Umbilical Stem Cell Therapy for Knees Reviews 2019

  • Journal List
  • Drug Des Devel Ther
  • v.15; 2021
  • PMC8462093

Drug Des Devel Ther. 2021; 15: 3927–3936.

Umbilical Cord Mesenchymal Stem Cell Therapy for Regenerative Treatment of Rheumatoid Arthritis: Opportunities and Challenges

Xiaolan Lv

oneDepartment of Laboratory Medicine, Liuzhou Motherhood and Child Healthcare Hospital, Liu Zhou, Guang Xi, People's Republic of China

Liming Wang

2Shaanxi Jiuzhou Biomedical Scientific discipline and Technology Group, Xi'an, Shaan Xi, People's Republic of China

XiaoRong Zou

3Section of Hematology, 986 Hospital of Fourth Armed forces Medical Academy, Xi'an, Shaan 11, People'due south Democracy of China

Shigao Huang

4Faculty of Health Sciences, University of Macau, Macau, People's Republic of China

Received 2021 Jun 3; Accepted 2021 Aug 26.

Abstract

Rheumatoid arthritis (RA) is an autoimmune affliction of unknown etiology with a high rate of disability. Traditional treatments for RA remain a challenging issue. For example, nonsteroidal anti-inflammatory drugs (NSAIDs) have no therapeutic effects on joint destruction, and the prominent side effects include gastrointestinal symptoms. RA is characterized by recurrence and bone attrition. Therefore, regenerative medicine and the use of umbilical cord mesenchymal stalk prison cell (UC-MSC) therapies take recently emerged as potential options. UC-MSCs are multifunctional stalk cells that are present in neonatal umbilical cord tissue and can differentiate into many kinds of cells, which have broad clinical application prospects in the tissue engineering science of bone, cartilage, musculus, tendon, ligament, nerve, liver, endothelium, and myocardium. Moreover, UC-MSCs accept advantages, such as convenient collection of materials and no upstanding disputes; thus, these cells take attracted increasing attention from researchers. However, in that location are few clinical studies regarding UC-MSC therapy for RA. In this paper, nosotros will review traditional drugs for RA treatment and then focus on UC-MSC therapy for RA, including preclinical and clinical UC-MSC applications for RA patients in the context of regenerative medicine. Finally, nosotros will summarize the challenges and perspectives of UC-MSCs every bit a potential therapeutic strategy for RA. This review volition assist to pattern and discover more potent and efficacious treatments for RA patients and assistance in advancing this form of cell therapy.

Keywords: cell-based therapies, rheumatoid arthritis, mesenchymal stem cells, umbilical cord, regenerative medicine

Graphical Abstruse

An external file that holds a picture, illustration, etc.  Object name is DDDT-15-3927-g0001.jpg

Introduction

Rheumatoid arthritis (RA) is a systemic, inflammatory autoimmune disease that mainly involves peripheral facet joint disease,1 which is the primary pathological characteristic, as well as joint synovial cell proliferation, inflammatory prison cell infiltration, and pannus.ii To date, traditional RA treatment mainly involves reducing the symptomatic inflammatory reaction and sequelae;3 conventional handling cannot satisfy the clinical requirement of achieving a curative event.4 Therefore, exploring more than effective and safer treatment options is increasingly important. Mesenchymal stalk cells (MSCs) are derived from early-development mesoderm pluripotent stem cells and take a high caste of cocky-renewal and multidirectional differentiation potential; MSCs widely exist in a multifariousness of human being tissues and can be cultured in vitro. Under specific weather condition, MSCs can differentiate into osteoblasts, nerve cells, adipose cells, muscle cells, and cardiomyocytes. These cells accept nifty application value in tissue engineering repair and in prison cell replacement therapy.5 First, MSCs can differentiate into bone and cartilage. 2d, these cells also inhibit immunity, convalesce inflammation, resist fibrosis, and promote vascular repair. Umbilical cord MSCs (UC-MSCs) are derived from homo umbilical cord blood and have MSC characteristics and functions. Therefore, UC-MSCs offer the hope of a new treatment selection for RA. In recent years, the use of UC-MSCs to treat rheumatic allowed diseases has garnered increasing attention,6–9 and the promising results of preclinical studies suggest that UC-MSCs may be a therapeutic method for regenerative medicine.vii,10,eleven However, there have been few studies regarding the furnishings of UC-MSCs on patients with RA who had recurrent symptoms afterwards long-term treatment with regular agents in the clinic. Withal, there is yet a pressing demand for examining efficiency and rubber considerations of UC-MSCs in clinical investigations. This review aims to summarize the current preclinical experimental and clinical studies of RA handling with UC-MSCs, examining the opportunities and challenges in research development progress.

Traditional Treatment Drugs for RA

According to the development time and principles, the current drugs for treating RA are divided into four generations: the starting time generation is nonsteroidal anti-inflammatory drugs (NSAIDs); the second generation is glucocorticoids; the third generation is disease-changing drugs (slow-interim antirheumatic drugs, SAARDs); and the quaternary generation are early biologic agents dominated past tumor necrosis factor (TNF)-α inhibitors. The following are the descriptions of the difficulties of these four drug types in RA treatment.

Offset, NSAIDs12 are the showtime-generation treatment for RA and represent drugs such as aspirin and diclofenac. These drugs inactivate cyclooxygenase (COX)-1 and COX-2 mainly through acetylation.thirteen NSAIDs have no therapeutic effect on joint devastation, and the prominent side effects are gastrointestinal symptoms. In contempo years, newly marketed specific COX-ii inhibitors, such as meloxicam and celecoxib, have not affected COX-L due to their specific inhibition of COX-2.14 Thus, this treatment can avoid the traditional gastrointestinal side effects of NSAIDs. Second, illness-modifying antirheumatic drugs (DMARDs)15 or SAARDs are third-generation therapies for RA. This form of drugs includes antimalarial drugs, gold preparations, and cytotoxic drugs. DMARDs mainly control the development of joint illness by reducing synovial inflammation and fifty-fifty play a role in repair, merely most of these agents accept serious side effects, such as airsickness, skin rash, leukocyte reductions, and liver and kidney function impairment. 3rd, glucocorticoids are the second-generation treatment for RA.xvi The mechanism of action is that glucocorticoids bind to glucocorticoid receptors and accomplish the nucleus, lowering the action of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and reducing the production of proinflammatory cytokines to effectively reduce inflammation. These drugs cannot block the progression of RA or joint destruction, and long-term apply can induce side furnishings such as infection, cortical hyperfunction, osteoporosis and hypertension. However, small doses and short courses of treatment tin can reduce symptoms through anti-inflammatory and anticleric effects.

