Next Article in Journal
Comprehensive Review on Bimolecular Fluorescence Complementation and Its Application in Deciphering Protein–Protein Interactions in Cell Signaling Pathways
Previous Article in Journal
Towards a Treatment for Leukodystrophy Using Cell-Based Interception and Precision Medicine
Previous Article in Special Issue
TSG-6 Inhibits the NF-κB Signaling Pathway and Promotes the Odontogenic Differentiation of Dental Pulp Stem Cells via CD44 in an Inflammatory Environment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy

1
The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China
2
Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Collage of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen 518060, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2024, 14(7), 858; https://doi.org/10.3390/biom14070858
Submission received: 28 May 2024 / Revised: 6 July 2024 / Accepted: 13 July 2024 / Published: 17 July 2024
(This article belongs to the Special Issue Recent Developments in Mesenchymal Stem Cells)

Abstract

:
Osteoarthritis (OA), a chronic joint disease affecting over 500 million individuals globally, is characterized by the destruction of articular cartilage and joint inflammation. Conventional treatments are insufficient for repairing damaged joint tissue, necessitating novel therapeutic approaches. Mesenchymal stem cells (MSCs), with their potential for differentiation and self-renewal, hold great promise as a treatment for OA. However, challenges such as MSC viability and apoptosis in the ischemic joint environment hinder their therapeutic effectiveness. Hydrogels with biocompatibility and degradability offer a three-dimensional scaffold that support cell viability and differentiation, making them ideal for MSC delivery in OA treatment. This review discusses the pathological features of OA, the properties of MSCs, the challenges associated with MSC therapy, and methods for hydrogel preparation and functionalization. Furthermore, it highlights the advantages of hydrogel-based MSC delivery systems while providing insights into future research directions and the clinical potential of this approach.

1. Introduction

Osteoarthritis (OA) is a prevalent chronic joint disease characterized by damaged articular cartilage tissue and the onset of joint inflammation, resulting in pain, functional limitations, and diminished quality of life for patients [1,2,3]. Conventional treatment methods are inadequate in repairing damaged joint cartilage tissue, highlighting the need for effective therapeutic approaches that facilitate joint tissue regeneration.
Mesenchymal stem cell (MSC) regenerative therapies have emerged as promising strategies for repairing damaged tissues and organs. MSCs can self-renew and differentiate into a variety of cell types, including chondrocytes and osteoblasts [4,5]. Moreover, their wide availability from diverse sources, ease of accessibility, and expandability in vitro make them a focal point in OA treatment research. However, MSC therapy for OA encounters significant challenges. For instance, the reduced viability and increased susceptibility to apoptosis of MSCs within the ischemic and hypoxic environment of the joint cavity hinder their therapeutic efficacy [6]. Additionally, the shear stress generated by the injection needle during MSC delivery into the joint cavity can compromise their viability [7]. Furthermore, individually injected MSCs may not fully differentiate into the desired cell types [8]. Henceforth, there is an urgent need for delivery techniques that ensure an optimal differentiation environment within the joint cavity while maintaining MSC viability during administration.
Hydrogels, as versatile carriers for drugs and cells, offer a three-dimensional scaffold structure that mimics the physiological environment of tissues. They have been demonstrated to be able to support cell viability and differentiation in various disease studies [9]. Furthermore, hydrogels exhibit excellent biocompatibility and biodegradability, making them an ideal choice for delivering MSCs in the treatment of osteoarthritis [10].
Here, we comprehensively reviewed the pathological features of OA, the properties of MSCs, and their applications in OA treatment. We also highlight the challenges associated with MSC therapy for OA. Subsequently, we delve into hydrogel preparation, functionalization methods, and their diverse biomedical applications. Finally, we discuss the advantages of hydrogels in delivering MSCs and review reported research cases involving applying hydrogels for MSC delivery in the treatment of OA. Additionally, we offer insights into future research directions and the promising clinical prospects of this approach.

2. Osteoarthritis (OA)

2.1. Pidemiological Characteristics and Physiopathological Mechanisms of OA

OA, a degeneration of the joints, primarily affects individuals aged 50 and older, with a higher incidence among females compared to males [11,12]. It is estimated that the global population of OA patients reaches as high as 500 million. By 2032, the prevalence of OA is projected to increase by 26.6% to 29.5% [13]. OA not only affects the patients’ health physically and mentally but also causes economic loss for both individuals and society. The average annual treatment costs per OA patient range from USD 700 to USD 15,600 [14].
OA is a chronic degenerative disease that impacts the whole joint, including bones, articular tissue, the synovium, subchondral bone, the meniscus, and ligaments. The primary characteristics of osteoarthritis involve the degeneration and deterioration of joint cartilage, the meniscus, and ligaments, along with inflammation and sclerosis of synovial tissues and the presence of subchondral bone cysts [15]. Traditionally viewed as a passive degenerative joint disease or a consequence of long-term mechanical wear and tear, emerging perspectives indicate that the OA process is active and dynamic. It is primarily driven by an imbalance between joint degradation and repair [16]. Initially, erosion starts on the cartilage surface and gradually penetrates into the calcified articular cartilage zone [17]. The avascular nature of cartilage tissue, coupled with limited chondrocyte proliferative capacity and impaired intrinsic repair mechanisms, exacerbates the deterioration of the cartilage layer [18,19]. Concurrently, the physical stress resulting from cartilage defects triggers pathological subchondral bone remodeling, further compromising the integrity of the cartilage layer. Ultimately within the cartilage microenvironment it contributes to osteophyte formation around the joint periphery [15]. (Figure 1).
Studies have demonstrated that the progression of OA is influenced by inflammatory factors, metalloproteinases, cellular senescence, estrogen, and biomechanical imbalances within the joint cartilage. These factors collectively contribute to local cartilage damage, osteophyte formation, subchondral bone remodeling, and excessive synovial proliferation. These pathological changes profoundly impact the patients’ quality of life and serve as crucial indicators of OA progression [20,21].
Figure 1. Schematic illustration of the physiopathology of OA. Part A indicates the healthy joint while Part B indicates the osteoarthritic joint. Reprinted with permission from Ref. [22]. Copyright 2022 Elsevier.
Figure 1. Schematic illustration of the physiopathology of OA. Part A indicates the healthy joint while Part B indicates the osteoarthritic joint. Reprinted with permission from Ref. [22]. Copyright 2022 Elsevier.
Biomolecules 14 00858 g001

2.2. Current Dilemma in OA Treatment

Currently, due to the avascular nature of articular cartilage tissues and the low self-renewing capacity of chondrocytes, structural damage to articular cartilage cannot self-heal. This is considered a fundamental factor in the progression and difficult-to-treat nature of OA [23]. There are no effective methods or drugs for the comprehensive treatment of OA. At present, treatment approaches for OA can be categorized into physical therapy, pharmacotherapy, and surgical intervention, as summarized in Table 1. However, the majority of current clinical drugs and treatment modalities only provide symptomatic relief by reducing joint inflammation and pain, thereby slowing down the progression of OA. They are unable to achieve complete healing of the damaged cartilage or cure OA [24].
Moreover, only a small number of patients achieve partial regeneration of articular cartilage through non-surgical treatments. However, the regeneration process is time-consuming, and the newly formed cartilage tissue often lacks adequate hardness and compressive strength [25]. It is crucial to be aware of the potential serious side effects associated with drug treatments. For instance, long-term oral administration of nonsteroidal anti-inflammatory drugs (NSAIDs) may lead to the development of gastrointestinal ulcers and bleeding. Moreover, drugs administered via intra-articular injection have limited residence time within the joint cavity, leading to low drug utilization and the need for repeated administration, thereby increasing the risk of treatment-related harm [26].
Surgical intervention is often necessary for patients with moderate to advanced OA. However, surgical treatments are invasive and may carry potential risks, such as infection and thrombosis, posing significant harm to the overall well-being of patients [27,28]. Therefore, there is an urgent need to develop new therapeutic drugs or approaches that can achieve cartilage repair and regeneration and ultimately cure OA.
Table 1. Common treatments for osteoarthritis.
Table 1. Common treatments for osteoarthritis.
TreamentsTherapeutic EffectsRef.
PharmacotherapyDiacereinInduces chondrogenesis; has analgesic, anti-inflammatory and antipyretic effects; and improves joint function in patients with osteoarthritis[29,30,31,32]
Chondroitin/glucosaminePain reliever, promotes cartilage regeneration
AcetaminophenPain reliever
OpioidsPain reliever
NSAIDsSuppresses the degradation of cartilage ECM, increases ECM anabolism, and reduces chondrocytes apoptosis
Physical modalitiesExercise, Tai ChiReduces weight load and maintains body balance[33,34,35,36,37,38,39,40]
CrutchesReduces joint loads
Acupuncture, balneotherapy/spa, hydrotherapy,
therapeutic ultrasound
Reduces local inflammatory stimuli by decreasing the expression of inflammatory factors, enhances the muscle strength around the knee to balance the stress
NMES, TENSRelieves pain, improves blood circulation, reduces edema, promotes bone and wound healing, etc.
Surgical treatmentsTotal joint
arthroplasty,
hemiarthroplasty,
arthroscopy
Reconstruction of joints to restore normal motor function[41,42]

3. Roles of MSCs in OA Therapy

3.1. Physiological Characteristics of MSCs

MSCs have long been of great interest in the fields of medical repairment, regeneration, and immune modulation. MSCs are non-phagocytic cells with a fibroblast-like appearance that express specific surface antigens [43,44] (Table 2). They possess potent self-renewal and multipotent differentiation capabilities, as well as remarkable immunomodulatory, anti-inflammatory, and homing properties, making them highly attractive in regenerative medicine [45,46]. The self-renewal capacity of MSCs allows them to maintain their population through cell division, while their multipotent differentiation ability enables them to differentiate into different cell types, including osteocytes, chondrocytes, and adipocytes [47,48]. Furthermore, their homing ability allows these stem cells to precisely target injured or treated areas, effectively enhancing the precision and efficiency of therapeutic interventions [49].
Initially, MSCs were primarily extracted from bone marrow. However, due to the challenges and invasiveness associated with bone marrow collection, researchers began exploring other tissue sources. Subsequently, MSCs were successfully isolated from adipose tissue, synovium, umbilical cord blood, periosteum, amniotic fluid, and membrane and perichondrium [50]. Some commonly used MSCs and their applications are summarized in Table 3. Among these, adipose-derived mesenchymal stem cells (AD-MSCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs) are currently the most widely used in clinical applications. This is because both AD-MSCs and BM-MSCs are relatively easy to culture and can be obtained in large quantities [51]. They both possess immunomodulatory properties and can mediate inflammation. Additionally, they exhibit low immunogenicity, which allows for their use in mismatched or even xenogeneic environments [52,53].
Studies have found that at the single-cell level, AD-MSCs exhibit lower transcriptional heterogeneity and immunogenicity compared to BM-MSCs [54]. Additionally, they demonstrate a stronger immunosuppressive capacity and higher multipotency [47]. Furthermore, adipose-derived MSCs exhibit a faster expansion rate and higher cell survival rates, and are more likely to retain their stem cell phenotype during the culture process [55].
Table 3. Varieties of MSCs and their biomedical applications.
Table 3. Varieties of MSCs and their biomedical applications.
MSCsOriginDifferentiation PotentialApplications
BM-MSCsBone marrowOsteocytes, chondrocytes, and adipocytesNonunion fractures, spinal cord injuries, and amyotrophic lateral sclerosis (ALS) [56,57,58,59,60,61]
Placenta-MSCsNewborn placental tissueOsteocytes, chondrocytes, adipocytes, and smooth muscle cellsMultiple sclerosis, knee osteoarthritis, preterm infant lung disease, and ovarian function restoration [62,63,64,65,66,67]
UC-MSCsIntervascular, perivascular, and subamniotic area of Wharton’s jellyOsteocytes, chondrocytes, and adipocytesTreatment of neurological disorders, cardiovascular diseases, and autoimmune diseases [68,69,70,71]
ADSCsAdipose tissueOsteocytes, adipocytes, chondrocytes, and smooth muscle cellsSkin regeneration, soft tissue repair, and treatment of diabetes [72,73,74,75]
Sy-MSCsSynovial fluid in the joint cavityOsteocytes, chondrocytes, adipocytes, muscle cells, and neuronsOsteoarthritis treatment, cartilage injuries, systemic autoimmune diseases, and tissue engineering [76,77,78,79,80,81]
DPSCsDental pulp tissue of permanent teeth, deciduous teeth, and wisdom teeth in adultsOsteocytes, chondrocytes, adipocytes, muscle cells, and neuronsDental treatment, neural repair, cardiovascular diseases, and bone tissue engineering [82,83,84,85,86]
AMSCsAmniotic membrane tissue from the placentaOsteocytes, chondrocytes, adipocytes, and smooth muscle cellsSkeletal tissue repair, autoimmune diseases, neurodegenerative diseases, liver diseases, and corneal repair [87,88,89,90,91]

3.2. The Therapeutic Potential of MSCs for OA

In clinical trials, MSCs have been increasingly utilized due to their stemness and tropic functions, which provide benefits. The stemness of MSCs refers to their ability to self-renew and differentiate into tissue-specific cells, including chondrocytes, adipocytes, and osteoblasts, which can replace cells in damaged tissues. The tropic functions of MSCs refers to their ability to generate a reparative milieu via cell-to-cell contact and paracrine secretion of various bioactive factors [92,93,94]. These functions promote the immunomodulation of inflammatory cells involved in tissue regeneration (e.g., macrophages, T cells, and mast cells) and their differentiation into endogenous progenitor cells (e.g., osteoprogenitors, chondroprogenitors, etc.) [95]. Due to their versatility, MSCs have been applied in the treatment of a variety of diseases. In the context of breast cancer, MSCs have been employed in conjunction with hematopoietic stem cell transplantation during chemotherapy to facilitate rapid hematopoietic recovery in patients [96]. In a study conducted by Rojas, it was observed in a mouse model that MSCs can migrate to the lungs and differentiate into lung-like cells [97]. This mechanism allows MSCs to replace damaged cells and potentially treat lung injuries. Furthermore, Jung’s research demonstrated that transfecting MSCs with insulin-like growth factor-1 (IGF-1) can provide protection to the myocardium of rats. The transfected MSCs were found to mitigate fibrosis and apoptosis, resulting in a reduction in the size of the infarcted area [98]. These studies demonstrate the broad range of applications for MSC therapy in biomedicine.
Subsequently, MSCs have shown remarkable effectiveness in treating liver disease [99], kidney injury [100], and cartilage tissue engineering [101]. Table 4 summarizes different types of MSCs applied in OA therapy. In the following sections, this article will focus on the application and mechanisms of MSCs in OA therapy by analyzing preclinical studies and clinical trials that explore the use of AD-MSCs and BM-MSCs in treating OA.

3.2.1. AD-MSCs

AD-MSCs have aroused significant interest in the treatment of osteoarthritis (OA) due to their easier accessibility compared to the traditionally used BM-MSCs. AD-MSCs offer advantages such as lower risk of donor site infection and pain, higher cell yield, and a better predictable differentiation pattern [111,112]. Additionally, studies have found that AD-MSCs exhibit histological similarities to chondroblasts in terms of extracellular matrix volume and composition, based on their chondrogenic potential. Jo et al. recruited 18 patients with knee osteoarthritis (KOA) and administered intra-articular injections of AD-MSCs for OA treatment. They found that the injection of 1.0 × 108 AD-MSCs into the joint cavity improved knee joint function and alleviated pain. Furthermore, they observed cartilage regeneration and a reduction in cartilage defects through the regeneration of hyaline-like articular cartilage [113]. Spasovski et al. also found that subcutaneous AD-MSCs can improve clinical symptoms in patients with osteoarthritis. They observed a reduction in pain at 3 months, with the best outcomes achieved at 6 months [114]. Additionally, studies indicated that intra-articular (IA) injection of AD-MSCs can significantly improve joint function and alleviate pain in patients with OA, with no reported adverse events [115]. The feasibility of utilizing AD-MSCs for the treatment of OA is well supported by these successful clinical cases. Studies indicate that the therapeutic mechanism underling the application of AD-MSCs in OA involves reducing pro-inflammatory cytokines and chemokines [116], inhibiting chondrocyte apoptosis [117], suppressing hypertrophy and fibrotic phenotype transition in chondrocytes [118,119], and concurrently decreasing collagenase expression [120].

3.2.2. BM-MSCs

BM-MSCs are another commonly used MSC therapy for OA. Kuroda et al. discovered that autologous transplantation of BM-MSCs promotes the repair of cartilage defects in young patients [121]. Recently, the safety of IA injection of BM-MSCs was confirmed in 12 OA patients. After a two-year follow-up, pain relief and improvement in cartilage quality were observed. In addition to promoting cartilage repair, BM-MSCs may also play a role in reducing inflammatory symptoms [122]. Zhang et al. found that co-culturing BM-MSCs with chondrocytes from patients with osteoarthritis increased the proliferation of osteoarthritic chondrocytes while suppressing their inflammatory activity [104]. Another phase I/IIa clinical trial demonstrated that autologous transplantation of BM-MSC can reduce synovial inflammation, improve knee joint function, and alleviate symptoms in patients with OA [123].

3.3. Difficulties of Applying MSCs in OA Treatment

The use of MSCs in OA treatment holds great promise, but several challenges must be overcome. Firstly, MSCs injected into the joint may be at high risk of starvation and death due to the avascular nature of cartilage tissue, which consequently diminishes the therapeutic efficacy of MSCs [124,125]. Additionally, injecting free MSCs without any supporting agents can be detrimental to the cells, as they are exposed to shear and stretching forces, as well as pressure changes caused by the needle, leading to a reduction in cell viability [22,126]. Furthermore, free MSCs are prone to migrate away from the injection site towards the surrounding tissues, resulting in fewer cells effectively delivering their therapeutic effects at the intended location over time [127]. Moreover, bare MSCs may not provide the ideal conditions necessary to unlock their full healing potential. Studies have shown that predifferentiated chondrogenic MSCs with transforming growth factor-b3 (TGF-b3) performed better than undifferentiated MSCs when implanted with a collagen scaffold into an ovine chronic defect model [128]. Lastly, both preclinical studies and clinical applications require a substantial number of MSCs [129]. Efficiently and rapidly expanding the MSC population is not only a critical issue for the bio-application of MSCs but also an urgent problem for other types of somatic stem cells. Therefore, there is an urgent need to optimize the delivery methods of MSCs into joints for OA therapy.

4. Bio-Application of Hydrogel Technologies

4.1. Characteristics of Hydrogel Technologies and Their Biomedical Application

Hydrogels, a class of unique polymer materials, are distinguished by their three-dimensional network structure formed by hydrophilic polymer chains (refer to Figure 2). These materials are known for their rapid water absorption and volumetric swelling while remaining insoluble in water [130]. The composition of hydrogels typically includes hydrophilic and hydrophobic groups, which are either chemically or physically crosslinked to create a stable network structure. This design endows hydrogels with exceptional water absorption capacity and shape-maintaining properties. Depending on the source of their monomers, hydrogels can be categorized into three types: natural hydrogels, synthetic hydrogels, and hybrid-origin hydrogels [131] (refer to Table 5).
With a specific physicochemical structure, hydrogels possess excellent properties, including biocompatibility, high water absorption, tunability, and biodegradability. Furthermore, hydrogels can be functionally modified to cater to specific application needs [146]. For example, the modification of polysaccharide-based hydrogels with polymers, including polyethylene, polyvinylpyrrolidone (PVP), and polyethylene glycol (PEG), can enhance their swelling/shrinkage responsiveness, mechanical strength, and adhesive properties (refer to Figure 3) [147]. The remarkable physicochemical attributes of hydrogels have led to their extensive application in the field of biomedicine. Currently, hydrogels are widely utilized in various applications, such as tissue engineering scaffolds, drug and cell delivery systems, controlled release systems, wound dressings, and biomedical devices. The utilization of hydrogels in these applications has been explored for various treatments, such as cancer treatment, wound healing, diabetes management, tissue regeneration, and osteoarthritis [148,149].

4.2. Biofabrication of Hydrogel

Hydrogels are commonly prepared using a variety of methods, each with distinct advantages, limitations, and ideal applications. These methods (refer to Table 6) include physical crosslinking, chemical crosslinking, photopolymerization, and enzymatic biocatalysis [130]. Physical crosslinking depends on non-covalent interactions between polymer chains. Techniques such as freeze-thawing create a porous structure by solidifying and recrystallizing the polymer, while ion condensation forms gels through the charge interactions of polyelectrolytes [150]. Chemical crosslinking involves the use of crosslinking agents like glutaraldehyde, diisocyanates, and natural crosslinkers such as tannic acid. These agents initiate or catalyze reactions within a polymer solution to form a crosslinked structure. This approach allows for precise control over the degree of crosslinking and the resulting physicochemical properties of hydrogels through adjusting the concentration of the crosslinking agent, the reaction time, and the temperature [151]. Photopolymerization employs photosensitive monomers and photoinitiators to initiate polymerization under ultraviolet (UV) or visible light, leading to the formation of a crosslinked network. Acrylic acid and acrylamide are commonly used as photopolymerizable monomers. The process involves dissolving these monomers and photoinitiators in a solvent, mixing them with a crosslinking agent, and exposing the prepolymer solution to light of a specific wavelength to form the hydrogel [152]. Enzymatic biocatalysis uses specific enzyme catalysts to induce crosslinking reactions within a polymer solution, a method often employed for natural polymer hydrogels such as gelatin and sodium alginate. This technique offers advantages such as mild reaction conditions, high selectivity, and sensitivity to bioactive substances [153]. In practical applications, the choice of hydrogel preparation method should be guided by the desired physicochemical properties, the degree of crosslinking, and the intended application environment of the hydrogel.

4.3. Functionalization of Hydrogel

Variations in the raw materials used to synthesize hydrogels result in a wide range of physicochemical properties, making it difficult for a single type of hydrogel to satisfy the diverse requirements of multiple application scenarios. Consequently, researchers frequently undertake functional modifications to expand the utility of hydrogels. Common strategies for hydrogel functionalization include covalent crosslinking, chemical modifications, immobilization of bioactive molecules, biomimetic modifications, and thermoresponsive alterations [147,156,157,158,159,160]. Through these diverse modification techniques, hydrogels can be endowed with enhanced functionalities, paving the way for versatile and intelligent applications. For instance, covalently crosslinking bioactive monomers with acrylamide monomers can significantly enhance the biocompatibility of acrylamide-based hydrogels [161]. Additionally, by immobilizing specific protein molecules, such as antibodies, onto the hydrogel surface, targeted interactions with particular tissues can be achieved [162]. Cristina Manferdini et al. have reviewed and summarized various strategies for functionalizing hydrogels tailored specifically for OA applications (refer to Table 7) [163].

4.4. Strategies Based on Combination of MSCs with Hydrogels for OA Treatment

Hydrogels, with their unique physicochemical properties, offer a promising solution for MSC delivery in OA treatment, effectively addressing several challenges associated with MSCs. Firstly, the internal porous structure of hydrogels provides an optimal environment for cell embedding, ensuring cell survival and functionality. For instance, Dong et al. fabricated a 3D hydrogel system of PEG-based hyperbranched multifunctional homopolymers. They found that that rat AD-MSCs embedded in the hydrogel maintained viability above 85% for two weeks (Figure 4). They also found that when the concentration of the polymer in the hydrogel system is less than 5%, variations in the polymer concentration have no effect on the cell viability [174]. Furthermore, within highly porous hydrogels, MSCs form clusters that enhance cell–cell interactions and prevent cells from diffusing away from the injection site. Karen E. Martin et al. developed a hydrogel-based strategy for MSC delivery for wound healing based on multi-arm poly (ethylene glycol) (PEG) macromers functionalized with maleimide end groups (such as 4-arm macromers, PEG-4MAL). They functionalized PEG-4MAL with an amide or ester to obtained PEG-4aMAL or PEG-4eMAL and constructed hydrogels with varying contents of PEG-4aMAL and PEG-4eMAL to determine the influence of hydrogel formulation on its property. They found that even on day 15, approximately 25% of the MSCs loaded within the 75% PEG-4eMAL hydrogel still resided at the injection site. However, MSCs loaded within the 0% PEG-4eMAL could not be detected on day 5 (Figure 5) [175]. Secondly, hydrogels enhance the anchorage of MSCs, which depend on a nucleated cellular product. Loading MSCs within hydrogels provides a three-dimensional microenvironment that promotes cell–cell interactions and facilitates ECM deposition [176], enabling rapid MSC adherence to the site of cartilage tissue damage. Additionally, encapsulating MSCs within hydrogels can significantly reduce the shear and stretching forces experienced during injection into the joint cavity. In a study led by Matthew A. Wagner and colleagues, the viability of cells in deionized water (as the control) was compared to that of cells in hydrogel during syringe needle flow, aiming to investigate whether hydrogels could effectively prevent cell membrane rupture and subsequently enhance cell viability. They found the viability of cells in the control group decreased by 76.14% from preinjection to postinjection, whereas the viability of cells encapsulated in hydrogel with a high concentration remained stable. Even in hydrogel with a low concentration, cell viability decreased by 19.38% from preinjection to postinjection, which was significantly lower than that observed in the control group [177]. Furthermore, hydrogels can be co-loaded with specific growth factors that promote cell proliferation or differentiation, as well as nutrients that sustain cellular metabolism. Paula Gonzalez-Fernandez et al. covalently conjugated glucose molecules to hyaluronic acid (HA) and used this HA–glucose as a scaffold for constructing an MSC-loaded hydrogel. The presence of ß-glucosidase in the joint cavity allows for the hydrolysis of glycosidic bonds between glucose moieties and hyaluronic acid, resulting in the release of glucose molecules to provide energy for MSCs. In their simulated in vitro experiments, they found that adding glucose to the hydrogel increased MSC viability by 71% [6]. Zhu et al. fabricated a hydrogel delivery system by crosslinking icariin, a component of traditional Chinese medicine known to promote ECM synthesis and enhance the differentiation of MSCs into chondrocytes, with HA. They discovered that the hydrogel incorporating icariin significantly improved the viability of BM-MSCs and induced their differentiation into chondrocytes. Using this carrier system to deliver BM-MSCs in an OA rat model promoted chondrogenesis, inhibited cartilage tissue degradation, and alleviated inflammatory symptoms. Importantly, the therapeutic effect of this carrier system in treating OA was superior to the direct injection of BM-MSCs (Figure 6) [178]. These approaches not only address issues such as insufficient cell proliferation or apoptosis caused by starvation within the joint cavity but also allow for the exploration of optimal therapeutic conditions through varying combinations. Besides, integration of MSCs with advanced gene manipulation techniques [179], such as RNA interference (RNAi) or other gene editing approaches [180] into hydrogels, represents another promising avenue for further improving MSC viability [181,182,183], differentiation capacity [184,185], and tissue repair efficacy [186,187]. Hydrogels serve as an effective platform for the localized and sustained delivery of gene expression modulators, thereby reducing the incidence of off-target effects and ensuring the prolonged regulation of gene expression at the intended target. This approach allows for precise spatiotemporal control over gene expression, enhancing the therapeutic efficacy and safety profile of gene-based interventions [188]. Numerous genes have been identified as pivotal for MSCs’ survival, trophic functions, differentiation potential, immunogenicity, and anti-inflammatory properties (refer to Table 8). For example, research by Shuo Wang and colleagues has highlighted the significance of Sirtuin 3 (Sirt3) in sustaining MSC viability under conditions of nutrient deprivation. They discovered that the downregulation of Sirt3 makes MSCs more susceptible to starvation-induced apoptosis [181]. Consequently, the integration of plasmids that overexpress Sirt3 into hydrogel scaffolds with MSCs, in conjunction with the controlled release mechanism of these plasmids by the scaffold, has the potential to induce a sustained and elevated expression level of Sirt3 in MSCs. This approach may confer upon MSCs a heightened resistance to apoptosis triggered by the nutrient-deprived environment within the joint space, thereby enhancing their viability and promoting cartilage tissue regeneration. Similarly, the targeted manipulation of specific gene expression might also improve the adherence of mesenchymal stem cells (MSCs) to the extracellular matrix (ECM) of cartilage [189], reduce the immunogenicity of MSCs [190,191], and promote their chondrogenic differentiation [192]. Indeed, achieving high transfection efficiency in a three-dimensional (3D) system such as a hydrogel may be more straightforward than in a two-dimensional (2D) system. Adriana M. Ledo and colleagues developed a 3D hydrogel system by integrating mesenchymal stem cells (MSCs) with nanocomplexes that contain plasmids encoding for SOX9 into the hydrogel scaffold. This 3D hydrogel system demonstrated significantly enhanced transfection efficiency for SOX9 compared to 2D systems. Moreover, the MSCs within this 3D hydrogel exhibited elevated expression levels of chondrogenic markers [193]. Lastly, hydrogels with porous polymeric networks have been demonstrated to mimic stem cell culture environments and promote cell–cell or cell–ECM interactions, providing an ideal medium for MSC expansion. A previous study showed that AD-MSCs cultured within a three-dimensional (3D) hydrogel scaffold enhanced the retention of a rejuvenated population of ASCs that were not senescent, as evidenced by increased expression of “stem-like” surface markers on MSCs compared to two-dimensional (2D) culture systems [194]. Amorn Pangjantuk and colleagues fabricated an alginate–hyaluronic acid (AL-HA) 3D hydrogel culture system. Compared to the 2D monolayer culture, this 3D culture system not only promoted the proliferation and survival of MSCs but also maintained their stemness more effectively. Importantly, MSCs grown within hydrogels exhibit a spherical cellular morphology (Figure 7) [195].

4.5. Recent Advances in Application of Combining MSCs with Hydrogel in OA Treatment

The versatility of MSCs has been extensively demonstrated in improving inflammation, preventing chondrocyte apoptosis, and promoting cartilage regeneration in the treatment of OA. In this section, we focus on recent advances in OA treatment that leverage both hydrogels and MSCs.
Currently, AD-MSCs and BM-MSCs are most commonly used in preclinical studies or clinical trials for OA therapy. Nevertheless, due to the limited availability of donor cells, researchers are exploring other potential sources of MSCs. It has been shown that the synovium and synovial fluid within joints contain MSCs [204,205]. MSCs derived from joint tissues are considered to have superior chondrogenic ability. For instance, Jun Li et al. developed a hyper-branched polyPEGDA/HA hydrogel incorporating arthroscopic fluid-derived MSCs (AFF-MSCs). They found that AFF-MSCs possess typical characteristics and properties of MSCs. The viability and DNA content of AFF-MSCs encapsulated in hydrogel respectively doubled and quadrupled over the course of 7 days. Encapsulation of AFF-MSCs promoted expression of chondrogenic markers, which was approximately more than four times that of MSCs cultured in dish. Encapsulated MSCs also exhibited a quadruple efficiency in repairing cartilage defects in rats compared to the control [206]. Additionally, mesenchymal stem cells derived from human umbilical cord blood [207,208] and synovium [209] have also been demonstrated to have therapeutic effects for OA when encapsulated in hydrogels.
In addition to expanding the sources of MSCs, there is growing interest in using MSCs derived from joint tissues and incorporating extracellular matrix (ECM) components and chondrogenic factors during hydrogel scaffold fabrication. It is believed that MSCs from joint tissues have greater potential for chondrogenic differentiation compared to those from other sources. The inclusion of ECM components endows the hydrogel with properties that mimic the natural ECM, facilitating MSC attachment and growth [210]. The incorporation of chondrogenic factors promotes the differentiation of MSCs into chondrocytes [170]. These elements within the hydrogel work synergistically to promote cartilage regeneration. In a study led by Shengbo Sang, a 3D hydrogel scaffold for regenerating cartilage was constructed by incorporating gelatin methacrylate (Gel-MA), chondroitin sulfate methacrylate (CS-MA), hyaluronic acid methacrylate (HA-MA), transforming growth factor-beta 1 (TGF-β1), and synovium-derived MSCs (SMSCs) (Figure 8). In this system, CS-MA and HA-MA are derivatives of chondroitin sulfate and hyaluronic acid, respectively, which are components of ECM. The incorporation of CS-MA and HA-MA (GelHACS-MA) significantly enhanced the proliferation of MSCs and facilitated cartilage repair in rats. Furthermore, the addition of TGF-β1 (GelHACS-MA + TGF-β1) further augmented the chondrogenic effect of GelHACS-MA [211].
Advanced OA is characterized not only by damaged cartilage tissue but also by abnormal subchondral bone structure, indicating the presence of osteochondral defects [28]. Therefore, simultaneous recovery of damaged cartilage and subchondral bone tissue is crucial for advanced OA therapy. Jason L. Guo and colleagues developed a bilayered hydrogel system consisting of two distinct layers with a height of 1.5 mm each. The upper layer was bio-conjugated with a chondrogenic peptide (GGGHAVDI) to induce chondrogenesis, while the lower layer was bio-conjugated with a glycine–histidine–lysine peptide derived from osteonectin to induce bone mineralization. Encapsulating MSCs into this bilayered hydrogel system effectively filled up the osteochondral defects in a rabbit model by regenerating both cartilage and bone tissues (Figure 9) [212].

5. Conclusions and Future Perspectives

Osteoarthritis (OA), a chronic degenerative condition, poses a significant health challenge for hundreds of millions of middle-aged and elderly individuals globally, with a particular impact on women [213]. Existing treatment options such as physical therapy, pharmacotherapy, and surgical intervention offer symptomatic relief but fail to address the underlying cause of the disease and can come with side effects and potential risks [27]. Consequently, researchers are pursuing innovative therapeutic strategies. Among these strategies, mesenchymal stem cell (MSC) therapy has shown considerable promise due to its self-renewal capacity, potential for multilineage differentiation, and low immunogenicity [94]. However, issues such as the low survival rates of MSCs within the joint cavity, their vulnerability to mechanical damage, and loss of stemness during in vitro expansion diminish the therapeutic effectiveness of MSCs and limit their biomedical applications [126,127,128,129].
The use of biodegradable and biocompatible hydrogels has emerged as an innovative strategy for delivering MSCs to the joints to enhance their viability and functionality [22]. These hydrogels simulate the three-dimensional structure of the extracellular matrix, providing an optimal environment for cell growth and protecting cells from mechanical injury [214]. However, there are still some concerns and limitations regarding the application of MSCs. For example, the low immunogenicity of MSCs is considered a crucial factor that cannot be overlooked in their biomedical use. Due to the large quantity required for tissue regeneration, most patients receive allogeneic MSCs during transplantation [215,216]. However, some recent studies found that allogeneic MSCs can elicit immune responses and therefore lead to rejection. Therefore, in addition to starvation and malnutrition, immunological rejection may also be a significant factor contributing to the low viability of MSCs in the articular cavity. Consequently, further investigations are warranted to explore the incorporation of immunosuppressive agents with MSCs into hydrogels for achieving high viability and retention of MSCs in OA treatment [217].
Besides, many studies use the term “mimic” to describe the hydrogels’ ability to provide an environment similar to the ECM of cartilage tissues for MSC survival and differentiation. However, in reality, human cartilage tissue is quite rigid and its stiffness exceeds that of commonly used hydrogels in preclinical studies [218]. Additionally, cartilage has a complex architecture, with distinct morphological and structural characteristics, even within the uncalcified cartilage zone [219]. The uncalcified cartilage zone can be divided into three zones based on their structural and constituent variance. For instance, in zone 1, collagen type II fibrils are densely packed, with a thin diameter of about 30–35 nm, and oriented parallel to the surface of the cartilage. In zone 2, the collagen fibrils are thin and oriented obliquely or perpendicularly to the articular surface. Zone 3 has the thickest collagen fibrils with a diameter of 40–80 nm and perpendicular orientation in uncalcified cartilage [220]. Until now, no studies have authentically fabricated hydrogels that structurally and constitutively resemble ECM. Therefore, most hydrogels differ significantly from the ECM of cartilage tissues despite incorporating components such as collagen and proteoglycan. Researchers should exercise caution when using terms like “mimic the ECM of cartilage.” One potential approach to fabricating hydrogels that mimic ECM is by applying 3D printing technology to print different layers similar to actual cartilage.
Lastly, although many kinds of MSCs derived from different tissues, such as adipose tissues, bone marrow, synovial fluid, and synovial tissues in the joint cavity, have been demonstrated as therapeutic for OA, there is still a lack of studies systematically comparing the accessibility, renewal capacity, chondrogenic differentiation potential, heterogeneity, and immunogenicity among these different types of MSCs. The most suitable MSCs for OA treatment should be identified before combining hydrogels and MSCs on a large scale in future treatments.
Although there are still challenges in applying hydrogel technology to enhance the therapeutic effect of MSCs in OA treatment, we believe, through these interdisciplinary research initiatives, that safer, more effective, and cost-efficient treatments for OA can be developed, thereby significantly improving the quality of life for patients.

Author Contributions

X.W., W.H. and H.H. wrote the main content of the manuscript; J.H., R.W., H.L. and Y.L. assisted in writing part of the manuscript. X.H. and G.W. drafted the outline, directed the writing, and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The present study was supported by grants from the Medical Research Fund of Qingyuan People’s Hospital (PA1c1ee205380526), the Special Funds of the Clinical Research from Qingyuan Hospital (QYRYCRC2023010), the Guangdong Provincial Medical Science and Technology Research Fund Project (A2023257; B2021209), the Qingyuan City Science and Technology Plan Project (2022KJJH031), and the Guangzhou Medical University Student Innovation Capability Enhancement Program for the year 2022 (02-408-240603070). And The APC was funded by the Medical Research Fund of Qingyuan People’s Hospital (PA1c1ee205380526).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wei, G.; Lu, K.; Umar, M.; Zhu, Z.; Lu, W.W.; Speakman, J.R.; Chen, Y.; Tong, L.; Chen, D. Risk of Metabolic Abnormalities in Osteoarthritis: A New Perspective to Understand Its Pathological Mechanisms. Bone Res. 2023, 11, 63. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, X.; Han, X.; Li, C.; Chen, Z.; Huang, H.; Chen, J.; Wu, C.; Fan, T.; Li, T.; Huang, W.; et al. 2D Materials for Bone Therapy. Adv. Drug Deliv. Rev. 2021, 178, 113970. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, C.; Yang, F.; Chen, R.; Yang, C.; Xiao, H.; Geng, B.; Xia, Y. TRPV Channels in Osteoarthritis: A Comprehensive Review. Biomolecules 2024, 14, 292. [Google Scholar] [CrossRef] [PubMed]
  4. Margiana, R.; Markov, A.; Zekiy, A.O.; Hamza, M.U.; Al-Dabbagh, K.A.; Al-Zubaidi, S.H.; Hameed, N.M.; Ahmad, I.; Sivaraman, R.; Kzar, H.H.; et al. Clinical Application of Mesenchymal Stem Cell in Regenerative Medicine: A Narrative Review. Stem Cell Res. Ther. 2022, 13, 366. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, L.; Han, X.; Qu, G.; Su, L.; Zhao, B.; Miao, J. A pH Probe Inhibits Senescence in Mesenchymal Stem Cells. Stem Cell Res. Ther. 2018, 9, 343. [Google Scholar] [CrossRef] [PubMed]
  6. Gonzalez-Fernandez, P.; Simula, L.; Jenni, S.; Jordan, O.; Allémann, E. Hyaluronan-Based Hydrogel Delivering Glucose to Mesenchymal Stem Cells Intended to Treat Osteoarthritis. Int. J. Pharm. 2024, 657, 124139. [Google Scholar] [CrossRef]
  7. Correa, S.; Grosskopf, A.K.; Lopez Hernandez, H.; Chan, D.; Yu, A.C.; Stapleton, L.M.; Appel, E.A. Translational Applications of Hydrogels. Chem. Rev. 2021, 121, 11385–11457. [Google Scholar] [CrossRef]
  8. Chen, J.; Pan, C.; Gao, Y.; Chen, Q.; An, X.; Liu, Z. Reactive Oxygen Species Scavenging Injectable Hydrogel Potentiates the Therapeutic Potential of Mesenchymal Stem Cells in Skin Flap Regeneration. ACS Appl. Mater. Interfaces 2024, 16, 17120–17128. [Google Scholar] [CrossRef]
  9. Kesharwani, P.; Alexander, A.; Shukla, R.; Jain, S.; Bisht, A.; Kumari, K.; Verma, K.; Sharma, S. Tissue Regeneration Properties of Hydrogels Derived from Biological Macromolecules: A Review. Int. J. Biol. Macromol. 2024, 271, 132280. [Google Scholar] [CrossRef]
  10. Gupta, A.; Lee, J.; Ghosh, T.; Nguyen, V.Q.; Dey, A.; Yoon, B.; Um, W.; Park, J.H. Polymeric Hydrogels for Controlled Drug Delivery to Treat Arthritis. Pharmaceutics 2022, 14, 540. [Google Scholar] [CrossRef]
  11. Zhang, H.; Lin, C.; Zeng, C.; Wang, Z.; Wang, H.; Lu, J.; Liu, X.; Shao, Y.; Zhao, C.; Pan, J.; et al. Synovial Macrophage M1 Polarisation Exacerbates Experimental Osteoarthritis Partially through R-Spondin-2. Ann. Rheum. Dis. 2018, 77, 1524–1534. [Google Scholar] [CrossRef] [PubMed]
  12. Guillemin, F.; Rat, A.C.; Mazieres, B.; Pouchot, J.; Fautrel, B.; Euller-Ziegler, L.; Fardellone, P.; Morvan, J.; Roux, C.H.; Verrouil, E.; et al. Prevalence of Symptomatic Hip and Knee Osteoarthritis: A Two-Phase Population-Based Survey. Osteoarthr. Cartil. 2011, 19, 1314–1322. [Google Scholar] [CrossRef] [PubMed]
  13. Yao, Q.; Wu, X.; Tao, C.; Gong, W.; Chen, M.; Qu, M.; Zhong, Y.; He, T.; Chen, S.; Xiao, G. Osteoarthritis: Pathogenic Signaling Pathways and Therapeutic Targets. Signal Transduct. Target. Ther. 2023, 8, 56. [Google Scholar] [CrossRef] [PubMed]
  14. Leifer, V.P.; Katz, J.N.; Losina, E. The Burden of OA-Health Services and Economics. Osteoarthr. Cartil. 2022, 30, 10–16. [Google Scholar] [CrossRef] [PubMed]
  15. Kim, J.-R.; Yoo, J.J.; Kim, H.A. Therapeutics in Osteoarthritis Based on an Understanding of Its Molecular Pathogenesis. Int. J. Mol. Sci. 2018, 19, 674. [Google Scholar] [CrossRef]
  16. Egloff, C.; Hügle, T.; Valderrabano, V. Biomechanics and Pathomechanisms of Osteoarthritis. Swiss Med. Wkly. 2012, 142, w13583. [Google Scholar] [CrossRef] [PubMed]
  17. Finnilä, M.A.J.; Das Gupta, S.; Turunen, M.J.; Hellberg, I.; Turkiewicz, A.; Lutz-Bueno, V.; Jonsson, E.; Holler, M.; Ali, N.; Hughes, V.; et al. Mineral Crystal Thickness in Calcified Cartilage and Subchondral Bone in Healthy and Osteoarthritic Human Knees. J. Bone Min. Res. 2022, 37, 1700–1710. [Google Scholar] [CrossRef] [PubMed]
  18. Hu, H.; Liu, W.; Sun, C.; Wang, Q.; Yang, W.; Zhang, Z.; Xia, Z.; Shao, Z.; Wang, B. Endogenous Repair and Regeneration of Injured Articular Cartilage: A Challenging but Promising Therapeutic Strategy. Aging Dis. 2021, 12, 886–901. [Google Scholar] [CrossRef] [PubMed]
  19. Schuurman, W.; Klein, T.J.; Dhert, W.J.A.; van Weeren, P.R.; Hutmacher, D.W.; Malda, J. Cartilage Regeneration Using Zonal Chondrocyte Subpopulations: A Promising Approach or an Overcomplicated Strategy? J. Tissue Eng. Regen. Med. 2015, 9, 669–678. [Google Scholar] [CrossRef]
  20. Loeser, R.F.; Collins, J.A.; Diekman, B.O. Ageing and the Pathogenesis of Osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 412–420. [Google Scholar] [CrossRef]
  21. Yuan, X.L.; Meng, H.Y.; Wang, Y.C.; Peng, J.; Guo, Q.Y.; Wang, A.Y.; Lu, S.B. Bone-Cartilage Interface Crosstalk in Osteoarthritis: Potential Pathways and Future Therapeutic Strategies. Osteoarthr. Cartil. 2014, 22, 1077–1089. [Google Scholar] [CrossRef] [PubMed]
  22. Gonzalez-Fernandez, P.; Rodríguez-Nogales, C.; Jordan, O.; Allémann, E. Combination of Mesenchymal Stem Cells and Bioactive Molecules in Hydrogels for Osteoarthritis Treatment. Eur. J. Pharm. Biopharm. 2022, 172, 41–52. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, Y.; Shah, K.M.; Luo, J. Strategies for Articular Cartilage Repair and Regeneration. Front. Bioeng. Biotechnol. 2021, 9, 770655. [Google Scholar] [CrossRef] [PubMed]
  24. Cai, X.; Yuan, S.; Zeng, Y.; Wang, C.; Yu, N.; Ding, C. New Trends in Pharmacological Treatments for Osteoarthritis. Front. Pharmacol. 2021, 12, 645842. [Google Scholar] [CrossRef] [PubMed]
  25. Szwed-Georgiou, A.; Płociński, P.; Kupikowska-Stobba, B.; Urbaniak, M.M.; Rusek-Wala, P.; Szustakiewicz, K.; Piszko, P.; Krupa, A.; Biernat, M.; Gazińska, M.; et al. Bioactive Materials for Bone Regeneration: Biomolecules and Delivery Systems. ACS Biomater. Sci. Eng. 2023, 9, 5222–5254. [Google Scholar] [CrossRef] [PubMed]
  26. Richard, M.J.; Driban, J.B.; McAlindon, T.E. Pharmaceutical Treatment of Osteoarthritis. Osteoarthr. Cartil. 2023, 31, 458–466. [Google Scholar] [CrossRef] [PubMed]
  27. DeRogatis, M.; Anis, H.K.; Sodhi, N.; Ehiorobo, J.O.; Chughtai, M.; Bhave, A.; Mont, M.A. Non-Operative Treatment Options for Knee Osteoarthritis. Ann. Transl. Med. 2019, 7, S245. [Google Scholar] [CrossRef] [PubMed]
  28. de l’Escalopier, N.; Anract, P.; Biau, D. Surgical Treatments for Osteoarthritis. Ann. Phys. Rehabil. Med. 2016, 59, 227–233. [Google Scholar] [CrossRef] [PubMed]
  29. Rannou, F.; Pelletier, J.-P.; Martel-Pelletier, J. Efficacy and Safety of Topical NSAIDs in the Management of Osteoarthritis: Evidence from Real-Life Setting Trials and Surveys. Semin. Arthritis Rheum. 2016, 45, S18–S21. [Google Scholar] [CrossRef]
  30. Hochberg, M.C.; Altman, R.D.; April, K.T.; Benkhalti, M.; Guyatt, G.; McGowan, J.; Towheed, T.; Welch, V.; Wells, G.; Tugwell, P. American College of Rheumatology 2012 Recommendations for the Use of Nonpharmacologic and Pharmacologic Therapies in Osteoarthritis of the Hand, Hip, and Knee. Arthritis Care Res. 2012, 64, 465–474. [Google Scholar] [CrossRef]
  31. Savvidou, O.; Milonaki, M.; Goumenos, S.; Flevas, D.; Papagelopoulos, P.; Moutsatsou, P. Glucocorticoid Signaling and Osteoarthritis. Mol. Cell Endocrinol. 2019, 480, 153–166. [Google Scholar] [CrossRef] [PubMed]
  32. Nees, T.A.; Rosshirt, N.; Zhang, J.A.; Reiner, T.; Sorbi, R.; Tripel, E.; Walker, T.; Schiltenwolf, M.; Hagmann, S.; Moradi, B. Synovial Cytokines Significantly Correlate with Osteoarthritis-Related Knee Pain and Disability: Inflammatory Mediators of Potential Clinical Relevance. J. Clin. Med. 2019, 8, 1343. [Google Scholar] [CrossRef] [PubMed]
  33. Messier, S.P.; Resnik, A.E.; Beavers, D.P.; Mihalko, S.L.; Miller, G.D.; Nicklas, B.J.; deVita, P.; Hunter, D.J.; Lyles, M.F.; Eckstein, F.; et al. Intentional Weight Loss in Overweight and Obese Patients with Knee Osteoarthritis: Is More Better? Arthritis Care Res. 2018, 70, 1569–1575. [Google Scholar] [CrossRef] [PubMed]
  34. Vincent, K.R.; Vasilopoulos, T.; Montero, C.; Vincent, H.K. Eccentric and Concentric Resistance Exercise Comparison for Knee Osteoarthritis. Med. Sci. Sports Exerc. 2019, 51, 1977–1986. [Google Scholar] [CrossRef] [PubMed]
  35. Franco, M.R.; Morelhão, P.K.; de Carvalho, A.; Pinto, R.Z. Aquatic Exercise for the Treatment of Hip and Knee Osteoarthritis. Phys. Ther. 2017, 97, 693–697. [Google Scholar] [CrossRef] [PubMed]
  36. Nguyen, C.; Lefèvre-Colau, M.-M.; Poiraudeau, S.; Rannou, F. Rehabilitation (Exercise and Strength Training) and Osteoarthritis: A Critical Narrative Review. Ann. Phys. Rehabil. Med. 2016, 59, 190–195. [Google Scholar] [CrossRef] [PubMed]
  37. Dias, J.M.; Cisneros, L.; Dias, R.; Fritsch, C.; Gomes, W.; Pereira, L.; Santos, M.L.; Ferreira, P.H. Hydrotherapy Improves Pain and Function in Older Women with Knee Osteoarthritis: A Randomized Controlled Trial. Braz. J. Phys. Ther. 2017, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  38. Dantas, L.O.; Osani, M.C.; Bannuru, R.R. Therapeutic Ultrasound for Knee Osteoarthritis: A Systematic Review and Meta-Analysis with Grade Quality Assessment. Braz. J. Phys. Ther. 2021, 25, 688–697. [Google Scholar] [CrossRef] [PubMed]
  39. Huang, Z.; Chen, J.; Ma, J.; Shen, B.; Pei, F.; Kraus, V.B. Effectiveness of Low-Level Laser Therapy in Patients with Knee Osteoarthritis: A Systematic Review and Meta-Analysis. Osteoarthr. Cartil. 2015, 23, 1437–1444. [Google Scholar] [CrossRef]
  40. Flynn, D.M. Chronic Musculoskeletal Pain: Nonpharmacologic, Noninvasive Treatments. Am. Fam. Phys. 2020, 102, 465–477. [Google Scholar]
  41. Lo, C.W.T.; Tsang, W.W.N.; Yan, C.H.; Lord, S.R.; Hill, K.D.; Wong, A.Y.L. Risk Factors for Falls in Patients with Total Hip Arthroplasty and Total Knee Arthroplasty: A Systematic Review and Meta-Analysis. Osteoarthr. Cartil. 2019, 27, 979–993. [Google Scholar] [CrossRef] [PubMed]
  42. Katz, J.N.; Brownlee, S.A.; Jones, M.H. The Role of Arthroscopy in the Management of Knee Osteoarthritis. Best Pract. Res. Clin. Rheumatol. 2014, 28, 143–156. [Google Scholar] [CrossRef] [PubMed]
  43. Ma, S.; Xie, N.; Li, W.; Yuan, B.; Shi, Y.; Wang, Y. Immunobiology of Mesenchymal Stem Cells. Cell Death Differ. 2014, 21, 216–225. [Google Scholar] [CrossRef] [PubMed]
  44. Li, X.; Wang, M.; Jing, X.; Guo, W.; Hao, C.; Zhang, Y.; Gao, S.; Chen, M.; Zhang, Z.; Zhang, X.; et al. Bone Marrow- and Adipose Tissue-Derived Mesenchymal Stem Cells: Characterization, Differentiation, and Applications in Cartilage Tissue Engineering. Crit. Rev. Eukaryot. Gene Expr. 2018, 28, 285–310. [Google Scholar] [CrossRef] [PubMed]
  45. Aprile, D.; Patrone, D.; Peluso, G.; Galderisi, U. Multipotent/Pluripotent Stem Cell Populations in Stromal Tissues and Peripheral Blood: Exploring Diversity, Potential, and Therapeutic Applications. Stem Cell Res. Ther. 2024, 15, 139. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, S.; Sun, S.; Fu, W.; Yang, Z.; Yao, H.; Zhang, Z. The Role and Prospects of Mesenchymal Stem Cells in Skin Repair and Regeneration. Biomedicines 2024, 12, 743. [Google Scholar] [CrossRef] [PubMed]
  47. Han, Y.; Yang, J.; Fang, J.; Zhou, Y.; Candi, E.; Wang, J.; Hua, D.; Shao, C.; Shi, Y. The Secretion Profile of Mesenchymal Stem Cells and Potential Applications in Treating Human Diseases. Signal Transduct. Target. Ther. 2022, 7, 92. [Google Scholar] [CrossRef] [PubMed]
  48. Widera, D. Recent Advances in Translational Adipose-Derived Stem Cell Biology. Biomolecules 2021, 11, 1660. [Google Scholar] [CrossRef] [PubMed]
  49. Ullah, M.; Liu, D.D.; Thakor, A.S. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience 2019, 15, 421–438. [Google Scholar] [CrossRef]
  50. Gnecchi, M.; Melo, L.G. Bone Marrow-Derived Mesenchymal Stem Cells: Isolation, Expansion, Characterization, Viral Transduction, and Production of Conditioned Medium. Methods Mol. Biol. 2009, 482, 281–294. [Google Scholar] [CrossRef]
  51. Abu-El-Rub, E.; Khaswaneh, R.R.; Almahasneh, F.A.; Almazari, R.; Alzu’bi, A. Adipose Tissue and Bone Marrow-Derived Mesenchymal Stem Cells Are Not Really the Same: Investigating the Differences in Their Immunomodulatory, Migratory, and Adhesive Profile. Biochem. Genet. 2024; Online ahead of print. [Google Scholar] [CrossRef]
  52. Aldrich, E.D.; Cui, X.; Murphy, C.A.; Lim, K.S.; Hooper, G.J.; McIlwraith, C.W.; Woodfield, T.B.F. Allogeneic Mesenchymal Stromal Cells for Cartilage Regeneration: A Review of in Vitro Evaluation, Clinical Experience, and Translational Opportunities. Stem Cells Transl. Med. 2021, 10, 1500–1515. [Google Scholar] [CrossRef] [PubMed]
  53. Adipose-Derived Stem Cells in Immune-Related Skin Disease: A Review of Current Research and Underlying Mechanisms. Available online: https://pubmed.ncbi.nlm.nih.gov/38331803/ (accessed on 27 May 2024).
  54. Tan, L.; Liu, X.; Dou, H.; Hou, Y. Characteristics and Regulation of Mesenchymal Stem Cell Plasticity by the Microenvironment—Specific Factors Involved in the Regulation of MSC Plasticity. Genes Dis. 2022, 9, 296–309. [Google Scholar] [CrossRef] [PubMed]
  55. Rosa, I.; Romano, E.; Fioretto, B.S.; Matucci-Cerinic, M.; Manetti, M. Adipose-Derived Stem Cells: Pathophysiologic Implications vs Therapeutic Potential in Systemic Sclerosis. World J. Stem Cells 2021, 13, 30–48. [Google Scholar] [CrossRef] [PubMed]
  56. Maldonado, V.V.; Patel, N.H.; Smith, E.E.; Barnes, C.L.; Gustafson, M.P.; Rao, R.R.; Samsonraj, R.M. Clinical Utility of Mesenchymal Stem/Stromal Cells in Regenerative Medicine and Cellular Therapy. J. Biol. Eng. 2023, 17, 44. [Google Scholar] [CrossRef] [PubMed]
  57. Shen, F.; Xiao, H.; Shi, Q. Mesenchymal Stem Cells Derived from the Fibrotic Tissue of Atrophic Nonunion or the Bone Marrow of Iliac Crest: A Donor-Matched Comparison. Regen. Ther. 2023, 24, 398–406. [Google Scholar] [CrossRef] [PubMed]
  58. Granero-Moltó, F.; Weis, J.A.; Miga, M.I.; Landis, B.; Myers, T.J.; O’Rear, L.; Longobardi, L.; Jansen, E.D.; Mortlock, D.P.; Spagnoli, A. Regenerative Effects of Transplanted Mesenchymal Stem Cells in Fracture Healing. Stem Cells 2009, 27, 1887–1898. [Google Scholar] [CrossRef] [PubMed]
  59. Cofano, F.; Boido, M.; Monticelli, M.; Zenga, F.; Ducati, A.; Vercelli, A.; Garbossa, D. Mesenchymal Stem Cells for Spinal Cord Injury: Current Options, Limitations, and Future of Cell Therapy. Int. J. Mol. Sci. 2019, 20, 2698. [Google Scholar] [CrossRef]
  60. Yousefifard, M.; Nasirinezhad, F.; Shardi Manaheji, H.; Janzadeh, A.; Hosseini, M.; Keshavarz, M. Human Bone Marrow-Derived and Umbilical Cord-Derived Mesenchymal Stem Cells for Alleviating Neuropathic Pain in a Spinal Cord Injury Model. Stem Cell Res. Ther. 2016, 7, 36. [Google Scholar] [CrossRef] [PubMed]
  61. Choi, M.R.; Kim, H.Y.; Park, J.-Y.; Lee, T.Y.; Baik, C.S.; Chai, Y.G.; Jung, K.H.; Park, K.S.; Roh, W.; Kim, K.S.; et al. Selection of Optimal Passage of Bone Marrow-Derived Mesenchymal Stem Cells for Stem Cell Therapy in Patients with Amyotrophic Lateral Sclerosis. Neurosci. Lett. 2010, 472, 94–98. [Google Scholar] [CrossRef]
  62. Moonshi, S.S.; Adelnia, H.; Wu, Y.; Ta, H.T. Placenta-Derived Mesenchymal Stem Cells for Treatment of Diseases: A Clinically Relevant Source. Adv. Ther. 2022, 5, 2200054. [Google Scholar] [CrossRef]
  63. Rhim, J.; Ha, C.-W.; Park, Y.-B.; Kim, J.-A.; Han, W.-J.; Choi, S.; Lee, K.; Park, H.; Park, H.-J. Cartilage Repair by Various Concentrations of Placenta-Derived Mesenchymal Stem Cells and Hyaluronic Acid Hydrogels in a Rabbit Model. Osteoarthr. Cartil. 2017, 25, S160. [Google Scholar] [CrossRef]
  64. Zhang, Y.; Zhong, Y.; Zou, L.; Liu, X. Significance of Placental Mesenchymal Stem Cell in Placenta Development and Implications for Preeclampsia. Front. Pharmacol. 2022, 13, 896531. [Google Scholar] [CrossRef]
  65. Wu, M.; Zhang, R.; Zou, Q.; Chen, Y.; Zhou, M.; Li, X.; Ran, R.; Chen, Q. Comparison of the Biological Characteristics of Mesenchymal Stem Cells Derived from the Human Placenta and Umbilical Cord. Sci. Rep. 2018, 8, 5014. [Google Scholar] [CrossRef] [PubMed]
  66. James, J.L.; Umapathy, A.; Srinivasan, S.; Barker, C.N.; Brooks, A.; Hearn, J.; Chhana, A.; Williams, E.; Sheppard, H.; McGlashan, S.R. The Chondrogenic Potential of First-Trimester and Term Placental Mesenchymal Stem/Stromal Cells. Cartilage 2021, 13, 544S–558S. [Google Scholar] [CrossRef]
  67. Vellasamy, S.; Sandrasaigaran, P.; Vidyadaran, S.; George, E.; Ramasamy, R. Isolation and Characterisation of Mesenchymal Stem Cells Derived from Human Placenta Tissue. World J. Stem Cells 2012, 4, 53–61. [Google Scholar] [CrossRef]
  68. Ahani-Nahayati, M.; Niazi, V.; Moradi, A.; Pourjabbar, B.; Roozafzoon, R.; Keshel, S.H.; Baradaran-Rafii, A. Umbilical Cord Mesenchymal Stem/Stromal Cells Potential to Treat Organ Disorders; An Emerging Strategy. Curr. Stem Cell Res. Ther. 2022, 17, 126–146. [Google Scholar] [CrossRef]
  69. Zhang, R.; Chen, H.; Zheng, Z.; Liu, Q.; Xu, L. Umbilical Cord-Derived Mesenchymal Stem Cell Therapy for Neurological Disorders via Inhibition of Mitogen-Activated Protein Kinase Pathway-Mediated Apoptosis. Mol. Med. Rep. 2015, 11, 1807–1812. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, Y.; Shen, H.; Ding, Y.; Yu, Y.; Shao, L.; Shen, Z. The Application of Umbilical Cord-Derived MSCs in Cardiovascular Diseases. J. Cell Mol. Med. 2021, 25, 8103–8114. [Google Scholar] [CrossRef] [PubMed]
  71. Abbaspanah, B.; Reyhani, S.; Mousavi, S.H. Applications of Umbilical Cord Derived Mesenchymal Stem Cells in Autoimmune and Immunological Disorders: From Literature to Clinical Practice. Curr. Stem Cell Res. Ther. 2021, 16, 454–464. [Google Scholar] [CrossRef]
  72. Gadelkarim, M.; Abushouk, A.I.; Ghanem, E.; Hamaad, A.M.; Saad, A.M.; Abdel-Daim, M.M. Adipose-Derived Stem Cells: Effectiveness and Advances in Delivery in Diabetic Wound Healing. Biomed. Pharmacother. 2018, 107, 625–633. [Google Scholar] [CrossRef]
  73. Sheng, L.; Yang, M.; Liang, Y.; Li, Q. Adipose Tissue-Derived Stem Cells (ADSCs) Transplantation Promotes Regeneration of Expanded Skin Using a Tissue Expansion Model. Wound Repair. Regen. 2013, 21, 746–754. [Google Scholar] [CrossRef]
  74. Cherubino, M.; Marra, K.G. Adipose-Derived Stem Cells for Soft Tissue Reconstruction. Regen. Med. 2009, 4, 109–117. [Google Scholar] [CrossRef] [PubMed]
  75. Yan, D.; Song, Y.; Zhang, B.; Cao, G.; Zhou, H.; Li, H.; Sun, H.; Deng, M.; Qiu, Y.; Yi, W.; et al. Progress and Application of Adipose-Derived Stem Cells in the Treatment of Diabetes and Its Complications. Stem Cell Res. Ther. 2024, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  76. Jeyaraman, M.; Muthu, S.; Jeyaraman, N.; Ranjan, R.; Jha, S.K.; Mishra, P. Synovium Derived Mesenchymal Stromal Cells (Sy-MSCs): A Promising Therapeutic Paradigm in the Management of Knee Osteoarthritis. Indian J. Orthop. 2022, 56, 1–15. [Google Scholar] [CrossRef]
  77. Yields and Chondrogenic Potential of Primary Synovial Mesenchymal Stem Cells Are Comparable between Rheumatoid Arthritis and Osteoarthritis Patients. Available online: https://pubmed.ncbi.nlm.nih.gov/28511664/ (accessed on 28 May 2024).
  78. Li, N.; Gao, J.; Mi, L.; Zhang, G.; Zhang, L.; Zhang, N.; Huo, R.; Hu, J.; Xu, K. Synovial Membrane Mesenchymal Stem Cells: Past Life, Current Situation, and Application in Bone and Joint Diseases. Stem Cell Res. Ther. 2020, 11, 381. [Google Scholar] [CrossRef]
  79. Jorgenson, K.D.; Hart, D.A.; Krawetz, R.; Sen, A. Production of Adult Human Synovial Fluid-Derived Mesenchymal Stem Cells in Stirred-Suspension Culture. Stem Cells Int. 2018, 2018, 8431053. [Google Scholar] [CrossRef]
  80. Insensitive Effects of Inflammatory Cytokines on the Reference Genes of Synovial Fluid Resident-Mesenchymal Stem Cells Derived from Rheumatoid Arthritis Patients. Available online: https://pubmed.ncbi.nlm.nih.gov/37894839/ (accessed on 28 May 2024).
  81. Meng, J.; Adkin, C.F.; Arechavala-Gomeza, V.; Boldrin, L.; Muntoni, F.; Morgan, J.E. The Contribution of Human Synovial Stem Cells to Skeletal Muscle Regeneration. Neuromuscul. Disord. 2010, 20, 6–15. [Google Scholar] [CrossRef]
  82. Botelho, J.; Cavacas, M.A.; Machado, V.; Mendes, J.J. Dental Stem Cells: Recent Progresses in Tissue Engineering and Regenerative Medicine. Ann. Med. 2017, 49, 644–651. [Google Scholar] [CrossRef]
  83. Chalisserry, E.P.; Nam, S.Y.; Park, S.H.; Anil, S. Therapeutic Potential of Dental Stem Cells. J. Tissue Eng. 2017, 8, 2041731417702531. [Google Scholar] [CrossRef] [PubMed]
  84. Potdar, P.D.; Jethmalani, Y.D. Human Dental Pulp Stem Cells: Applications in Future Regenerative Medicine. World J. Stem Cells 2015, 7, 839–851. [Google Scholar] [CrossRef]
  85. Al-Maswary, A.A.; O’Reilly, M.; Holmes, A.P.; Walmsley, A.D.; Cooper, P.R.; Scheven, B.A. Exploring the Neurogenic Differentiation of Human Dental Pulp Stem Cells. PLoS ONE 2022, 17, e0277134. [Google Scholar] [CrossRef]
  86. Song, B.; Jiang, W.; Alraies, A.; Liu, Q.; Gudla, V.; Oni, J.; Wei, X.; Sloan, A.; Ni, L.; Agarwal, M. Bladder Smooth Muscle Cells Differentiation from Dental Pulp Stem Cells: Future Potential for Bladder Tissue Engineering. Stem Cells Int. 2016, 2016, 6979368. [Google Scholar] [CrossRef]
  87. Corradetti, B.; Meucci, A.; Bizzaro, D.; Cremonesi, F.; Lange Consiglio, A. Mesenchymal Stem Cells from Amnion and Amniotic Fluid in the Bovine. Reproduction 2013, 145, 391–400. [Google Scholar] [CrossRef] [PubMed]
  88. Kim, E.Y.; Lee, K.-B.; Kim, M.K. The Potential of Mesenchymal Stem Cells Derived from Amniotic Membrane and Amniotic Fluid for Neuronal Regenerative Therapy. BMB Rep. 2014, 47, 135–140. [Google Scholar] [CrossRef] [PubMed]
  89. Liu, Q.-W.; Ying, Y.-M.; Zhou, J.-X.; Zhang, W.-J.; Liu, Z.; Jia, B.-B.; Gu, H.-C.; Zhao, C.-Y.; Guan, X.-H.; Deng, K.-Y.; et al. Human Amniotic Mesenchymal Stem Cells-Derived IGFBP-3, DKK-3, and DKK-1 Attenuate Liver Fibrosis through Inhibiting Hepatic Stellate Cell Activation by Blocking Wnt/β-Catenin Signaling Pathway in Mice. Stem Cell Res. Ther. 2022, 13, 224. [Google Scholar] [CrossRef]
  90. Navas, A.; Magaña-Guerrero, F.S.; Domínguez-López, A.; Chávez-García, C.; Partido, G.; Graue-Hernández, E.O.; Sánchez-García, F.J.; Garfias, Y. Anti-Inflammatory and Anti-Fibrotic Effects of Human Amniotic Membrane Mesenchymal Stem Cells and Their Potential in Corneal Repair. Stem Cells Transl. Med. 2018, 7, 906–917. [Google Scholar] [CrossRef] [PubMed]
  91. Amniotic Membrane Mesenchymal Cells-Derived Factors Skew T Cell Polarization toward Treg and Downregulate Th1 and Th17 Cells Subsets. Available online: https://pubmed.ncbi.nlm.nih.gov/25348066/ (accessed on 28 May 2024).
  92. Nombela-Arrieta, C.; Ritz, J.; Silberstein, L.E. The Elusive Nature and Function of Mesenchymal Stem Cells. Nat. Rev. Mol. Cell Biol. 2011, 12, 126–131. [Google Scholar] [CrossRef]
  93. Maged, G.; Abdelsamed, M.A.; Wang, H.; Lotfy, A. The Potency of Mesenchymal Stem/Stromal Cells: Does Donor Sex Matter? Stem Cell Res. Ther. 2024, 15, 112. [Google Scholar] [CrossRef] [PubMed]
  94. Ding, D.-C.; Shyu, W.-C.; Lin, S.-Z. Mesenchymal Stem Cells. Cell Transpl. 2011, 20, 5–14. [Google Scholar] [CrossRef]
  95. Samsonraj, R.M.; Raghunath, M.; Nurcombe, V.; Hui, J.H.; van Wijnen, A.J.; Cool, S.M. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl. Med. 2017, 6, 2173–2185. [Google Scholar] [CrossRef]
  96. Koç, O.N.; Gerson, S.L.; Cooper, B.W.; Dyhouse, S.M.; Haynesworth, S.E.; Caplan, A.I.; Lazarus, H.M. Rapid Hematopoietic Recovery after Coinfusion of Autologous-Blood Stem Cells and Culture-Expanded Marrow Mesenchymal Stem Cells in Advanced Breast Cancer Patients Receiving High-Dose Chemotherapy. J. Clin. Oncol. 2000, 18, 307–316. [Google Scholar] [CrossRef] [PubMed]
  97. Rojas, M.; Xu, J.; Woods, C.R.; Mora, A.L.; Spears, W.; Roman, J.; Brigham, K.L. Bone Marrow-Derived Mesenchymal Stem Cells in Repair of the Injured Lung. Am. J. Respir. Cell Mol. Biol. 2005, 33, 145–152. [Google Scholar] [CrossRef] [PubMed]
  98. Jung, S.; Kim, J.-H.; Yim, C.; Lee, M.; Kang, H.J.; Choi, D. Therapeutic Effects of a Mesenchymal Stem Cell-based Insulin-like Growth Factor-1/Enhanced Green Fluorescent Protein Dual Gene Sorting System in a Myocardial Infarction Rat Model. Mol. Med. Rep. 2018, 18, 5563–5571. [Google Scholar] [CrossRef] [PubMed]
  99. Hu, C.; Wu, Z.; Li, L. Mesenchymal Stromal Cells Promote Liver Regeneration through Regulation of Immune Cells. Int. J. Biol. Sci. 2020, 16, 893–903. [Google Scholar] [CrossRef] [PubMed]
  100. Peired, A.J.; Sisti, A.; Romagnani, P. Mesenchymal Stem Cell-Based Therapy for Kidney Disease: A Review of Clinical Evidence. Stem Cells Int. 2016, 2016, 4798639. [Google Scholar] [CrossRef] [PubMed]
  101. Richardson, S.M.; Kalamegam, G.; Pushparaj, P.N.; Matta, C.; Memic, A.; Khademhosseini, A.; Mobasheri, R.; Poletti, F.L.; Hoyland, J.A.; Mobasheri, A. Mesenchymal Stem Cells in Regenerative Medicine: Focus on Articular Cartilage and Intervertebral Disc Regeneration. Methods 2016, 99, 69–80. [Google Scholar] [CrossRef] [PubMed]
  102. Huang, S.; Song, X.; Li, T.; Xiao, J.; Chen, Y.; Gong, X.; Zeng, W.; Yang, L.; Chen, C. Pellet Coculture of Osteoarthritic Chondrocytes and Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells with Chitosan/Hyaluronic Acid Nanoparticles Promotes Chondrogenic Differentiation. Stem Cell Res. Ther. 2017, 8, 264. [Google Scholar] [CrossRef] [PubMed]
  103. Muñoz-Criado, I.; Meseguer-Ripolles, J.; Mellado-López, M.; Alastrue-Agudo, A.; Griffeth, R.J.; Forteza-Vila, J.; Cugat, R.; García, M.; Moreno-Manzano, V. Human Suprapatellar Fat Pad-Derived Mesenchymal Stem Cells Induce Chondrogenesis and Cartilage Repair in a Model of Severe Osteoarthritis. Stem Cells Int. 2017, 2017, 4758930. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Q.; Chen, Y.; Wang, Q.; Fang, C.; Sun, Y.; Yuan, T.; Wang, Y.; Bao, R.; Zhao, N. Effect of Bone Marrow-Derived Stem Cells on Chondrocytes from Patients with Osteoarthritis. Mol. Med. Rep. 2016, 13, 1795–1800. [Google Scholar] [CrossRef]
  105. Li, K.; Yan, G.; Huang, H.; Zheng, M.; Ma, K.; Cui, X.; Lu, D.; Zheng, L.; Zhu, B.; Cheng, J.; et al. Anti-Inflammatory and Immunomodulatory Effects of the Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells on Osteoarthritis via M2 Macrophages. J. Nanobiotechnol. 2022, 20, 38. [Google Scholar] [CrossRef]
  106. Vahedi, P.; Roshangar, L.; Jarolmasjed, S.; Shafaei, H.; Samadi, N.; Soleimanirad, J. Effect of Low-Intensity Pulsed Ultrasound on Regenerative Potential of Transplanted ASCs €“PCL Construct in Articular Cartilage Defects in Sheep. Indian J. Anim. Sci. 2016, 86, 111–1114. [Google Scholar] [CrossRef]
  107. Lopa, S.; Mondadori, C.; Mainardi, V.L.; Talò, G.; Costantini, M.; Candrian, C.; Święszkowski, W.; Moretti, M. Translational Application of Microfluidics and Bioprinting for Stem Cell-Based Cartilage Repair. Stem Cells Int. 2018, 2018, 6594841. [Google Scholar] [CrossRef] [PubMed]
  108. Fink, T.; Rasmussen, J.G.; Emmersen, J.; Pilgaard, L.; Fahlman, Å.; Brunberg, S.; Josefsson, J.; Arnemo, J.M.; Zachar, V.; Swenson, J.E.; et al. Adipose-Derived Stem Cells from the Brown Bear (Ursus Arctos) Spontaneously Undergo Chondrogenic and Osteogenic Differentiation in Vitro. Stem Cell Res. 2011, 7, 89–95. [Google Scholar] [CrossRef] [PubMed]
  109. Derfoul, A.; Perkins, G.L.; Hall, D.J.; Tuan, R.S. Glucocorticoids Promote Chondrogenic Differentiation of Adult Human Mesenchymal Stem Cells by Enhancing Expression of Cartilage Extracellular Matrix Genes. Stem Cells 2006, 24, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  110. Jin, Y.; Xu, M.; Zhu, H.; Dong, C.; Ji, J.; Liu, Y.; Deng, A.; Gu, Z. Therapeutic Effects of Bone Marrow Mesenchymal Stem Cells-Derived Exosomes on Osteoarthritis. J. Cell Mol. Med. 2021, 25, 9281–9294. [Google Scholar] [CrossRef]
  111. Molnar, V.; Pavelić, E.; Vrdoljak, K.; Čemerin, M.; Klarić, E.; Matišić, V.; Bjelica, R.; Brlek, P.; Kovačić, I.; Tremolada, C.; et al. Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review. Genes 2022, 13, 949. [Google Scholar] [CrossRef]
  112. Wei, Z.-J.; Wang, Q.-Q.; Cui, Z.-G.; Inadera, H.; Jiang, X.; Wu, C.-A. Which Is the Most Effective One in Knee Osteoarthritis Treatment from Mesenchymal Stem Cells Obtained from Different Sources?—A Systematic Review with Conventional and Network Meta-Analyses of Randomized Controlled Trials. Ann. Transl. Med. 2021, 9, 452. [Google Scholar] [CrossRef] [PubMed]
  113. Jo, C.H.; Lee, Y.G.; Shin, W.H.; Kim, H.; Chai, J.W.; Jeong, E.C.; Kim, J.E.; Shim, H.; Shin, J.S.; Shin, I.S.; 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 2014, 32, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
  114. Spasovski, D.; Spasovski, V.; Baščarević, Z.; Stojiljković, M.; Vreća, M.; Anđelković, M.; Pavlović, S. Intra-Articular Injection of Autologous Adipose-Derived Mesenchymal Stem Cells in the Treatment of Knee Osteoarthritis. J. Gene Med. 2018, 20, e3002. [Google Scholar] [CrossRef]
  115. Zhu, C.; Wu, W.; Qu, X. Mesenchymal Stem Cells in Osteoarthritis Therapy: A Review. Am. J. Transl. Res. 2021, 13, 448–461. [Google Scholar]
  116. Cheng, J.-H.; Hsu, C.-C.; Hsu, S.-L.; Chou, W.-Y.; Wu, Y.-N.; Kuo, C.-E.A.; Hsu, T.-C.; Shiu, L.-Y.; Jhan, S.-W. Adipose-Derived Mesenchymal Stem Cells-Conditioned Medium Modulates the Expression of Inflammation Induced Bone Morphogenetic Protein-2, -5 and -6 as Well as Compared with Shockwave Therapy on Rat Knee Osteoarthritis. Biomedicines 2021, 9, 1399. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, R.; Meng, F.; Zhang, Q.; Zou, Z.; Xiao, K.; Zhu, T.; Li, H.; Zhang, W.; Ma, J.; Ma, J. Allogeneic Adipose-Derived Mesenchymal Stem Cells Promote the Expression of Chondrocyte Redifferentiation Markers and Retard the Progression of Knee Osteoarthritis in Rabbits. Am. J. Transl. Res. 2021, 13, 632–645. [Google Scholar] [PubMed]
  118. Song, Y.; Du, H.; Dai, C.; Zhang, L.; Li, S.; Hunter, D.J.; Lu, L.; Bao, C. Human Adipose-Derived Mesenchymal Stem Cells for Osteoarthritis: A Pilot Study with Long-Term Follow-up and Repeated Injections. Regen. Med. 2018, 13, 295–307. [Google Scholar] [CrossRef] [PubMed]
  119. Kouroupis, D.; Willman, M.A.; Best, T.M.; Kaplan, L.D.; Correa, D. Infrapatellar Fat Pad-Derived Mesenchymal Stem Cell-Based Spheroids Enhance Their Therapeutic Efficacy to Reverse Synovitis and Fat Pad Fibrosis. Stem Cell Res. Ther. 2021, 12, 44. [Google Scholar] [CrossRef] [PubMed]
  120. Platas, J.; Guillén, M.I.; del Caz, M.D.P.; Gomar, F.; Mirabet, V.; Alcaraz, M.J. Conditioned Media from Adipose-Tissue-Derived Mesenchymal Stem Cells Downregulate Degradative Mediators Induced by Interleukin-1β in Osteoarthritic Chondrocytes. Mediat. Inflamm. 2013, 2013, 357014. [Google Scholar] [CrossRef] [PubMed]
  121. Kuroda, R.; Ishida, K.; Matsumoto, T.; Akisue, T.; Fujioka, H.; Mizuno, K.; Ohgushi, H.; Wakitani, S.; Kurosaka, M. Treatment of a Full-Thickness Articular Cartilage Defect in the Femoral Condyle of an Athlete with Autologous Bone-Marrow Stromal Cells. Osteoarthr. Cartil. 2007, 15, 226–231. [Google Scholar] [CrossRef] [PubMed]
  122. Orozco, L.; Munar, A.; Soler, R.; Alberca, M.; Soler, F.; Huguet, M.; Sentís, J.; Sánchez, A.; García-Sancho, J. Treatment of Knee Osteoarthritis with Autologous Mesenchymal Stem Cells: Two-Year Follow-up Results. Transplantation 2014, 97, e66–e68. [Google Scholar] [CrossRef] [PubMed]
  123. 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. 2019, 8, 746–757. [Google Scholar] [CrossRef] [PubMed]
  124. Carlo, D.; Stefano, B.; Nicholas, E.; Roberto, V.; Elena, M.S.; Bruno, M. Mesenchymal Stem Cells Injection in Hip Osteoarthritis: Preliminary Results. Acta Biomed. 2019, 90, 75–80. [Google Scholar] [CrossRef]
  125. Denoeud, C.; Luo, G.; Paquet, J.; Boisselier, J.; Wosinski, P.; Moya, A.; Diallo, A.; Larochette, N.; Marinesco, S.; Meiller, A.; et al. Enzyme-Controlled, Nutritive Hydrogel for Mesenchymal Stromal Cell Survival and Paracrine Functions. Commun. Biol. 2023, 6, 1266. [Google Scholar] [CrossRef]
  126. Amer, M.H.; Rose, F.R.A.J.; Shakesheff, K.M.; Modo, M.; White, L.J. Translational Considerations in Injectable Cell-Based Therapeutics for Neurological Applications: Concepts, Progress and Challenges. NPJ Regen. Med. 2017, 2, 23. [Google Scholar] [CrossRef] [PubMed]
  127. Levy, O.; Kuai, R.; Siren, E.M.J.; Bhere, D.; Milton, Y.; Nissar, N.; De Biasio, M.; Heinelt, M.; Reeve, B.; Abdi, R.; et al. Shattering Barriers toward Clinically Meaningful MSC Therapies. Sci. Adv. 2020, 6, eaba6884. [Google Scholar] [CrossRef] [PubMed]
  128. Music, E.; Futrega, K.; Doran, M.R. Sheep as a Model for Evaluating Mesenchymal Stem/Stromal Cell (MSC)-Based Chondral Defect Repair. Osteoarthr. Cartil. 2018, 26, 730–740. [Google Scholar] [CrossRef] [PubMed]
  129. Sabapathy, V.; Kumar, S. hiPSC-Derived iMSCs: NextGen MSCs as an Advanced Therapeutically Active Cell Resource for Regenerative Medicine. J. Cell Mol. Med. 2016, 20, 1571–1588. [Google Scholar] [CrossRef] [PubMed]
  130. Ahmed, E.M. Hydrogel: Preparation, Characterization, and Applications: A Review. J. Adv. Res. 2015, 6, 105–121. [Google Scholar] [CrossRef] [PubMed]
  131. Ahmad, Z.; Salman, S.; Khan, S.A.; Amin, A.; Rahman, Z.U.; Al-Ghamdi, Y.O.; Akhtar, K.; Bakhsh, E.M.; Khan, S.B. Versatility of Hydrogels: From Synthetic Strategies, Classification, and Properties to Biomedical Applications. Gels 2022, 8, 167. [Google Scholar] [CrossRef] [PubMed]
  132. Ho, T.-C.; Chang, C.-C.; Chan, H.-P.; Chung, T.-W.; Shu, C.-W.; Chuang, K.-P.; Duh, T.-H.; Yang, M.-H.; Tyan, Y.-C. Hydrogels: Properties and Applications in Biomedicine. Molecules 2022, 27, 2902. [Google Scholar] [CrossRef] [PubMed]
  133. Balakrishnan, B.; Jayakrishnan, A. Self-Cross-Linking Biopolymers as Injectable in Situ Forming Biodegradable Scaffolds. Biomaterials 2005, 26, 3941–3951. [Google Scholar] [CrossRef] [PubMed]
  134. Mahinroosta, M.; Jomeh Farsangi, Z.; Allahverdi, A.; Shakoori, Z. Hydrogels as Intelligent Materials: A Brief Review of Synthesis, Properties and Applications. Mater. Today Chem. 2018, 8, 42–55. [Google Scholar] [CrossRef]
  135. Catoira, M.C.; Fusaro, L.; Di Francesco, D.; Ramella, M.; Boccafoschi, F. Overview of Natural Hydrogels for Regenerative Medicine Applications. J. Mater. Sci. Mater. Med. 2019, 30, 115. [Google Scholar] [CrossRef]
  136. Vijayavenkataraman, S.; Vialli, N.; Fuh, J.Y.H.; Lu, W.F. Conductive Collagen/Polypyrrole-b-Polycaprolactone Hydrogel for Bioprinting of Neural Tissue Constructs. Int. J. Bioprint 2019, 5, 229. [Google Scholar] [CrossRef] [PubMed]
  137. Bonelli, N.; Poggi, G.; Chelazzi, D.; Giorgi, R.; Baglioni, P. Poly(Vinyl Alcohol)/Poly(Vinyl Pyrrolidone) Hydrogels for the Cleaning of Art. J. Colloid. Interface Sci. 2019, 536, 339–348. [Google Scholar] [CrossRef] [PubMed]
  138. Saffer, E.M.; Tew, G.N.; Bhatia, S.R. Poly(Lactic Acid)-Poly(Ethylene Oxide) Block Copolymers: New Directions in Self-Assembly and Biomedical Applications. Curr. Med. Chem. 2011, 18, 5676–5686. [Google Scholar] [CrossRef] [PubMed]
  139. Atta, S.; Khaliq, S.; Islam, A.; Javeria, I.; Jamil, T.; Athar, M.M.; Shafiq, M.I.; Ghaffar, A. Injectable Biopolymer Based Hydrogels for Drug Delivery Applications. Int. J. Biol. Macromol. 2015, 80, 240–245. [Google Scholar] [CrossRef]
  140. Rodríguez Rodríguez, R.; Garcia, Z.; Jimenez-Palomar, I.; Avalos, J.; Espinosa-Andrews, H. Development of Gelatin/Chitosan/PVA Hydrogels: Thermal Stability, Water State, Viscoelasticity, and Cytotoxicity Assays. J. Appl. Polym. Sci. 2019, 136, 47149. [Google Scholar] [CrossRef]
  141. Nagase, K.; Yamato, M.; Kanazawa, H.; Okano, T. Poly(N-Isopropylacrylamide)-Based Thermoresponsive Surfaces Provide New Types of Biomedical Applications. Biomaterials 2018, 153, 27–48. [Google Scholar] [CrossRef] [PubMed]
  142. McMasters, J.; Poh, S.; Lin, J.B.; Panitch, A. Delivery of Anti-Inflammatory Peptides from Hollow PEGylated Poly(NIPAM) Nanoparticles Reduces Inflammation in an Ex Vivo Osteoarthritis Model. J. Control Release 2017, 258, 161–170. [Google Scholar] [CrossRef] [PubMed]
  143. Lu, K.-Y.; Lin, Y.-C.; Lu, H.-T.; Ho, Y.-C.; Weng, S.-C.; Tsai, M.-L.; Mi, F.-L. A Novel Injectable in Situ Forming Gel Based on Carboxymethyl Hexanoyl Chitosan/Hyaluronic Acid Polymer Blending for Sustained Release of Berberine. Carbohydr. Polym. 2019, 206, 664–673. [Google Scholar] [CrossRef] [PubMed]
  144. Arunkumar, P.; Indulekha, S.; Vijayalakshmi, S.; Srivastava, R. Poly (Caprolactone) Microparticles and Chitosan Thermogels Based Injectable Formulation of Etoricoxib for the Potential Treatment of Osteoarthritis. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 61, 534–544. [Google Scholar] [CrossRef]
  145. Agnihotri, S.A.; Aminabhavi, T.M. Novel Interpenetrating Network Chitosan-Poly(Ethylene Oxide-g-Acrylamide) Hydrogel Microspheres for the Controlled Release of Capecitabine. Int. J. Pharm. 2006, 324, 103–115. [Google Scholar] [CrossRef]
  146. Chai, Q.; Jiao, Y.; Yu, X. Hydrogels for Biomedical Applications: Their Characteristics and the Mechanisms behind Them. Gels 2017, 3, 6. [Google Scholar] [CrossRef] [PubMed]
  147. Yang, Q.; Peng, J.; Xiao, H.; Xu, X.; Qian, Z. Polysaccharide Hydrogels: Functionalization, Construction and Served as Scaffold for Tissue Engineering. Carbohydr. Polym. 2022, 278, 118952. [Google Scholar] [CrossRef] [PubMed]
  148. Caló, E.; Khutoryanskiy, V.V. Biomedical Applications of Hydrogels: A Review of Patents and Commercial Products. Eur. Polym. J. 2015, 65, 252–267. [Google Scholar] [CrossRef]
  149. Gan, X.; Wang, X.; Huang, Y.; Li, G.; Kang, H. Applications of Hydrogels in Osteoarthritis Treatment. Biomedicines 2024, 12, 923. [Google Scholar] [CrossRef] [PubMed]
  150. Waresindo, W.X.; Luthfianti, H.R.; Priyanto, A.; Hapidin, D.A.; Edikresnha, D.; Aimon, A.H.; Suciati, T.; Khairurrijal, K. Freeze–Thaw Hydrogel Fabrication Method: Basic Principles, Synthesis Parameters, Properties, and Biomedical Applications. Mater. Res. Express 2023, 10, 024003. [Google Scholar] [CrossRef]
  151. Wang, W.; Liu, W.; Wu, J.; Liu, M.; Wang, Y.; Liu, H.; Liu, J. Preparation and Characterization of Particle-Filled Microgels by Chemical Cross-Linking Based on Zein and Carboxymethyl Starch for Delivering the Quercetin. Carbohydr. Polym. 2024, 323, 121375. [Google Scholar] [CrossRef] [PubMed]
  152. Mironi-Harpaz, I.; Wang, D.Y.; Venkatraman, S.; Seliktar, D. Photopolymerization of Cell-Encapsulating Hydrogels: Crosslinking Efficiency versus Cytotoxicity. Acta Biomater. 2012, 8, 1838–1848. [Google Scholar] [CrossRef] [PubMed]
  153. Moreira Teixeira, L.S.; Feijen, J.; van Blitterswijk, C.A.; Dijkstra, P.J.; Karperien, M. Enzyme-Catalyzed Crosslinkable Hydrogels: Emerging Strategies for Tissue Engineering. Biomaterials 2012, 33, 1281–1290. [Google Scholar] [CrossRef]
  154. Li, Z.; Lu, F.; Liu, Y. A Review of the Mechanism, Properties, and Applications of Hydrogels Prepared by Enzymatic Cross-Linking. J. Agric. Food Chem. 2023, 71, 10238–10249. [Google Scholar] [CrossRef]
  155. Nicol, E. Photopolymerized Porous Hydrogels. Biomacromolecules 2021, 22, 1325–1345. [Google Scholar] [CrossRef]
  156. Oliva, N.; Conde, J.; Wang, K.; Artzi, N. Designing Hydrogels for On-Demand Therapy. Acc. Chem. Res. 2017, 50, 669–679. [Google Scholar] [CrossRef] [PubMed]
  157. Guimarães, C.F.; Ahmed, R.; Marques, A.P.; Reis, R.L.; Demirci, U. Engineering Hydrogel-Based Biomedical Photonics: Design, Fabrication, and Applications. Adv. Mater. 2021, 33, e2006582. [Google Scholar] [CrossRef] [PubMed]
  158. Dhand, A.P.; Galarraga, J.H.; Burdick, J.A. Enhancing Biopolymer Hydrogel Functionality through Interpenetrating Networks. Trends Biotechnol. 2021, 39, 519–538. [Google Scholar] [CrossRef]
  159. Shi, W.; Huang, J.; Fang, R.; Liu, M. Imparting Functionality to the Hydrogel by Magnetic-Field-Induced Nano-Assembly and Macro-Response. ACS Appl. Mater. Interfaces 2020, 12, 5177–5194. [Google Scholar] [CrossRef]
  160. Wang, Y.; Katyal, P.; Montclare, J.K. Protein-Engineered Functional Materials. Adv. Healthc. Mater. 2019, 8, e1801374. [Google Scholar] [CrossRef]
  161. Hacker, M.C.; Nawaz, H.A. Multi-Functional Macromers for Hydrogel Design in Biomedical Engineering and Regenerative Medicine. Int. J. Mol. Sci. 2015, 16, 27677–27706. [Google Scholar] [CrossRef] [PubMed]
  162. Correa, S.; Meany, E.L.; Gale, E.C.; Klich, J.H.; Saouaf, O.M.; Mayer, A.T.; Xiao, Z.; Liong, C.S.; Brown, R.A.; Maikawa, C.L.; et al. Injectable Nanoparticle-Based Hydrogels Enable the Safe and Effective Deployment of Immunostimulatory CD40 Agonist Antibodies. Adv. Sci. 2022, 9, e2103677. [Google Scholar] [CrossRef]
  163. Manferdini, C.; Gabusi, E.; Saleh, Y.; Lenzi, E.; D’Atri, G.; Ricotti, L.; Lisignoli, G. Mesenchymal Stromal Cells Laden in Hydrogels for Osteoarthritis Cartilage Regeneration: A Systematic Review from In Vitro Studies to Clinical Applications. Cells 2022, 11, 3969. [Google Scholar] [CrossRef]
  164. Lolli, A.; Sivasubramaniyan, K.; Vainieri, M.L.; Oieni, J.; Kops, N.; Yayon, A.; van Osch, G.J.V.M. Hydrogel-Based Delivery of antimiR-221 Enhances Cartilage Regeneration by Endogenous Cells. J. Control Release 2019, 309, 220–230. [Google Scholar] [CrossRef]
  165. Li, H.; Feng, F.; Bingham, C.O., 3rd; Elisseeff, J.H. Matrix Metalloproteinases and Inhibitors in Cartilage Tissue Engineering. J. Tissue Eng. Regen. Med. 2012, 6, 144–154. [Google Scholar] [CrossRef]
  166. Snyder, T.N.; Madhavan, K.; Intrator, M.; Dregalla, R.C.; Park, D. A Fibrin/Hyaluronic Acid Hydrogel for the Delivery of Mesenchymal Stem Cells and Potential for Articular Cartilage Repair. J. Biol. Eng. 2014, 8, 10. [Google Scholar] [CrossRef]
  167. Yan, X.; Yang, B.; Chen, Y.; Song, Y.; Ye, J.; Pan, Y.; Zhou, B.; Wang, Y.; Mao, F.; Dong, Y.; et al. Anti-Friction MSCs Delivery System Improves the Therapy for Severe Osteoarthritis. Adv. Mater. 2021, 33, e2104758. [Google Scholar] [CrossRef] [PubMed]
  168. Whitworth, D.J.; Frith, J.E.; Frith, T.J.R.; Ovchinnikov, D.A.; Cooper-White, J.J.; Wolvetang, E.J. Derivation of Mesenchymal Stromal Cells from Canine Induced Pluripotent Stem Cells by Inhibition of the TGFβ/Activin Signaling Pathway. Stem Cells Dev. 2014, 23, 3021–3033. [Google Scholar] [CrossRef] [PubMed]
  169. Weißenberger, M.; Weißenberger, M.H.; Wagenbrenner, M.; Heinz, T.; Reboredo, J.; Holzapfel, B.M.; Rudert, M.; Groll, J.; Evans, C.H.; Steinert, A.F. Different Types of Cartilage Neotissue Fabricated from Collagen Hydrogels and Mesenchymal Stromal Cells via SOX9, TGFB1 or BMP2 Gene Transfer. PLoS ONE 2020, 15, e0237479. [Google Scholar] [CrossRef] [PubMed]
  170. Kilmer, C.E.; Walimbe, T.; Panitch, A.; Liu, J.C. Incorporation of a Collagen-Binding Chondroitin Sulfate Molecule to a Collagen Type I and II Blend Hydrogel for Cartilage Tissue Engineering. ACS Biomater. Sci. Eng. 2022, 8, 1247–1257. [Google Scholar] [CrossRef]
  171. Michalopoulos, E.; Knight, R.L.; Korossis, S.; Kearney, J.N.; Fisher, J.; Ingham, E. Development of Methods for Studying the Differentiation of Human Mesenchymal Stem Cells under Cyclic Compressive Strain. Tissue Eng. Part C Methods 2012, 18, 252–262. [Google Scholar] [CrossRef] [PubMed]
  172. Aisenbrey, E.A.; Bryant, S.J. Mechanical Loading Inhibits Hypertrophy in Chondrogenically Differentiating hMSCs within a Biomimetic Hydrogel. J. Mater. Chem. B 2016, 4, 3562–3574. [Google Scholar] [CrossRef]
  173. Wang, W.; Wan, Y.; Fu, T.; Zhou, T.; Tang, X.; Wu, H.; Liu, C.; Jagodzinski, M. Effect of Cyclic Compression on Bone Marrow Mesenchymal Stromal Cells in Tissue Engineered Cartilage Scaffold. J. Biomed. Mater. Res. A 2019, 107, 1294–1302. [Google Scholar] [CrossRef] [PubMed]
  174. Dong, Y.; Qin, Y.; Dubaa, M.; Killion, J.; Gao, Y.; Zhao, T.; Zhou, D.; Duscher, D.; Geever, L.; Gurtner, G.C.; et al. A Rapid Crosslinking Injectable Hydrogel for Stem Cell Delivery, from Multifunctional Hyperbranched Polymers via RAFT Homopolymerization of PEGDA. Polym. Chem. 2015, 6, 6182–6192. [Google Scholar] [CrossRef]
  175. Martin, K.E.; Hunckler, M.D.; Chee, E.; Caplin, J.D.; Barber, G.F.; Kalelkar, P.P.; Schneider, R.S.; García, A.J. Hydrolytic Hydrogels Tune Mesenchymal Stem Cell Persistence and Immunomodulation for Enhanced Diabetic Cutaneous Wound Healing. Biomaterials 2023, 301, 122256. [Google Scholar] [CrossRef]
  176. He, J.; Zhang, N.; Zhu, Y.; Jin, R.; Wu, F. MSC Spheroids-Loaded Collagen Hydrogels Simultaneously Promote Neuronal Differentiation and Suppress Inflammatory Reaction through PI3K-Akt Signaling Pathway. Biomaterials 2021, 265, 120448. [Google Scholar] [CrossRef] [PubMed]
  177. Wagner, M.A.; Marks, W.H.; Bhatia, S.K. Hydrogel Encapsulation to Improve Cell Viability during Syringe Needle Flow. J. Long-Term Eff. Med. Implants 2014, 24, 151–162. [Google Scholar] [CrossRef] [PubMed]
  178. Zhu, Y.; Ye, L.; Cai, X.; Li, Z.; Fan, Y.; Yang, F. Icariin-Loaded Hydrogel Regulates Bone Marrow Mesenchymal Stem Cell Chondrogenic Differentiation and Promotes Cartilage Repair in Osteoarthritis. Front. Bioeng. Biotechnol. 2022, 10, 755260. [Google Scholar] [CrossRef] [PubMed]
  179. Kováč, J.; Priščáková, P.; Gbelcová, H.; Heydari, A.; Žiaran, S. Bioadhesive and Injectable Hydrogels and Their Correlation with Mesenchymal Stem Cells Differentiation for Cartilage Repair: A Mini-Review. Polymers 2023, 15, 4228. [Google Scholar] [CrossRef] [PubMed]
  180. Wang, L.L.; Burdick, J.A. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies. Adv. Healthc. Mater. 2017, 6, 1601041. [Google Scholar] [CrossRef] [PubMed]
  181. Wang, S.; Zhang, C.; Niyazi, S.; Zheng, L.; Li, J.; Zhang, W.; Xu, M.; Rong, R.; Yang, C.; Zhu, T. A Novel Cytoprotective Peptide Protects Mesenchymal Stem Cells against Mitochondrial Dysfunction and Apoptosis Induced by Starvation via Nrf2/Sirt3/FoxO3a Pathway. J. Transl. Med. 2017, 15, 33. [Google Scholar] [CrossRef] [PubMed]
  182. Goyal, U.; Ta, M. A Novel Role of Vitronectin in Promoting Survival of Mesenchymal Stem Cells under Serum Deprivation Stress. Stem Cell Res. Ther. 2020, 11, 181. [Google Scholar] [CrossRef] [PubMed]
  183. Ko, K.I.; Coimbra, L.S.; Tian, C.; Alblowi, J.; Kayal, R.A.; Einhorn, T.A.; Gerstenfeld, L.C.; Pignolo, R.J.; Graves, D.T. Diabetes Reduces Mesenchymal Stem Cells in Fracture Healing through a TNFα-Mediated Mechanism. Diabetologia 2015, 58, 633–642. [Google Scholar] [CrossRef] [PubMed]
  184. He, X.; Wang, H.; Jin, T.; Xu, Y.; Mei, L.; Yang, J. TLR4 Activation Promotes Bone Marrow MSC Proliferation and Osteogenic Differentiation via Wnt3a and Wnt5a Signaling. PLoS ONE 2016, 11, e0149876. [Google Scholar] [CrossRef]
  185. Feng, M.; Liu, W.; Ding, J.; Qiu, Y.; Chen, Q. Sonic Hedgehog Induces Mesenchymal Stromal Cell Senescence-Associated Secretory Phenotype and Chondrocyte Apoptosis in Human Osteoarthritic Cartilage. Front. Cell Dev. Biol. 2021, 9, 716610. [Google Scholar] [CrossRef]
  186. Zhang, S.; Hu, P.; Liu, T.; Li, Z.; Huang, Y.; Liao, J.; Hamid, M.R.; Wen, L.; Wang, T.; Mo, C.; et al. Kartogenin Hydrolysis Product 4-Aminobiphenyl Distributes to Cartilage and Mediates Cartilage Regeneration. Theranostics 2019, 9, 7108–7121. [Google Scholar] [CrossRef] [PubMed]
  187. Aizman, I.; Tirumalashetty, B.J.; McGrogan, M.; Case, C.C. Comparison of the Neuropoietic Activity of Gene-Modified versus Parental Mesenchymal Stromal Cells and the Identification of Soluble and Extracellular Matrix-Related Neuropoietic Mediators. Stem Cell Res. Ther. 2014, 5, 29. [Google Scholar] [CrossRef]
  188. Wang, Y.; Malcolm, D.W.; Benoit, D.S.W. Controlled and Sustained Delivery of siRNA/NPs from Hydrogels Expedites Bone Fracture Healing. Biomaterials 2017, 139, 127–138. [Google Scholar] [CrossRef] [PubMed]
  189. Zhu, H.; Mitsuhashi, N.; Klein, A.; Barsky, L.W.; Weinberg, K.; Barr, M.L.; Demetriou, A.; Wu, G.D. The Role of the Hyaluronan Receptor CD44 in Mesenchymal Stem Cell Migration in the Extracellular Matrix. Stem Cells 2006, 24, 928–935. [Google Scholar] [CrossRef] [PubMed]
  190. Oh, J.Y.; Kim, H.; Lee, H.J.; Lee, K.; Barreda, H.; Kim, H.J.; Shin, E.; Bae, E.-H.; Kaur, G.; Zhang, Y.; et al. MHC Class I Enables MSCs to Evade NK-Cell-Mediated Cytotoxicity and Exert Immunosuppressive Activity. Stem Cells 2022, 40, 870–882. [Google Scholar] [CrossRef] [PubMed]
  191. Cho, W.J.; Mittal, S.K.; Chauhan, S.K. Mesenchymal Stromal Cells Suppress T-Cell-Mediated Delayed-Type Hypersensitivity via ALCAM-CD6 Interaction. Stem Cells Transl. Med. 2023, 12, 221–233. [Google Scholar] [CrossRef] [PubMed]
  192. Huang, D.; Li, Y.; Ma, Z.; Lin, H.; Zhu, X.; Xiao, Y.; Zhang, X. Collagen Hydrogel Viscoelasticity Regulates MSC Chondrogenesis in a ROCK-Dependent Manner. Sci. Adv. 2023, 9, eade9497. [Google Scholar] [CrossRef] [PubMed]
  193. Ledo, A.M.; Vining, K.H.; Alonso, M.J.; Garcia-Fuentes, M.; Mooney, D.J. Extracellular Matrix Mechanics Regulate Transfection and SOX9-Directed Differentiation of Mesenchymal Stem Cells. Acta Biomater. 2020, 110, 153–163. [Google Scholar] [CrossRef] [PubMed]
  194. Hodge, J.G.; Robinson, J.L.; Mellott, A.J. Novel Hydrogel System Eliminates Subculturing and Improves Retention of Nonsenescent Mesenchymal Stem Cell Populations. Regen. Med. 2023, 18, 23–36. [Google Scholar] [CrossRef]
  195. Pangjantuk, A.; Kaokaen, P.; Kunhorm, P.; Chaicharoenaudomrung, N.; Noisa, P. 3D Culture of Alginate-Hyaluronic Acid Hydrogel Supports the Stemness of Human Mesenchymal Stem Cells. Sci. Rep. 2024, 14, 4436. [Google Scholar] [CrossRef]
  196. Gao, X.; Liang, X.; Liu, B.; Hong, Y.; He, H.; Shen, Y.; Chen, J.; Huang, X.; Hu, B.; Li, W.; et al. Downregulation of ALKBH5 Rejuvenates Aged Human Mesenchymal Stem Cells and Enhances Their Therapeutic Efficacy in Myocardial Infarction. FASEB J. 2023, 37, e23294. [Google Scholar] [CrossRef] [PubMed]
  197. Chen, F.; Wang, S.; Zeng, C.; Tang, S.; Gu, H.; Wang, Z.; Li, J.; Feng, P.; Zhang, Y.; Wang, P.; et al. Silencing circSERPINE2 Restrains Mesenchymal Stem Cell Senescence via the YBX3/PCNA/P21 Axis. Cell Mol. Life Sci. 2023, 80, 325. [Google Scholar] [CrossRef] [PubMed]
  198. An, X.; Li, L.; Chen, Y.; Luo, A.; Ni, Z.; Liu, J.; Yuan, Y.; Shi, M.; Chen, B.; Long, D.; et al. Mesenchymal Stem Cells Ameliorated Glucolipotoxicity in HUVECs through TSG-6. Int. J. Mol. Sci. 2016, 17, 483. [Google Scholar] [CrossRef] [PubMed]
  199. Zhang, W.-Q.; Huang, S.-H.; Huang, X.; Li, J.-H.; Ye, P.; Xu, J.; Zheng, P.-Z.; Shen, H.-Y.; Huang, J.-R. Regulation of Human Mesenchymal Stem Cell Differentiation by TREM-2. Hum. Immunol. 2016, 77, 476–482. [Google Scholar] [CrossRef]
  200. Yoon, D.S.; Lee, K.-M.; Choi, Y.; Ko, E.A.; Lee, N.-H.; Cho, S.; Park, K.H.; Lee, J.-H.; Kim, H.-W.; Lee, J.W. TLR4 Downregulation by the RNA-Binding Protein PUM1 Alleviates Cellular Aging and Osteoarthritis. Cell Death Differ. 2022, 29, 1364–1378. [Google Scholar] [CrossRef]
  201. Yang, Y.; Liu, S.; He, C.; Lv, T.; Zeng, L.; Zhang, F.; Chen, H.; Zhao, R.C. LncRNA LYPLAL1-AS1 Rejuvenates Human Adipose-Derived Mesenchymal Stem Cell Senescence via Transcriptional MIRLET7B Inactivation. Cell Biosci. 2022, 12, 45. [Google Scholar] [CrossRef] [PubMed]
  202. Zhu, Y.; Zhang, X.; Gu, R.; Liu, X.; Wang, S.; Xia, D.; Li, Z.; Lian, X.; Zhang, P.; Liu, Y.; et al. LAMA2 Regulates the Fate Commitment of Mesenchymal Stem Cells via Hedgehog Signaling. Stem Cell Res. Ther. 2020, 11, 135. [Google Scholar] [CrossRef]
  203. Hu, S.; Li, J.; Xu, X.; Liu, A.; He, H.; Xu, J.; Chen, Q.; Liu, S.; Liu, L.; Qiu, H.; et al. The Hepatocyte Growth Factor-Expressing Character Is Required for Mesenchymal Stem Cells to Protect the Lung Injured by Lipopolysaccharide In Vivo. Stem Cell Res. Ther. 2016, 7, 66. [Google Scholar] [CrossRef] [PubMed]
  204. Tang, H.-C.; Chen, W.-C.; Chiang, C.-W.; Chen, L.-Y.; Chang, Y.-C.; Chen, C.-H. Differentiation Effects of Platelet-Rich Plasma Concentrations on Synovial Fluid Mesenchymal Stem Cells from Pigs Cultivated in Alginate Complex Hydrogel. Int. J. Mol. Sci. 2015, 16, 18507–18521. [Google Scholar] [CrossRef]
  205. Hori, J.; Deie, M.; Kobayashi, T.; Yasunaga, Y.; Kawamata, S.; Ochi, M. Articular Cartilage Repair Using an Intra-Articular Magnet and Synovium-Derived Cells. J. Orthop. Res. 2011, 29, 531–538. [Google Scholar] [CrossRef]
  206. Li, J.; Huang, Y.; Song, J.; Li, X.; Zhang, X.; Zhou, Z.; Chen, D.; Ma, P.X.; Peng, W.; Wang, W.; et al. Cartilage Regeneration Using Arthroscopic Flushing Fluid-Derived Mesenchymal Stem Cells Encapsulated in a One-Step Rapid Cross-Linked Hydrogel. Acta Biomater. 2018, 79, 202–215. [Google Scholar] [CrossRef] [PubMed]
  207. Chung, J.Y.; Song, M.; Ha, C.-W.; Kim, J.-A.; Lee, C.-H.; Park, Y.-B. Comparison of Articular Cartilage Repair with Different Hydrogel-Human Umbilical Cord Blood-Derived Mesenchymal Stem Cell Composites in a Rat Model. Stem Cell Res. Ther. 2014, 5, 39. [Google Scholar] [CrossRef] [PubMed]
  208. Park, Y.-B.; Ha, C.-W.; Lee, C.-H.; Yoon, Y.C.; Park, Y.-G. Cartilage Regeneration in Osteoarthritic Patients by a Composite of Allogeneic Umbilical Cord Blood-Derived Mesenchymal Stem Cells and Hyaluronate Hydrogel: Results from a Clinical Trial for Safety and Proof-of-Concept with 7 Years of Extended Follow-Up. Stem Cells Transl. Med. 2017, 6, 613–621. [Google Scholar] [CrossRef] [PubMed]
  209. Kim, J.-K.; Bae, H.C.; Ro, D.H.; Lee, S.; Lee, M.C.; Han, H.-S. Enhancement of Cartilage Regeneration of Synovial Stem Cells/Hydrogel by Using Transglutaminase-4. Tissue Eng. Part A 2021, 27, 761–770. [Google Scholar] [CrossRef] [PubMed]
  210. Geckil, H.; Xu, F.; Zhang, X.; Moon, S.; Demirci, U. Engineering Hydrogels as Extracellular Matrix Mimics. Nanomedicine 2010, 5, 469–484. [Google Scholar] [CrossRef] [PubMed]
  211. Sang, S.; Mao, X.; Cao, Y.; Liu, Z.; Shen, Z.; Li, M.; Jia, W.; Guo, Z.; Wang, Z.; Xiang, C.; et al. 3D Bioprinting Using Synovium-Derived MSC-Laden Photo-Cross-Linked ECM Bioink for Cartilage Regeneration. ACS Appl. Mater. Interfaces 2023, 15, 8895–8913. [Google Scholar] [CrossRef] [PubMed]
  212. Guo, J.L.; Kim, Y.S.; Koons, G.L.; Lam, J.; Navara, A.M.; Barrios, S.; Xie, V.Y.; Watson, E.; Smith, B.T.; Pearce, H.A.; et al. Bilayered, Peptide-Biofunctionalized Hydrogels for in Vivo Osteochondral Tissue Repair. Acta Biomater. 2021, 128, 120–129. [Google Scholar] [CrossRef] [PubMed]
  213. Abramoff, B.; Caldera, F.E. Osteoarthritis: Pathology, Diagnosis, and Treatment Options. Med. Clin. N. Am. 2020, 104, 293–311. [Google Scholar] [CrossRef] [PubMed]
  214. Tibbitt, M.W.; Anseth, K.S. Hydrogels as Extracellular Matrix Mimics for 3D Cell Culture. Biotechnol. Bioeng. 2009, 103, 655–663. [Google Scholar] [CrossRef]
  215. Ankrum, J.; Karp, J.M. Mesenchymal Stem Cell Therapy: Two Steps Forward, One Step Back. Trends Mol. Med. 2010, 16, 203–209. [Google Scholar] [CrossRef]
  216. Ankrum, J.A.; Ong, J.F.; Karp, J.M. Mesenchymal Stem Cells: Immune Evasive, Not Immune Privileged. Nat. Biotechnol. 2014, 32, 252–260. [Google Scholar] [CrossRef] [PubMed]
  217. Ge, W.; Jiang, J.; Baroja, M.L.; Arp, J.; Zassoko, R.; Liu, W.; Bartholomew, A.; Garcia, B.; Wang, H. Infusion of Mesenchymal Stem Cells and Rapamycin Synergize to Attenuate Alloimmune Responses and Promote Cardiac Allograft Tolerance. Am. J. Transpl. 2009, 9, 1760–1772. [Google Scholar] [CrossRef] [PubMed]
  218. Carballo, C.B.; Nakagawa, Y.; Sekiya, I.; Rodeo, S.A. Basic Science of Articular Cartilage. Clin. Sports Med. 2017, 36, 413–425. [Google Scholar] [CrossRef] [PubMed]
  219. Vincent, T.L.; McClurg, O.; Troeberg, L. The Extracellular Matrix of Articular Cartilage Controls the Bioavailability of Pericellular Matrix-Bound Growth Factors to Drive Tissue Homeostasis and Repair. Int. J. Mol. Sci. 2022, 23, 6003. [Google Scholar] [CrossRef] [PubMed]
  220. Gahunia, H.K.; Pritzker, K.P.H. Structure and Function of Articular Cartilage. In Articular Cartilage of the Knee: Health, Disease and Therapy; Gahunia, H.K., Gross, A.E., Pritzker, K.P.H., Babyn, P.S., Murnaghan, L., Eds.; Springer: New York, NY, USA, 2020; pp. 3–70. ISBN 978-1-4939-7587-7. [Google Scholar]
Figure 2. Classification of hydrogels. Reprinted with permission from Ref. [132]. Copyright 2022 MDPI.
Figure 2. Classification of hydrogels. Reprinted with permission from Ref. [132]. Copyright 2022 MDPI.
Biomolecules 14 00858 g002
Figure 3. Schematic of strategies for modifying natural polysaccharide hydrogels according to their functional groups. Reprinted with permission from Ref. [147]. Copyright 2022 Elsevier.
Figure 3. Schematic of strategies for modifying natural polysaccharide hydrogels according to their functional groups. Reprinted with permission from Ref. [147]. Copyright 2022 Elsevier.
Biomolecules 14 00858 g003
Figure 4. Viability analysis of rASCs in the hydrogels up to 14 days. (A) Green staining indicates live cells and red indicates dead cells. (Scale bars represent 100 μm). (B) Statistics of (A). Adapted with permission from Ref. [174]. Copyright 2010 Royal Society of Chemistry.
Figure 4. Viability analysis of rASCs in the hydrogels up to 14 days. (A) Green staining indicates live cells and red indicates dead cells. (Scale bars represent 100 μm). (B) Statistics of (A). Adapted with permission from Ref. [174]. Copyright 2010 Royal Society of Chemistry.
Biomolecules 14 00858 g004
Figure 5. Hydrolytic hydrogels promote MSC retention at site of injection. (A) Schematic of MSC retention studies. (B) IVIS images of MSCfluc after injection. (C) Normalizing the bioluminescence of transplanted MSCfluc over time. *** p < 0.001. (D) MSCfluc retention over time. (The different line represents each individual sample). Reprinted with permission from Ref. [175]. Copyright 2023 Elsevier.
Figure 5. Hydrolytic hydrogels promote MSC retention at site of injection. (A) Schematic of MSC retention studies. (B) IVIS images of MSCfluc after injection. (C) Normalizing the bioluminescence of transplanted MSCfluc over time. *** p < 0.001. (D) MSCfluc retention over time. (The different line represents each individual sample). Reprinted with permission from Ref. [175]. Copyright 2023 Elsevier.
Biomolecules 14 00858 g005
Figure 6. IA injection of BMSCs embedding PHa@I hydrogels prevents cartilage degeneration. (A) Schedule of in vivo experiment. (B) Hematoxylin eosin staining of cartilage. (C) Safranin O-fast green staining of cartilage. (D) Osteoarthritis Research Society International (OARSI) scores. (E) Markin scores. (“*” vs the PBS group, * p < 0.05, ** p < 0.01, and *** p < 0.001; “#” vs the PHa@I Group, # p < 0.05 and ## p < 0.01; “&” vs the BMSC Group, & p < 0.05). Reprinted with permission from Ref. [178]. Copyright © 2022 Zhu, Ye, Cai, Li, Fan and Yang.
Figure 6. IA injection of BMSCs embedding PHa@I hydrogels prevents cartilage degeneration. (A) Schedule of in vivo experiment. (B) Hematoxylin eosin staining of cartilage. (C) Safranin O-fast green staining of cartilage. (D) Osteoarthritis Research Society International (OARSI) scores. (E) Markin scores. (“*” vs the PBS group, * p < 0.05, ** p < 0.01, and *** p < 0.001; “#” vs the PHa@I Group, # p < 0.05 and ## p < 0.01; “&” vs the BMSC Group, & p < 0.05). Reprinted with permission from Ref. [178]. Copyright © 2022 Zhu, Ye, Cai, Li, Fan and Yang.
Biomolecules 14 00858 g006
Figure 7. Morphology of MSCs in 3D culture hydrogels. Reprinted with permission from Ref. [195]. Copyright © Amorn Pangjantuk, Palakorn Kaokaen, Phongsakorn Kunhorm, Nipha Chaicharoenaudomrung, and Parinya Noisa 2024.
Figure 7. Morphology of MSCs in 3D culture hydrogels. Reprinted with permission from Ref. [195]. Copyright © Amorn Pangjantuk, Palakorn Kaokaen, Phongsakorn Kunhorm, Nipha Chaicharoenaudomrung, and Parinya Noisa 2024.
Biomolecules 14 00858 g007
Figure 8. Schematic illustration of the GelHACS-MA hydrogel design. (A) Fabrication of Gel-MA. (B) Fabrication of HA-MA. (C) Fabrication of CS-MA. (D) Fabrication of GelHACS-MA hydrogels. (E) Isolation of SMSCs. (F) Design strategy illustration. Adapted with permission from the Ref. [211]. Copyright 2023 American Chemical Society.
Figure 8. Schematic illustration of the GelHACS-MA hydrogel design. (A) Fabrication of Gel-MA. (B) Fabrication of HA-MA. (C) Fabrication of CS-MA. (D) Fabrication of GelHACS-MA hydrogels. (E) Isolation of SMSCs. (F) Design strategy illustration. Adapted with permission from the Ref. [211]. Copyright 2023 American Chemical Society.
Biomolecules 14 00858 g008
Figure 9. Schematic illustration of regenerating the osteochondral tissue by a bilayered, tissue-specific hydrogel system. Reprinted with permission from Ref. [212]. Copyright 2021 Elsevier.
Figure 9. Schematic illustration of regenerating the osteochondral tissue by a bilayered, tissue-specific hydrogel system. Reprinted with permission from Ref. [212]. Copyright 2021 Elsevier.
Biomolecules 14 00858 g009
Table 2. Identification methods for MSCs.
Table 2. Identification methods for MSCs.
International Society for Cellular Therapy (ISCT) Definition of MSC Properties
Plastic adhesion or notYes (in standard culture conditions)
Specific antigenCD105+ CD73+ CD90+ Stro-1+, CD29+, CD44+, CD73+, CD146+, and SSEA4+,
CD14-, CD34-, CD45- or CD79a-, CD11b- or HLA-DR-, CD19-
In vitro diffenentiated abilityAdipocytes, osteoblasts, chondroblasts
Table 4. Different kinds of MSCs for OA therapy.
Table 4. Different kinds of MSCs for OA therapy.
PropertyKinds of MSCsModelOutcome of OA TreatmentRef.
Anti-
inflammation
IPFP-ASCsHumanPromoting chondrogenic differentiation and preventing articular cartilage thickening and inflammation[102]
Supra-hASCMouseReducing OA-associated knee inflammation and cartilage degenerative grade[103]
BM-MSCsHumanPromoting cell proliferation of chondrocytes and inhibiting inflammatory activity in osteoarthritis[104]
hUCMSCs-EVsHumanPromoting the polarization of M2-type macrophages, reducing the infammatory of cytokines (IL-10) response[105]
Cartilage regenerationIPFP-ASCsSheepPromoting the expression of cartilage genes[106]
BM-MSCsHumanInducing chondrogenic differentiation[107]
Sc-ASCsBearPromoting chondrogenic differentiation[108]
BM-MSCsHumanPromoting chondrogenic differentiation by enhancing the expression of cartilage extracellular matrix genes[109]
BM-MSCsRatPrevents cell apoptosis and inhibits senescence of chondrocytes by reducing the IL-1β level and improving the inflammation in joints[110]
Table 5. Types of hydrogels and characterizations.
Table 5. Types of hydrogels and characterizations.
Types of HydrogelsCharacterizationsRef.
Natural hydrogels①Polysaccharide: hyaluronic acid, chondroitin sulfate, chitin, chitosan, cellulose, starch, gum, alginate, and carrageenan1. Low immune response
2. Low toxicity response
3. Non-toxic and non-immunogenic degradation products
4. Poor stability, rapid degradation
5. Relatively low mechanical strength
[130,133,134,135]
②Protein-based materials: gelatin, collagen, fibroin, sericin
③Polyphenols: lignin
④Organic polyester/inorganic polyester: polyphthalamide
⑤Polyanhydride: polyadipic acid
⑥Biopolymer: nucleic acid, DNA
Synthetic hydrogels①Polycaprolactone-(PCL)
②Polyvinylpyrrolidone-(PVP)
③Polylactic acid- (PLA)
④Polyethylene glycol-(PEG)
⑤Polyvinyl alcohol- (PVA)
1. Providing customized performance characteristics
2. Controlability, reproducibility, and excellent mechanical performance
3. Poor compatibility with host tissues
4. Low biological activity
[136,137,138,139,140,141,142]
Hybrid-origin hydrogels①Carboxymethyl chitosan-(CHC)
②Hyaluronic acid-(HA)
1. pH-dependent drug release characteristics
2. Inhibition of cell apoptosis
[143,144,145]
①Chitosan
②Polycaprolactone microspheres
1. Dual functionality of supplementing mucus and storing drugs
2. Prolonging drug residence time in the body
①Semi-polyacrylonitrile chitosan-poly(acrylamide-ethylene oxide) hydrogel microspheresUsed for encapsulation and delivery of anticancer drugs
Table 6. Hydrogel preparation methods.
Table 6. Hydrogel preparation methods.
Hydrogel Preparation MethodAdvantageDisadvantageRef.
Chemical crosslinking①High degree of crosslinking and stability
②Highly adjustable
③Wide range of applicability
①Biotoxicity
②Toxic substances need to be cleared.
③Long reaction time and complex preparation
[132]
Physical crosslinking①Mildly reactive and environmentally friendly
②Prepared at room temperature
③Gel structures with reversible properties can be prepared
①Poor gel stabilization
②Sensitive to temperature and ionic concentration conditions and structural instability
③The preparation process can be complex.
[132]
Enzymatic crosslinking①Good biocompatibility
②Mild chemical reaction, sensitive to biologically active substances
③It can be prepared under physiological conditions.
①Enzyme stability and activity are easily affected.
②The enzyme-catalyzed reaction rate is slower and the preparation time is longer.
③The range of applicability is limited by the available enzymes and substrates.
[154]
Photopolymerization crosslinking①The preparation process is simple and easy to operate.
②A high degree of crosslinking can be achieved in a relatively short period of time.
③Better spatial and temporal control
①Possible phototoxicity to organisms
②Limited by the depth of light penetration and the rate of reaction
③Technical and equipment support is required for photosensitive monomer selection and light source control.
[155]
Table 7. The mechanisms of hydrogels in the treatment of osteoarthritis (OA). Reprinted with permission Ref. [163]. Copyright 2022 MDPI.
Table 7. The mechanisms of hydrogels in the treatment of osteoarthritis (OA). Reprinted with permission Ref. [163]. Copyright 2022 MDPI.
Hydrogel TypeCell Type and LoadingChondrogenic Inducting FactorsMain ResultsRef.
Fibrin/hyaluronan hydrogelHuman BMSCs TGF-β1Increasing COL2, ACAN, and GAG levels[164]
10% PEGDAGoat BMSCsTGF-β1Increasing COL2 and GAG level[165]
Fibrin MeHAHuman MSCsN.I.Increasing SOX9 level[166]
DNA supramolecularRabbit BMSCsN.IIncreasing COL2, SOX9 and ACAN level, decreasing COL1 and COL10 levels[167]
PEG–hyaluronic acid (HA)Canine MSCs TGF-β3Increaing proteoglycan and GAG levels[168]
Collagen type 1Human BMSCs NoIncreasing COL2 and GAG levels and condroitin sulfate[169]
Chondroitin sulfate (CS)Rabbit BMSCsTGF-β3Increasing GAG and COL2 levels[170]
Collagen and alginateHuman MSCsNoIncreasing CBFA-1, Sox9, and aggrecan levels[171]
Chondroitin sulfate (CS) and PEGHuman MSCsN.I.Increasing collagen II gene expression [172]
ChitosanRat BMMSCsN.I.Promoting chondrogenesis markers expression (Sox9, aggrecan, and collagen II)[173]
Table 8. Genes that are pivotal for MSCs’ properties.
Table 8. Genes that are pivotal for MSCs’ properties.
Gene SymbolKinds of MSCsGene FunctionsGene ManipulationDiseasesRef.
Sirt3 (sirtuin 3)BMSCsAgainst starvation-induced apoptosisKnockdownIn vitro[181]
ALKBH5 (AlkB homolog 5)BMSCsInducer of aging in MSCsKnockdownMyocardial
infarction
[196]
circSERPINE2 (serpin family E member 2)BMSCsInducer of aging in MSCsKnockdownOsteoarthritis[197]
NICD1 (notch receptor 1)BMSCsEnhanced neuropoietic effectsKnockdownIschemic stroke
and Parkinson’s
[187]
ALCAM (activated leukocyte cell adhesion molecule)BMSCsInhibiting the activation and proliferation of allogeneic CD4+ T cellsKnockdownAllograft
rejection,
autoimmune diseases
[191]
SHH (Sonic hedgehog signaling molecule)OA-MSCInducer of aging in MSCsKnockdownOsteoarthritis[185]
FOXO1 (forkhead box O1)BMSCsAgainst TNF-α-induced apoptosis in MSCsKnockdownDiabetes [183]
TSG-6 (tumor necrosis factor-α-stimulated protein 6)HUC-MSCsAgainst cellular damage caused by high sugar and fatKnockdownDiabetes [198]
TREM-2 (triggering receptor expressed on myeloid cells 2)MSCsCritical for MSCs’ pluripotency and immunomodulatory capacityKnockdownIn vitro[199]
TLR4 (Toll-like receptor 4)BMSCsPromote proliferation and osteogenic differentiation of MSCsKnockdownFracture
healing, osteoporosis
[184]
VTN (vitronectin)WJ-MSCsAgainst starvation-induced apoptosisKnockdownIschemic diseases
and wound healing
[182]
RPS6KA2 (ribosomal protein S6 kinase A2)BMSC and UC-MSC Critical for repairing cartilage defectsKnockdownOsteoarthritis[186]
PUM1 (Pumilio RNA binding family member 1)BMSCs Against aging of MSCKnockdownOsteoarthritis[200]
LYPLAL1-AS1 (LYPLAL1 antisense RNA 1)hADSCsAgainst aging of MSCOverexpressionSenile disease[201]
LAMA2 (laminin subunit alpha 2)hASCs
and
hBMMSCs
Inhibiting of osteogenic differentiation but promoting adipogenic differentiation of MSCsKnockdownBone defect diseases[202]
HGF (hepatocyte growth factor)BMSCsRepair lung endothelial cell functionKnockdownAcute lung injury[203]
CD44 (CD44 molecule (IN blood group))BMSCsMediates cell adhesion to ECM, promotes cell migrationKnockdownTissue damage and graft fibrosis[189]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, X.; He, W.; Huang, H.; Han, J.; Wang, R.; Li, H.; Long, Y.; Wang, G.; Han, X. Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy. Biomolecules 2024, 14, 858. https://doi.org/10.3390/biom14070858

AMA Style

Wang X, He W, Huang H, Han J, Wang R, Li H, Long Y, Wang G, Han X. Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy. Biomolecules. 2024; 14(7):858. https://doi.org/10.3390/biom14070858

Chicago/Turabian Style

Wang, Xiangjiang, Wentao He, Hao Huang, Jiali Han, Ruren Wang, Hongyi Li, Ying Long, Guiqing Wang, and Xianjing Han. 2024. "Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy" Biomolecules 14, no. 7: 858. https://doi.org/10.3390/biom14070858

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop