances, the individual will thus be diagnosed with OA (5).

OA is also referred to as degenerative arthritis due to its tendency to develop as a person ages, as well as the fact that it is characterized by the loss of the cartilage that leads to constant friction followed by the eventual deformation of the bones (1,8). In such events, the patient will experience inflammation and subsequently, pain and stiffness to the joints (7,8). OA can affect any joint in the body, but often occurs in the knees, small joints of the fingers, lower back, neck and hips (9). Though it was typically accepted that it occurs as part of the aging process, there are other causes of OA including congenital bone deformities, joint overuse, traumatic injuries, obesity and genetic diseases, such as Paget's disease and diabetes (7,10,11).

2. Challenges of conventional treatment methods for OA

The primary process that underlies the development of OA is the substantial degeneration of the structures within the articular cartilage, thus causing severe pain and reduced mobility. Unfortunately, the treatment options available for patients with OA are palliative measures rather than curative. Symptom management, with a focus on halting or slowing the progression of OA range from physical and nonsurgical therapies to surgery, including: i) Exercise programs for muscle strengthening and weight loss, and the use of supporting devices such as braces; ii) pharmacological interventions to alleviate pain; and iii) surgical interventions (12,13). However, there are certain challenges and limitations to all of these approaches as discussed in the following subsections.

Physical measures

A basic attempt to treat OA involves the introduction of an exercise program to strengthen the muscles surrounding the affected joints, and to promote weight loss if required. With regards to knee OA, where the knees act as the pillar of support to the human body, a study demonstrated the association between muscle weakness, particularly the quadriceps, and the development of knee OA (14). In addition to weak muscle strength, obesity which is suggested to be secondary to inactivity, is well-established to favor the development of knee OA through increased leverage, whereby the risk of knee OA is reported to increase by 36% with every 2 units of body mass index (BMI) gained, and the likelihood of developing knee OA by 4.2x in individuals with a BMI >30 kg/m2 (15,16). Hence, it was recommended that patients with OA increase the amount of exercise they do, such as weight lifting and strength training to increase muscle strength, as this has been proven to reduce pain and improve physical function, as well as aid in weight-loss (17-20). However, the pain and physical restrictions that come with OA often act as hurdles that keep OA patients from implementing and sustaining such activities. In addition, maintaining a healthy weight is another challenge faced by patients with OA, as it requires (often substantial) changes to their lifestyle, long term determination, and commitment to achieve noticeable results.

Use of braces

The general purpose of the braces is to provide support, as well as align and immobilize the area of the affected the joint (21). They prevent and correct deformities, thereby improving function and assisting in slowing the progression of the disease (22). There are several categories of braces available for various purposes. The unloader knee brace is used specifically for patients with OA to alleviate pain and improve physical function. Other categories of braces include the prophylactic knee brace, which is used to provide protection of the healthy knees against injuries during athletic activities; the patellofemoral knee brace which is used for anterior knee pain; and the functional knee brace is used to improve stability of an unstable knee in ligament injuries, such as a torn anterior cruciate ligament (ACL) or post-ACL reconstruction (22). Studies have reported that the use of braces has beneficial effects for patients with OA by reducing the pain they experience, improving the physical function of the affected joint, as well as delaying the need for surgical interventions (23,24). However, despite the benefits of these braces, it is only able to provide short-term pain relief, and are inefficient for long-term management (25). Furthermore, the efficacy of the knee braces varies between patients with OA; as indicated in certain studies, the use of braces lacks symptomatic relief, may fit poorly, and may cause discomfort when wearing the braces as well as skin irritation (26,27).

Medication

The use of pharmacological treatments for OA is often considered as a supplement in cases of severe OA, following failure to relieve symptoms by non-pharmacological methods (28). Some of the drugs used are analgesics, such as acetaminophen and opioids, NSAIDs and COX-2 inhibitors (29). Although the complementary usage of drugs and non-pharmacological regimen has been shown to be most effective for pain management of OA, there are safety concerns regarding the adverse effects of the drugs on the human body, such as liver toxicity as well as renal, cardiovascular and gastrointestinal side effects (29).

Surgical intervention

Surgery is considered as the final resort, when both pharmacological and non-pharmacological regimens fail to relieve the symptoms in patients with severe OA, particularly when their joints have entered a state of severe damage, causing unbearable pain, as well as deterioration of function in the affected patient. The types of surgical treatments for OA include arthroscopic lavage and debridement, cartilage repair, osteotomies and joint arthroplasty (30). Arthroscopic lavage and debridement are often performed as an initial surgical option that entails the removal of fragments of the meniscus, loose cartilage or osteophytes, and shaving of rough cartilage, and has been shown to alleviate pain and improve physical function (31). However, it has been demonstrated in randomized controlled trials that arthroscopic surgery was no more effective than a placebo surgery for treating knee OA. In the early 2000's, two seminal studies by Kirkley et al (32) and Moseley et al (33), made an impact on the use of arthroscopy for OA, in which the authors reported that patients with OA who received arthroscopy lavage and debridement did not show any improvements in the pain score and physical functions compared with a placebo group who underwent a sham surgery, a group that received optimized therapy, and a group that underwent physical therapy. The publication of these two landmark studies was followed by the update of the guidelines for the treatment of OA by the United Kingdom's National Institute for Health and Care Excellence, which no longer recommends arthroscopy as a treatment for OA (1).

The final resort for a patient with OA who has progressed to the most severe grade would be a total joint replacement. However, the procedure for joint replacements may cause unbearable pain and requires a long duration for rehabilitation. The adverse outcomes observed in patients who undergo total knee replacement surgery include myocardial infarction, infections and pulmonary embolism (34).

3. Stem cell therapy

The inconsistency of palliative treatments for OA highlight the need for a more reliable and curative approach that targets the root cause of OA; the degeneration of articular cartilage. Hence, the notion of stem cell therapy has galvanized intensive investigation into its potential use for treatment of OA, due to its regenerative properties. Owing to their excellent self-renewing capacity as well the ability to differentiate into >200 cell types, the use of stem cells in cellular therapy has ushered in an exciting new epoch for the fields of regenerative medicine with grounds for optimism to address the present unmet medical needs to treat a variety of degenerative diseases, including OA (35). At present, three types of stem cells are commonly studied with regard to stem cell therapy: Embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult stem cells, and these are discussed below.

ESCs

ESCs are considered to be totipotent stem cells derived from the fertilized zygote cell, wherein the embryo is usually 4-5 days old (35,36). The use of embryonic stem cells has generated ethical concerns, particularly with regard to how they are obtained (37). Hence, it is restricted for use in biomedical research only, and is to date, illegal for use as a treatment of any diseases, as their remains a notable bone of contention over the considerable ethical issues that arise, given that an embryo must be aborted to obtain the ESCs (37,38).

iPSCs

As one of the major revolutions in stem cell research, the identification and interest in research on iPSCs was spurred on by the ethical issues raised over the sourcing of ESCs (39,40). iPSCs were first discovered in 2006 by Takahashi and Yamanaka (39) who successfully reprogrammed the terminally differentiated fibroblast to an iPSC via introduction of four transcription factors, the so called Yamanaka factors; Sox2, Oct3/4, Klf4 and c-Myc (39,41). Similar to ESCs, iPSCs exhibit a high degree of pluripotency, with the additional benefit of circumventing the ethical concerns regarding the use of ESCs.

Despite its initial promise as a potential substitute for ESCs however, the transition to iPSC research for clinical applications highlighted several obstacles inherent to the use of iPSCs for cellular therapy, which includes genomic instability, immunogenicity, teratoma formation and clonal variations amongst iPSCs derived from the same donor cells, thus raising major concerns over the safety of their use clinically (40,42,43).

Adult stem cells

Adult stem cells are usually found in differentiated cells of specific tissues after birth, and are further categorized into hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) (44). The regeneration of the damaged tissues using adult stem cells was greeted as a breakthrough in relatively recent years, and has exhibited encouraging outcomes when used for treatment of several chronic degenerative conditions, such as degenerative disc disease, Parkinson's disease and amyotrophic lateral sclerosis. Numerous studies have demonstrated the safety and efficacy of adult stem cells for the treatment of several diseases (45-47).

HSCs

HSCs are the building blocks for the production of blood cells, including the erythrocyte and leukocyte lineages, as well as platelets (48,49). HSCs are found in the bone marrow and the umbilical cord, and are now primarily used for the treatment of the majority of disorders of blood cells, including primary immune deficiencies, congenital cytopenia, and storage and metabolic disorders (50,51). The transplantation of HSCs has been shown to ameliorate bone lesions, a decline in cognitive and central nervous function, as well as improving the survival of children diagnosed with Hurler's syndrome (the most severe form of mucopolysaccharidoses) (52). Furthermore, the use of HSCs has been deemed to be curative in the treatment of sickle cell diseases (53). There have been attempts to assess the efficacy of HSCs for the treatment of OA, wherein a study by Abdelmoaty et al (54) showed that patients who received repeated injections of autologous peripheral blood stem cells experienced improvements in physical function as well as improved articular cartilage quality. At present, the only FDA-approved stem cell products consist of hematopoietic progenitor cells that are derived from umbilical cord blood, solely for the treatment of blood disorders involving the hematopoietic system (55).

MSCs

MSCs are multipotent stem cells that are found ubiquitously throughout the musculoskeletal system in the human body, and can differentiate into various cell types including, but not limited to, osteoblasts, chondrocytes, adipocytes, astrocytes and cardiomyocytes (56-61). They can be isolated from the HSCs based on their ability to adhere readily to the plastic surfaces of tissue culture plates (56). MSCs can be derived from various tissues in the body, including bone marrow, adipose tissue, synovium, umbilical cord blood, dental pulp, amniotic fluid, dermis and peripheral blood (62-67). The characterization of MSCs are defined based on a guideline proposed by the International Society for Cellular Therapy (ISCT); the minimum criteria being that cells exhibit expression of specific markers, including CD44, CD90 and CD105, but lack expression of CD34, DC45 and CD133, as these are markers of HSCs (68). The revelation of the intrinsic nature of MSCs to regenerate and differentiate into chondrocytes has increased interest in the investigation of MSCs, and they show potential as an excellent alternative treatment option for OA. One study that used bone marrow aspirates in treating knee OA has successfully entered clinical trial phase 4, and was registered in ClinicalTrials.gov (Identifier, NCT03289416). In this clinical trial, undifferentiated cells found in the bone marrow aspirate concentrate (BMA) were shown to promote healing of damaged tissue, and aid in growth, repair and tissue regeneration. Whereas the full benefits of BMA remain to be elucidated, studies have shown that this treatment can relieve pain, and improve healing in articular cartilage and bone grafts (69). Although MSCs can be derived from numerous tissues in the body as mentioned previously, the two types of MSCs that are widely studied for the treatment of OA are bone marrow-derived MSCs (BMSCs) and adipose-derived stem cells (ASCs) (70,71).

4. Types of MSC therapy for the treatment of OA

BMSCs

BMSCs are a population of fibroblast-like cells that reside in the stroma of the bone marrow (66). These MSCs were initially isolated from the bone marrow aspirate of the iliac crest, before the subsequent emergence of MSCs derived from other tissues, such as adipose tissue, umbilical cord and amniotic fluid (72,73). Currently, BMSCs are regarded as the gold standard and remain the most frequently investigated cell type, as they are hypothesized to possess higher potential for chondrogenic differentiation (70). There are 58 registered clinical trials in Clinicaltrials.gov (as of March 2021), on the use of BMSCs for treatment of osteoarthritis using the key words ‘osteoarthritis’ and ‘bone marrow stem cells’. However, only a handful of studies have been completed and have published their results (74-81).

ASCs

ASCs are stem cells isolated from adipose tissues. They were first identified as an MSC in 2001(82), following which, they have been widely studied for their potential therapeutic value in regenerative medicine and tissue engineering (63,83-85). Although ASCs exhibit similarities to the BMSCs, there are several distinct characteristics between these 2 types of stem cells, such as their differentiation potential and the complement of cell surface markers. BMSCs express CD106 (a marker that is involved in MSCs-mediated immunosuppression and the binding of hematopoietic progenitor cells) which was found to be absent on ASCs (86,87) and the ASCs express CD49d (α4 integrin that is involved in facilitating leukocyte migration) which was not detected on BMSCs (86). Unlike BMSCs, ASCs can be obtained in high yields from adipose tissues, which can be found abundantly in the body (84). It has been estimated that MSCs account for 0.001-0.004% of the bone marrow aspirate cells, whereas ASCs account for ~2% of the lipoaspirate cells (84). The isolation of ASCs can be performed via liposuction aspirates or from subcutaneous adipose tissue fragments, and is less invasive compared to BMSCs (63). As of March 2021, 52 registered clinical studies were found on Clinicaltrials.gov using the key words ‘osteoarthritis’ and ‘adipose stem cells’.

5. General advantages of MSCs over other types of stem cells for therapeutic purposes

MSCs are favored over the other types of stem cells, as they exhibit numerous advantages for therapeutic purposes, such as their relative abundance, ease of isolation, their multilineage differential potential, lower risk of malignant transformation, immunomodulatory properties and the lack of ethical issues.

Abundance and ease of isolation

Previous reports have suggested that MSCs originate from the perivascular niche, thus making it possible to isolate them from various tissues in the body such as bone marrow, adipose tissue, peripheral blood, the placenta and the umbilical cord (72,73). However, the bone marrow and subcutaneous adipose tissues remain the preferential sources of obtaining MSCs, due to their relative abundance in the human body, particularly in the subcutaneous adipose tissue (88).

Multilineage differential potential

MSCs can be differentiated into various cell lineages. Over the years, in addition to the production of osteocytes, chondrocytes and adipocytes from MSCs, studies have also successfully induced MSCs to differentiate into oligodendrocytes (89-91), insulin-producing cells (71,92,93) and cardiomyocytes (94), highlighting their potential for the treatment of various degenerative diseases, including diabetes mellitus (95), cardiovascular diseases (96,97) and bone diseases (98).

Lower risk of malignant transformation

MSCs are endogenously programmed to exhibit limited proliferation capacity in cultures, after which cells enter a state of senescence, preventing their ability to divide; this is termed the ‘Hayflick limit’ (99). Senescence is defined as a stress response that results in the arrest of cell proliferation, thus preventing the propagation of damaged cells and lowering the risk of malignant transformation in the body (100).

Immunomodulatory properties

Another characteristic of MSCs that contributes to the advantage of using MSCs is that they have been shown to exhibit immunomodulatory properties, wherein they secrete anti-inflammatory cytokines to suppress both the adaptive and innate immune responses, thus permitting their use as universal donor cells without the need for immunosuppressants (101-103). This is due to the presence of unique surface markers that permit the MSCs to remain undetected by the immune system, including the lack of expression of major histocompatibility complex class II and co-stimulatory cluster of differentiation (CD) molecules such as CD40 ligand, CD40, CD80 and CD86 (104-107). This highlights the possibility for the use of allogeneic MSCs to treat patients who do not meet the criteria for autologous stem cell therapy.

Lack of ethical issues

Unlike ESCs, MSCs can be derived from various tissues in the body and hence, the ethical concerns associated with ESCs do not apply to MSCs. Although ESCs have received significant interest due to their high-degree of pluripotency, the use of ESCs in clinical applications remains controversial due to the safety concerns over teratoma formation, as well as the ethical issues with regard to sourcing (108,109).

6. MSC therapy offers analgesic, chondroprotective and regenerative properties when used for the treatment of OA

Studies on MSC therapy for OA that have been performed globally to evaluate their safety and efficacy, ranging from proof-of-concept studies to randomized controlled clinical trials, and have yielded positive results. Additionally, there are no studies showing notable side effects of the use of BMSCs and ASCs, and they have seen progressive improvements when used to reduce pain, physical function, stabilization of cartilage defects and even the thickening of articular cartilage in patients with OA. Table I summarizes the randomized controlled trials of BMSCs and ASCs as a treatment for OA performed between 2010 and 2020. The general procedures used are as shown in Fig. 1. Collectively, the studies highlight their beneficial properties, exhibiting analgesic, chondroprotective and anatomical regenerative properties.

Table I

Summary of randomized clinical trials of BMSCs and ASCs for the treatment of knee osteoarthritis between 2010 and 2020.

Table I

Summary of randomized clinical trials of BMSCs and ASCs for the treatment of knee osteoarthritis between 2010 and 2020.

Author, yearMSC source/typeType of studyExperimental designInterventionMeasurementOutcomes(Refs.)
Lamo-Espinosa et al, 2016Autologous BMSCCase series (n=32). Final follow-up, 12 monthsMulticenter, randomized, controlled, phase I/II clinical trialAdministration, Single intra-articular injection of MSCs. Cell dose, 1x107 cells (low-dose), and 1x108 cells (high-dose).VAS, WOMAC, X-ray and MRIPain reduction at 3 months. The group of patients injected with 1x108 cells exhibited the best response to MSC treatment, which reduced pain and preserved knee joint space at month 12.(75)
Bastos et al, 2018Autologous BMSCCase series (n=18). Final follow up, 12 monthsDouble-blinded, randomized, phase I clinical trialAdministration, Single intra-articular injection of MSCs vs. MSCs/PRP. Cell dose, Not mentionedKOOSMSCs alone or in combination with PRP were safe and resulted in significant clinical improvement in patients with knee OA for up to 12 months.(136)
Bastos et al, 2020Autologous BMSCCase series (n=47). Final follow up, 12 monthsDouble-blinded, randomized, controlled clinical trialAdministration, Single intra-articular injection of MSCs vs. MSCs/PRP vs. Corticosteroid. Cell dose, 4x107 cellsKOOS and Knee ROMMSCs/PRP groups showed the highest percentages of improvement for the KOOS domains and global score.(137)
Emadedin et al, 2018Autologous BMSCCase series (n=47). Final follow up, 6 monthsTriple-blinded, randomized, placebo-controlled phase I/II clinical trialAdministration, Single intra-articular injection of MSCs. Cell dose, 4x107 cellsVAS and WOMACSignificantly reduced pain in 6 months.(138)
Hernigou et al, 2018Autologous BMSCCase series (n=30). Final follow up, 12 yearsProspective, randomized, controlled trialAdministration, Single intra-articular injection of MSCs. Cell dose, 40 ml of bone marrow graft with an average of 6.5x103 MSCs/ml.Knee society score, radiograph and MRIReduction in pain, increase in cartilage volume, and reduction in bone marrow lesions in patients who received MSC treatment.(139)
Vega et al, 2015Allogeneic BMSCCase series (n=30). Final follow up, 12 monthsMulticentered, randomized, controlled phase I/II clinical trial.Administration, Single intra-articular injection of MSCs. Cell dose, 4x107 cells.VAS, WOMAC, Lesquesne algofunctional indices, SF-12 and MRIAllogeneic BMSC transplantation is safe and feasible with no major adverse outcomes; provides pain relief and significantly improves cartilage quality.(77)
Gupta et al, 2016Allogeneic BMSCCase series (n=60). Final follow up, 12 monthsRandomized, double-blind, multicentric, placebo-controlled, phase II studyAdministration, Single intra-articular injection of MSCs Cell dose, 2.5x107 cells, 5x107, 7.5x107, and 1.5x108 cellsVAS, WOMAC, ICOAP, X-ray and MRIMaximum symptoms reduction in patients treated with 25x106 cells, at 12 months. However, no improvements in the morphology of cartilage were observed in any of the groups of patients.(140)
Song et al, 2018Autologous ASCCase series (n=18). Final follow up, 96 weeksDouble-blinded, randomized, phase I/IIa clinical trialAdministration, Repeated intra-articular injection of MSCs. Phase l, 1x107 cells (low-dose), 2x107 cells (mid-dose), and 5x107 cells (high-dose), repeated; Phase II, 5x107 cells (high-dose)WOMAC, NRS-11, SF-36 and MRISignificant reduction in pain and increase in volume of cartilage at week 48 onwards, particularly in the high dose group. Furthermore, the cartilage increased to a higher degree after the third injection.(141)
Freitag et al, 2019Autologous ASCCase series (n=30). Final follow up, 12 monthsUnblinded randomized controlled trialAdministration, Single or two intra-articular injections of MSCs. Cell dose, 1x107 cellsWOMAC, KOOS, NPRS and MRISignificant reduction in pain and functional improvements in all patients who received the treatment. OA did not progress by the time of the final follow-up period in 89% of the patients who received two-injections.(85)
Lee et al, 2019Autologous ASCsCase series (n=24). Final follow up, 6 monthsDouble-blinded, randomized, placebo-controlled phase IIb clinical trialAdministration, Single intra-articular injections of MSCs. Cell dose, 1x108 cellsVAS, WOMAC, KOOS, physical examination and MRIReduced pain and preserved size of cartilage defect of medial femoral condyle over 6 months.(142)

[i] MSC, Mesenchymal stem cell; BMSC, bone marrow-derived stem cell; ASC, adipose-derived stem cell; PRP, platelet-rich plasma; IL, interleukin; OA, osteoarthritis; TKA, total knee arthroplasty; MRI, magnetic resonance imaging; VAS, Visual Analogue Scale; WOMAC, The Western Ontario and McMaster Universities osteoarthritis index; KOOS, Knee Injury and Osteoarthritis Outcome score; Knee ROM, Knee range of motion; SF-12, 12-item Short Form health survey; SF-36, 36-item Short Form health survey; WORMS, Whole Organ Magnetic Resonance Imaging Score; NRS-11, The 11-point Numerical Rating Scale; ICOAP, The Measure of Intermittent and Constant Osteoarthritis Pain; NPRS, Numeric Pain Rating Scale

Analgesic and chondroprotective effects

Although the specific molecular mechanisms by which MSCs exert an analgesic effect remain to be elucidated, it is hypothesized that they revolve around its immunomodulatory effects, by inducing the synthesis of anti-inflammatory cytokines, such as IL-10, and downregulating the production of pro-inflammatory cytokines, such as IL-1, IL-6, TNFα and interferon (IFN)-γ (110-114). MSCs are able to secrete numerous soluble growth factors and cytokines, including TNFα-stimulated gene/protein 6 (TSG-6), prostaglandin E2 (PGE-2) and indoleamine 2,3-dioxygenase (IDO), which contribute to their ability to mitigate pain (103,115-117). It has been shown that the presence of MSCs results in the production of TSG-6, leading to inhibition of the toll-like receptors-2/nuclear factor κ-light-chain-enhancer of activated B cells signaling pathway, followed by the subsequent downregulation of inflammatory mediators, such as nitric oxide, TNFα and IL-1 (103,115). Upregulation of PGE-2 by MSCs leads to inhibition of the IFN-γ, inducing the differentiation of M1-type macrophages to M2-type macrophages (116). Similarly, the production of IDO by MSCs also promotes the conversion of M1 macrophages to M2 macrophages (117).

The homing migratory ability of MSCs to sites of injury and inflammation provides beneficial relief in both BMSC and saline-treated knee in patients with bilateral OA (118-120).

Chondrocyte regenerative effect

The regenerative effect of MSCs on the cartilage has been shown to involve the high expression of several genes responsible for inducing chondrogenesis, and the subsequent development of normal cartilage. These genes include the production of thrombospondin-2, which promotes the Notch signaling pathway; the production of bone morphogenetic protein 2, which induces the SMAD signaling pathway; and the Indian hedgehog signaling pathway, which promotes the expression of SOX9 followed by increased expression of the Col2a1 gene, thus stimulating the production of proteoglycans and type II collagen, all of which are involved in cartilage regeneration (121-123). The likely mechanism by which MSCs exert their anti-inflammatory and cartilage regenerative effects is summarized in Fig. 2.

Several studies have highlighted the analgesic effects of MSCs is addition to their cartilage regenerative effects; however, follow-up is usually limited to 6-12 months post-treatment. The long-term potential of MSC therapy may thus be underrated, as the structural changes required for a prominent effect take at least a year to occur (79,124).

In general, numerous reports have shown that as little as a single injection of a 1x106 cell dose is sufficient for initiation of their analgesic effects, although eliciting chondrocyte regeneration response in cartilage requires a much higher dose of at least 1x108 cells (81,125). Hence, higher quality randomized, controlled clinical studies with larger cohorts are required to strengthen the evidence and evaluate the quality of its therapeutic outcomes. Additionally, established protocols for consideration of the optimal dose, time of intervention, method of delivery and safety precautions also require extensive study.

7. Challenges involved in the use of MSC therapy for OA

Despite the promising aspects for the use of MSCs clinically, this approach is considered relatively new and is surrounded by challenges involved in its use for OA, and these are discussed below.

Effects of the donor's health condition and age on stem cell properties

The differentiation potential and proliferation capacity of the MSCs are known to be affected by patients requiring autologous MSC therapy. Certain metabolic conditions, such as diabetes and obesity, can generate microenvironmental cues that predispose the differentiation potential of MSCs towards adipocyte differentiation rather than chondrocyte differentiation (126-128). Besides, the proliferative capacity of MSCs decreased with age (126).

Instability of chondrocyte-like phenotypes

One of the major challenges accompanying the use of MSCs for treatment of OA is the unsustainable cellular and hyaline cartilage phenotype of differentiated chondrocytes. Previous studies have documented the possible involvement of these cells in the development of heterotopic ossification, a process where bone formation occurs in non-skeletal tissues (129-132). These studies reported on the transient secretion of type II collagen from the MSCs, followed by the up-regulated expression of collagen type X, matrix metalloproteinase and alkaline phosphatase activity, thus indicating a shift from the chondrogenic to a hypertrophic phenotype that precedes osteogenesis, a phenomenon that does not normally occur amongst the chondrocytes found in the hyaline cartilage in the joints.

Limited replicative lifespan

As mentioned earlier, the MSCs will enter a state of senescence whereby they lose their ability to proliferate. This is a double-edged characteristic, as it may lower the risk of malignant tumors, but limit the therapeutic use of stem cells. It was documented that the MSCs exhibited abnormalities in the morphology of the cells, such as enlargement, reduced expression of specific surface markers, and finally senescence as they reached higher passage numbers (133,134). The entry into a state of senescence in MSCs was shown to affect the differential potential of the cells, the immunomodulation capability and their migratory ability (133). Although cryopreservation of MSCs can be used to address this issue, the process comes with its own challenges; a decrease in viability, colony forming units and integrin expression were observed after cryopreservation and thawing of the cells (135).

Technical challenges

MSC therapy requires highly skilled professionals, as the culturing of MSCs must be performed with utmost care to prevent contamination of the cells. Additionally, considerably more research is required in order to establish a clear protocol for the isolation, expansion, differentiation and pre-conditioning of MSCs, and to determine the appropriate concentration of MSCs for use in patients with OA.

Social

While stem cell therapy may offer an attractive option for treatment of currently uncurable diseases, the costs are currently considerably high, owing to the need to cover the cost of the individual harvesting, isolation, and expansion of cells in a sterile facility. Additionally, the increasing popularity of stem cell therapy has warranted the use widespread biobanking, and an increased access to the various highly multipotent stem cells, which could be at risk of exploitation. Hence, policies on biobanking of stem cells must be regulated to address the possible issues regarding its usage, control as well as patients' consent.

8. Conclusions

The emergence of stem cell-based therapies has brought about novel avenues to address the, as of yet, unmet curative treatment for various degenerative disorders, including OA. The multipotency of the MSCs, along with its self-renewal and immunomodulatory properties, availability and ease of isolation highlight the potential use of MSCs for cellular therapeutic approaches in OA, and promising results have been demonstrated in various pre-clinical and clinical trials. However, several challenges are involved in this process, and this requires standardized solutions before they can be recommended clinically. Efforts on investigating and establishing protocols to increase the stability of the chondrocyte-like phenotype of the MSCs is required to raise the success rates of MSC-based treatment in patients with OA, and to lower the cost. In addition, the appropriate concentration of stem cells for specific treatments, and the long-term follow-ups of patients with OA treated with MSCs should be performed to investigate the long-term safety and efficacy of MSC-based therapy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

SJQL and NKW both wrote and revised the article. Both authors read and approved the final manuscript. Data sharing is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

National Clinical Guideline Centre (UK): Osteoarthritis-Care and Μanagement in Αdults. London: National Institute for Health and Care Excellence (UK), 2014.

2 

James SL, Abate D, Hassen Abate K, Abay SM, Abbafati C, Abbasi N, Abbastabar H, Abd-Allah F, Abdela J, Abdelalim A, et al: Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 392:1789–1858. 2018.PubMed/NCBI View Article : Google Scholar

3 

Blagojevic M, Jinks C, Jeffery A and Jordan KP: Risk factors for onset of osteoarthritis of the knee in older adults: A systemic review and meta-analysis. Osteoarthritis Cartilage. 18:24–33. 2010.PubMed/NCBI View Article : Google Scholar

4 

Loeser RF: Molecular mechanisms of cartilage destruction: Mechanics, inflammatory mediators, and aging collide. Arthritis Rheum. 54:1357–1360. 2006.PubMed/NCBI View Article : Google Scholar

5 

Ng HY, Alvin Lee KX and Shen KX: Articular cartilage: Structure, composition, injuries and repair. JSM Bone Jt Dis. 1(1010)2017.

6 

Ostrowska M, Maśliński W, Prochorec-Sobieszek M, Nieciecki M and Sudoł-Szopińska I: Cartilage and bone damage in rheumatoid arthritis. Reumatologia. 56:111–120. 2018.PubMed/NCBI View Article : Google Scholar

7 

Mahajan A, Verma S and Tandon V: Osteoarthritis. J Assoc Physicians India. 53:634–641. 2005.PubMed/NCBI

8 

Houard X, Goldring MB and Berenbaum F: Homeostatic mechanisms in articular cartilage and role of inflammation in osteoarthritis. Curr Rheumatol Rep. 15(375)2013.PubMed/NCBI View Article : Google Scholar

9 

Gignac MAM, Irvin E, Cullen K, Van Eerd D, Beaton DE, Mahood Q, McLeod C and Backman CL: Men and women's occupational activities and the risk of developing osteoarthritis of the knee, hip, or hands: A systematic review and recommendations for future research. Arthritis Care Res (Hoboken). 72:378–396. 2020.PubMed/NCBI View Article : Google Scholar

10 

Michael JW, Schlüter-Brust KU and Eysel P: The epidemiology, etiology, diagnosis, and treatment of osteoarthritis of the knee. Dtsch Arztebl Int. 107:152–162. 2010.PubMed/NCBI View Article : Google Scholar

11 

Helliwell P: Osteoarthritis and Paget's disease. Br J Rheumatol. 34:1061–1063. 1995.PubMed/NCBI View Article : Google Scholar

12 

Mora JC, Przkora R and Cruz-Almeida Y: Knee osteoarthritis: Pathophysiology and current treatment modalities. J Pain Res. 11:2189–2196. 2018.PubMed/NCBI View Article : Google Scholar

13 

de l'Escalopier N, Anract P and Biau D: Surgical treatments for osteoarthritis. Ann Phys Rehabil Med. 59:227–233. 2016.PubMed/NCBI View Article : Google Scholar

14 

Øiestad BE, Juhl CB, Eitzen I and Thorlund JB: Knee extensor muscle weakness is a risk factor for development of knee osteoarthritis. A systematic review and meta-analysis. Osteoarthritis Cartilage. 23:171–177. 2015.PubMed/NCBI View Article : Google Scholar

15 

March LM and Bagga H: Epidemiology of osteoarthritis in Australia. Med J Aust. 180:S6–S10. 2004.PubMed/NCBI View Article : Google Scholar

16 

Messier SP: Obesity and osteoarthritis: Disease genesis and nonpharmacologic weight management. Rheum Dis Clin North Am. 34:713–729. 2008.PubMed/NCBI View Article : Google Scholar

17 

Vincent KR and Vincent HK: Resistance exercise for knee osteoarthritis. PM R. 4 (Suppl 5):S45–S52. 2012.PubMed/NCBI View Article : Google Scholar

18 

Rogers MW and Wilder FV: The effects of strength training among persons with hand osteoarthritis: A two-year follow-up study. J Hand Ther. 20:244–249; quiz 250. 2007.PubMed/NCBI View Article : Google Scholar

19 

Latham N and Liu CJ: Strength training in older adults: The benefits for osteoarthritis. Clin Geriatr Med. 26:445–459. 2010.PubMed/NCBI View Article : Google Scholar

20 

Aguiar GC, Rocha SG, da Silva Rezende GA, do Nascimento MR and Scalzo PL: Effects of resistance training in individuals with knee osteoarthritis. Fisioter Mov. 29:589–596. 2016.

21 

Thoumie P, Marty M, Avouac B, Pallez A, Vaumousse A, Pipet LPT, Monroche A, Graveleau N, Bonnin A, Amor CB and Coudeyre E: Effect of unloading brace treatment on pain and function in patients with symptomatic knee osteoarthritis: The ROTOR randomized clinical trial. Sci Rep. 8(10519)2018.PubMed/NCBI View Article : Google Scholar

22 

Chew KT, Lew HL, Date E and Fredericson M: Current evidence and clinical applications of therapeutic knee braces. Am J Phys Med Rehabil. 86:678–686. 2007.PubMed/NCBI View Article : Google Scholar

23 

Lee PY, Winfield TG, Harris SR, Storey E and Chandratreya A: Unloading knee brace is a cost-effective method to bridge and delay surgery in unicompartmental knee arthritis. BMJ Open Sport Exerc Med. 2(e000195)2017.PubMed/NCBI View Article : Google Scholar

24 

Ostrander RV, Leddon CE, Hackel JG, O'Grady CP and Roth CA: Efficacy of unloader bracing in reducing symptoms of knee osteoarthritis. Am J Orthop (Belle Mead NJ). 45:306–311. 2016.PubMed/NCBI

25 

Wilson B, Rankin H and Barnes CL: Long-term results of an unloader brace in patients with unicompartmental knee osteoarthritis. Orthopedics. 34:e334–e337. 2011.PubMed/NCBI View Article : Google Scholar

26 

Yu SP, Williams M, Eyles JP, Chen JS, Makovey J and Hunter DJ: Effectiveness of knee bracing in osteoarthritis: Pragmatic trial in a multidisciplinary clinic. Int J Rheum Dis. 19:279–286. 2016.PubMed/NCBI View Article : Google Scholar

27 

Squyer E, Stamper DL, Hamilton DT, Sabin JA and Leopold SS: Unloader knee braces for osteoarthritis: Do patients actually wear them? Clin Orthop Relat Res. 471:1982–1991. 2013.PubMed/NCBI View Article : Google Scholar

28 

Nejati P, Farzinmehr A and Moradi-Lakeh M: The effect of exercise therapy on knee osteoarthritis: A randomized clinical trial. Med J Islam Repub Iran. 29(186)2015.PubMed/NCBI

29 

Reid MC, Eccleston C and Pillemer K: Management of chronic pain in older adults. BMJ. 350(h532)2015.PubMed/NCBI View Article : Google Scholar

30 

Rönn K, Reischl N, Gautier E and Jacobi M: Current surgical treatment of knee osteoarthritis. Arthritis. 2011(454873)2011.PubMed/NCBI View Article : Google Scholar

31 

Wang X, Wanyan P, Wang JM, Tian JH, Hu L, Shen XP and Yang KH: A randomized, controlled trial to assess the efficacy of arthroscopic debridement in combination with oral medication versus oral medication in patients with gouty knee arthritis. Indian J Surg. 77 (Suppl 2):S628–S634. 2015.PubMed/NCBI View Article : Google Scholar

32 

Kirkley A, Birmingham TB, Litchfield RB, Giffin JR, Willits KR, Wong CJ, Feagan BG, Donner A, Griffin SH, D'Ascanio LM, et al: A randomized trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 359:1097–1107. 2008.PubMed/NCBI View Article : Google Scholar

33 

Moseley JB, O'Malley K, Petersen NJ, Menke TJ, Brody BA, Kuykendall DH, Hollingsworth JC, Ashton CM and Wray NP: A controlled trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 347:81–88. 2002.PubMed/NCBI View Article : Google Scholar

34 

Choi HG, Kwon BC, Kim JI and Lee JK: Total knee arthroplasty reduces the risk of mortality in osteoarthritis patients up to 12 years: A Korean national cohort longitudinal follow-up study. J Orthop Surg (Hong Kong). 28(2309499020902589)2020.PubMed/NCBI View Article : Google Scholar

35 

Rajabzadeh N, Fathi E and Farahzadi R: Stem cell-based regenerative medicine. Stem Cell Investig. 6(19)2019.PubMed/NCBI View Article : Google Scholar

36 

Landry DW and Zucker HA: Embryonic death and the creation of human embryonic stem cells. J Clin Invest. 114:1184–1186. 2004.PubMed/NCBI View Article : Google Scholar

37 

Ede V and Obeagu EI: Ethical issues in human embryonic stem cell research: A christian perspective. Int J Med Sci Dent Res. 1:8–14. 2019.

38 

Lo B and Parham L: Ethical issues in stem cell research. Endocr Rev. 30:204–213. 2009.PubMed/NCBI View Article : Google Scholar

39 

Takahashi K and Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. 2006.PubMed/NCBI View Article : Google Scholar

40 

Shi Y, Inoue H, Wu JC and Yamanaka S: Induced pluripotent stem cell technology: A decade of progress. Nat Rev Drug Discov. 16:115–130. 2017.PubMed/NCBI View Article : Google Scholar

41 

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 131:861–872. 2007.PubMed/NCBI View Article : Google Scholar

42 

Gutierrez-Aranda I, Ramos-Mejia V, Bueno C, Munoz-Lopez M, Real PJ, Mácia A, Sanchez L, Ligero G, Garcia-Parez JL and Menendez P: Human induced pluripotent stem cells develop teratoma more efficiently and faster than human embryonic stem cells regardless the site of injection. Stem Cells. 28:1568–1570. 2010.PubMed/NCBI View Article : Google Scholar

43 

Zhang M, Wang L, An K, Cai J, Li G, Yang C, Liu H, Du F, Han X, Zhang Z, et al: Lower genomic stability of induced pluripotent stem cells reflects increased non-homologous end joining. Cancer Commun (Lond). 38(49)2018.PubMed/NCBI View Article : Google Scholar

44 

Christensen R and Serakinci N: Adult Stem Cells. In: Encyclopedia of Cancer. Springer, Heidelberg, pp1-5, 2015.

45 

Centeno C, Markle J, Dodson E, Stemper I, Williams CJ, Hyzy M, Ichim T and Freeman M: Treatment of lumbar degenerative disc disease-associated radicular pain with culture-expanded autologous mesenchymal stem cells: A pilot study on safety and efficacy. J Transl Med. 15(197)2017.PubMed/NCBI View Article : Google Scholar

46 

Berry JD, Cudkowicz ME, Windebank AJ, Staff NP, Owegi M, Nicholson K, McKenna-Yasek D, Levy YS, Abramov N, Kaspi H, et al: NurOwn, phase 2, randomized, clinical trial in patients with ALS: Safety, clinical, and biomarker results. Neurology. 93:e2294–e2305. 2019.PubMed/NCBI View Article : Google Scholar

47 

Riecke J, Johns KM, Cai C, Vahidy FS, Parsha K, Furr-Stimming E, Schiess M and Savitz SI: A meta-analysis of mesenchymal stem cells in animal models of Parkinson's disease. Stem Cells Dev. 24:2082–2090. 2015.PubMed/NCBI View Article : Google Scholar

48 

Ng AP and Alexander WS: Haematopoietic stem cells: Past, present and future. Cell Death Discov. 3(17002)2017.PubMed/NCBI View Article : Google Scholar

49 

Yamamoto R, Wilkinson AC and Nakauchi H: Changing concepts in hematopoietic stem cells. Science. 362:895–896. 2018.PubMed/NCBI View Article : Google Scholar

50 

Domen J, Wagers A and Weissman IL: Bone marrow (Hematopoietic) stem cells. Stem Cell Information: The National Institutes of Health resource for stem cell research: 13-34, 2006.

51 

Morgan RA, Gray D, Lomova A and Kohn DB: Hematopoietic stem cell gene therapy: Progress and lessons learned. Cell Stem Cell. 21:574–590. 2017.PubMed/NCBI View Article : Google Scholar

52 

Taylor M, Khan S, Stapleton M, Wang J, Chen J, Wynn R, Yabe H, Chinen Y, Boelens JJ, Mason RW, et al: Hematopoietic stem cell transplantation for mucopolysaccharidoses: Past, present, and future. Biol Blood Marrow Transplant. 25:e226–e246. 2019.PubMed/NCBI View Article : Google Scholar

53 

Walters MC: Update of hematopoietic cell transplantation for sickle cell disease. Curr Opin Hematol. 22:227–233. 2015.PubMed/NCBI View Article : Google Scholar

54 

Abdelmoaty N, Alattar E, Ahmed K, Ibrahim Y and Darwish B: Regenerative power of autologous peripheral blood stem cell injection in knee osteoarthritis by a non-invasive approach-MRI Study. Ann Rheum Dis. 73(1066)2014.

55 

U.S Food and Drug Administration: Approved Cellular and Gene Therapy Products, 2021.

56 

Prockop DJ: Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 276:71–74. 1997.PubMed/NCBI View Article : Google Scholar

57 

Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, et al: Cardiomyocytes can be generated from marrow stromal cells in vitro. J Clin Invest. 103:697–705. 1999.PubMed/NCBI View Article : Google Scholar

58 

Wakitani S, Saito T and Caplan AI: Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve. 18:1417–1426. 1995.PubMed/NCBI View Article : Google Scholar

59 

Nuttall ME, Patton AJ, Olivera DL, Nadeau DP and Gowen M: Human trabecular bone cells are able to express both osteoblastic and adipocytic phenotype: Implications for osteopenic disorders. J Bone Miner Res. 13:371–382. 1998.PubMed/NCBI View Article : Google Scholar

60 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999.PubMed/NCBI View Article : Google Scholar

61 

Bianco P, Robey PG and Simmons PJ: Mesenchymal stem cells: Revisiting history, concepts, and assays. Cell Stem Cell. 2:313–319. 2008.PubMed/NCBI View Article : Google Scholar

62 

Roufosse CA, Direkze NC, Otto WR and Wright NA: Circulating mesenchymal stem cells. Int J Biochem Cell Biol. 36:585–597. 2004.PubMed/NCBI View Article : Google Scholar

63 

Lindroos B, Suuronen R and Miettinen S: The potential of adipose stem cells in regenerative medicine. Stem Cell Rev Rep. 7:269–291. 2011.PubMed/NCBI View Article : Google Scholar

64 

Erices A, Conget P and Minguell JJ: Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 109:235–242. 2000.PubMed/NCBI View Article : Google Scholar

65 

Gronthos S, Mankani M, Brahim J, Robey PG and Shi S: Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 97:13625–13630. 2000.PubMed/NCBI View Article : Google Scholar

66 

Haniffa MA, Wang XN, Holtick U, Rae M, Isaacs JD, Dickinson AM, Hilkens CM and Collin MP: Adult human fibroblasts are potent immunoregulatory cells and functionally equivalent to mesenchymal stem cells. J Immunol. 179:1595–1604. 2007.PubMed/NCBI View Article : Google Scholar

67 

Sessarego N, Parodi A, Podesta M, Benvenuto F, Mogni M, Raviolo V, Lituania M, Kunkl A, Ferlazzo G, Bricarelli FD, et al: Multipotent mesenchymal stromal cells from amniotic fluid: Solid perspectives for clinical application. Haematologica. 93:339–346. 2008.PubMed/NCBI View Article : Google Scholar

68 

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop DJ and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International society for cellular therapy position statement. Cytotherapy. 8:315–317. 2006.PubMed/NCBI View Article : Google Scholar

69 

Gianakos AL, Sun L, Patel JN, Adams DM and Liporace FA: Clinical application of concentrated bone marrow aspirate in orthopaedics: A systematic review. World J Orthop. 8:491–506. 2017.PubMed/NCBI View Article : Google Scholar

70 

Mohamed-Ahmed S, Fristad I, Lie SA, Suliman S, Mustafa K, Vindenes H and Idris SB: Adipose-derived and bone marrow mesenchymal stem cells: A donor-matched comparison. Stem Cell Res Ther. 9(168)2018.PubMed/NCBI View Article : Google Scholar

71 

Gabr MM, Zakaria MM, Refaie AF, Abdel-Rahman EA, Reda AM, Ali SS, Khater SM, Ashamallah SA, Ismail AM, Ismail HEA, et al: From human mesenchymal stem cells to insulin-producing cells: Comparison between bone marrow- and adipose tissue-derived cells. Biomed Res Int. 2017(3854232)2017.PubMed/NCBI View Article : Google Scholar

72 

da Silva Meirelles L, Chagastelles PC and Nardi NB: Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 119:2204–2213. 2006.PubMed/NCBI View Article : Google Scholar

73 

Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, et al: A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 3:301–313. 2008.PubMed/NCBI View Article : Google Scholar

74 

Orozco L, Munar A, Soler R, Alberca M, Soler F, Huguet M, Sentís J, Sánchez A and García-Sancho J: Treatment of knee osteoarthritis with autologous mesenchymal stem cells: A pilot study. Transplantation. 95:1535–1541. 2013.PubMed/NCBI View Article : Google Scholar

75 

Lamo-Espinosa JM, Mora G, Blanco JF, Granero-Moltó F, Nuñez-Córdoba JM, Sánchez-Echenique C, Bondía JM, Aquerreta JD, Andreu EJ, Ornilla E, et al: Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: Multicenter randomized controlled clinical trial (phase I/II). J Transl Med. 14(246)2016.PubMed/NCBI View Article : Google Scholar

76 

Garay-Mendoza D, Villarreal-Martínez L, Garza-Bedolla A, Pérez-Garza DM, Acosta-Olivo C, Vilchez-Cavazos F, Diaz-Hutchinson C, Gómez-Almaguer D, Jaime-Pérez JC and Mancías-Guerra C: The effect of intra-articular injection of autologous bone marrow stem cells on pain and knee function in patients with osteoarthritis. Int J Rheum Dis. 21:140–147. 2018.PubMed/NCBI View Article : Google Scholar

77 

Vega A, Martín-Ferrero MA, Del Canto F, Alberca M, García V, Munar A, Orozco L, Soler R, Fuertes JJ, Huguet M, et al: Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: A randomized controlled trial. Transplantation. 99:1681–1690. 2015.PubMed/NCBI View Article : Google Scholar

78 

Shadmanfar S, Labibzadeh N, Emadedin M, Jaroughi N, Azimian V, Mardpour S, Kakroodi FA, Bolurieh T, Hosseini SE, Chehrazi M, et al: Intra-articular knee implantation of autologous bone marrow-derived mesenchymal stromal cells in rheumatoid arthritis patients with knee involvement: Results of a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy. 20:499–506. 2018.PubMed/NCBI View Article : Google Scholar

79 

Al-Najar M, Khalil H, Al-Ajlouni J, Al-Antary E, Hamdan M, Rahmeh R, Alhattab D, Samara O, Yasin M, Abdullah AA, et al: Intra-articular injection of expanded autologous bone marrow mesenchymal cells in moderate and severe knee osteoarthritis is safe: A phase I/II study. J Orthop Surg Res. 12(190)2017.PubMed/NCBI View Article : Google Scholar

80 

Anz AW, Hubbard R, Rendos NK, Everts PA, Andrews JR and Hackel JG: Bone marrow aspirate concentrate is equivalent to platelet-rich plasma for the treatment of knee osteoarthritis at 1 year: A prospective, randomized trial. Orthop J Sports Med. 2(2325967119900958)2020.PubMed/NCBI View Article : Google Scholar

81 

Chahal J, Gómez-Aristizábal A, Shestopaloff K, Bhatt S, Chaboureau A, Fazio A, Chisholm J, Weston A, Chiovitti J, Keating A, et al: Bone marrow mesenchymal stromal cell treatment in patients with osteoarthritis results in overall improvement in pain and symptoms and reduces synovial inflammation. Stem Cells Transl Med. 8:746–757. 2019.PubMed/NCBI View Article : Google Scholar

82 

Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP and Hedrick MH: Multilineage cells from human adipose tissue: Implications for cell-based therapies 2. Tissue Eng. 7:211–228. 2001.PubMed/NCBI View Article : Google Scholar

83 

Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P and Hedrick MH: Human adipose tissue is a source of multipotent stem cells. Raff M (ed). Mol Biol Cell. 13:4279–4295. 2002.PubMed/NCBI View Article : Google Scholar

84 

Coughlin RP, Oldweiler A, Mickelson DT and Moorman CT III: Adipose-derived stem cell transplant technique for degenerative joint disease. Arthrosc Tech. 6:e1761–e1766. 2017.PubMed/NCBI View Article : Google Scholar

85 

Freitag J, Bates D, Wickham J, Shah K, Huguenin L, Tenen A, Paterson K and Boyd R: Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: A randomized controlled trial. Regen Med. 14:213–230. 2019.PubMed/NCBI View Article : Google Scholar

86 

De Ugarte DA, Alfonso Z, Zuk PA, Elbarbary A, Zhu M, Ashjian P, Benhaim P, Hedrick MH and Fraser JK: Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow. Immunol Lett. 89:267–270. 2003.PubMed/NCBI View Article : Google Scholar

87 

Yang ZX, Han ZB, Ji YR, Wang YW, Liang L, Chi Y, Yang SG, Li LN, Luo WF, Li JP, et al: CD106 identifies a subpopulation of mesenchymal stem cells with unique immunomodulatory properties. PLoS One. 8(e59354)2013.PubMed/NCBI View Article : Google Scholar

88 

Krampera M, Marconi S, Pasini A, Galiè M, Rigotti G, Mosna F, Tinelli M, Lovato L, Anghileri E, Andreini A, et al: Induction of neural-like differentiation in human mesenchymal stem cells derived from bone marrow, fat, spleen and thymus. Bone. 40:382–390. 2007.PubMed/NCBI View Article : Google Scholar

89 

Kennea NL, Waddington SN, Chan J, O'Donoghue K, Yeung D, Taylor DL, Al-Allaf FA, Pirianov G, Themis M, Edwards AD, et al: Differentiation of human fetal mesenchymal stem cells into cells with an oligodendrocyte phenotype. Cell Cycle. 8:1069–1079. 2009.PubMed/NCBI View Article : Google Scholar

90 

Shah S, Yin PT, Uehara TM, Chueng ST, Yang L and Lee KB: Guiding stem cell differentiation into oligodendrocytes using graphene-nanofiber hybrid scaffolds. Adv Mater. 26:3673–3680. 2014.PubMed/NCBI View Article : Google Scholar

91 

Steffenhagen C, Dechant FX, Oberbauer E, Furtner T, Weidner N, Küry P, Aigner L and Rivera FJ: Mesenchymal stem cells prime proliferating adult neural progenitors toward an oligodendrocyte fate. Stem Cells Dev. 21:1838–1851. 2012.PubMed/NCBI View Article : Google Scholar

92 

Allahverdi A, Abroun S, Jafarian A, Soleimani M, Taghikhani M and Eskandari F: Differentiation of human mesenchymal stem cells into insulin producing cells by using a lentiviral vector carrying PDX1. Cell J. 17:231–242. 2015.PubMed/NCBI View Article : Google Scholar

93 

Chen LB, Jiang XB and Yang L: Differentiation of rat marrow mesenchymal stem cells into pancreatic islet beta-cells. World J Gastroenterol. 10:3016–3020. 2004.PubMed/NCBI View Article : Google Scholar

94 

Singh A, Singh A and Sen D: Mesenchymal stem cells in cardiac regeneration: A detailed progress report of the last 6 years (2010-2015). Stem Cell Res Ther. 7(82)2016.PubMed/NCBI View Article : Google Scholar

95 

Solis MA, Moreno Velásquez I, Correa R and Huang LLH: Stem cells as a potential therapy for diabetes mellitus: A call-to-action in Latin America. Diabetol Metab Syndr. 11(20)2019.PubMed/NCBI View Article : Google Scholar

96 

Sun R, Li X, Liu M, Zeng Y, Chen S and Zhang P: Advances in stem cell therapy for cardiovascular disease (Review). Int J Mol Med. 38:23–29. 2016.PubMed/NCBI View Article : Google Scholar

97 

Terashvili M and Bosnjak ZJ: Stem cell therapies in cardiovascular disease. J Cardiothorac Vasc Anesth. 33:209–222. 2019.PubMed/NCBI View Article : Google Scholar

98 

Paspaliaris V and Kolios G: Stem cells in osteoporosis: From biology to new therapeutic approaches. Stem Cells Int. 2019(1730978)2019.PubMed/NCBI View Article : Google Scholar

99 

Mercado-Sáenz S, Ruiz-Gómez MJ, Morales-Moreno F and Martínez-Morillo M: Cellular aging: Theories and technological influence. Brazilian Arch Biol Technol. 53:1319–1332. 2010.

100 

Shay JW and Wright WE: Role of telomeres and telomerase in cancer. Semin Cancer Biol. 21:349–353. 2011.PubMed/NCBI View Article : Google Scholar

101 

Keating A: Mesenchymal stromal cells: New directions. Cell Stem Cell. 10:709–716. 2012.PubMed/NCBI View Article : Google Scholar

102 

Le Blanc K and Mougiakakos D: Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol. 12:383–396. 2012.PubMed/NCBI View Article : Google Scholar

103 

Prockop DJ and Oh JY: Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol Ther. 20:14–20. 2012.PubMed/NCBI View Article : Google Scholar

104 

Ryan JM, Barry FP, Murphy JM and Mahon BP: Mesenchymal stem cells avoid allogeneic rejection. J Inflamm (Lond). 2(8)2005.PubMed/NCBI View Article : Google Scholar

105 

Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, Simonetti DW, Deans RJ and McIntosh KR: T cell responses to allogeneic human mesenchymal stem cells: Immunogenicity, tolerance, and suppression. J Biomed Sci. 12:47–57. 2005.PubMed/NCBI View Article : Google Scholar

106 

Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S and Gianni AM: Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 99:3838–3843. 2002.PubMed/NCBI View Article : Google Scholar

107 

Tse WT, Pendleton JD, Beyer WM, Egalka MC and Guinan EC: Suppression of allogeneic T-cell proliferation by human marrow stromal cells: Implications in transplantation. Transplantation. 75:389–397. 2003.PubMed/NCBI View Article : Google Scholar

108 

Lee H, Shamy GA, Elkabetz Y, Schofield CM, Harrsion NL, Panagiotakos G, Socci ND, Tabar V and Studer L: Directed differentiation and transplantation of human embryonic stem cell-derived motoneurons. Stem Cells. 25:1931–1939. 2007.PubMed/NCBI View Article : Google Scholar

109 

Su W, Zhou M, Zheng Y, Fan Y, Wang L, Han Z, Kong D, Zhao RC, Wu JC, Xiang R and Li Z: Bioluminescence reporter gene imaging characterize human embryonic stem cell-derived teratoma formation. J Cell Biochem. 112:840–848. 2011.PubMed/NCBI View Article : Google Scholar

110 

Aggarwal S and Pittenger MF: Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 105:1816–1822. 2005.PubMed/NCBI View Article : Google Scholar

111 

Li H and Fu X: Mechanisms of action of mesenchymal stem cells in cutaneous wound repair and regeneration. Cell Tissue Res. 348:371–377. 2012.PubMed/NCBI View Article : Google Scholar

112 

Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A and Leroux MA: Concise review: Role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 1:142–149. 2012.PubMed/NCBI View Article : Google Scholar

113 

Williams CG, Kim TK, Taboas A, Malik A, Manson P and Elisseeff J: In vitro chondrogenesis of bone marrow-derived mesenchymal stem cells in a photopolymerizing hydrogel. Tissue Eng. 9:679–688. 2003.PubMed/NCBI View Article : Google Scholar

114 

Zhang W, Ge W, Li C, You S, Liao L, Han Q, Deng W and Zhao RC: Effects of mesenchymal stem cells on differentiation, maturation, and function of human monocyte-derived dendritic cells. Stem Cells Dev. 13:263–271. 2004.PubMed/NCBI View Article : Google Scholar

115 

Choi H, Lee RH, Bazhanov N, Oh JY and Prockop DJ: Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in resident macrophages. Blood. 118:330–338. 2011.PubMed/NCBI View Article : Google Scholar

116 

Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, Robey PG, Leelahavanichkul K, Koller BH, Brown JM, et al: Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 15:42–49. 2009.PubMed/NCBI View Article : Google Scholar

117 

François M, Romieu-Mourez R, Li M and Galipeau J: Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation. Mol Ther. 20:187–195. 2012.PubMed/NCBI View Article : Google Scholar

118 

Shapiro SA, Kazmerchak SE, Heckman MG, Zubair AC and O'Connor MI: A prospective, single-blind, placebo-controlled trial of bone marrow aspirate concentrate for knee osteoarthritis. Am J Sports Med. 45:82–90. 2017.PubMed/NCBI View Article : Google Scholar

119 

Kanaya A, Deie M, Adachi N, Nishimori M, Yanada S and Ochi M: Intra-articular injection of mesenchymal stromal cells in partially torn anterior cruciate ligaments in a rat model. Arthroscopy. 23:610–617. 2007.PubMed/NCBI View Article : Google Scholar

120 

Eseonu OI and De Bari C: Homing of mesenchymal stem cells: Mechanistic or stochastic? Implications for targeted delivery in arthritis. Rheumatology (Oxford). 54:210–218. 2015.PubMed/NCBI View Article : Google Scholar

121 

Horie M, Choi H, Lee RH, Reger RL, Ylostalo J, Muneta T, Sekiya I and Prockop DJ: Intra-articular injection of human mesenchymal stem cells (MSCs) promote rat meniscal regeneration by being activated to express Indian hedgehog that enhances expression of type II collagen. Osteoarthritis Cartilage. 20:1197–1207. 2012.PubMed/NCBI View Article : Google Scholar

122 

Jeong SY, Ha J, Lee M, Jin HJ, Kim DH, Choi SJ, Oh W, Yang YS, Kim JS, Kim BG, et al: Autocrine action of thrombospondin-2 determines the chondrogenic differentiation potential and suppresses hypertrophic maturation of human umbilical cord blood-derived mesenchymal stem cells. Stem Cells. 33:3291–3303. 2015.PubMed/NCBI View Article : Google Scholar

123 

Scarfì S: Use of bone morphogenetic proteins in mesenchymal stem cell stimulation of cartilage and bone repair. World J Stem Cells. 8:1–12. 2016.PubMed/NCBI View Article : Google Scholar

124 

Oliver KS, Bayes M, Crane D and Pathikonda C: Clinical outcome of bone marrow concentrate in knee osteoarthritis. J Prolotherapy. 7:937–946. 2015.

125 

Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, Kim JE, Shim H, Shin JS, Shin IS, et al: Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: A proof-of-concept clinical trial. Stem Cells. 32:1254–1266. 2014.PubMed/NCBI View Article : Google Scholar

126 

Dexheimer V, Mueller S, Braatz F and Richter W: Reduced reactivation from dormancy but maintained lineage choice of human mesenchymal stem cells with donor age. PLoS One. 6(e22980)2011.PubMed/NCBI View Article : Google Scholar

127 

Ferone A and Messina G: Sera of overweight patients alter adipogenesis and osteogenesis of bone marrow mesenchymal stromal cells, a phenomenon that also persists in weight loss individuals. J Stem Cell Res Ther. 6(1000347)2016.

128 

Mansilla E, Díaz Aquino V, Zambn D, Marin GH, Mártire K, Roque G, Ichim T, Riordan NH, Patel A, Sturla F, et al: Could metabolic syndrome, lipodystrophy, and aging be mesenchymal stem cell exhaustion syndromes? Stem Cells Int. 2011(943216)2011.PubMed/NCBI View Article : Google Scholar

129 

Johnstone B, Hering TM, Caplan AI, Goldberg VM and Yoo JU: In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 238:265–272. 1998.PubMed/NCBI View Article : Google Scholar

130 

Tatebe M, Nakamura R, Kagami H, Okada K and Ueda M: Differentiation of transplanted mesenchymal stem cells in a large osteochondral defect in rabbit. Cytotherapy. 7:520–530. 2005.PubMed/NCBI View Article : Google Scholar

131 

Pelttari K, Winter A, Steck E, Goetzke K, Hennig T, Ochs BG, Aigner T and Richter W: Premature induction of hypertrophy during in vitro chondrogenesis of human mesenchymal stem cells correlates with calcification and vascular invasion after ectopic transplantation in SCID mice. Arthritis Rheum. 54:3254–3266. 2006.PubMed/NCBI View Article : Google Scholar

132 

Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO and Pittenger MF: Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng. 4:415–428. 1998.PubMed/NCBI View Article : Google Scholar

133 

Turinetto V, Vitale E and Giachino C: Senescence in human mesenchymal stem cells: Functional changes and implications in stem cell-based therapy. Int J Mol Sci. 17(1164)2016.PubMed/NCBI View Article : Google Scholar

134 

Gu Y, Li T, Ding Y, Sun L, Tu T, Zhu W, Hu J and Sun X: Changes in mesenchymal stem cells following long-term culture in vitro. Mol Med Rep. 13:5207–5215. 2016.PubMed/NCBI View Article : Google Scholar

135 

Irioda AC, Cassilha R, Zocche L, Francisco JC, Cunha RC, Ferreira PE, Guarita-Souza LC, Ferreira RJ, Mogharbel BF, Garikipati VN, et al: Human adipose-derived mesenchymal stem cells cryopreservation and thawing decrease α4-Integrin expression. Stem Cells Int. 2016(2562718)2016.PubMed/NCBI View Article : Google Scholar

136 

Bastos R, Mathias M, Andrade R, Bastos R, Balduino A, Schott V, Rodeo S and Espregueira-Mendes J: Intra-articular injections of expanded mesenchymal stem cells with and without addition of platelet-rich plasma are safe and effective for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 11:3342–3350. 2018.PubMed/NCBI View Article : Google Scholar

137 

Bastos R, Mathias M, Andrade R, Amaral RJFC, Schott V, Balduino A, Bastos R, Miguel Oliveira J, Reis RL, Rodeo S and Espregueira-Mendes J: Intra-articular injection of culture-expanded mesenchymal stem cells with or without addition of platelet-rich plasma is effective in decreasing pain and symptoms in knee osteoarthritis: A controlled, double-blind clinical trial. Knee Surg Sports Traumatol Arthrosc. 28:1989–1999. 2020.PubMed/NCBI View Article : Google Scholar

138 

Emadedin M, Labibzadeh N, Liastani MG, Karimi A, Jaroughi N, Bolurieh T, Hosseini SE, Baharvand H and Aghdami N: Intra-articular implantation of autologous bone marrow-derived mesenchymal stromal cells to treat knee osteoarthritis: A randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy. 20:1238–1246. 2018.PubMed/NCBI View Article : Google Scholar

139 

Hernigou P, Auregan JC, Dubory A, Flouzat-Lachaniette CH, Chevallier N and Rouard H: Subchondral stem cell therapy versus contralateral total knee arthroplasty for osteoarthritis following secondary osteonecrosis of the knee. Int Orthop. 42:2563–2571. 2018.PubMed/NCBI View Article : Google Scholar

140 

Gupta PK, Chullikana A, Rengasamy M, Shetty N, Pandey V, Agarwal V, Wagh SY, Vellotare PK, Damodaran D, Viswanathan P, et al: Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®): Preclinical and clinical trial in osteoarthritis of the knee joint. Arthritis Res Ther. 18(301)2016.PubMed/NCBI View Article : Google Scholar

141 

Song Y, Du H, Dai C, Zhang L, Li S, Hunter DJ, Lu L and Bao C: Human adipose-derived mesenchymal stem cells for osteoarthritis: A pilot study with long-term follow-up and repeated injections. Regen Med. 13:295–307. 2018.PubMed/NCBI View Article : Google Scholar

142 

Lee WS, Kim HJ, Kim KI, Kim GB and Jin W: Intra-Articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: A phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med. 8:504–511. 2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2021
Volume 15 Issue 2

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Loo SJ and Loo SJ: Advantages and challenges of stem cell therapy for osteoarthritis (Review). Biomed Rep 15: 67, 2021
APA
Loo, S.J., & Loo, S.J. (2021). Advantages and challenges of stem cell therapy for osteoarthritis (Review). Biomedical Reports, 15, 67. https://doi.org/10.3892/br.2021.1443
MLA
Loo, S. J., Wong, N. K."Advantages and challenges of stem cell therapy for osteoarthritis (Review)". Biomedical Reports 15.2 (2021): 67.
Chicago
Loo, S. J., Wong, N. K."Advantages and challenges of stem cell therapy for osteoarthritis (Review)". Biomedical Reports 15, no. 2 (2021): 67. https://doi.org/10.3892/br.2021.1443