Finally, early biological agents, mainly TNF-α inhibitors, are the 4th generation of drugs for the handling of RA.17 Because of the advantages of high pharmacological selectivity and low toxicity and side effects, biologics will have wide application prospects. Amongst the biologics used for the treatment of RA, the most clinically studied are TNF-α inhibitors. Three TNF-α inhibitors, etanercept, infliximab and adalimumab, are currently approved for the treatment of RA.18 The most common agin reactions are injection-site reactions and infection. Infliximab, developed by Centocor, is used for the treatment of patients with early RA,19 and the common adverse reactions are infection and allergic reactions in some patients.20 Adalimumab, developed by Abbott, is the first homo TNF antagonist and has adverse reactions such equally nasopharyngitis and upper respiratory tract infection, and patients are prone to tuberculosis recurrence.14,21 In addition, compared with the utilize of methotrexate (MTX; 30.nine%), the risk of opportunistic infections and invasive melanoma increased by fifty% in patients treated with TNF antagonists (forty.1%) compared with those treated with TNF antagonists.22 These findings limit the broad application of biological agents to some extent.

Advantages of UC-MSC Therapy for RA

MSCs are multipotent cells derived from the eye, peripheral blood, umbilical cord blood, muscle, lung, trabecular os, intestinal tract, kidney, liver, pancreas, synovium, skin, and even the brain and tin can exist a promising form of regenerative medicine.23 In recent years, MSCs have been widely used in the treatment of various diseases.24 In People's republic of china, MSCs accept been used to treat some clinically refractory diseases, such every bit spinal string injury, cognitive palsy, amyotrophic lateral sclerosis, systemic lupus erythematosus, systemic sclerosis, Crohn's disease, stroke, diabetes, diabetic foot, and cirrhosis, among other atmospheric condition.25–27 Clinical practice shows that MSCs are effective treatments for many diseases, such cotransplantation with hematopoietic stem cells, the enhancement of hematopoietic role, and implantation with hematopoietic stalk cell grafts to treat graft-versus-host affliction (GVHD).24 In clinical research, the chief types of MSCs are bone marrow-derived MSCs (BM-MSCs), adipose-derived mesenchymal stromal cells (Advertizement-MSCs) and UC-MSCs.

RA is an inexorably progressive lung disease of unknown origin.28,29 The prognosis is poor, limited treatment options are bachelor, and the median survival remains simply 3–5 years.30 Despite the use of pirfenidone and nintedanib for the treatment of RA, MSCs have potential equally therapies for RA.3 In recent years, based on the biological characteristics of MSCs, much research has been done on the part of MSCs in autoimmune diseases.31 In particular, MSCs can reduce the incidence of such diseases, particularly RA, can also reduce inflammation later disease onset and can delay further disease development.32 MSCs have immunoregulatory roles, regulating the expression of cytokines by T cells, B cells, dendritic cells (DCs), and natural killer (NK) cells.33,34 However, RA is mainly caused by autoimmune disease mediated past T cells and involves B cells, cytokines, apoptosis, proteases, and other factors.35 Therefore, the use of MSCs to intervene in the pathological basis of RA-mediated inflammation has become a viable method. Near 100 studies have been conducted to establish experimental models for MSC treatment of RA. In most experimental models, the efficacy of MSC treatment was demonstrated past significantly reducing the consecration and progression of experimental arthritis. The most widely used model is the collagen-induced arthritis (CIA) model.36 These promising models pave the mode for MSC treatment of human RA affliction. Currently, 9 clinical trials are active, and as a effect, the clinical data are not publicly available, although the MSCs are known to have included l% UC-MSCs, 25% Ad-MSCs, and 25% BM-MSCs.9

Compared with BM-MSCs and Advertizing-MSCs, UC-MSCs have the following advantages: cell surface expression of major histocompatibility circuitous I (MHC I) is lower, and MHCII is not expressed, then UC-MSCs seldom cause immune rejection. Additionally, the growth environment of UC-MSCs more than pure than that of other MSCs, these cells have stronger abilities to proliferate and differentiate and are piece of cake to culture in vitro. Furthermore, UC-MSCs are conveniently derived from umbilical cords, so there are fewer ethical bug, and they exercise not hands transmit disease.37

Preclinical Experiments

Regulation of the Immune Response

UC-MSCs mediate the differentiation, proliferation, and activation of T cell subsets in various ways and inhibit B cell proliferation and differentiation, every bit well equally the maturation of DCs and NK jail cell activeness to reach various immunomodulatory effects.6 Therefore, UC-MSCs may be a viable method to intervene in the pathological footing of RA-associated inflammation. Contempo studies on the handling of RA suggest that UC-MSCs suppress the various inflammatory effects of fibroblast-similar synoviocytes (FLSs) and T cells in vitro and attenuate the development of CIA in vivo.38 MSCs have low immunogenicity and express MHC-I molecules just non MHC-II molecules and costimulatory molecules such every bit B7-i, B7-2, CD40, CD40, and Fas ligand. MSCs are not restricted by MHC molecules, so they play an immunomodulatory role in vivo.39 Researchers40 accept demonstrated that UC-MSCs have an immunomodulatory effect on allogeneic T lymphocytes and can inhibit the proliferation and changes in T lymphocytes stimulated by other jail cell subsets and cytokines secreted past T cells in the context of allowed responses. The effects of UC-MSCs on the immune regulation of Th17 cells in RA patients accept been reported by Wang et al.41 The results showed that UC-MSCs could significantly reduce the numbers of Th17 cells and the expression of cytokines and reduce the expression of ROR-γt and serum levels IL-17, and there was a good correlation with the dose. UC-MSCs were observed in vitro in a mixed cell civilisation with peripheral blood mononuclear cells (PBMCs) from RA patients. In vitro, the Th17 level was downregulated in the highly active group, merely no like regulation was observed in the normal and mildly agile groups, suggesting that the immunoregulatory effect of UC-MSCs on Th17 cells may be regulated by other factors. Gu et al,42 ended that UC-MSCs could increase the peripheral blood Treg proportion in CIA rats and inhibit secretion by Th17 cells and the action of neutrophils. UC-MSCs can also subtract the release of proinflammatory factors and induce immune reconstruction, in addition to reducing the immune inflammatory reaction. Therefore, UC-MSCs may provide a new way to care for RA. Another intriguing feature of MSCs is that they tin can escape allowed recognition and inhibit immune responses and could get a potential tool for immunomodulatory cell therapy in immune-mediated diseases.43

Injury Repair

UC-MSCs accept potent proliferative abilities and multidirectional properties and tin can differentiate into developed cells in vivo or in vitro under appropriate environments.44 UC-MSCs have the capacity for tissue repair and regeneration. UC-MSCs secrete a range of growth factors, such as hepatocyte growth factor (HGF), vascular endothelial growth gene (VEGF), stromal cell-derived factor-1 (SDF-1), keratinocyte growth factor (KGF), fibroblast growth factor (FGF), and insulin-similar growth factor-1 (IGF-1), to facilitate proliferation and tissue damage repair.45,46 MSCs take the potential to be pluripotent and capable of giving rise to a diverseness of cell types. The properties of diverse factors, growth factors, and extracellular vesicles can be transmitted.47 NF-κB and other cellular signaling pathways can be afflicted to treat diseases.48 After intravenous injection, UC-MSCs can differentiate into alveolar epithelial cells and pulmonary vascular endothelial cells.49 Additionally, UC-MSCs can play a part in injury repair. There is bear witness that angiopoietin-1 (ANG-one) and angiopoietin-two (ANG-two) are secreted with KGF to repair harm. MSCs have homing abilities, can drift to injured sites, and can differentiate into local components of injured sites and assist tissue regeneration past secreting chemokines, cytokines, and growth factors45. Hence, UC-MSCs can be widely used in articular cartilage repair.

Reduction in the Inflammatory Response

UC-MSCs play a role in inflammation at the site of injury to mitigate the inflammatory response. Active migration to the injured site for repair is called the "render" of the MSCs' "nest part," and a strong tendency has been demonstrated in animate being experiments in vivo under various microenvironmental weather condition.50,51 Researchers have explored how the immunomodulatory effects of nodules are mediated by UC-MSCs in vitro.52–55 UC-MSCs can express a variety of MSC markers and a variety of stem cell-related genes, are cells with the potential for multidirectional differentiation, and have low immunogenicity, which may be due to the depression level of MHC-I antigens and no expression of MHC-Two antigens or FasL. The furnishings of these cells are not related to the expression of costimulatory molecules associated with T jail cell activation.56 UC-MSCs significantly reduce inflammation by extensively downregulating inflammatory products or cytotoxic mediators that human activity on allowed cells and increment the level of the anti-inflammatory cytokine IL-x.57 In addition to suppressing the immune response, UC-MSCs induce the production of IL-ten-secreting regulatory T cells (Tregs) during local inflammation. Il-ten, a characteristic Treg cytokine, plays an important part in controlling autoantigen-responsive T cells in vivo, and these Tregs inhibit the effects of effector T cells on antigens. UC-MSCs extensively reduce cytokines and chemokines and direct actuate macrophages.58

Greish et al59 used UC-MSCs to treat an RA rat model induced with complete Freund'southward adjuvant (CFA), and the results showed that the group of rats that received hematopoietic stem cells (HSCs) and the group of rats that received UC-MSCs both showed improvements in arthritis symptoms subsequently 21 days of treatment, and arthritis symptoms completely disappeared after 34 days. The mean diameter in the UC-MSC group was but half that of the control grouping and of the MTX group, and that of the UC-MSC grouping was 0.half dozen mm smaller than that of the HSC group. Serum levels of TNF-α, IFN-γ, and IL-1 in the UC-MSC group were significantly decreased compared with those of the command group and the MTX group, while the expression of IL-10 was increased. Hematoxylin-eosin (HE) staining showed that UC-MSCs could significantly reduce leukocyte infiltration and synovial tissue hyperplasia in the articular cavity. Masson staining showed edema in the intrasynovial vessels in near all arthritic rats, with vacuoles forming in the vessel walls, ultimately leading to hemorrhagic necrosis. Additionally, the UC-MSC group likewise showed a reduction in extensive fibrosis of the articulation space, which verified that UC-MSCs could improve the symptoms of CFA-induced RA, and the machinery of activeness may be through regulating the expression of cytokines and improving pathological changes.

Clinical Enquiry

At present, many universities, medical institutions, scientific research institutions, and enterprises have launched dozens of MSC treatments for RA affliction. Related clinical studies are proceeding. Past July 2020, according to ClinicalTrials.gov, an international clinical trial registry, multiple clinical trials of UC-MSCs for the handling of RA have been recorded. There have been as many as six application cases, which have been organized by domestic units and Korean affiliates, using UC-MSCs either alone or combined with DMARDs. The interventions, outcomes, measures, and study designs were analyzed. Information on the clinical trial Phase Is shown in Table 1. Domestic research is more often than not included. There was one clinical trial away with applicants from Korea ({"type":"clinical-trial","attrs":{"text":"NCT03618784","term_id":"NCT03618784"}}NCT03618784). The written report designs were similar to the domestic studies, and the evaluation included erythrocyte sedimentation charge per unit (ESR), Korean Health Assessment Questionnaire (KHAQ), clinical affliction activeness alphabetize (CDAI), 100 mm pain VAS and changes in cytokines.

Tabular array 1

Clinical Trial of Umbilical Cord Mesenchymal Stem Cells Therapy of RA from ClinicalTrials.gov

NCT Number Interventions Upshot Measures Historic period Phases Enrollment Written report Designs
{"blazon":"clinical-trial","attrs":{"text":"NCT02643823","term_id":"NCT02643823"}}NCT02643823 hUC-MSC+DMARDs; DMARDs SAE; RA Serology; DAS 28 Index 18–eighty Phase 1 forty Allocation: Randomized
Intervention Model: Parallel Assignment
Masking: None (Open Label)
Primary Purpose: Treatment
{"type":"clinical-trial","attrs":{"text":"NCT01985464","term_id":"NCT01985464"}}NCT01985464 hUC-MSC AE;28-DAS; EULAR; QOL; HAQ; at months 3 and 12 of CRP; ESR anti-CAM; RF; EULAR ≥18 Phase 1|Phase 2 20 Allocation: North/A
Intervention Model: Single Group Assignment
Masking: None (Open Characterization)
Master Purpose: Handling
{"type":"clinical-trial","attrs":{"text":"NCT01547091","term_id":"NCT01547091"}}NCT01547091 UC-MSCs; DMARDs; UCMSC+DMARDs Safety of MSC handling; RA Serology; DAS 28 Index; Patient'southward assessment of pain. xviii–lxx Phase ane|Phase two 200 Resource allotment: Not-Randomized
Intervention Model: Unmarried Group Assignment
Masking: None (Open up Label)
Master Purpose: Handling
{"type":"clinical-trial","attrs":{"text":"NCT03828344","term_id":"NCT03828344"}}NCT03828344 hUC-MSC;
Placebo
Frequency of AE; SAE
All following indexes at Calendar week 12 and Week 24:ACR20; ACR50; ACR70; DAS28-CRP; HAQ-DI score; SDAI; RF; anti-CCP
18–70 Phase 1 16 Allocation: Randomized
Intervention Model: Parallel Consignment
Masking: Quadruple (Participant, Intendance Provider, Investigator, Outcomes Assessor)
Master Purpose: Treatment
{"type":"clinical-trial","attrs":{"text":"NCT03798028","term_id":"NCT03798028"}}NCT03798028 UC-MSCs BT; HGB; FVC; DLCO; ACR 20; ACR 50; ACR lxx; WBC; PC; FVC DLCO; Lung CT; 6MWT 18–lxx Not Applicable 250 Allocation: Randomized
Intervention Model: Parallel Assignment
Masking: Single (Participant)
Primary Purpose: Treatment
{"type":"clinical-trial","attrs":{"text":"NCT03618784","term_id":"NCT03618784"}}NCT03618784 FURESTEM-RA Inj; Sterile saline AE; ACR20,l,70; EULAR; DAS28-ESR; KHAQ; CDAI; 100mm Hurting VAS; consumed amount of rescue medicine; Modify in Cytokine (TNF-a, IL-1b, IL-4, IL-6, IL-8, IL-10, IL-13, IL-17A, IL-21, IL-22) 19–80 Phase i|Phase two 33 Allocation: Randomized
Intervention Model: Parallel Assignment
Masking: Quadruple (Participant, Intendance Provider, Investigator, Outcomes Assessor)
Master Purpose: Treatment

In most clinical studies, UC-MSCs extensively reduced cytokines and chemokines and directly activated macrophages.8 The pathogenesis of RA and its immune mechanisms are related to cytokine disorders. UC-MSCs are widely bachelor, and their immune rejection is low. Based on preclinical studies, UC-MSCs have very skilful immune regulation and are able to repair tissue impairment. Therefore, the clinical application of prison cell therapy for RA has achieved a certain outcome. Many National Clinical Trials (NCTs) registered on ClinicalTrials.gov are shown in Table one. Most of the studies are from China, and only two FURESTEM-RA Inj trials are from Korea, the latter of which evaluated full general indices such as ACR20/50/70, EULAR, DAS28-ESR, and KHAQ and detected changes in cytokines (TNF-α, IL-1β, IL-4, IL-6, IL-viii, IL-10, IL-13, IL-17A, IL-21, and IL-22).lx In S Korea, a multicenter randomized double-blind parallel placebo-controlled phase I/IIa RA clinical trial ({"type":"clinical-trial","attrs":{"text":"NCT03618784","term_id":"NCT03618784"}}NCT03618784) administered allogeneic UC-MSCs (named FURESTEM-RA Inj) to 33 RA patients. Clinical and safety parameters were monitored for one month following the infusion of UC-MSCs. No serious adverse events or major abnormalities in serum chemical or hematologic profiles were observed. Early clinical studies of the use of UC-MSCs in RA patients have shown promising safety profiles and could significantly improve symptoms and repair damaged tissue.seven,61 Phase 1 clinical trials have been conducted to evaluate the rubber of UC-MSC therapy.

Our previous study was registered at ClinicalTrials.gov ({"blazon":"clinical-trial","attrs":{"text":"NCT01547091","term_id":"NCT01547091"}}NCT01547091),viii in which UC-MSCs were intravenously injected into RA patients. A full of 172 patients were divided into ii groups: those who received DMARDs or those who received DMARDs plus UC-MSCs (4×107 cells per treatment). Diet, sleep, physical strength, fatigue, and other clinical symptoms were examined, and pregnant improvements were observed. For the UC-MSC treatment group, all patients met the ACR20 standard. Twelve patients met the ACR70 criteria, and the DAS 28 score was less than two.6. The levels of complement C3 and C4, routine blood tests, liver and kidney part, and serum immunoglobulin levels showed no significant changes earlier and after treatment. No serious adverse effects occurred during the treatment menstruation, and then UC-MSCs were considered to exhibit immune regulation, prevent the release of inflammatory mediators, reduce soft tissue injury and play a role in other important effects that could alleviate clinical RA symptoms. Thus, safety has been demonstrated for the clinical handling of RA. Additionally, nosotros conducted prospective stage I/II studies in x juvenile idiopathic arthritis (JIA) children56 less than 17 years of age and 64 RA patients7 anile 18 to 64 years. The patients were treated with a twoscore mL of a UC-MSC suspension product (two × 107 cells/20 mL) via intravenous injection. The ESR, C reactive protein (CRP) levels, and rheumatoid gene (RF) levels at 1 year and 3 years after treatment and anti-CCP at iii years afterward handling were significantly lower than those pretreatment (P < 0.05). One yr and three years after treatment, The HAQ and DAS28 decreased in comparison to the pretreatment levels (P < 0.05). Overall, UC-MSC therapy tin be a safe, effective, and viable therapeutic selection for RA patients. Liu and colleague showed that UC-MSCs have therapeutic potential in the handling of RA. The results showed that UC-MSCs suppressed the various inflammatory furnishings of FLSs and T cells. In addition, the immunosuppressive activity of UC-MSCs could be prolonged by the participation of Tregs.

Challenges and Perspectives

Umbilical cord blood and tissue are platonic sources of MSCs because they can be obtained by noninvasive means more hands than from os marrow.60 These cells take a lower relative adventure of viral contamination and can be cryopreserved for autologous transplantation. However, the success rate of cultured MSCs from umbilical string blood is not loftier, with i written report suggesting simply half dozen%, while the success charge per unit of cultured MSCs from umbilical cord claret can reach 100%. Therefore, in recent years, the umbilical cord has go an important source of MSCs. Although the physiological characteristics and mechanisms of action of MSCs accept been deeply studied, there are still many challenges to be solved in the treatment of RA. Now, at that place is no unified plan for stem cell transplantation to care for this illness, such equally pretransplantation medication, transplanted cell numbers, and the optimal timing of treatments. The safety of the handling and the suitable population still need long-term follow-upwards.

Genetic stability is affected by donors, tissue sources, and culture weather during distension, storage conditions, and the time of passage.62,63 Stultz et al,64 examined the chromosomal stability of BM-MSCs from different donors and algebraically analyzed the results. The results showed that there were more than abnormal karyotypes in the 3rd generation than in the fifth and 7th generations, and the abnormal karyotypes gradually decreased with increasing generations through genetic modeling of inheritance. According to different donor ages in the elderly and immature groups, the algebraic model showed that donors from the elderly group were characterized mainly by chromosome translocation, while donors from the young group were characterized by aneuploidy, but in both the elderly group and the young grouping, at that place were no significant differences in chromosomes; chromosomal abnormality occurred mainly in the early stages of the algebraic model, and with more than passage times, there were more mature and stable the chromosomes. Duarte and colleagues65 compared pre- and mail cryopreservation of the human umbilical vein to assess the influence of chromosome status on MSCs and found that nonclone chromosome abnormality was detected after frozen storage in liquid nitrogen, and the 3 short artillery of a chromosome were inverted when the cells were stored for two months.

At present, the MSCs used to study regeneration technology mainly include BM-MSCs and UC-MSCs.7,10,l Cord blood is from the umbilical string near the fetal side of the placenta. This blood is rich in stem cells and progenitor cells and has the potential for multidirectional differentiation. There is no upstanding problem in treatments with cord blood stalk cells. Cathay is a populous country with abundant availability of umbilical string blood. Due to the lack of understanding and a scientific research platform, the evolution of umbilical string blood has been neglected. BM-MSCs have more sufficient sources and are more than convenient to collect than other types of MSCs and do not limited MHC II molecules, so they cannot identify diverse antigenic mechanisms. Due to a lack of allowed activation in two mutual stimulatory signal transduction pathways and immune tolerance, the incidence of GVHD is depression.66 There are also archaic cells, such as endothelial progenitor cells, in umbilical string blood, which are benign to vascular reconstruction and to improving blood supply at the site of injuries, equally such cells play an auxiliary part in repair. Compared with BM-MSCs, UC-MSCs are more than primitive and have stronger amplification abilities. The application of UC-MSCs does not involve ethical issues, and obtaining umbilical string blood from newborns does not cause any impairment to newborns. Because umbilical cord blood is protected by the placental bulwark, it is less likely to crusade viral infections during transplantation than os marrow transplantation. Moreover, cord blood stalk cells are extremely pure. String claret is easy to obtain and is an available resource. UC-MSC therapy involves no upstanding issues and no tissue antigenicity and has a simple transplantation route, obvious and reliable efficacy, and no adverse side effects. UC-MSCs tin promote the directional activation of stem cells and accelerate nerve function repair.

A large number of studies accept confirmed that UC-MSCs play a role in RA disease, but their mechanism remains unclear. There are challenges in the use of UC-MSC therapy in clinical trials. For example, a multiple center randomized controlled trial (RCT) needs to be performed; a large sample size and clinical trials need to be synthetic to verify the therapeutic consequence; there is no unified standard for the optimal dose, optimal time, treatment frequency, approach, treatment conditions and indications of UC-MSC therapy for RA; and whether in that location volition be other potential side effects is a problem that needs to be observed and solved in future research. In summary, UC-MSCs are studied not but in the laboratory only also in the clinic. All studies showed excellent efficacy, especially those utilizing UC-MSCs, which are a potential feasible method of RA treatment.

Conclusion

UC-MSCs evidence skilful efficacy and tolerability in RA patients and have emerged equally a promising alternative in the direction of RA. The pathogenesis of RA is related to disorders of allowed mechanisms and cytokines. UC-MSCs are widely bachelor and have low immunogenic responses, and the limitations, such as the lack of traditional stem cell sources, allogeneic rejection, and ideals, have been overcome. As UC-MSCs have achieved a certain efficacy and few side effects in clinical practice, they are worthy of further large-sample RCTs for future evaluation.

Funding Statement

This enquiry was supported by the Guangxi Zhuang Autonomous Region self-funded project (NO: Z20190359) and the Projection of Liuzhou Science and Technology Agency (No: 2018BJ10502).

Abbreviations

BM, os marrow; BM-MSCs, bone marrow mesenchymal stalk cells; RA, rheumatoid arthritis; UC-MSCs, Umbilical string mesenchymal stalk cells; DAS 28, Disease Activity Score 28-joint; AE, adverse events; ESR, erythrocyte sedimentation charge per unit; RF rheumatoid factor; DMARDs, Drug Rheumatoid Arthritis With Disease-Modifying Drugs; SAE, Serious Adverse Events; DAS28-CRP, disease action score 28-joint count using C reactive protein; HAQ-DI, health cess questionnaire disability index; SDAI, Simplified Disease Activity Index; anti-CCP, anti-cyclic citrullinated peptide; HGB, hemoglobin; CIA, collagen-induced arthritis; FLSs, fibroblast-like synoviocytes; FVC, forced vital capacity; ACR, American College of Rheumatology.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Otón T, Carmona Fifty. The epidemiology of established rheumatoid arthritis. Best Pract Res Clin Rheumatol . 2019;33(five):101477. doi:10.1016/j.berh.2019.101477 [PubMed] [CrossRef] [Google Scholar]

2. Goldring SR. Pathogenesis of os erosions in rheumatoid arthritis. Curr Opin Rheumatol . 2002;fourteen(4):406–410. doi:10.1097/00002281-200207000-00013 [PubMed] [CrossRef] [Google Scholar]

iii. Subesinghe S, Bechman K, Rutherford AI, Goldblatt D, Galloway JB. A systematic review and metaanalysis of antirheumatic drugs and vaccine immunogenicity in rheumatoid arthritis. J Rheumatol . 2018;45(6):733–744. doi:10.3899/jrheum.170710 [PubMed] [CrossRef] [Google Scholar]

4. El-Gabalawy H. The preclinical stages of RA: lessons from human studies and animal models. Best Pract Res Clin Rheumatol . 2009;23(1):49–58. doi:10.1016/j.berh.2008.xi.004 [PubMed] [CrossRef] [Google Scholar]

5. Chang NC. Autophagy and stem cells: self-eating for self-renewal. Front Cell Develop Biol . 2020;8:138. doi:ten.3389/fcell.2020.00138 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Ross CL, Ang DC, Almeida-Porada Chiliad. Targeting mesenchymal stromal cells/pericytes (MSCs) with Pulsed Electromagnetic Field (PEMF) has the potential to treat rheumatoid arthritis. Front end Immunol . 2019;ten:266. doi:10.3389/fimmu.2019.00266 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

7. Wang 50, Huang Due south, Li S, et al. Efficacy and rubber of umbilical cord mesenchymal stem cell therapy for rheumatoid arthritis patients: a prospective phase I/Ii report. Drug Des Devel Ther . 2019;13:4331–4340. doi:10.2147/DDDT.S225613 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

8. Wang 50, Wang L, Cong X, et al. Human umbilical cord mesenchymal stem cell therapy for patients with agile rheumatoid arthritis: safety and efficacy. Stem Cells Dev . 2013;22(23):3192–3202. [PubMed] [Google Scholar]

ix. Lopez-Santalla G, Fernandez-Perez R, Garin MI. Mesenchymal stalk/stromal cells for rheumatoid arthritis treatment: an update on clinical applications. Cells . 2020;9(8):1852. doi:10.3390/cells9081852 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

10. Coelho A, Alvites RD, Branquinho MV, Guerreiro SG, Maurício Ac. Mesenchymal stem cells (MSCs) as a potential therapeutic strategy in COVID-nineteen patients: literature inquiry. Front Cell Develop Biol . 2020;8:602647. doi:10.3389/fcell.2020.602647 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

eleven. Wang L, Huang S, Dang Y, et al. Cord claret-derived cytokine-induced killer cellular therapy plus radiation therapy for esophageal cancer: a case report. Medicine . 2014;93(28):e340. doi:ten.1097/MD.0000000000000340 [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]

12. Wienecke T, Gøtzsche PCJ. Paracetamol versus nonsteroidal anti‐inflammatory drugs for rheumatoid arthritis. Cochrane Database Syst Rev . 2004;(1):CD003789. doi:10.1002/14651858.CD003789.pub2. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

thirteen. Tiano HF, Loftin CD, Akunda J, et al. Deficiency of either cyclooxygenase (COX)-1 or COX-2 alters epidermal differentiation and reduces mouse peel tumorigenesis. Cancer Res . 2002;62(12):3395–3401. [PubMed] [Google Scholar]

fourteen. Lovell DJ, Ruperto N, Goodman S, et al. Adalimumab with or without methotrexate in juvenile rheumatoid arthritis. N Engl J Med . 2008;359(viii):810–820. [PubMed] [Google Scholar]

xv. Schett G, Emery P, Tanaka Y, et al. Tapering biologic and conventional DMARD therapy in rheumatoid arthritis: current prove and futurity directions. Ann Rheum Dis . 2016;75(eight):1428–1437. [PubMed] [Google Scholar]

xvi. Castillo J, Milani C, Mendez-Allwood DJE. Ofatumumab, a second-generation anti-CD20 monoclonal antibiotic, for the treatment of lymphoproliferative and autoimmune disorders. Skillful Opin Investig Drugs . 2009;eighteen(iv):491–500. [PubMed] [Google Scholar]

17. Song S-NJ, Iwahashi M, Tomosugi North, et al. Comparative evaluation of the furnishings of treatment with tocilizumab and TNF-α inhibitors on serum hepcidin, anemia response and disease activeness in rheumatoid arthritis patients. Arthritis Res Ther . 2013;15(v):one–10. [PMC free article] [PubMed] [Google Scholar]

18. Downey CJ. Serious infection during etanercept, infliximab and adalimumab therapy for rheumatoid arthritis: a literature review. J Rheum Dis . 2016;19(6):536–550. [PubMed] [Google Scholar]

19. Smolen JS, Van Der Heijde DM, St. Clair EW, et al. Predictors of articulation damage in patients with early rheumatoid arthritis treated with high‐dose methotrexate with or without concomitant infliximab: results from the ASPIRE trial. Arthritis Rheum . 2006;54(iii):702–710. [PubMed] [Google Scholar]

xx. St. Clair EW, Van Der Heijde DM, Smolen JS, et al. Combination of infliximab and methotrexate therapy for early rheumatoid arthritis: a randomized, controlled trial. Arthritis Rheum 2004;50(11):3432–3443. [PubMed] [Google Scholar]

21. Bartelds GM, Wijbrandts CA, Nurmohamed MT, et al. Clinical response to adalimumab: relationship to anti-adalimumab antibodies and serum adalimumab concentrations in rheumatoid arthritis. Ann Rheum Dis . 2007;66(7):921–926. [PMC gratis article] [PubMed] [Google Scholar]

22. Raaschou P, Simard JF, Holmqvist M, Askling J. Rheumatoid arthritis, anti-tumour necrosis gene therapy, and risk of cancerous melanoma: nationwide population based prospective accomplice study from Sweden. BMJ . 2013;346(f1939). [PubMed] [Google Scholar]

23. Vizoso FJ, Eiro N, Costa Fifty, et al. Mesenchymal stem cells in homeostasis and systemic diseases: hypothesis, evidences, and therapeutic opportunities. Int J Mol Sci . 2019;20(15):3738. doi:x.3390/ijms20153738 [PMC gratis commodity] [PubMed] [CrossRef] [Google Scholar]

24. Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther . 2019;10(1). doi:x.1186/s13287-018-1105-9 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

25. Li Y, Jin D, Xie West, et al. Mesenchymal stem cells-derived exosomes: a possible therapeutic strategy for osteoporosis. Curr Stalk Prison cell Res Ther . 2018;13(five):362–368. doi:10.2174/1574888X13666180403163456 [PubMed] [CrossRef] [Google Scholar]

26. Qu J, Zhang H. Roles of mesenchymal stalk cells in spinal cord injury. Stem Cells Int . 2017;2017:5251313. doi:10.1155/2017/5251313 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

27. Zhou T, Li H-Y, Liao C, Lin W, Lin S. Clinical efficacy and safety of mesenchymal stem cells for systemic lupus erythematosus. Stem Cells Int . 2020;2020:6518508. doi:10.1155/2020/6518508 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

28. Ferro F, Elefante Eastward, Luciano N, Talarico R, Todoerti M. One year in review 2017: novelties in the treatment of rheumatoid arthritis. Clin Exp Rheumatol . 2017;35(5):721–734. [PubMed] [Google Scholar]

29. Silvagni E, Di Battista M, Bonifacio AF, Zucchi D, Governato K, Scirè CA. One yr in review 2019: novelties in the treatment of rheumatoid arthritis. Clin Exp Rheumatol . 2019;37(4):519–534. [PubMed] [Google Scholar]

30. Cheung TT, McInnes IB. Hereafter therapeutic targets in rheumatoid arthritis? Semin Immunopathol . 2017;39(iv):487–500. doi:10.1007/s00281-017-0623-3 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

31. Luo Y, Wu Westward, Gu J, et al. Human being gingival tissue-derived MSC suppress osteoclastogenesis and os erosion via CD39-adenosine betoken pathway in autoimmune arthritis. EBioMedicine . 2019;43:620–631. doi:10.1016/j.ebiom.2019.04.058 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

32. Luque-Campos Northward, Contreras-López RA, Jose Paredes-Martínez G, et al. Mesenchymal stem cells improve rheumatoid arthritis progression by controlling memory T prison cell response. Front end Immunol . 2019;10:798. doi:x.3389/fimmu.2019.00798 [PMC gratis commodity] [PubMed] [CrossRef] [Google Scholar]

33. Liu R, Zhao P, Tan Due west, Zhang M. Cell therapies for refractory rheumatoid arthritis. Clin Exp Rheumatol . 2018;36(5):911–919. [PubMed] [Google Scholar]

34. Haikal SM, Abdeltawab NF, Rashed LA, Abd El-Galil TI, Elmalt HA, Amin MA. Combination therapy of mesenchymal stromal cells and interleukin-iv attenuates rheumatoid arthritis in a collagen-induced murine model. Cells . 2019;8(8):823. doi:10.3390/cells8080823 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

35. Li E, Zhang Z, Jiang B, Yan Fifty, Park JW, Xu RH. Generation of mesenchymal stalk cells from human embryonic stem cells in a complete serum-costless condition. Int J Biol Sci . 2018;xiv(13):1901–1909. doi:10.7150/ijbs.25306 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

36. Hynes K, Brilliant R, Proudman S, Haynes D, Gronthos Southward, Bartold M. Immunomodulatory backdrop of mesenchymal stalk cell in experimental arthritis in rat and mouse models: a systematic review. Semin Arthritis Rheum . 2016;46(1):1–nineteen. doi:ten.1016/j.semarthrit.2016.02.008 [PubMed] [CrossRef] [Google Scholar]

37. Andrzejewska A, Lukomska B, Janowski Grand. Concise review: mesenchymal stem cells: from roots to boost. Stalk Cells . 2019;37(7):855–864. doi:x.1002/stem.3016 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

38. Liu Y, Mu R, Wang S, et al. Therapeutic potential of man umbilical string mesenchymal stem cells in the handling of rheumatoid arthritis. Arthritis Res Ther . 2010;12(vi):R210. doi:10.1186/ar3187 [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]

39. Berglund AK, Fisher MB, Cameron KA, Poole EJ, Schnabel LV. Transforming growth factor-β2 downregulates Major Histocompatibility Complex (MHC) I and MHC II surface expression on equine bone marrow-derived mesenchymal stalk cells without altering other phenotypic cell surface markers. Front end Vet Sci . 2017;4:84. doi:10.3389/fvets.2017.00084 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

xl. Karahuseyinoglu S, Cinar O, Kilic Due east, et al. Biology of stem cells in human umbilical cord stroma: in situ and in vitro surveys. Stalk Cells . 2007;25(2):319–331. doi:10.1634/stemcells.2006-0286 [PubMed] [CrossRef] [Google Scholar]

41. Wang Q, Li X, Luo J, et al. The allogeneic umbilical string mesenchymal stem cells regulate the function of T helper 17 cells from patients with rheumatoid arthritis in an in vitro co-culture system. BMC Musculoskelet Disord . 2012;13(ane):249. doi:10.1186/1471-2474-thirteen-249 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

42. Gu J, Lin CM, Gu W, et al. [Immunomodulatory event of UC-MSC on function of immunocytes of rats with collagen type II induced arthritis]. Zhongguo Shi Yan Xue Ye Xue Za Zhi . 2014;22(1):166–170. [Chinese] [PubMed] [Google Scholar]

43. Zhao Q, Ren H, Han Z. Mesenchymal stalk cells: immunomodulatory capability and clinical potential in immune diseases. J Cell Immunother . 2016;2(i):iii–20. doi:ten.1016/j.jocit.2014.12.001 [CrossRef] [Google Scholar]

44. Shou Chiliad, Huang Y, Qi B, et al. Induction of mesenchymal stem cell differentiation in the absence of soluble inducer for cutaneous wound regeneration by a chitin nanofiber-based hydrogel. J Tissue Eng Regen Med . 2018;12(ii):e867–e880. doi:10.1002/term.2400 [PubMed] [CrossRef] [Google Scholar]

45. Fu X, Liu Thou, Halim A, Ju Y, Luo Q, Vocal AG. Mesenchymal stem cell migration and tissue repair. Cells . 2019;viii(8):784. [PMC free commodity] [PubMed] [Google Scholar]

46. Chen 50, Carlton M, Chen X, et al. Result of fibronectin, FGF-2, and BMP4 in the stemness maintenance of BMSCs and the metabolic and proteomic cues involved. Stem Cell Res Ther . 2021;12(1):165. doi:10.1186/s13287-021-02227-vii [PMC free article] [PubMed] [CrossRef] [Google Scholar]

47. Xu L, Liu Y, Sun Y, et al. Tissue source determines the differentiation potentials of mesenchymal stem cells: a comparative study of man mesenchymal stem cells from os marrow and adipose tissue. Stem Cell Res Ther . 2017;8(one):275. doi:ten.1186/s13287-017-0716-ten [PMC free article] [PubMed] [CrossRef] [Google Scholar]

48. Zhang B, Luo Q, Halim A, Ju Y, Morita Y, Song G. Directed differentiation and paracrine mechanisms of mesenchymal stalk cells: potential implications for tendon repair and regeneration. Curr Stem Cell Res Ther . 2017;12(6):447–454. doi:10.2174/1574888X12666170502102423 [PubMed] [CrossRef] [Google Scholar]

49. Vocal H, Yin Z, Wu T, et al. Enhanced effect of tendon stem/progenitor cells combined with tendon-derived decellularized extracellular matrix on tendon regeneration. Jail cell Transplant . 2018;27(11):1634–1643. doi:10.1177/0963689718805383 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

50. Wu Y, Wang J, Scott PG, Tredget EE. Bone marrow-derived stalk cells in wound healing: a review. Wound Repair Regen . 2007;xv(Suppl s1):S18–26. doi:10.1111/j.1524-475X.2007.00221.x [PubMed] [CrossRef] [Google Scholar]

51. Herdrich BJ, Lind RC, Liechty KW. Multipotent adult progenitor cells: their office in wound healing and the treatment of dermal wounds. Cytotherapy . 2008;10(6):543–550. doi:10.1080/14653240802345820 [PubMed] [CrossRef] [Google Scholar]

52. Cha J, Falanga V. Stalk cells in cutaneous wound healing. Clin Dermatol . 2007;25(i):73–78. doi:10.1016/j.clindermatol.2006.ten.002 [PubMed] [CrossRef] [Google Scholar]

53. Fu X, Li H. Mesenchymal stem cells and skin wound repair and regeneration: possibilities and questions. Jail cell Tissue Res . 2009;335(ii):317–321. doi:10.1007/s00441-008-0724-3 [PubMed] [CrossRef] [Google Scholar]

54. Suh Due west, Kim KL, Kim JM, et al. Transplantation of endothelial progenitor cells accelerates dermal wound healing with increased recruitment of monocytes/macrophages and neovascularization. Stem Cells . 2005;23(ten):1571–1578. doi:10.1634/stemcells.2004-0340 [PubMed] [CrossRef] [Google Scholar]

55. Volk SW, Radu A, Zhang L, Liechty KW. Stromal progenitor cell therapy corrects the wound-healing defect in the ischemic rabbit ear model of chronic wound repair. Wound Repair Regen . 2007;xv(5):736–747. doi:10.1111/j.1524-475X.2007.00277.x [PubMed] [CrossRef] [Google Scholar]

56. Wei Z, Yuan J, Wang G, Ocansey DKW, Xu Z, Mao FJSCI. Regulatory effect of mesenchymal stem cells on t cell phenotypes in autoimmune diseases. Stem Cell Int . 2021;2021:5583994. doi:10.1155/2021/5583994 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

57. Mattar P, Bieback K. Comparing the immunomodulatory properties of os marrow, adipose tissue, and birth-associated tissue mesenchymal stromal cells. Front Immunol . 2015;vi:560. [PMC free commodity] [PubMed] [Google Scholar]

58. Song J-Y, Kang HJ, Hong JS, et al. Umbilical cord-derived mesenchymal stalk cell extracts reduce colitis in mice by re-polarizing intestinal macrophages. Sci Rep . 2017;7(1):1–11. [PMC free commodity] [PubMed] [Google Scholar]

59. Greish S, Abogresha N, Abdel-Hady Z, Zakaria E, Ghaly M, Hefny One thousand. Human being umbilical cord mesenchymal stalk cells equally treatment of adjuvant rheumatoid arthritis in a rat model. Earth J Stem Cells . 2012;iv(10):101–109. doi:10.4252/wjsc.v4.i10.101 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

60. Park EH, Lim HS, Lee S, et al. Intravenous infusion of umbilical cord blood-derived mesenchymal stem cells in rheumatoid arthritis: a phase Ia clinical trial. Stem Cells Transl Med . 2018;7(9):636–642. doi:10.1002/sctm.18-0031 [PMC costless commodity] [PubMed] [CrossRef] [Google Scholar]

61. Qi T, Gao H, Dang Y, Huang S, Peng M. Cervus and cucumis peptides combined umbilical cord mesenchymal stem cells therapy for rheumatoid arthritis. Medicine . 2020;99(28):e21222. doi:ten.1097/MD.0000000000021222 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

62. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi Thou. Cell therapy using allogeneic os marrow mesenchymal stalk cells prevents tissue harm in collagen-induced arthritis. Arthritis Rheum . 2007;56(4):1175–1186. doi:10.1002/art.22511 [PubMed] [CrossRef] [Google Scholar]

63. Sensebé L, Tarte K, Galipeau J, et al. Express acquisition of chromosomal aberrations in human developed mesenchymal stromal cells. Cell Stem Jail cell . 2012;10(one):ix–10. doi:10.1016/j.stem.2011.12.005 [PubMed] [CrossRef] [Google Scholar]

64. Stultz BG, McGinnis Thou, Thompson EE, Lo Surdo JL, Bauer SR, Hursh DA. Chromosomal stability of mesenchymal stromal cells during in vitro civilisation. Cytotherapy . 2016;18(iii):336–343. doi:10.1016/j.jcyt.2015.11.017 [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]

65. Duarte DM, Cornélio DA, Corado C, et al. Chromosomal characterization of cryopreserved mesenchymal stalk cells from the human subendothelium umbilical string vein. Regen Med . 2012;7(ii):147–157. doi:10.2217/rme.11.113 [PubMed] [CrossRef] [Google Scholar]

66. Zheng G, Liu Y, Jing Q, Zhang L. Differentiation of human umbilical cord-derived mesenchymal stalk cells into hepatocytes in vitro. Biomed Mater Eng . 2015;25(one Suppl):145–157. doi:x.3233/BME-141249 [PubMed] [CrossRef] [Google Scholar]


Articles from Drug Design, Development and Therapy are provided here courtesy of Dove Press


moonhibed1982.blogspot.com

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8462093/

0 Response to "Umbilical Stem Cell Therapy for Knees Reviews 2019"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel