Journal of Stem Cells Research Development & Therapy Category: Medical Type: Short Review

Mesenchymal Stem Cells in the Treatment of Knee Osteoarthritis

Vitorio Peric1*, Tomislav Kottek2, Vilim Molnar1,3, Vid Matisic1, Fabijan Cukelj1,4, Dragan Primorac1,3,4,5

1 St. Catherine Specialty Hospital, 49210 Zabok/ 10000 Zagreb, Croatia
2 School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
3 School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
4 University of Split, Medical School, 21000 Split, Croatia
5 Eberly college of science, The Pennsylvania State University, University Park, State College, 16802 PA, United states

*Corresponding Author(s):
Vitorio Peric
St. Catherine Specialty Hospital, 49210 Zabok/ 10000 Zagreb, Croatia
Tel:+385 955315388,
Email:vitorioperic@gmail.com

Received Date: Sep 04, 2020
Accepted Date: Sep 17, 2020
Published Date: Sep 23, 2020

Abstract

Osteoarthritis is the most common musculoskeletal progressive disease, affecting 303 million people worldwide. The prevalence of knee OA among adults 60 years-of-age and older is approximately 10% in men and 13% in women. Although surgical treatment still provides the gold standard in knee osteoarthritis therapy, mesenchymal stem cell therapy is increasingly gaining the attention of physicians. Therefore, a thorough understanding of osteoarthritis pathogenesis is essential for successful treatment. Mesenchymal stem cells are adult stem cells that can be found in bone marrow, adipose tissue, peripheral blood, skeletal muscle, hearth and umbilical cord. Their characteristics include self-renewal, proliferation and differentiation into several cell types. This review provides an insight into the research conducted so far with special reference to the comparison of different types of mesenchymal stem cells in the knee osteoarthritis treatment.

Keywords

Adipose tissue; Bone marrow; Knee osteoarthritis; Mesenchymal stem cells; Placental tissue

INTRODUCTION

Osteoarthritis (OA) is the most common progressive musculoskeletal disorder, affecting 303 million people worldwide [1]. Although OA can affect any joint in the body, it most commonly affects weight-bearing hip and knee joints [2]. The prevalence of knee OA among adults 60 years-of-age or older is approximately 10% in men and 13% in women and is constantly increasing due to the growing number of obese population, longer life expectancy and aging population [3].

The role of cytokines, chemokines, miRNA, gene expression and imbalance between anabolic and catabolic pathways is crucial in understanding the pathogenesis of OA [4]. Knee OA results in structural modifications in articular cartilage and the subchondral bone, but also Hoffa's pad, synovia, ligaments and muscles [4]. Therefore, OA can be observed and interpreted as a whole joint disease.

Patients suffering from OA present with pain, stiffness, swelling and reduced range of motion in the affected joint, all of which result in reduced quality of life and mental health impairment due to chronic pain [5]. While joint replacement surgery is the gold standard for knee OA treatment, new treatment options have drawn clinicians’ attention as a potentially preferable alternative method for OA treatment. These methods can be divided into pharmacological and non-pharmacological therapies. An unavoidable part of successful OA treatment is physical therapy and bodyweight reduction, i.e. reducing the load in the joints that present major non-pharmacological methods. Optimal pharmacologic treatment for pain and disability caused by OA should be tailored in accordance with individual patient needs and their comorbidities. The emerging field of pharmacogenomics provides the clinician with new options to guide precision treatment with a maximal therapeutic effect and minimal risk to the patient [6,7]. A new class of drugs called S/DMOADs (symptomatic or disease-modifying osteoarthritic drugs), including chondroitin sulfate and glucosamine, are commonly used in pharmacological therapy for OA, while injections of autologous mesenchymal stem cells, Hyaluronic Acid (HA), Platelet-Rich Plasma (PRP) are becoming increasingly used methods [8]. In this short commentary review, we will focus on current findings on mesenchymal stem cells in the treatment of knee OA.

MESENCHYMAL STEM CELL TREATMENT OF KNEE OSTEOARTHRITIS

Stem cells emerged as a promising line of treatment for OA due to their characteristics, such as self-renewal, proliferation and differentiation into several cell types [9]. Mesenchymal Stem Cells (MSCs) are adult stem cells present in various tissues throughout the body. For instance, they can be found in bone marrow, adipose tissue, peripheral blood, skeletal muscle, heart and umbilical cord [10]. Their ability to differentiate towards osteoblasts, chondrocytes and adipocytes, together with generating immunomodulatory and paracrine mechanisms around damaged tissue have put them in the spotlight in the field of regenerative medicine [11]. The mechanisms by which MSCs repair damaged cartilage are inhibition of cell apoptosis, reduction of inflammation by suppressing activation, proliferation and infiltration of macrophages, T and B- lymphocytes; secretion of trophic, chondrogenic, angiogenic, anti-fibrotic and anti-catabolic factors [12]. Many studies have been conducted with the aim of selecting the best source of MSCs. Factors such as the amount of harvest volume, cell isolation procedure, the regenerative capacity of certain cells and the risks of harvesting procedure should be assessed [13]. A systematic review conducted in 2018 included 28 scientific articles and confirmed the safety of MSCs in the treatment of various musculoskeletal pathologies [14].

Bone-marrow mesenchymal stem cells

Bone-Marrow Mesenchymal Stem Cells (BM-MSCs) are usually obtained by aspiration from the posterior or anterior iliac crest. Since it is estimated that only about 0.001% of nucleated cells from BM aspirate are MSCs, density-gradient centrifugation of the aspirate is needed to produce a Bone Marrow Aspirate Concentrate (BMAC) [15]. This process increases not only the number of MSCs but also hematopoietic stem cells and platelets containing growth factors, important for stem cell migration and chondrogenesis [16]. The observed clinical results of BM-MSCs therapy are generally positive. A recent meta-analysis indicated improvement in pain level measured by the Visual Analogue Scale (VAS), International Knee Documentation Committee (IKDC) function score, Tegner Activity Scale and Lysholm Knee score when compared to respective results before treatment with BM-MSCs [17]. A recent literature review of clinical data published between 2014 and 2019 regarding intraarticular autologous BM-MSCs injections also showed predominantly positive results, pointing out that a moderate-high number of cells (40 × 106) achieves optimal responses in individuals with grade ≥ 2 knee OA on the Kellgren-Lawrence scale, while lower (24 × 106) and higher (100 × 106) cell numbers, despite showing significant improvement, are associated with a larger number of adverse effects, such as persistent knee pain and swelling [18]. Although the intraarticular injection is the most commonly used method of BM-MSCs application, a recently published clinical trial determined that the BM-MSCs injection in the subchondral bone of an osteoarthritic knee is more effective to postpone total knee arthroplasty comparing to intraarticular injection [19].

Adipose-derived mesenchymal stem cells

Adipose-Derived Mesenchymal Stem Cells (AD-MSCs) are usually obtained from abdominal subcutaneous adipose tissue by lipoaspiration [20]. Those procedures are less invasive compared to BM-MSCs extraction procedures. Moreover, adipose tissue contains 500 times more MSCs compared to the same volume of bone marrow [13,21]. Adipose tissue provides a significant, easily accessible source of cells contained in stromal vascular fraction (SVF) for prompt administration and provides a compelling amount of cells from which multipotent AD-MSCs can be isolated [22]. At present, AD-MSCs in the form of Stromal Vascular Fraction from Lipoaspirate (SVF-LA) or Stromal Vascular Fraction from Microfragmented Lipoaspirate (SVF-MLA) are used in most clinical trials and treatment protocols. A study that analyzed cell phenotypes within CD45 - fraction in these two samples (SVF-LA and SVF-MLA) identified the following cell types: Endothelial Progenitor Cells (EPC), endothelial mature cells, pericytes, transitional pericytes, and Supra Adventitial-Adipose Stromal Cells (SA-ASC), with a surprising and intriguing result of increased EPC number, and reduction of leukocytes and SA-ASC in SVF-MLA compared with SVF-LA [23].

When applied intraarticularly, AD-MSCs therapy showed significant clinical improvement in numerous studies. Improvements in clinical scores such as The Knee Injury and Osteoarthritis Outcome Score (KOOS), The Western Ontario and McMaster Universities Arthritis Index (WOMAC), Timed Up and Go test (TUG) and VAS (both in resting and in movement), as well as an increase in range of motion (ROM), were observed [24-26]. Also, structural analysis, measured by MRI Osteoarthritis Knee Scores (MOAKS), showed a significant rate of cartilage loss regression and less osteophyte formation comparing the group treated with AD-MCSs with the group treated conservatively [27,28]. Furthermore, studies have shown that intraarticular application of AD-MSCs has an impact on proteoglycan synthesis in the cartilage of patients with knee OA. Measured by the dGEMRIC index (delayed gadolinium-enhanced magnetic resonance imaging of cartilage), hyaline cartilage Glycosaminoglycan (GAG) content significantly increased six and twelve months after the treatment [29]. The observed effects of AD-MSCs were visible in a two-year follow-up period after the initial intraarticular intervention [30]. A recent study showed a systemic effect on circulating immune cells after intraarticular application of AD-MSCs in patients suffering from knee OA. The increased percentage of regulatory T cells, as well as transitional B cells, persisted for at least 3 months after treatment, whilst the monocyte level decreased and remained low at 3 months after AD-MSC injection. The studies, therefore, indicate that AD-MSCs treatment provides a long-lasting clinical and systemic immunomodulatory effect [25,31]. Current concepts that include co-administration of AD-MSCs alongside HA or PRP could offer interesting results. It is stated that HA provides an environment in which AD-MSCs can more easily adhere to the target area around the lesion and differentiate into cells needed to build damaged bone and cartilage components whereas PRP contains highly concentrated platelets and a wide range of growth factors providing AD-MSCs proliferation [32,33] (Table 1).

Author

Year of publication

Number of participants

MSC harvest location

Outcomes

Hernigou et al. [19]

2020

60 (120 knees)

BM-MSCs

VAS pain scores were decreased 12 months after the initial treatment with BM-MSCs applied intraarticularly and into the subchondral bone. The results were significantly better in the subchondral MSCs group and the effect had been seen for 24 months. Regression of bone marrow lesions and synovitis scores (MRI based) was also noticed in the subchondral group after 24 months.

Fodor et al. [24]

2016

6

AD-MSCs

(SVF)

Significant improvement in WOMAC and VAS scores after 12 months. Increased ROM and TUG 3 months after the initial procedure.

Pers et al. [25]

2016

18

AD-MSCs (SVF)

Reduction in pain level (VAS) and WOMAC in all three groups 6 months after treatment with statistically significant results only in the low dose group (2x106 cells injected).

Hudetz et al. [26]

2019

20

MFAT

(SVF)

Improvement in KOOS and reduction in pain level (VAS), as well as WOMAC, 12 months after treatment

Freitag et al. [27]

2019

30

AD-MSCs

SVF

Patients were divided into 3 groups. One injection group (100x106 AD-MSCs at baseline) and two injection group (100 x106 AD-MSCs at baseline and 6 months) showed improvement in pain level (NPRS), WOMAC and KOOS comparing to the control group.

Higuchi et al. [28]

2020

34 (57 knees)

AD-MSCs

Patients were injected approximately 1x108 AD-MSCs in the affected knee. Improvements in VAS score and KOOS scale and subscales including KOOS-pain, KOOS-symptom, KOOS-QOL, and KOOS-ADL were registered 6 months after therapy.

Hudetz et al. [29]

2017

17 (32 knees)

MFAT (SVF)

The increase in GAG content was measured using dGEMRIC. Any increase of more than 15% is considered a relevant change. Out of the 331 total measurements, 175 showed improvement in GAG content by 52.9% 12 months after the treatment.

Boric et al. [30]

2019

10 (18 knees)

MFAT (SVF)

Patients were assessed 24 months after AD-MSCs administration as a continuation of the research of Hudetz et al., conducted in 2017. Results were evaluated using an indirect approach that involves estimating GAG content by dGEMRIC and a direct approach based on the VAS score. Out of 19 conducted measurements, 12 showed improvements compared to baseline measurements. 7 measurements showed a decrease in GAG content compared to baseline. VAS score improved in all patients.

Castellanos et al. [34]

2019

20

AMUC

The decrease in knee pain at 12 and 24 weeks after the treatment, improvement in physical function and stiffness (measured by WOMAC) 12 weeks after the treatment.

Khalifeh Soltani et al. [35]

2019

20

PLMSCs

Improvements in clinical measures of pain, symptoms, ADL, S/R (measured by KOOS) and ROM compared with saline injection group

Farr et al. [36]

2019

200

ASA

Improvements in pain (VAS), activities of daily living (KOOS) compared with saline in HA groups

Ryu et al. [37]

2020

52

BMAC and hUCB-MSCs

Improvements in VAS, IKDC, KOOS without significant differences between the two groups

Table 1: Clinical outcomes from different sources of MSCs used in clinical studies reviewed in the article. BM-MSCs - Bone Marrow-Mesenchymal Stem Cells; VAS - Visual Analog Scale; MSCs - Mesenchymal Stem Cells; AD-MSCs - Adipose-Derived Mesenchymal Stem Cells; SVF - Stromal Vascular Fraction; WOMAC -The Western Ontario and McMaster Universities Arthritis Index; ROM - Range Of Motion; TUG - Timed Up and Go; KOOS - The Knee Injury and Osteoarthritis Outcome Score; MFAT - Microfragmented Adipose Tissue; NPRS - Numeric Pain Rating Scale; QOL - Quality of Life; ADL - Activities Of Daily Living; GAG -Glycosaminoglycans; dGEMRIC - delayed Gadolinium (Gd)-Enhanced Magnetic Resonance Imaging of Cartilage; AMUC - Amniotic Membrane/Umbilical Cord Particulate; PLMSCs - Placental Derived Mesenchymal Stem Cells; S/R - Sport and Recreation; ASA - Amniotic Suspension Allograft; HA – Hyaluronic Acid; BMAC - Bone Marrow Aspirate Concentrate; hUCB - MSCs- Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells; IKDC - International Knee Documentation Committee.

Placental tissue

Placental tissue also represents a source of stem cells. Neonatal MSCs can be isolated from amniotic fluid, amnion, chorion, umbilical cord tissue, and blood. Containing not only MSCs, but also a collagen-rich structural matrix, epithelial cells, fibroblasts and several biologically active factors, placental tissue has been successfully used as a treatment for burns and wounds, and recent data suggest potential benefits in orthopedic [34,38]. Although the number of trials is still small, the described outcomes are generally positive. The available literature demonstrated that allogenic placental mesenchymal stem cells are safe and efficient in terms of clinical improvements regarding knee ROM, quality of life, the activity of daily living, sport/recreational activity, pain reduction and even chondral thickness in a 24-week follow-up period [35]. Amniotic tissue, a distinct placental tissue, contains stromal cells which have the chondrogenic and osteogenic capacity, and also provides a rich source of HA and proteoglycans, essential for cartilage structure [39]. A randomized controlled single-blind study including 200 patients with moderate knee OA evaluated the efficacy of symptom modulation with Amniotic Suspension Allograft (ASA) injection compared with saline and HA. Patients receiving ASA presented with both statistically significant and clinically meaningful improvements compared with the other two groups [36]. Another study compared the effects of BMAC and allogeneic human umbilical cord blood-derived MSC implantation in the osteoarthritic knee, and results showed that there is no significant difference in clinical outcome between the two methods [37]. Even though, more studies are required to investigate potential risks of allogeneic cell implantations before such methods find a place in everyday clinical practice.

CONCLUSION

In recent years, due to the increasing use of MSCs in clinical practice around the world, scientific research about MSCs has become increasingly extensive and relevant. Although the results so far are promising, pointing out the advantages of MSCs treatment such as reduction of patient’s knee pain, regression of affected joint damage, less invasiveness and shorter hospital stay, additional studies that would address the optimal procedure steps, timing and number of injections, as well as patient selection criteria are still needed to establish these methods in everyday clinical practice. Novel methods, including a combination of currently available therapeutic approaches and different administration methods (intraarticular, subchondral) could offer better treatment outcomes in the future.

Comparing the two most commonly used sources of MSCs for the knee OA treatment: AD-MSCs showed to be more easily harvested than BM-MSCs, with less invasive and less painful harvesting procedures. Furthermore, adipose tissue provided a higher count of MSCs when compared to the same harvested volume of bone marrow. In vitro studies show different regenerative capacities of AD-MSCs and BM-MSCs, however, the in vivo effect is still measured in a subjective, symptom-specific outcome rather than a thorough biochemical, histological and pathophysiologic effect on OA [40]. More studies are required to address and compare the regenerative capacity of different sources of MSCs before definitive conclusions can be drawn. Also, the benefits of placental tissue should be further investigated, together with potential side effects from allogeneic transplants.

The main limitations of the conducted studies include lack of standardization associated with patient selection and assessment parameters, as well as undefined and insufficiently long post-treatment follow-up period. Furthermore, there is still no sufficiently effective, inexpensive and reliable method, by which the number of applied MSCs could be determined. Such a method could serve as the foundation of advances in personalized medicine and could provide even greater advances in OA therapy. 

While joint replacement surgery still represents the gold standard in the treatment of OA, MSCs therapy provides a possibly great alternative and is assumed that it will take a major role in future OA treatment.

AUTHOR’S CONTRIBUTION

VP, TK and VMo reviewed the literature and drafted the manuscript, VMa, F? and DP critically revised the manuscript. All of the authors approved the final version of the manuscript.

FUNDING

This research received no external funding.

ACKNOWLEDGEMENTS

No funding was received to support the research or data analysis in this article.

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

REFERENCES

  1. Kloppenburg M, Berenbaum F (2020) Osteoarthritis year in review 2019: Epidemiology and therapy. Osteoarthr Cartil 28: 242-248.
  2. Bortoluzzi A, Furini F, Scirè CA (2018) Osteoarthritis and its management - Epidemiology, nutritional aspects and environmental factors. Autoimmun Rev 17: 1097-1104.
  3. Moghimi N, Rahmani K, Delpisheh A, Saidi A, Azadi NA, et al. (2019) Risk factors of knee osteoarthritis: A case-control study. Pakistan J Med Sci 35: 636-640.
  4. Primorac D, Molnar V, Rod E, Jele? Ž, ?ukelj F, et al. (2020) Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes (Basel) 11: 854.
  5. Park H, Kim H, Lee Y (2020) Knee osteoarthritis and its association with mental health and health-related quality of life: A nationwide cross-sectional study. Geriatr Gerontol Int 20: 379-383.
  6. Primorac D, Bach-Rojecky L, Va?unec D, Juginovi? A, Žuni? K, et al. (2020) Pharmacogenomics at the center of precision medicine: Challenges and perspective in an era of Big Data. Pharmacogenomics 21: 141-156.
  7. Höppner W, Primorac D (2016) Pharmacogenetics in clinical practice: Experience with 16 commonly used drugs. St. Catherine Hospital, Zagreb, Berlin, Hamburg.
  8. Hudetz D, Jele? Ž, Rod E, Bori? I, Ple?ko M, et al. (2019) The Future of Cartilage Repair. In: Bodiroga-Vukobrat N, et al. (eds), Personalized Medicine in Healthcare Systems: Legal, Medical and Economic Implications. Cham: Springer International Publishing. Pg no: 375-411.
  9. Kim GB, Shon O-J (2020) Current perspectives in stem cell therapies for osteoarthritis of the knee. Yeungnam Univ J Med 37: 149-158.
  10. Väänänen HK (2005) Mesenchymal stem cells. Ann Med 37: 469-479.
  11. Freitag J, Bates D, Boyd R, Shah K, Barnard A, et al. (2016) Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy - a review. BMC Musculoskelet Disord 17: 230.
  12. Mancuso P, Raman S, Glynn A, Barry F, Murphy JM (2019) Mesenchymal Stem Cell Therapy for Osteoarthritis: The Critical Role of the Cell Secretome. Front Bioeng Biotechnol 7: 9.
  13. Shariatzadeh M, Song J, Wilson SL (2019) The efficacy of different sources of mesenchymal stem cells for the treatment of knee osteoarthritis. Cell Tissue Res 378: 399-410.
  14. McIntyre JA, Jones IA, Han B, Vangsness CT (2018) Intra-articular Mesenchymal Stem Cell Therapy for the Human Joint: A Systematic Review. Am J Sports Med 46: 3550-3563.
  15. Kasten P, Beyen I, Egermann M, Suda AJ, Moghaddam AA, et al. (2008) Instant stem cell therapy: Characterization and concentration of human mesenchymal stem cells in vitro. Eur Cells Mater 16: 47-55.
  16. Madry H, Gao L, Eichler H, Orth P, Cucchiarini M (2017) Bone Marrow Aspirate Concentrate-Enhanced Marrow Stimulation of Chondral Defects. Stem Cells Int 2017: 1-13.
  17. Awad ME, Hussein KA, Helwa I, Abdelsamid MF, Aguilar-Perez A, et al. (2019) Meta-Analysis and Evidence Base for the Efficacy of Autologous Bone Marrow Mesenchymal Stem Cells in Knee Cartilage Repair: Methodological Guidelines and Quality Assessment. Stem Cells Int 2019: 1-15.
  18. Doyle EC, Wragg NM, Wilson SL (2020) Intraarticular injection of bone marrow-derived mesenchymal stem cells enhances regeneration in knee osteoarthritis. Knee Surg Sport Traumatol Arthrosc.
  19. Hernigou P, Bouthors C, Bastard C, Flouzat Lachaniette CH, Rouard H, et al. (2020) Subchondral bone or intra-articular injection of bone marrow concentrate mesenchymal stem cells in bilateral knee osteoarthritis: What better postpone knee arthroplasty at fifteen years? A randomized study. Int Orthop 2020.
  20. Lu L, Dai C, Zhang Z, Du H, Li S, et al. (2019) Treatment of knee osteoarthritis with intra-articular injection of autologous adipose-derived mesenchymal progenitor cells: A prospective, randomized, double-blind, active-controlled, phase IIb clinical trial. Stem Cell Res Ther 10: 143.
  21. Damia E, Chicharro D, Lopez S, Cuervo B, Rubio M, et al. (2018) Adipose-Derived Mesenchymal Stem Cells: Are They a Good Therapeutic Strategy for Osteoarthritis? Int J Mol Sci 19: 1926.
  22. Bora P, Majumdar AS (2017) Adipose tissue-derived stromal vascular fraction in regenerative medicine: A brief review on biology and translation. Stem Cell Res Ther 8: 145.
  23. Polancec D, Zenic L, Hudetz D, Boric I, Jelec Z, et al. (2019) Immunophenotyping of a Stromal Vascular Fraction from Microfragmented Lipoaspirate Used in Osteoarthritis Cartilage Treatment and Its Lipoaspirate Counterpart. Genes (Basel) 10: 474.
  24. Fodor PB, Paulseth SG (2016) Adipose Derived Stromal Cell (ADSC) Injections for Pain Management of Osteoarthritis in the Human Knee Joint. Aesthetic Surg J 36: 229-236.
  25. Pers Y-M, Rackwitz L, Ferreira R, Pullig O, Delfour C, et al. (2016) Adipose Mesenchymal Stromal Cell-Based Therapy for Severe Osteoarthritis of the Knee: A Phase I Dose-Escalation Trial. Stem Cells Transl Med 5: 847-856.
  26. Hudetz D, Bori? I, Rod E, Jele? Ž, Kunovac B, et al. (2019) Early results of intra-articular micro-fragmented lipoaspirate treatment in patients with late stages knee osteoarthritis: A prospective study. Croat Med J 60: 227-236.
  27. Freitag J, Bates D, Wickham J, Shah K, Huguenin L, et al. (2019) Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: A randomized controlled trial. Regen Med 14: 213-230.
  28. Higuchi J, Yamagami R, Matsumoto T, Terao T, Inoue K, et al. (2020) Associations of clinical outcomes and MRI findings in intra-articular administration of autologous adipose-derived stem cells for knee osteoarthritis. Regen Ther 14: 332-340.
  29. Hudetz D, Bori? I, Rod E, Jele? Ž, Radi? A, et al. (2017) The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tissue on Proteoglycan Synthesis in Patients with Knee Osteoarthritis. Genes (Basel) 8: 270.
  30. Bori? I, Hudetz D, Rod E, Jele? Ž, Vrdoljak T, et al. (2019) A 24-Month Follow-Up Study of the Effect of Intra-Articular Injection of Autologous Microfragmented Fat Tissue on Proteoglycan Synthesis in Patients with Knee Osteoarthritis. Genes (Basel) 10: 1051.
  31. Saeed H, Ahsan M, Saleem Z, Iqtedar M, Islam M, et al. (2016) Mesenchymal stem cells (MSCs) as skeletal therapeutics-an update. J Biomed Sci 23: 41.
  32. Pak J, Lee JH, Kartolo WA, Lee SH (2016) Cartilage Regeneration in Human with Adipose Tissue-Derived Stem Cells: Current Status in Clinical Implications. Biomed Res Int 2016: 4702674.
  33. Taniguchi Y, Yoshioka T, Sugaya H, Gosho M, Aoto K, et al. (2019) Growth factor levels in leukocyte-poor platelet-rich plasma and correlations with donor age, gender, and platelets in the Japanese population. J Exp Orthop 6: 4.
  34. Castellanos R, Tighe S (2019) Injectable Amniotic Membrane/Umbilical Cord Particulate for Knee Osteoarthritis: A Prospective, Single-Center Pilot Study. Pain Med 20: 2283-2291.
  35. Soltani SK, Forogh B, Ahmadbeigi N, Kharazi HH, Fallahzadeh K, et al. (2019) Safety and efficacy of allogenic placental mesenchymal stem cells for treating knee osteoarthritis: a pilot study. Cytotherapy 21: 54-63.
  36. Farr J, Gomoll AH, Yanke AB, Strauss EJ, Mowry KC (2019) A Randomized Controlled Single-Blind Study Demonstrating Superiority of Amniotic Suspension Allograft Injection Over Hyaluronic Acid and Saline Control for Modification of Knee Osteoarthritis Symptoms. J Knee Surg 32: 1143-1154.
  37. Ryu DJ, Jeon YS, Park JS, Bae GC, Kim J, et al. (2020) Comparison of Bone Marrow Aspirate Concentrate and Allogenic Human Umbilical Cord Blood Derived Mesenchymal Stem Cell Implantation on Chondral Defect of Knee: Assessment of Clinical and Magnetic Resonance Imaging Outcomes at 2-Year Follow-Up. Cell Transplant 29: 096368972094358.
  38. McIntyre JA, Jones IA, Danilkovich A, Vangsness CT (2018) The Placenta: Applications in Orthopaedic Sports Medicine. Am J Sports Med 46: 234-247.
  39. Hannon C, Yanke A, Farr J (2019) Amniotic Tissue Modulation of Knee Pain-A Focus on Osteoarthritis. J Knee Surg 32: 26-36.
  40. Li CY, Wu XY, Tong JB, Yang XX, Zhao JL, , et al. (2015) Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther 6: 55.

Citation: Peri? V, Kottek T, Molnar V, Matiši? V, ?ukelj F, et al. (2020) Mesenchymal Stem Cells in the Treatment of Knee Osteoarthritis. J Stem Cell Res Dev Ther 6: 050.

Copyright: © 2020  Vitorio Peric, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!