1
|
Sourugeon Y, Boffa A, Perucca Orfei C, de Girolamo L, Magalon J, Sánchez M, Tischer T, Filardo G, Laver L. Cell-based therapies have disease-modifying effects on osteoarthritis in animal models: A systematic review by the ESSKA Orthobiologic Initiative. Part 3: Umbilical cord, placenta, and other sources for cell-based injectable therapies. Knee Surg Sports Traumatol Arthrosc 2025; 33:1695-1708. [PMID: 39302089 PMCID: PMC12022835 DOI: 10.1002/ksa.12472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE This systematic review aimed to investigate in animal models the presence of disease-modifying effects driven by non-bone marrow-derived and non-adipose-derived products, with a particular focus on umbilical cord and placenta-derived cell-based therapies for the intra-articular injective treatment of osteoarthritis (OA). METHODS A systematic review was performed on three electronic databases (PubMed, Web of Science and Embase) according to PRISMA guidelines. The results were synthesised to investigate disease-modifying effects in preclinical animal studies comparing injectable umbilical cord, placenta, and other sources-derived products with OA controls. The risk of bias was assessed using the SYRCLE tool. RESULTS A total of 80 studies were included (2314 animals). Cell therapies were most commonly obtained from the umbilical cord in 33 studies and placenta/amniotic tissue in 18. Cell products were xenogeneic in 61 studies and allogeneic in the remaining 19 studies. Overall, 25/27 (92.6%) of studies on umbilical cord-derived products documented better results compared to OA controls in at least one of the following outcomes: macroscopic, histological and/or immunohistochemical findings, with 19/22 of studies (83.4%) show positive results at the cartilage level and 4/6 of studies (66.7%) at the synovial level. Placenta-derived injectable products documented positive results in 13/16 (81.3%) of the studies, 12/15 (80.0%) at the cartilage level, and 2/4 (50.0%) at the synovial level, but 2/16 studies (12.5%) found overall worse results than OA controls. Other sources (embryonic, synovial, peripheral blood, dental pulp, cartilage, meniscus and muscle-derived products) were investigated in fewer preclinical studies. The risk of bias was low in 42% of items, unclear in 49%, and high in 9% of items. CONCLUSION Interest in cell-based injectable therapies for OA treatment is soaring, particularly for alternatives to bone marrow and adipose tissue. While expanded umbilical cord mesenchymal stem cells reported auspicious disease-modifying effects in preventing OA progression in animal models, placenta/amniotic tissue also reported deleterious effects on OA joints. Lower evidence has been found for other cellular sources such as embryonic, synovial, peripheral blood, dental-pulp, cartilage, meniscus, and muscle-derived products. LEVEL OF EVIDENCE Level II.
Collapse
Affiliation(s)
- Yosef Sourugeon
- Division of Surgery, Orthopaedics DepartmentChaim Sheba Medical CentreRamat GanIsrael
| | - Angelo Boffa
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'AmbrogioMilanItaly
| | - Jeremy Magalon
- INSERM, NRA, C2VNAix Marseille UniversityMarseilleFrance
- SAS RemedexMarseilleFrance
- Cell Therapy Laboratory, Hôpital De La Conception, AP‐HMMarseilleFrance
| | - Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas VitoriaVitoria‑GasteizSpain
- Arthroscopic Surgery Unit, Hospital Vithas VitoriaVitoria‐GasteizSpain
| | - Thomas Tischer
- Department of Orthopaedic and Trauma SurgeryMalteser WaldkrankenhausErlangenGermany
- Department of Orthopaedic SurgeryUniversity of RostockRostockGermany
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of SurgeryService of Orthopaedics and Traumatology, EOCLuganoSwitzerland
- Faculty of Biomedical SciencesUniversità Della Svizzera ItalianaLuganoSwitzerland
| | - Lior Laver
- Arthrosport ClinicTel‑AvivIsrael
- Rappaport Faculty of Medicine, Technion University Hospital (IsraelInstitute of Technology)HaifaIsrael
- Department of OrthopaedicsHillel Yaffe Medical Center (HYMC)HaderaIsrael
| |
Collapse
|
2
|
Trivedi AH, Wang VZ, McClain EJ, Vyas PS, Swink IR, Snell ED, Cheng BC, DeMeo PJ. The Categorization of Perinatal Derivatives for Orthopedic Applications. Biomedicines 2024; 12:1544. [PMID: 39062117 PMCID: PMC11274709 DOI: 10.3390/biomedicines12071544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Musculoskeletal (MSK) pathology encompasses an array of conditions that can cause anything from mild discomfort to permanent injury. Their prevalence and impact on disability have sparked interest in more effective treatments, particularly within orthopedics. As a result, the human placenta has come into focus within regenerative medicine as a perinatal derivative (PnD). These biologics are sourced from components of the placenta, each possessing a unique composition of collagens, proteins, and factors believed to aid in healing and regeneration. This review aims to explore the current literature on PnD biologics and their potential benefits for treating various MSK pathologies. We delve into different types of PnDs and their healing effects on muscles, tendons, bones, cartilage, ligaments, and nerves. Our discussions highlight the crucial role of immune modulation in the healing process for each condition. PnDs have been observed to influence the balance between anti- and pro-inflammatory factors and, in some cases, act as biologic scaffolds for tissue growth. Additionally, we assess the range of PnDs available, while also addressing gaps in our understanding, particularly regarding biologic processing methods. Although certain PnD biologics have varying levels of support in orthopedic literature, further clinical investigations are necessary to fully evaluate their impact on human patients.
Collapse
Affiliation(s)
- Amol H. Trivedi
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
- Drexel University College of Medicine, Drexel University, University City Campus, Philadelphia, PA 19104, USA
| | - Vicki Z. Wang
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward J. McClain
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Praveer S. Vyas
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Isaac R. Swink
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward D. Snell
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Boyle C. Cheng
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Patrick J. DeMeo
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| |
Collapse
|
3
|
Ehioghae M, Vippa TK, Askins D, Slusarczyk S, Bobo E, Montoya A, Anderson D, Robinson CL, Kaye AD, Urits I. Exploring Orthopedic Stem-Cell Approaches for Osteoarthritis Management: Current Trends and Future Horizons. Curr Pain Headache Rep 2024; 28:27-35. [PMID: 38010488 DOI: 10.1007/s11916-023-01191-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a prevalent and debilitating condition characterized by joint degeneration and pain. Current treatment options aim to alleviate symptoms and slow disease progression but lack curative potential. Stem cell therapies have emerged as a promising alternative. This article explores the epidemiology, pathophysiology, clinical manifestations of hip and knee OA, and the evolving role of stem cell therapies in their treatment. RECENT FINDINGS The global prevalence of OA, with knee OA being the most common form, has fueled the demand for stem cell therapies. Despite limited robust evidence supporting their efficacy, clinical trials investigating stem-cell treatments for OA have reported encouraging radiological and clinical improvements. Stem cell therapies offer potential disease-modifying benefits through immunomodulatory actions, growth factor secretion, and chondrogenic capabilities. Adipose-derived mesenchymal stem cells (ADMSCs) have shown promise in clinical trials for OA treatment, offering potential pain relief and functional improvement. ADMSCs possess advantages such as accessibility and a favorable safety profile, making them a viable option for OA management. Although other stem-cell types, including human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), have been used in OA treatment, ADMSCs have demonstrated superior outcomes. By providing a comprehensive overview of the evolving landscape of stem cell therapies for hip and knee OA, this article highlights the potential of stem-cell treatments to address the limitations of current therapies. However, further research is required to establish their long-term efficacy, identify optimal stem-cell types, and develop standardized protocols.
Collapse
Affiliation(s)
| | | | | | | | - Elena Bobo
- Tulane University School of Medicine, New Orleans, USA
| | - Alexis Montoya
- University of Arizona College of Medicine-Phoenix, Phoenix, USA
| | | | | | - Alan D Kaye
- Louisiana State University Health Shreveport, Shreveport, USA
| | | |
Collapse
|
4
|
Abou-Shanab AM, Gaser OA, Salah RA, El-Badri N. Application of the Human Amniotic Membrane as an Adjuvant Therapy for the Treatment of Hepatocellular Carcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:129-146. [PMID: 38036871 DOI: 10.1007/5584_2023_792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related morbidity and mortality worldwide. Current therapeutic approaches suffer significant side effects and lack of clear understanding of their molecular targets. Recent studies reported the anticancer effects, immunomodulatory properties, and antiangiogenic effects of the human amniotic membrane (hAM). hAM is a transparent protective membrane that surrounds the fetus. Preclinical studies showed pro-apoptotic and antiproliferative properties of hAM treatment on cancer cells. Herein, we present the latest findings of the application of the hAM in combating HCC tumorigenesis and the underlying molecular pathogenies and the role of transforming growth factor-beta (TGFβ), P53, WNT/beta-catenin, and PI3K/AKT pathways. The emerging clinical applications of hAM in cancer therapy provide evidence for its diverse and unique features and suitability for the management of a wide range of pathological conditions.
Collapse
Affiliation(s)
- Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Ola A Gaser
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt.
| |
Collapse
|
5
|
Huang C, Zhao Y, Lin S, Li L, Guo X, Yumiseba S, Yang JD, Hariri R, Ye Q, He S, Kilcoyne A. Characterization of human placenta-derived exosome (pExo) as a potential osteoarthritis disease modifying therapeutic. Arthritis Res Ther 2023; 25:229. [PMID: 38017556 PMCID: PMC10683254 DOI: 10.1186/s13075-023-03219-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE Human placenta-derived exosomes (pExo) were generated, characterized, and evaluated as a therapeutic candidate for the treatment of osteoarthritis (OA). METHODS pExo was generated from full-term human placenta tissues by sequential centrifugation, purification, and sterile filtration. Upon analysis of particle size, cytokine composition, and exosome marker expression, pExo was further tested in cell-based assays to examine its effects on human chondrocytes. In vivo therapeutic efficacies were evaluated in a medial meniscal tear/medial collateral ligament tear (MCLT + MMT) rat model, in which animals received pExo injections intraarticularly and weight bearing tests during in-life stage while histopathology and immunohistochemistry were performed as terminal endpoints. RESULTS pExo displayed typical particle size, expressed maker proteins of exosome, and contained proteins with pro-proliferative, pro-anabolic, anti-catabolic, or anti-inflammatory activities. In vitro, pExo promoted chondrocyte migration and proliferation dose-dependently, which may involve its activation of cell growth-related signaling pathways. Expression of inflammatory and catabolic genes induced in a cellular OA model was significantly suppressed by pExo. In the rat OA model, pExo alleviated pain burden, restored cartilage degeneration, and downregulated expressions of pro-inflammatory, catabolic, or apoptotic proteins in a dose-dependent manner. CONCLUSIONS Our study demonstrates that pExo has multiple potential therapeutic effects including symptom control and disease modifying characteristics. This may make it an attractive candidate for further development as an anti-OA therapeutic.
Collapse
Affiliation(s)
- Chenfei Huang
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Yuechao Zhao
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA.
| | - Shengchen Lin
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Lin Li
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Xuan Guo
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | | | - Jeng-Dar Yang
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Robert Hariri
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Qian Ye
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Shuyang He
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Adrian Kilcoyne
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| |
Collapse
|
6
|
Lin ASP, Reece DS, Thote T, Sridaran S, Stevens HY, Willett NJ, Guldberg RE. Intra-articular delivery of micronized dehydrated human amnion/chorion membrane reduces degenerative changes after onset of post-traumatic osteoarthritis. Front Bioeng Biotechnol 2023; 11:1224141. [PMID: 37744252 PMCID: PMC10512062 DOI: 10.3389/fbioe.2023.1224141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Micronized dehydrated human amnion/chorion membrane (mdHACM) has reduced short term post-traumatic osteoarthritis (PTOA) progression in rats when delivered 24 h after medial meniscal transection (MMT) and is being investigated for clinical use as a disease modifying therapy. Much remains to be assessed, including its potential for longer-term therapeutic benefit and treatment effects after onset of joint degeneration. Objectives: Characterize longer-term effects of acute treatment with mdHACM and determine whether treatment administered to joints with established PTOA could slow or reverse degeneration. Hypotheses: Acute treatment effects will be sustained for 6 weeks, and delivery of mdHACM after onset of joint degeneration will attenuate structural osteoarthritic changes. Methods: Rats underwent MMT or sham surgery (left leg). mdHACM was delivered intra-articularly 24 h or 3 weeks post-surgery (n = 5-7 per group). Six weeks post-surgery, animals were euthanized and left tibiae scanned using equilibrium partitioning of an ionic contrast agent microcomputed tomography (EPIC-µCT) to structurally quantify joint degeneration. Histology was performed to examine tibial plateau cartilage. Results: Quantitative 3D µCT showed that cartilage structural metrics (thickness, X-ray attenuation, surface roughness, exposed bone area) for delayed mdHACM treatment limbs were significantly improved over saline treatment and not significantly different from shams. Subchondral bone mineral density and thickness for the delayed treatment group were significantly improved over acute treated, and subchondral bone thickness was not significantly different from sham. Marginal osteophyte degenerative changes were decreased with delayed mdHACM treatment compared to saline. Acute treatment (24 h post-surgery) did not reduce longer-term joint tissue degeneration compared to saline. Histology supported µCT findings and further revealed that while delayed treatment reduced cartilage damage, chondrocytes displayed qualitatively different morphologies and density compared to sham. Conclusion: This study provides insight into effects of intra-articular delivery timing relative to PTOA progression and the duration of therapeutic benefit of mdHACM. Results suggest that mdHACM injection into already osteoarthritic joints can improve joint health, but a single, acute mdHACM injection post-injury does not prevent long term osteoarthritis associated with meniscal instability. Further work is needed to fully characterize the durability of therapeutic benefit in stable osteoarthritic joints and the effects of repeated injections.
Collapse
Affiliation(s)
- Angela S. P. Lin
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States
| | - David S. Reece
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Tanushree Thote
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Sanjay Sridaran
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Hazel Y. Stevens
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Nick J. Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States
| | - Robert E. Guldberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States
| |
Collapse
|
7
|
Hofmann N, Rennekampff HO, Salz AK, Börgel M. Preparation of human amniotic membrane for transplantation in different application areas. FRONTIERS IN TRANSPLANTATION 2023; 2:1152068. [PMID: 38993896 PMCID: PMC11235369 DOI: 10.3389/frtra.2023.1152068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/20/2023] [Indexed: 07/13/2024]
Abstract
The human amniotic membrane (hAM) is the inner layer of the placenta and plays protective and nutritional roles for the fetus during pregnancy. It contains multiple growth factors and proteins that mediate unique regenerative properties and enhance wound healing in tissue regeneration. Due to these characteristics hAM has been successfully utilized in ophthalmology for many decades. This material has also found application in a variety of additional therapeutic areas. Particularly noteworthy are the extraordinary effects in the healing of chronic wounds and in the treatment of burns. But hAM has also been used successfully in gynecology, oral medicine, and plastic surgery and as a scaffold for in vitro cell culture approaches. This review aims to summarize the different graft preparation, preservation and storage techniques that are used and to present advantages and disadvantages of these methods. It shows the characteristics of the hAM according to the processing and storage methods used. The paper provides an overview of the currently mainly used application areas and raises new application possibilities. In addition, further preparation types like extracts, homogenates, and the resulting treatment alternatives are described.
Collapse
Affiliation(s)
- Nicola Hofmann
- German Society for Tissue Transplantation (DGFG) gGmbH, Hannover, Germany
| | - Hans-Oliver Rennekampff
- Klinik für Plastische Chirurgie, Hand- und Verbrennungschirurgie, Rhein-Maas Klinikum GmbH, Würselen, Germany
| | | | - Martin Börgel
- German Society for Tissue Transplantation (DGFG) gGmbH, Hannover, Germany
| |
Collapse
|
8
|
Thada RR, Debata M, Mandal S, Gunasekaran D, Mohan VD, Chandrasekaran N, Sivagnanam UT. In vitro and Ex vivo characterization of nanonized amniotic membrane particles: An untapped modality for ocular surface reconstruction. Exp Eye Res 2023; 231:109471. [PMID: 37086963 DOI: 10.1016/j.exer.2023.109471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/21/2023] [Accepted: 04/08/2023] [Indexed: 04/24/2023]
Abstract
The pristine Human Amniotic Membrane (HAM) has portrayed outstanding potential as scaffold for ocular surface reconstruction and regeneration. However, in treatment procedures where the supporting membrane matrix of HAM is not obligatory and only the bioactive molecules are vital, the surgical practise of HAM grafting causes redundant trauma and economic burden to the patient. Hence, in our laboratory we have attempted to break down HAM to nanoscale particles and validate its potential as a competent ocular therapeutic agent; by conducting a comparative analysis between the fresh, lyophilized, micronized and Nanonized Amniotic Membrane (NAM) particles. Our results evidently showcased that the prepared NAM particles was <100 nm and the major biomolecules such as collagen and hyaluronic acid were well retained. Further, the NAM particles eluted significantly higher amounts of proteins and growth factors while maintaining its stability and isotonicity when stored at 4 °C. Its biostability was assayed in the presence of lysozyme enzyme. Its remarkable ability to promote cell proliferation in rabbit corneal cells and negative cytotoxicity is an added advantage for ocular application. The ocular biocompatibility of NAM, evaluated by the ex vivo assessment of corneal thickness, transparency, histopathology, immunohistochemistry and corneal permeability clearly indicated its suitability for ophthalmic applications.
Collapse
Affiliation(s)
- Raja Rajeshwari Thada
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India; Department of Leather Technology, (Housed at CSIR-Central Leather Research Institute), Alagappa College of Technology, Anna University, Chennai, India.
| | - Mayadhar Debata
- Advanced Materials Technology Department, CSIR-Institute of Minerals and Materials Technology, Bhubaneswar, Odisha, India.
| | - Shuvam Mandal
- Advanced Materials Technology Department, CSIR-Institute of Minerals and Materials Technology, Bhubaneswar, Odisha, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India.
| | - Deebasuganya Gunasekaran
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India; Department of Leather Technology, (Housed at CSIR-Central Leather Research Institute), Alagappa College of Technology, Anna University, Chennai, India.
| | - Vimala Devi Mohan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India; Department of Leather Technology, (Housed at CSIR-Central Leather Research Institute), Alagappa College of Technology, Anna University, Chennai, India.
| | | | | |
Collapse
|
9
|
Jahanafrooz Z, Bakhshandeh B, Behnam Abdollahi S, Seyedjafari E. Human amniotic membrane as a multifunctional biomaterial: recent advances and applications. J Biomater Appl 2023; 37:1341-1354. [PMID: 36331116 DOI: 10.1177/08853282221137609] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The developing fetus is wrapped by a human amniotic membrane or amnion. Amnion is a promising human tissue allograft in clinical application because of its chemical composition, collagen-based, and mechanical properties of the extracellular matrix. In addition, amnion contains cells and growth factors; therefore, meets the essential parameters of tissue engineering. No donor morbidity, easy processing and storage, fewer ethical issue, anti-inflammatory, antioxidant, antibacterial, and non-immunogenic properties are other advantages of amnion usage. For these reasons, amnion can resolve some bottlenecks in the regenerative medicine issues such as tissue engineering and cell therapy. Over the last decades, biomedical applications of amnion have evolved from a simple sheet for skin or cornea repair to high-technology applications such as amnion nanocomposite, powder, or hydrogel for the regeneration of cartilage, muscle, tendon, and heart. Furthermore, amnion has anticancer as well as drug/cell delivery capacity. This review highlights various ancient and new applications of amnion in research and clinical applications, from regenerative medicine to cancer therapy, focusing on articles published during the last decade that also revealed information regarding amnion-based products. Challenges and future perspectives of the amnion in regenerative medicine are also discussed.
Collapse
|
10
|
Aratikatla A, Maffulli N, Rodriguez HC, Gupta M, Potty AG, Gupta A. Allogenic Perinatal Tissue for Musculoskeletal Regenerative Medicine Applications: A Systematic Review. Biomedicines 2022; 10:3173. [PMID: 36551929 PMCID: PMC9775213 DOI: 10.3390/biomedicines10123173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/26/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Producing tremendous amounts of stress and financial burden on the global patient population and healthcare systems around the world, most current modalities of treatment for musculoskeletal ailments often do not address the etiopathogenetic causes of these disorders. Regenerative medicine for musculoskeletal disorders relies on orthobiologics derived from either allogenic or autologous sources. Multiple drawbacks are associated with autologous sources, including donor-site morbidity, a dearth of studies, and variability in both patient reported and clinical/functional outcomes. On the other hand, allogenic sources address several of these concerns, and continue to be a suitable source of mesenchymal stem cells (MSCs). This review qualitatively reports both the preclinical and clinical outcomes of publications studying the applications of umbilical cord (-derived Wharton's jelly), amniotic suspension allograft, amniotic membrane, and amniotic fluid in musculoskeletal medicine. A systematic review was conducted utilizing the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines on studies published between January 2010 and October 2022 that used allogeneic perinatal tissues. Further randomized controlled clinical studies are necessary to properly evaluate the safety and efficacy of these tissues in orthopedic surgery.
Collapse
Affiliation(s)
- Adarsh Aratikatla
- The Royal College of Surgeons in Ireland, D02 YN77 Dublin 2, Ireland
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, 84084 Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Ortopedica” Department, Hospital of Salerno, 84124 Salerno, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent ST5 5BG, UK
| | - Hugo C. Rodriguez
- Department of Orthopaedic Surgery, Larkin Community Hospital, South Miami, FL 33143, USA
| | - Manu Gupta
- Polar Aesthetics Dental & Cosmetic Centre, Noida 201301, UP, India
| | - Anish G. Potty
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
| | - Ashim Gupta
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
- Future Biologics, Lawrenceville, GA 30043, USA
- BioIntegrate, Lawrenceville, GA 30043, USA
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, UP, India
- Regenerative Orthopaedics, Noida 201301, UP, India
| |
Collapse
|
11
|
Sawvell E, Wright N, Ode G, Mercuri J. Perinatal Tissue-Derived Allografts and Stromal Cells for the Treatment of Knee Osteoarthritis: A Review of Preclinical and Clinical Evidence. Cartilage 2022; 13:184-199. [PMID: 36398763 PMCID: PMC9924983 DOI: 10.1177/19476035221137725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The use of perinatal-derived tissues and mesenchymal stromal cells (MSCs) as alternative treatment options to corticosteroid and hyaluronic acid injections has been gaining popularity. However, their ability to attenuate osteoarthritic (OA) symptoms while also slowing the progression of the disease remains controversial. Thus, the objective of this article is to summarize the results from both preclinical and clinical studies evaluating the efficacy of perinatal-derived tissue allografts and MSCs for the treatment of OA. DESIGN A comprehensive literature search was conducted on databases including Pubmed, ScienceDirect, and Google Scholar beginning in March 2020 for both preclinical and clinical studies evaluating perinatal-derived tissues and MSCs in OA. Eighteen studies met the inclusion criteria and were used for this review. RESULTS Both animal models and early human clinical trials demonstrated that perinatal tissues could reduce joint inflammation and pain as well as improve range of motion and function in OA. Perinatal tissue-derived MSCs in animal studies have shown the potential to support chondrocyte proliferation while also decreasing inflammatory gene and protein expression. Limited clinical results suggest perinatal tissue-derived MSC sources may also be a viable alternative or adjunct to hyaluronic acid in reducing pain and symptoms in an arthritic joint. CONCLUSIONS Perinatal tissue-derived allografts and MSCs have promise as potential therapeutics for mitigating OA progression. However, further research is warranted to fully define the therapeutic mechanism(s) of action and safety of these biological therapies.
Collapse
Affiliation(s)
- Emily Sawvell
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA
| | - Noah Wright
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA
| | - Gabriella Ode
- Department of Orthopaedic Surgery,
Prisma Health–Upstate, Greenville, SC, USA
| | - Jeremy Mercuri
- Laboratory of Orthopaedic Tissue
Regeneration & Orthobiologics, Department of Bioengineering, Clemson University,
Clemson, SC, USA,Frank H. Stelling and C. Dayton Riddle
Orthopaedic Education and Research Laboratory, Clemson University Biomedical
Engineering Innovation Campus, Greenville, SC, USA,Jeremy Mercuri, Laboratory of Orthopaedic
Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson
University, 401-5 Rhodes Engineering Research Center, Clemson, SC 29634, USA.
| |
Collapse
|
12
|
Wang Y, Liu L, Le Z, Tay A. Analysis of Nanomedicine Efficacy for Osteoarthritis. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Ling Liu
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
- Tissue Engineering Programme National University of Singapore Singapore 117510 Singapore
| |
Collapse
|
13
|
Gupta A. Amniotic Suspension Allograft for Treatment of Knee Osteoarthritis. Biomedicines 2022; 10:2658. [PMID: 36289919 PMCID: PMC9599686 DOI: 10.3390/biomedicines10102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/21/2022] Open
Abstract
Osteoarthritis (OA) is an immensely pervasive joint disorder-typically concerning large weight-bearing joints-affecting over 30 million people in the United States, with this number predicted to reach 67 million by 2030 [...].
Collapse
Affiliation(s)
- Ashim Gupta
- Regenerative Orthopaedics, Noida 201301, UP, India;
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, UP, India
- Future Biologics, Noida 201301, UP, India
- BioIntegrate, Lawrenceville, GA 30043, USA
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
| |
Collapse
|
14
|
Gupta A. Allogenic Amniotic Tissue for Treatment of Knee and Hip Osteoarthritis. Pharmaceuticals (Basel) 2022; 15:404. [PMID: 35455401 PMCID: PMC9031613 DOI: 10.3390/ph15040404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) impacts millions of people and places a high burden on healthcare systems in the United States. Current treatment modalities have limitations and do not address underlying pathology. Lately, there has been an immense growth in the use of biologics, including perinatal allogenic tissues for orthopedic regenerative medicine applications. Amniotic tissue is an exciting new alternative for such applications. Despite several published studies that reported its use for treatment of ophthalmic conditions and complex wounds, there are limited clinical studies evaluating its safety and efficacy in treating patients suffering with knee or hip OA. In this manuscript, I focused on three prospective clinical studies which evaluated the safety and efficacy of amniotic tissue in patients suffering with moderate knee or hip OA. The results from these studies presented the scientific community with much needed, well-executed, and prospective clinical trials. Though these trials demonstrated that administration of amniotic tissue in knee or hip joint is safe and potentially effective, more multi-center, prospective, double-blinded, randomized controlled trials are warranted to further establish the efficacy of amniotic tissue to mitigate symptoms of knee and hip OA to ultimately justify its clinical use.
Collapse
Affiliation(s)
- Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA;
- BioIntegrate, Lawrenceville, GA 30043, USA
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
- Veterans in Pain (V.I.P.), Valencia, CA 91354, USA
- General Therapeutics, Cleveland Heights, OH 44118, USA
| |
Collapse
|
15
|
Meadows MC, Elisman K, Nho SJ, Mowry K, Safran MR. A Single Injection of Amniotic Suspension Allograft Is Safe and Effective for Treatment of Mild to Moderate Hip Osteoarthritis: A Prospective Study. Arthroscopy 2022; 38:325-331. [PMID: 33940131 DOI: 10.1016/j.arthro.2021.04.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of our study was to examine the effects of a commercially available amniotic suspension allograft (ASA) (ReNu, Organogenesis, Canton, MA) in a patient population with moderate osteoarthritis of the hip. METHODS Ten patients with symptomatic hip osteoarthritis, defined as Tonnis grade 1 or 2 on radiographic examination, were prospectively enrolled. Each patient received a single image-guided injection of ASA into the hip joint. Patient-reported outcomes measures, including the 12-item International Hip Outcome Tool, Modified Harris Hip Score, and Single Assessment Numeric Evaluation scores were recorded at baseline, 6 months, and 12 months postinjection. A linear regression model was performed to detect differences in outcome scores from baseline. RESULTS Nine patients had complete 12-month data available for analysis. One patient failed treatment and underwent arthroplasty at 2 months postinjection. The cohort includes 5 males and 4 females, aged 47-67. International Hip Outcome Tool scores demonstrated a significant improvement between baseline and 12 months (P = .02). Single Assessment Numeric Evaluation scores demonstrated a significant difference between baseline and 6 months (P < .01), as well as between baseline and 12 months (P < .01). Modified Harris Hip Scores demonstrated a significant difference between baseline and 6 months (P = .02) and between baseline and 12 months (P = .01). There were no major adverse events in the course of the study period. CONCLUSION This study demonstrates promising results for relief of pain and improvement in patient-reported outcomes with intra-articular ASA in patients with moderate osteoarthritis of the hip for up to one year, although the exact mechanism of action remains unknown. LEVEL OF EVIDENCE: IV, case series.
Collapse
Affiliation(s)
- Molly C Meadows
- Stanford University Department of Orthopaedic Surgery, Redwood City, California, U.S.A..
| | - Katia Elisman
- Stanford University Department of Orthopaedic Surgery, Redwood City, California, U.S.A
| | - Shane J Nho
- Midwest Orthopedics at Rush, Chicago, Illinois, U.S.A
| | | | - Marc R Safran
- Stanford University Department of Orthopaedic Surgery, Redwood City, California, U.S.A
| |
Collapse
|
16
|
Elkhenany H, El-Derby A, Abd Elkodous M, Salah RA, Lotfy A, El-Badri N. Applications of the amniotic membrane in tissue engineering and regeneration: the hundred-year challenge. Stem Cell Res Ther 2022; 13:8. [PMID: 35012669 PMCID: PMC8744057 DOI: 10.1186/s13287-021-02684-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
The amniotic membrane (Amnio-M) has various applications in regenerative medicine. It acts as a highly biocompatible natural scaffold and as a source of several types of stem cells and potent growth factors. It also serves as an effective nano-reservoir for drug delivery, thanks to its high entrapment properties. Over the past century, the use of the Amnio-M in the clinic has evolved from a simple sheet for topical applications for skin and corneal repair into more advanced forms, such as micronized dehydrated membrane, amniotic cytokine extract, and solubilized powder injections to regenerate muscles, cartilage, and tendons. This review highlights the development of the Amnio-M over the years and the implication of new and emerging nanotechnology to support expanding its use for tissue engineering and clinical applications.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, 12582, Giza, Egypt
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Azza El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, 12582, Giza, Egypt
| | - Mohamed Abd Elkodous
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, 12582, Giza, Egypt
| | - Radwa A Salah
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, 12582, Giza, Egypt
| | - Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, 12582, Giza, Egypt.
| |
Collapse
|
17
|
Anderson LE, Pearson JJ, Brimeyer AL, Temenoff JS. Injection of Micronized Human Amnion/Chorion Membrane Results in Increased Early Supraspinatus Muscle Regeneration in a Chronic Model of Rotator Cuff Tear. Ann Biomed Eng 2021; 49:3698-3710. [PMID: 34766224 DOI: 10.1007/s10439-021-02880-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 10/18/2021] [Indexed: 01/08/2023]
Abstract
Surgical repair of severe rotator cuff tear often results in retear due to unaddressed muscle degeneration. The objective of this study was to test the regenerative potential of micronized dehydrated Human Amnion/Chorion Membrane (dHACM), in a clinically relevant delayed reattachment model of rotator cuff repair. Micronized dHACM was injected into rat supraspinatus muscle during tendon re-attachment surgery, three weeks after original tendon injury. One week after material injection, inflammatory and mesenchymal stem cell infiltration into supraspinatus muscles was assessed via flow cytometry. Histological methods were utilized to assess structural and regenerative changes in muscle one and three weeks after material injection. Micronized dHACM injection resulted in increased M1-like macrophages (17.1 [Formula: see text] fold change over contralateral controls) and regenerating muscle fibers (4.3% vs 1.7% in saline treated muscles) one week after injection compared to saline treated muscles. Tendon reattachment itself exhibited intrinsic healing in this model, demonstrated by a general return of muscle weight and reduced fibrosis. Our results indicate that injection of micronized dHACM may initiate an inflammatory response in degenerated muscle that promotes early muscle regeneration, and that our animal model may be a suitable platform for studying treatments in muscle at early timepoints, before intrinsic healing occurs.
Collapse
Affiliation(s)
- Leah E Anderson
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, 315 Ferst Dr., Atlanta, GA, 30332, USA
| | - Joseph J Pearson
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, 315 Ferst Dr., Atlanta, GA, 30332, USA
| | - Alexandra L Brimeyer
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, 315 Ferst Dr., Atlanta, GA, 30332, USA
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, 315 Ferst Dr., Atlanta, GA, 30332, USA.
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
18
|
A novel placental tissue biologic, PTP-001, inhibits inflammatory and catabolic responses in vitro and prevents pain and cartilage degeneration in a rat model of osteoarthritis. Osteoarthritis Cartilage 2021; 29:1203-1212. [PMID: 34023528 DOI: 10.1016/j.joca.2021.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Characterization of a novel human placental tissue-derived biologic, PTP-001, which is in development as a candidate therapeutic for the treatment of osteoarthritis symptoms and pathophysiology. METHODS Human placental tissues from healthy donors were prepared as a particulate formulation, PTP-001. PTP-001 extracts were assayed for the presence of disease-relevant biofactors which could have beneficial effects in treating osteoarthritis. PTP-001 eluates were tested in human chondrocyte cultures to determine effects on the production of a key collagen-degrading matrix metalloproteinase, MMP-13. PTP-001 eluates were also assessed for anti-inflammatory potential in human monocyte/macrophage cultures, as well as for growth-stimulating anabolic effects in human synoviocytes. The in vivo effects of PTP-001 on joint pain and histopathology were evaluated in a rat model of osteoarthritis induced surgically by destabilization of the medial meniscus. RESULTS PTP-001 was found to contain an array of beneficial growth factors, cytokines and anti-inflammatory molecules. PTP-001 eluates dose-dependently inhibited the production of chondrocyte MMP-13, and the secretion of proinflammatory cytokines from monocyte/macrophage cultures. PTP-001 eluates also promoted proliferation of cultured synovial cells. In a rat osteoarthritis model, PTP-001 significantly reduced pain responses throughout 6 weeks post-dosing. The magnitude and duration of pain reduction following a single intraarticular treatment with PTP-001 was comparable to that observed for animals treated with a corticosteroid (active control). For rats dosed twice with PTP-001, significant reductions in cartilage histopathology scores were observed. CONCLUSIONS PTP-001 represents a promising biologic treatment for osteoarthritis, with a multi-modal mechanism of action that may contribute to symptom management and disease modification.
Collapse
|
19
|
The effect of Scrophularia striata on cell attachment and biocompatibility of decellularized bovine pericardia. Cell Tissue Bank 2021; 23:261-269. [PMID: 34173897 DOI: 10.1007/s10561-021-09939-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
Since using tissue transplantation has faced limitations all over the world, regenerative medicine has introduced decellularized tissues as natural scaffolds and researchers are trying to improve their efficiency and function. In this study, to increase cell attachment and ultimately cell proliferation on decellularized bovine pericardia, scrophularia striata extract was used. Scrophularia striata is an Iranian traditional medicinal plant. For this aim after decellularization of bovine pericardium and analysis of its morphology, it was incubated in scrophularia striata solution. Next, isolated human adipose-derived mesenchymal stem cells were cultured on the tissue. Finally, MTT assay, nitric oxide assay, and scanning electron microscopy observation were performed. MTT showed an increase in cell survival after treating the tissue with the plant extract after 48 h in a dose dependent manner significantly. The survival of cells in 0.5%, 2.5%, and 5% groups was about 5, 10 and 15 folds higher in comparison to control groups, respectively. Additionally, nitric oxide secretion in 2.5% and 5% samples was three and five folds higher than that in control group, respectively. Moreover, SEM observation indicated an impressive and dose-dependent effect of using Scrophularia striata on tissue biocompatibility. The results of this study showed that using Scrophularia striata increased cell viability and cell attachment on decellularized pericardia which could pave the way for the use of natural extracts of medicinal plants to reduce unwanted effects and make desired changes in decellularized tissues.
Collapse
|