1
|
van Rest KLC, Jeffrey ST, Kaestner L, Gudde A, Oosthuysen A, Roovers JWR, Guler Z. Evaluation of Electrospun Poly-4-Hydroxybutyrate as Biofunctional and Degradable Scaffold for Pelvic Organ Prolapse in a Vaginal Sheep Model. Macromol Biosci 2025; 25:e2400412. [PMID: 40008865 PMCID: PMC11995834 DOI: 10.1002/mabi.202400412] [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: 08/28/2024] [Revised: 12/15/2024] [Indexed: 02/27/2025]
Abstract
Pelvic organ prolapse (POP) affects many women, especially after menopause. POP occurs due to the descent of weakened supportive tissue. Current prolapse surgeries have high failure rates, due to disturbed wound healing caused by lower tissue regeneration and estrogen depletion. Absorbable poly-4-hydroxybutyrate (P4HB) knit implants exhibited improved cell and tissue response leading to less complications from prolapse surgery. This study aims to enhance wound healing and improve surgical outcomes by using an electrospun (ES) P4HB scaffold (ES P4HB) that emulates natural tissue structure. Further 17β-estradiol (E2)-a prominent wound healing factor-is incorporated into the scaffold (ES P4HB-E2). Parous Dohne Merino sheep underwent posterior vaginal wall implantation of either P4HB (n = 6) or 17β-estradiol relasing P4HB-E2 (n = 6) scaffolds, or underwent native tissue repair (NTR) (n = 4). Vaginal explants were compared for short-term host response in terms of gross necropsy, histomorphology, biomechanics, tissue-integration, and degradation of P4HB at 3-months post-implantation. Both scaffolds show promising results with enhanced mechanical properties and increased macrophage infiltration compared to NTR, but without differences between scaffolds. Thus, it seems electrospun P4HB scaffolds already improve tissue integration and healing. Further long-term studies are needed before these scaffolds can be used in clinical practice.
Collapse
Affiliation(s)
- Krista L. C. van Rest
- Amsterdam UMC Location University of AmsterdamDepartment of Obstetrics and GynecologyAmsterdam1105 AZThe Netherlands
- Amsterdam Reproduction and Development Research InstituteAmsterdam1105 AZThe Netherlands
| | - Stephen T. Jeffrey
- Department of Obstetrics and GynecologyGroote Schuur HospitalUniversity of Cape TownCape Town7925South Africa
| | - Lisa Kaestner
- Department of UrologyGroote Schuur HospitalUniversity of Cape TownCape Town7925South Africa
| | - Aksel Gudde
- Amsterdam UMC Location University of AmsterdamDepartment of Obstetrics and GynecologyAmsterdam1105 AZThe Netherlands
- Amsterdam Reproduction and Development Research InstituteAmsterdam1105 AZThe Netherlands
| | - Anel Oosthuysen
- Cardiovascular Research UnitDivision of Cardiothoracic SurgeryUniversity of Cape TownCape Town7925South Africa
| | - Jan‐Paul W. R. Roovers
- Amsterdam UMC Location University of AmsterdamDepartment of Obstetrics and GynecologyAmsterdam1105 AZThe Netherlands
- Amsterdam Reproduction and Development Research InstituteAmsterdam1105 AZThe Netherlands
| | - Zeliha Guler
- Amsterdam UMC Location University of AmsterdamDepartment of Obstetrics and GynecologyAmsterdam1105 AZThe Netherlands
- Amsterdam Reproduction and Development Research InstituteAmsterdam1105 AZThe Netherlands
| |
Collapse
|
2
|
Jingyue X, Zhiwei Z, Jirui W, Fei F, Jiang W, Yali M. Application and research progress in composite stem cell materials of pelvic floor reconstruction. Eur J Obstet Gynecol Reprod Biol 2025; 307:49-54. [PMID: 39883984 DOI: 10.1016/j.ejogrb.2025.01.018] [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] [Received: 01/19/2024] [Revised: 10/05/2024] [Accepted: 01/08/2025] [Indexed: 02/01/2025]
Abstract
As an important tool for the surgical treatment of pelvic floor dysfunction, the safety of mesh must be guaranteed. Although the short-term curative effect of most synthetic mesh is satisfactory, complications often occur due to its material. Planting stem cells on mesh through specific methods may resolve the problems of mesh with poor biocompatibility or an uncontrollable rate o degradation. Based on recent research, this paper summarizes the research progress of stem cells composited with common mesh materials, such as polypropylene, polylactic acid and acellular matrix.
Collapse
Affiliation(s)
- Xiao Jingyue
- Department of Obstetrics and Gynaecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China; West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhao Zhiwei
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| | - Wen Jirui
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| | - Fang Fei
- Deep Underground Space Medical Centre, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wu Jiang
- Deep Underground Space Medical Centre, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Miao Yali
- Department of Obstetrics and Gynaecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
Paul K, Darzi S, O'Connell CD, Hennes DMZB, Rosamilia A, Gargett CE, Werkmeister JA, Mukherjee S. 3D Printed Mesh Geometry Modulates Immune Response and Interface Biology in Mouse and Sheep Model: Implications for Pelvic Floor Surgery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405004. [PMID: 39297316 PMCID: PMC11923936 DOI: 10.1002/advs.202405004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/13/2024] [Indexed: 03/21/2025]
Abstract
Pelvic organ prolapse (POP) is a highly prevalent yet neglected health burden for women. Strengthening the pelvic floor with bioactive tissue-engineered meshes is an emerging concept. This study investigates tissue regenerative design parameters, including degradability, porosity, and angulation, to develop alternative degradable melt electrowritten (MEW) constructs for surgical applications of POP. MEW constructs were fabricated in hierarchical geometries by two-way stacking of the fibers with three different inter layer angles of 90°, 45°, or 22.5°. Implants printed at 22.5° have higher tensile strength under dry conditions and show better vaginal fibroblast (VF) attachment in vitro. In vivo assessment using preclinical mouse and ovine models demonstrates more effective degradation and improved tissue integration in 22.5° angular meshes compared to 90° and 45° meshes, with evidence of neo-collagen deposition within implants at 6 weeks. The pattern and geometry of the layered MEW implants also influence the foreign body response, wherein the anti-inflammatory phenotype shows a greater ratio of anti-inflammatory CD206+ M2 macrophages/pro-inflammatory CCR7+ M1 macrophages. This presents an attractive strategy for improving the design and fabrication of next-generation vaginal implants for pelvic reconstructive surgery.
Collapse
Affiliation(s)
- Kallyanashis Paul
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Saeedeh Darzi
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Cathal D. O'Connell
- Biofab3D@ACMDSt Vincent's HospitalMelbourneVIC3065Australia
- Discipline of Electrical and Biomedical EngineeringSchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| | - David M. Z. B. Hennes
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
- Pelvic Floor Disorders UnitMonash HealthClaytonVIC3168Australia
| | - Anna Rosamilia
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
- Pelvic Floor Disorders UnitMonash HealthClaytonVIC3168Australia
| | - Caroline E. Gargett
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Jerome A Werkmeister
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Shayanti Mukherjee
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| |
Collapse
|
4
|
Sadiasa A, Werkmeister JA, Gurung S, Gargett CE. Steps towards the clinical application of endometrial and menstrual fluid mesenchymal stem cells for the treatment of gynecological disorders. Expert Opin Biol Ther 2025:1-23. [PMID: 39925343 DOI: 10.1080/14712598.2025.2465826] [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: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The human endometrium is a highly regenerative tissue that contains mesenchymal stem/stromal cells (MSCs). These MSCs are sourced via office-based biopsies and menstrual fluid, providing a less invasive and readily available option for cell-based therapies. This review provides an update on endometrial-derived MSCs as a treatment option for gynecological diseases. AREAS COVERED This narrative review covers the characterization and therapeutic mechanisms of endometrium biopsy-derived MSCs (eMSCs) and menstrual fluid-derived mesenchymal stromal cells (MenSCs), highlighting similarities and differences. It also covers studies of their application in preclinical animal models and in clinical trials as potential cell-based therapies for gynecological diseases. EXPERT OPINION eMSCs and MenSCs from a homologous tissue source have the potential to promote regenerative activity as a treatment for gynecological diseases. Both eMSCs and MenSCs demonstrate therapeutic benefits through their paracrine activity in tissue regeneration, immunomodulation, angiogenesis, and mitigating fibrosis. Further research is essential to establish standardized isolation and characterization protocols, particularly for heterogeneous MenSCs, and to fully understand their mechanisms of action. Implementing SUSD2 magnetic bead sorting for purifying eMSCs from endometrial tissues and menstrual fluid is crucial for their use in future cell-based therapies. Optimization of production, storage, and delivery methods will maximize their therapeutic effectiveness.
Collapse
Affiliation(s)
- Alexander Sadiasa
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
5
|
Arul Murugan P, Bellare J. Design and development of vaginal wall mimicking poly( ϵ-caprolactone) based nanofibrous prosthetic mesh for pelvic organ prolapse: evaluation of biocompatibility and antibacterial ability. Biomed Mater 2025; 20:025013. [PMID: 39715589 DOI: 10.1088/1748-605x/ada2d0] [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] [Received: 06/20/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024]
Abstract
Mechanical non-conformance of conventionally used transvaginal non-degradable meshes has led to complications such as organ perforation, dyspareunia caused by mesh stiffness and stress shielding. In this study, we have solved the dire need to mimic the mechanical properties of the vaginal wall by designing and developing a soft and elastic mesh made of polycaprolactone (PCL), citric acid modified polyethylene glycol (PEGC) and zinc oxide (ZnO) prepared through electrospinning and testedin vitroandin vivo. The mesh containing 90:10:0.1 of PCL, PEGC and ZnO (PEGC-15 0.1ZnO mesh) conforms to the mechanical properties of the vaginal wall of the pelvic floor, has a burst strength of ∼35 N even after gamma-sterilization and 28 d of degradation inin vitro.In vitrostudies using adipose-derived stem cells revealed that the PCL-PEGC-15 0.1ZnO meshes were biocompatible and supported higher collagen production than commercial mesh.An in vitrobacterial adhesion study showed a 2-log reduction compared to commercially available mesh for prolapse treatment. Initial biocompatibility assessment in a rabbit model also showed that the PCL-PEGC-15 0.1ZnO mesh is biocompatible and supports fibrosis throughout the mesh. The softness and flexibility of the PCL-PEGC-15 0.1ZnO mesh based onin vitrotrials and initialin vivotrials show that the mesh has a potential clinical impact for pelvic floor repair treatment.
Collapse
Affiliation(s)
- Preethi Arul Murugan
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
- Wadhwani Research Center for Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| |
Collapse
|
6
|
Chen SH, Kuo HJ, Chou PY, Tsai CH, Chen SH, Yao YC, Liu SJ. Engineered, Radially Aligned, Gradient-Metformin-Eluting Nanofiber Dressings Accelerate Burn-Wound Healing. Int J Nanomedicine 2024; 19:11463-11477. [PMID: 39530106 PMCID: PMC11552508 DOI: 10.2147/ijn.s492244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Deep, second- and third-degree burn injuries may lead to irreversible damage to the traumatized tissue and to coagulation or thrombosis of the microvessels, further compromising wound healing. Engineered, morphologically gradient drug-eluting nanofiber dressings promote wound healing by mimicking tissue structure and providing sustained drug delivery, which is particularly beneficial for wound management. Methods This study exploited a resorbable, radially aligned nanofiber dressing that provides the sustained gradient release of metformin at the wound site using a pin-ring electrospinning technique and a differential membrane-thickness approach. Results The experimental results suggested that the electrospun nanofibrous dressings exhibited uniform and radially oriented fiber distributions. In vitro, these dressings offered an extended release of metformin for 30 d. The incorporation of water-soluble metformin significantly enhanced the hydrophilicity of the nanofiber membranes. Moreover, the in vivo burn-wound-healing model of rats showed that the radially aligned gradient metformin-eluting poly(lactic-co-glycolic acid) (PLGA) nanofibers exhibited significantly superior healing capability compared to the pristine PLGA, metformin-eluting, and control dressings. Histological images showed that the mesh/nanofibers produced no adverse effects. Conclusion The findings in this study emphasize the potential of resorbable, radially aligned nanofiber dressings as advanced wound care solutions, offering broad applicability and meaningful clinical impact.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Department of Plastic and Reconstructive Surgery, Chang-Gung Memorial Hospital, Chang-Gung University and Medical College, Taoyuan, Taiwan
| | - Hsiao-Jui Kuo
- Department of Plastic and Reconstructive Surgery, Chang-Gung Memorial Hospital, Chang-Gung University and Medical College, Taoyuan, Taiwan
| | - Pang-Yun Chou
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
| | - Chia-Hsuan Tsai
- Department of Plastic and Reconstructive Surgery, Chang-Gung Memorial Hospital, Keelung Branch, Chang-Gung University and Medical College, Keelung, Taiwan
| | - Shih-Hsien Chen
- Department of Plastic and Reconstructive Surgery, Chang-Gung Memorial Hospital, Chang-Gung University and Medical College, Taoyuan, Taiwan
| | - Yi-Chen Yao
- Department of Mechanical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
| |
Collapse
|
7
|
Barcena AJR, Mishra A, Bolinas DKM, Martin BM, Melancon MP. Integration of Electrospun Scaffolds and Biological Polymers for Enhancing the Delivery and Efficacy of Mesenchymal Stem/Stromal Cell Therapies. FRONT BIOSCI-LANDMRK 2024; 29:228. [PMID: 38940050 PMCID: PMC11725061 DOI: 10.31083/j.fbl2906228] [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: 03/19/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach for a variety of diseases due to their immunomodulatory and tissue regeneration capabilities. Despite their potential, the clinical application of MSC therapies is hindered by limited cell retention and engraftment at the target sites. Electrospun scaffolds, with their high surface area-to-volume ratio and tunable physicochemical properties, can be used as platforms for MSC delivery. However, synthetic polymers often lack the bioactive cues necessary for optimal cell-scaffold interactions. Integrating electrospun scaffolds and biological polymers, such as polysaccharides, proteins, and composites, combines the mechanical integrity of synthetic materials with the bioactivity of natural polymers and represents a strategic approach to enhance cell-scaffold interactions. The molecular interactions between MSCs and blended or functionalized scaffolds have been examined in recent studies, and it has been shown that integration can enhance MSC adhesion, proliferation, and paracrine secretion through the activation of multiple signaling pathways, such as FAK/Src, MAPK, PI3K/Akt, Wnt/β-catenin, and YAP/TAZ. Preclinical studies on small animals also reveal that the integration of electrospun scaffolds and natural polymers represents a promising approach to enhancing the delivery and efficacy of MSCs in the context of regenerating bone, cartilage, muscle, cardiac, vascular, and nervous tissues. Future research should concentrate on identifying the distinct characteristics of the MSC niche, investigating the processes involved in MSC-scaffold interactions, and applying new technologies in stem cell treatment and biofabrication to enhance scaffold design. Research on large animal models and collaboration among materials scientists, engineers, and physicians are crucial to translating these advancements into clinical use.
Collapse
Affiliation(s)
- Allan John R. Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Archana Mishra
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dominic Karl M. Bolinas
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Benjamin M. Martin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
8
|
Chen YP, Lo TS, Chien YH, Kuo YH, Liu SJ. In Vitro and In Vivo Drug Release from a Nano-Hydroxyapatite Reinforced Resorbable Nanofibrous Scaffold for Treating Female Pelvic Organ Prolapse. Polymers (Basel) 2024; 16:1667. [PMID: 38932015 PMCID: PMC11207985 DOI: 10.3390/polym16121667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Pelvic prolapse stands as a substantial medical concern, notably impacting a significant segment of the population, predominantly women. This condition, characterized by the descent of pelvic organs, such as the uterus, bladder, or rectum, from their normal positions, can lead to a range of distressing symptoms, including pelvic pressure, urinary incontinence, and discomfort during intercourse. Clinical challenges abound in the treatment landscape of pelvic prolapse, stemming from its multifactorial etiology and the diverse array of symptoms experienced by affected individuals. Current treatment options, while offering relief to some extent, often fall short in addressing the full spectrum of symptoms and may pose risks of complications or recurrence. Consequently, there exists a palpable need for innovative solutions that can provide more effective, durable, and patient-tailored interventions for pelvic prolapse. We manufactured an integrated polycaprolactone (PCL) mesh, reinforced with nano-hydroxyapatite (nHA), along with drug-eluting poly(lactic-co-glycolic acid) (PLGA) nanofibers for a prolapse scaffold. This aims to offer a promising avenue for enhanced treatment outcomes and improved quality of life for individuals grappling with pelvic prolapse. Solution extrusion additive manufacturing and electrospinning methods were utilized to prepare the nHA filled PCL mesh and drug-incorporated PLGA nanofibers, respectively. The pharmaceuticals employed included metronidazole, ketorolac, bleomycin, and estrone. Properties of fabricated resorbable scaffolds were assessed. The in vitro release characteristics of various pharmaceuticals from the meshes/nanofibers were evaluated. Furthermore, the in vivo drug elution pattern was also estimated on a rat model. The empirical data show that nHA reinforced PCL mesh exhibited superior mechanical strength to virgin PCL mesh. Electrospun resorbable nanofibers possessed diameters ranging from 85 to 540 nm, and released effective metronidazole, ketorolac, bleomycin, and estradiol, respectively, for 9, 30, 3, and over 30 days in vitro. Further, the mesh/nanofiber scaffolds also liberated high drug levels at the target site for more than 28 days in vivo, while the drug concentrations in blood remained low. This discovery suggests that resorbable scaffold can serve as a viable option for treating female pelvic organ prolapse.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Tsia-Shu Lo
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
| | - Yu-Han Chien
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-H.C.); (Y.-H.K.)
| | - Yi-Hua Kuo
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-H.C.); (Y.-H.K.)
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-H.C.); (Y.-H.K.)
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| |
Collapse
|
9
|
Zhang D, Xu D, Huang X, Wei Y, Tang F, Qin X, Liang W, Liang Z, Jin L, Wang H, Wang H. Puerarin-Loaded Electrospun Patches with Anti-Inflammatory and Pro-Collagen Synthesis Properties for Pelvic Floor Reconstruction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308590. [PMID: 38509840 DOI: 10.1002/advs.202308590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/02/2024] [Indexed: 03/22/2024]
Abstract
Pelvic organ prolapse (POP) is one of the most common pelvic floor dysfunction disorders worldwide. The weakening of pelvic connective tissues initiated by excessive collagen degradation is a leading cause of POP. However, the patches currently used in the clinic trigger an unfavorable inflammatory response, which often leads to implantation failure and the inability to simultaneously reverse progressive collagen degradation. Therefore, to overcome the present challenges, a new strategy is applied by introducing puerarin (Pue) into poly(l-lactic acid) (PLLA) using electrospinning technology. PLLA improves the mechanical properties of the patch, while Pue offers intrinsic anti-inflammatory and pro-collagen synthesis effects. The results show that Pue is released from PLLA@Pue in a sustained manner for more than 20 days, with a total release rate exceeding 80%. The PLLA@Pue electrospun patches also show good biocompatibility and low cytotoxicity. The excellent anti-inflammatory and pro-collagen synthesis properties of the PLLA@Pue patch are demonstrated both in vitro in H2O2-stimulated mouse fibroblasts and in vivo in rat abdominal wall muscle defects. Therefore, it is believed that this multifunctional electrospun patch integrating anti-inflammatory and pro-collagen synthesis properties can overcome the limitations of traditional patches and has great prospects for efficient pelvic floor reconstruction.
Collapse
Affiliation(s)
- Di Zhang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Dong Xu
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xiaobo Huang
- Department of Ophthalmology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yingqi Wei
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fuxin Tang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xiusen Qin
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Weiwen Liang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Zhongping Liang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, China
| | - Lin Jin
- International Joint Research Laboratory for Biomedical Nanomaterials of Henan, Zhoukou Normal University, Zhoukou, 466001, China
| | - Hui Wang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Huaiming Wang
- Department of General Surgery (Colorectal Surgery), Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| |
Collapse
|
10
|
Seifalian A, Digesu A, Khullar V. The use of animal models in preclinical investigations for the development of a surgical mesh for pelvic organ prolapse. Int Urogynecol J 2024; 35:741-758. [PMID: 38358519 PMCID: PMC11052796 DOI: 10.1007/s00192-024-05741-3] [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] [Received: 08/17/2023] [Accepted: 01/20/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION AND HYPOTHESIS Polypropylene (PP) mesh for the treatment of pelvic organ prolapse (POP) has raised substantial concerns over long-term complications, leading to its ban in multiple countries. In response, emerging materials are being explored as alternatives for prolapse surgery. Preclinical animal models have historically played a pivotal role in validating medical devices, prior to clinical trials. Successful translation of these materials necessitates the identification of suitable animal models that replicate the female human pelvis and its biomechanical properties. Preclinical in vivo testing assesses the safety of surgical mesh and treatment efficacy in preventing POP recurrence. METHODS The research critically reviews animal models used for preclinical pelvic mesh testing over the last decade and proposes a promising model for future preclinical studies. RESULTS Rats were the most common mammal used for toxicity and biocompatibility investigations through abdominal implantation. Although non-human primates serve as a gold standard for efficacy testing, ethical considerations limit their use owing to their close biological and cognitive resemblance to humans. Consequently, sheep were the most preferred large animal model owing to their reproductive system similarities and propensity for spontaneous POP following parity. CONCLUSION The study contributes valuable insights into the selection of appropriate animal models for preclinical pelvic mesh testing, offering guidance that is crucial for enhancing the safety and efficacy of novel surgical interventions in the treatment of POP.
Collapse
Affiliation(s)
- Amelia Seifalian
- Department of Urogynaecology, Imperial College London, London, UK.
| | - Alex Digesu
- Department of Urogynaecology, Imperial College London, London, UK
| | - Vikram Khullar
- Department of Urogynaecology, Imperial College London, London, UK
| |
Collapse
|
11
|
Liu J, Li F, Ouyang Y, Su Z, Chen D, Liang Z, Zhang Z, Lin R, Luo T, Guo L. Naringin-induced M2 macrophage polarization facilitates osteogenesis of BMSCs and improves cranial bone defect healing in rat. Arch Biochem Biophys 2024; 753:109890. [PMID: 38246327 DOI: 10.1016/j.abb.2024.109890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
Osteoimmunology has uncovered the critical role of the immune microenvironment in the bone healing process, with macrophages playing a central part in generating immune responses via chemokine production. Naringin, a flavanone glycoside extracted from various plants, has been shown to promote osteoblast differentiation, thereby enhancing bone formation and mitigating osteoporosis progression. Current research on the osteogenic mechanism primarily focuses on the direct impact of naringin on mesenchymal stem cells, while its indirect immunoregulatory effects remain elusive. In this study, we investigated the bone defect-enhancing effects of varying naringin concentrations in vivo using a cranial bone defect model in Sprague-Dawley rats. We assessed the osteoimmune modulation capacity of naringin by exposing lipopolysaccharide (LPS)-induced RAW 264.7 macrophages to different doses of naringin. To further elucidate the underlying osteogenic enhancement mechanism, Bone Marrow Stromal Cells (BMSCs) derived from mice were treated with conditioned media from naringin-treated macrophages. Our findings indicated that naringin promotes M2 phenotype polarization in macrophages, as evidenced by the downregulation of pro-inflammatory cytokines Inducible Nitric Oxide Synthase (iNOS), interleukin (IL)-1β, and Tumor Necrosis Factor (TNF)-α, and the upregulation of anti-inflammatory cytokine Transforming growth factor (TGF)-β. Transcriptome analysis revealed that differentially expressed genes were significantly enriched in osteoblast differentiation and anti-inflammatory response pathways in naringin-pretreated macrophages, with the cytokines signaling pathway being upregulated. The conditioned media from naringin-treated macrophages stimulated the expression of osteogenic-related genes Alkaline phosphatase (Alp), osteocalcin (Ocn), osteopontin (Opn), and Runt-related transcription factor (Runx) 2, as well as protein expression in BMSCs. In conclusion, naringin alleviates macrophage inflammation by promoting M2 phenotype polarization, which in turn enhances the osteogenic differentiation of BMSCs, contributing to its bone healing effects in vivo. These results suggest that naringin holds significant potential for improving bone defect healing through osteoimmune modulation.
Collapse
Affiliation(s)
- Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Fuyao Li
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Zitian Liang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Ruofei Lin
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tao Luo
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China.
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Aslan E, Maytalman E, Nemutlu Samur D, Köle E, Günizi ÖC. An in vitro pilot study investigating placenta-derived mesenchymal stem cell coating on polypropylene mesh materials. Int Urogynecol J 2024; 35:553-559. [PMID: 38206335 DOI: 10.1007/s00192-023-05687-y] [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] [Received: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION AND HYPOTHESIS Polypropylene meshes (PM) used in pelvic organ prolapse surgery are being withdrawn from the market. Although concerns about the usage of PMs in stress incontinence surgery have been raised, it is still one of the best methods of curing stress urinary incontinence. With advancements in stem cell-based therapies, especially mesenchymal stem cells (MSCs), it is believed that coating the synthetic meshes with MSCs may minimize excessive tissue reactions ultimately leading to clinical problems such as pain, erosion or extrusion of the implanted material. In our study we tried to show the possibility of coating the PM with placenta-derived MSCs. METHODS Mesenchymal stem cells obtained from six placentas were isolated, cultured, and identified. MSCs were then soaked in either fibronectin or collagen prior to co-culturing with strips of PMs. One group is used as a control, and hence was not pretreated before co-culturing. Specimens were fixed and stained with both Gram and hematoxylin and eosin and marked with Vybran Dil and DAPI. All preparations were examined under a light microscope. The IMAGEJ program was utilized to determine the surface area of meshes coated with MSCs. RESULTS We clearly showed that PMs can be coated successfully with placenta-derived MSCs. The percentage of the coated area is significantly increased when meshes were pretreated with fibronectin or collagen (p<0.0001). CONCLUSIONS Placenta-derived MSCs can successfully coat PMs. The immunomodulatory properties of MSCs, which may be of great advantage in preventing the side effects of meshes, should be tested by in vivo and hopefully human studies before clinical applications.
Collapse
Affiliation(s)
- Erdogan Aslan
- Faculty of Medicine, Department of Obstetrics and Gynecology, Alanya Alaaddin Keykubat University, Payallar Mh. Atatürk Cd. Yasemin Villaları, No:111/L-4, Alanya, Antalya, Türkiye.
| | - Erkan Maytalman
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| | - Dilara Nemutlu Samur
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| | - Emre Köle
- Faculty of Medicine, Department of Obstetrics and Gynecology, Alanya Alaaddin Keykubat University, Payallar Mh. Atatürk Cd. Yasemin Villaları, No:111/L-4, Alanya, Antalya, Türkiye
| | - Özlem Ceren Günizi
- Faculty of Medicine, Department of Pharmacology, Alanya Alaaddin Keykubat University, Alanya, Antalya, Türkiye
| |
Collapse
|
13
|
Xiong S, Zhang Y, Zeng J, Zhou J, Liu S, Wei P, Liu H, Yi F, Wan Z, Xiong L, Zhang B, Li J. DLP fabrication of HA scaffold with customized porous structures to regulate immune microenvironment and macrophage polarization for enhancing bone regeneration. Mater Today Bio 2024; 24:100929. [PMID: 38229884 PMCID: PMC10789648 DOI: 10.1016/j.mtbio.2023.100929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/24/2023] [Accepted: 12/23/2023] [Indexed: 01/18/2024] Open
Abstract
The immune microenvironment plays a pivotal role in osteoanagenesis. Biomaterials can modulate osteogenic efficacy by inducing specific local immune reactions. As 3D-printing technology advances, digital light projection printing has emerged as a promising method for creating large scale, high-precision biomaterial scaffolds. By adjusting the solid content and the sintering conditions during printing, the pore size of biomaterials can be meticulously controlled. Yet, the systematic influence of pore size on the immune microenvironment remains uncharted. We fabricated 3D-printed hydroxyapatite bioceramic scaffolds with three distinct pore sizes: 400 μm, 600 μm, and 800 μm. Our study revealed that scaffolds with a pore size of 600 μm promote macrophage M2 polarization, which is achieved by upregulating interferon-beta and HIF-1α production. When these materials were implanted subcutaneously in rats and within rabbit skulls, we observed that the 600 μm scaffolds notably improved the long-term inflammatory response, fostered vascular proliferation, and augmented new bone growth. This research paves the way for innovative therapeutic strategies for treating large segmental bone defects in clinical settings.
Collapse
Affiliation(s)
- Shilang Xiong
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Yinuo Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Jianhua Zeng
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jingyu Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shiwei Liu
- Department of Orthopedics, Ganzhou People's Hospital No.16, Mei Guan Road, Zhang Gong District, Ganzhou, Jiangxi, 341000, China
| | - Peng Wei
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hantian Liu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Feng Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zongmiao Wan
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Long Xiong
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Jingtang Li
- Department of Traumatology, Jiangxi Provincial People's Hospital the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
14
|
Darzi S, Alappadan J, Paul K, Mazdumder P, Rosamilia A, Truong YB, Gargett C, Werkmeister J, Mukherjee S. Immunobiology of foreign body response to composite PLACL/gelatin electrospun nanofiber meshes with mesenchymal stem/stromal cells in a mouse model: Implications in pelvic floor tissue engineering and regeneration. BIOMATERIALS ADVANCES 2023; 155:213669. [PMID: 37980818 DOI: 10.1016/j.bioadv.2023.213669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/21/2023]
Abstract
Pelvic Organ Prolapse (POP) is a common gynaecological disorder where pelvic organs protrude into the vagina. While transvaginal mesh surgery using non-degradable polymers was a commonly accepted treatment for POP, it has been associated with high rates of adverse events such as mesh erosion, exposure and inflammation due to serious foreign body response and therefore banned from clinical use after regulatory mandates. This study proposes a tissue engineering strategy using uterine endometrium-derived mesenchymal stem/stromal cells (eMSC) delivered with degradable poly L-lactic acid-co-poly ε-caprolactone (PLACL) and gelatin (G) in form of a composite electrospun nanofibrous mesh (P + G nanomesh) and evaluates the immunomodulatory mechanism at the material interfaces. The study highlights the critical acute and chronic inflammatory markers along with remodelling factors that determine the mesh surgery outcome. We hypothesise that such a bioengineered construct enhances mesh integration and mitigates the Foreign Body Response (FBR) at the host interface associated with mesh complications. Our results show that eMSC-based nanomesh significantly increased 7 genes associated with ECM synthesis and cell adhesion including, Itgb1, Itgb2, Vcam1, Cd44, Cdh2, Tgfb1, Tgfbr1, 6 genes related to angiogenesis including Ang1, Ang2, Vegfa, Pdgfa, Serpin1, Cxcl12, and 5 genes associated with collagen remodelling Col1a1, Col3a1, Col6a1, Col6a2, Col4a5 at six weeks post-implantation. Our findings suggest that cell-based tissue-engineered constructs potentially mitigate the FBR response elicited by biomaterial implants. From a clinical perspective, this construct provides an alternative to current inadequacies in surgical outcomes by modulating the immune response, inducing angiogenesis and ECM synthesis during the acute and chronic phases of the FBR.
Collapse
Affiliation(s)
- Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Janet Alappadan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Permita Mazdumder
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Anna Rosamilia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; Pelvic Floor Disorders Unit, Monash Health, Clayton, VIC 3168, Australia
| | | | - Caroline Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Jerome Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
15
|
Geoghegan N, O'Loughlin M, Delaney C, Rochfort KD, Kennedy M, Kolagatla S, Podhorska L, Rodriguez BJ, Florea L, Kelleher SM. Controlled degradation of polycaprolactone-based micropillar arrays. Biomater Sci 2023; 11:3077-3091. [PMID: 36876330 PMCID: PMC10152922 DOI: 10.1039/d3bm00165b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Herein we demonstrate the fabrication of arrays of micropillars, achieved through the combination of direct laser writing and nanoimprint lithography. By combining two diacrylate monomers, polycaprolactone dimethacrylate (PCLDMA) and 1,6-hexanediol diacrylate (HDDA), two copolymer formulations that, owing to the varying ratios of the hydrolysable ester functionalities present in the polycaprolactone moiety, can be degraded in the presence of base in a controllable manner. As such, the degradation of the micropillars can be tuned over several days as a function of PCLDMA concentration within the copolymer formulations, and the topography greatly varied over a short space of time, as visualised using scanning electron microscopy and atomic force microscopy. Crosslinked neat HDDA was used as a control material, demonstrating that the presence of the PCL was responsible for the ability of the microstructures to degrade in the controlled manner. In addition, the mass loss of the crosslinked materials was minimal, demonstrating the degradation of microstructured surfaces without loss of bulk properties was possible. Moreover, the compatibility of these crosslinked materials with mammalian cells was explored. The influence of both indirect and direct contact of the materials with A549 cells was assessed by profiling indices reflective of cytotoxicity such as morphology, adhesion, metabolic activity, oxidative balance, and release of injury markers. No significant changes in the aforementioned profile were observed in the cells cultured under these conditions for up to 72 h, with the cell-material interaction suggesting these materials may have potential in microfabrication contexts towards biomedical application purposes.
Collapse
Affiliation(s)
- Niamh Geoghegan
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Mark O'Loughlin
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Colm Delaney
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Meabh Kennedy
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Srikanth Kolagatla
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lucia Podhorska
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Brian J Rodriguez
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Larisa Florea
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Susan M Kelleher
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
16
|
Polypropylene Pelvic Mesh: What Went Wrong and What Will Be of the Future? Biomedicines 2023; 11:biomedicines11030741. [PMID: 36979721 PMCID: PMC10045074 DOI: 10.3390/biomedicines11030741] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Background: Polypropylene (PP) pelvic mesh is a synthetic mesh made of PP polymer used to treat pelvic organ prolapse (POP). Its use has become highly controversial due to reports of serious complications. This research critically reviews the current management options for POP and PP mesh as a viable clinical application for the treatment of POP. The safety and suitability of PP material were rigorously studied and critically evaluated, with consideration to the mechanical and chemical properties of PP. We proposed the ideal properties of the ‘perfect’ synthetic pelvic mesh with emerging advanced materials. Methods: We performed a literature review using PubMed/Medline, Embase, Cochrane Library (Wiley) databases, and ClinicalTrials.gov databases, including the relevant keywords: pelvic organ prolapse (POP), polypropylene mesh, synthetic mesh, and mesh complications. Results: The results of this review found that although PP is nontoxic, its physical properties demonstrate a significant mismatch between its viscoelastic properties compared to the surrounding tissue, which is a likely cause of complications. In addition, a lack of integration of PP mesh into surrounding tissue over longer periods of follow up is another risk factor for irreversible complications. Conclusions: PP mesh has caused a rise in reports of complications involving chronic pain and mesh exposure. This is due to the mechanical and physicochemical properties of PP mesh. As a result, PP mesh for the treatment of POP has been banned in multiple countries, currently with no alternative available. We propose the development of a pelvic mesh using advanced materials including emerging graphene-based nanocomposite materials.
Collapse
|
17
|
Sueters J, Groenman FA, Bouman MB, Roovers JPW, de Vries R, Smit TH, Huirne JAF. Tissue Engineering Neovagina for Vaginoplasty in Mayer-Rokitansky-Küster-Hauser Syndrome and Gender Dysphoria Patients: A Systematic Review. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:28-46. [PMID: 35819292 DOI: 10.1089/ten.teb.2022.0067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: Vaginoplasty is a surgical solution to multiple disorders, including Mayer-Rokitansky-Küster-Hauser syndrome and male-to-female gender dysphoria. Using nonvaginal tissues for these reconstructions is associated with many complications, and autologous vaginal tissue may not be sufficient. The potential of tissue engineering for vaginoplasty was studied through a systematic bibliography search. Cell types, biomaterials, and signaling factors were analyzed by investigating advantages, disadvantages, complications, and research quantity. Search Methods: A systematic search was performed in Medline, EMBASE, Web of Science, and Scopus until March 8, 2022. Term combinations for tissue engineering, guided tissue regeneration, regenerative medicine, and tissue scaffold were applied, together with vaginoplasty and neovagina. The snowball method was performed on references and a Google Scholar search on the first 200 hits. Original research articles on human and/or animal subjects that met the inclusion (reconstruction of vaginal tissue and tissue engineering method) and no exclusion criteria (not available as full text; written in foreign language; nonoriginal study article; genital surgery other than neovaginal reconstruction; and vaginal reconstruction with autologous or allogenic tissue without tissue engineering or scaffold) were assessed. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) checklist, the Newcastle-Ottawa Scale, and the Gold Standard Publication Checklist were used to evaluate article quality and bias. Outcomes: A total of 31 out of 1569 articles were included. Data extraction was based on cell origin and type, biomaterial nature and composition, host species, number of hosts and controls, neovaginal size, replacement fraction, and signaling factors. An overview of used tissue engineering methods for neovaginal formation was created, showing high variance of cell types, biomaterials, and signaling factors and the same topics were rarely covered multiple times. Autologous vaginal cells and extracellular matrix-based biomaterials showed preferential properties, and stem cells carry potential. However, quality confirmation of orthotopic cell-seeded acellular vaginal matrix by clinical trials is needed as well as exploration of signaling factors for vaginoplasty. Impact statement General article quality was weak to sufficient due to unreported cofounders and incomplete animal study descriptions. Article quality and heterogenicity made identification of optimal cell types, biomaterials, or signaling factors unreliable. However, trends showed that autologous cells prevent complications and compatibility issues such as healthy cell destruction, whereas stem cells prevent cross talk (interference of signaling pathways by signals from other cell types) and rejection (but need confirmation testing beyond animal trials). Natural (orthotopic) extracellular matrix biomaterials have great preferential properties that encourage future research, and signaling factors for vascularization are important for tissue engineering of full-sized neovagina.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Freek A Groenman
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Centre of Expertise on Gender Dysphoria, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Mark-Bram Bouman
- Centre of Expertise on Gender Dysphoria, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Jan Paul W Roovers
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Theo H Smit
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Department of Medical Biology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Research Institute Reproduction and Development, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Li L, Li Q, Gui L, Deng Y, Wang L, Jiao J, Hu Y, Lan X, Hou J, Li Y, Lu D. Sequential gastrodin release PU/n-HA composite scaffolds reprogram macrophages for improved osteogenesis and angiogenesis. Bioact Mater 2023; 19:24-37. [PMID: 35415312 PMCID: PMC8980440 DOI: 10.1016/j.bioactmat.2022.03.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Wound healing is a highly orchestrated process involving a variety of cells, including immune cells. Developing immunomodulatory biomaterials for regenerative engineering applications, such as bone regeneration, is an appealing strategy. Herein, inspired by the immunomodulatory effects of gastrodin (a bioactive component in traditional Chinese herbal medicine), a series of new immunomodulatory gastrodin-comprising biodegradable polyurethane (gastrodin-PU) and nano-hydroxyapatite (n-HA) (gastrodin-PU/n-HA) composites were developed. RAW 264.7 macrophages, rat bone marrow mesenchymal stem cells (rBMSCs), and human umbilical vein endothelial cells (HUVECs) were cultured with gastrodin-PU/n-HA containing different concentrations of gastrodin (0.5%, 1%, and 2%) to decipher their immunomodulatory effects on osteogenesis and angiogenesis in vitro. Results demonstrated that, compared with PU/n-HA, gastrodin-PU/n-HA induced macrophage polarization toward the M2 phenotype, as evidenced by the higher expression level of pro-regenerative cytokines (CD206, Arg-1) and the lower expression of pro-inflammatory cytokines (iNOS). The expression levels of osteogenesis-related factors (BMP-2 and ALP) in the rBMSCs and angiogenesis-related factors (VEGF and BFGF) in the HUVECs were significantly up-regulated in gastrodin-PU/n-HA/macrophage-conditioned medium. The immunomodulatory effects of gastrodin-PU/n-HA to reprogram macrophages from a pro-inflammatory (M1) phenotype to an anti-inflammatory and pro-healing (M2) phenotype were validated in a rat subcutaneous implantation model. And the 2% gastrodin-PU/n-HA significantly decreased fibrous capsule formation and enhanced angiogenesis. Additionally, 2% gastrodin-PU/n-HA scaffolds implanted in the rat femoral condyle defect model showed accelerated osteogenesis and angiogenesis. Thus, the novel gastrodin-PU/n-HA scaffold may represent a new and promising immunomodulatory biomaterial for bone repair and regeneration. A new immunomodulatory gastrodin-PU/n-HA biomaterial has been developed. The gastrodin-PU/n-HA triggered M2 macrophage polarization. The osteogenesis and angiogenesis were enhanced in response to the local immune microenvironment. The findings prove a therapeutic strategy in bone defect and other inflammatory osteoimmune disorders.
Collapse
Affiliation(s)
- Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Li Gui
- Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, 650011, China
| | - Yi Deng
- School of Chemical Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Lu Wang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jianlin Jiao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Yingrui Hu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Xiaoqian Lan
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650000, China
| | - Jianhong Hou
- Department of Orthopaedics, The Third People's Hospital of Yunnan Province, Kunming, 650011, China
- Corresponding author.
| | - Yao Li
- Department of Stomatology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
- Corresponding author.
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
- Corresponding author.
| |
Collapse
|
19
|
Lin M, Lu Y, Chen J. Tissue-engineered repair material for pelvic floor dysfunction. Front Bioeng Biotechnol 2022; 10:968482. [PMID: 36147522 PMCID: PMC9485870 DOI: 10.3389/fbioe.2022.968482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Pelvic floor dysfunction (PFD) is a highly prevalent urogynecology disorder affecting many women worldwide, with symptoms including pelvic organ prolapse (POP), stress urinary incontinence (SUI), fecal incontinence, and overactive bladder syndrome (OAB). At present, the clinical treatments of PFD are still conservative and symptom-based, including non-surgical treatment and surgery. Surgical repair is an effective and durable treatment for PFD, and synthetic and biological materials can be used to enforce or reinforce the diseased tissue. However, synthetic materials such as polypropylene patches caused a series of complications such as mesh erosion, exposure, pain, and inflammation. The poor mechanical properties and high degradation speed of the biomaterial meshes resulted in poor anatomical reduction effect and limitation to clinical application. Therefore, the current treatment options are suboptimal. Recently, tissue-engineered repair material (TERM) has been applied to repair PFD and could markedly improve the prognosis of POP and SUI repair surgery in animal models. We review the directions and progression of TERM in POP and SUI repair. Adipose-derived stem cells (ADSCs) and endometrial mesenchymal stem cells (eMSCs) appear to be suitable cell types for scaffold seeding and clinical implantation. The multidisciplinary therapy approach to tissue engineering is a promising direction for tissue repair. More and longer follow-up studies are needed before determining cell types and materials for PFD repair.
Collapse
Affiliation(s)
- Meina Lin
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University) and Liaoning Key Laboratory of Reproductive Health, Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China
| | - Yongping Lu
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University) and Liaoning Key Laboratory of Reproductive Health, Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China
- *Correspondence: Yongping Lu, ; Jing Chen,
| | - Jing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yongping Lu, ; Jing Chen,
| |
Collapse
|
20
|
Chou PY, Lee D, Chen SH, Liao CT, Lo LJ, Liu SJ. 3D-printed/electrospun bioresorbable nanofibrous drug-eluting cuboid frames for repair of alveolar bone defects. Int J Pharm 2022; 615:121497. [DOI: 10.1016/j.ijpharm.2022.121497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/27/2021] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
|
21
|
Francés-Herrero E, Lopez R, Hellström M, de Miguel-Gómez L, Herraiz S, Brännström M, Pellicer A, Cervelló I. OUP accepted manuscript. Hum Reprod Update 2022; 28:798-837. [PMID: 35652272 PMCID: PMC9629485 DOI: 10.1093/humupd/dmac025] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/13/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To provide the optimal milieu for implantation and fetal development, the female reproductive system must orchestrate uterine dynamics with the appropriate hormones produced by the ovaries. Mature oocytes may be fertilized in the fallopian tubes, and the resulting zygote is transported toward the uterus, where it can implant and continue developing. The cervix acts as a physical barrier to protect the fetus throughout pregnancy, and the vagina acts as a birth canal (involving uterine and cervix mechanisms) and facilitates copulation. Fertility can be compromised by pathologies that affect any of these organs or processes, and therefore, being able to accurately model them or restore their function is of paramount importance in applied and translational research. However, innate differences in human and animal model reproductive tracts, and the static nature of 2D cell/tissue culture techniques, necessitate continued research and development of dynamic and more complex in vitro platforms, ex vivo approaches and in vivo therapies to study and support reproductive biology. To meet this need, bioengineering is propelling the research on female reproduction into a new dimension through a wide range of potential applications and preclinical models, and the burgeoning number and variety of studies makes for a rapidly changing state of the field. OBJECTIVE AND RATIONALE This review aims to summarize the mounting evidence on bioengineering strategies, platforms and therapies currently available and under development in the context of female reproductive medicine, in order to further understand female reproductive biology and provide new options for fertility restoration. Specifically, techniques used in, or for, the uterus (endometrium and myometrium), ovary, fallopian tubes, cervix and vagina will be discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase databases was conducted to identify relevant studies published between January 2000 and September 2021. The search terms included: bioengineering, reproduction, artificial, biomaterial, microfluidic, bioprinting, organoid, hydrogel, scaffold, uterus, endometrium, ovary, fallopian tubes, oviduct, cervix, vagina, endometriosis, adenomyosis, uterine fibroids, chlamydia, Asherman’s syndrome, intrauterine adhesions, uterine polyps, polycystic ovary syndrome and primary ovarian insufficiency. Additional studies were identified by manually searching the references of the selected articles and of complementary reviews. Eligibility criteria included original, rigorous and accessible peer-reviewed work, published in English, on female reproductive bioengineering techniques in preclinical (in vitro/in vivo/ex vivo) and/or clinical testing phases. OUTCOMES Out of the 10 390 records identified, 312 studies were included for systematic review. Owing to inconsistencies in the study measurements and designs, the findings were assessed qualitatively rather than by meta-analysis. Hydrogels and scaffolds were commonly applied in various bioengineering-related studies of the female reproductive tract. Emerging technologies, such as organoids and bioprinting, offered personalized diagnoses and alternative treatment options, respectively. Promising microfluidic systems combining various bioengineering approaches have also shown translational value. WIDER IMPLICATIONS The complexity of the molecular, endocrine and tissue-level interactions regulating female reproduction present challenges for bioengineering approaches to replace female reproductive organs. However, interdisciplinary work is providing valuable insight into the physicochemical properties necessary for reproductive biological processes to occur. Defining the landscape of reproductive bioengineering technologies currently available and under development for women can provide alternative models for toxicology/drug testing, ex vivo fertility options, clinical therapies and a basis for future organ regeneration studies.
Collapse
Affiliation(s)
| | | | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lucía de Miguel-Gómez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- Fundación IVI, IVI-RMA Global, Valencia, Spain
| | - Sonia Herraiz
- Fundación IVI, IVI-RMA Global, Valencia, Spain
- Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Antonio Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- IVI Roma Parioli, IVI-RMA Global, Rome, Italy
| | | |
Collapse
|
22
|
Marinaro F, Silva JM, Barros AA, Aroso IM, Gómez-Blanco JC, Jardin I, Lopez JJ, Pulido M, de Pedro MÁ, Reis RL, Sánchez-Margallo FM, Casado JG, López E. A Fibrin Coating Method of Polypropylene Meshes Enables the Adhesion of Menstrual Blood-Derived Mesenchymal Stromal Cells: A New Delivery Strategy for Stem Cell-Based Therapies. Int J Mol Sci 2021; 22:13385. [PMID: 34948187 PMCID: PMC8706515 DOI: 10.3390/ijms222413385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Polypropylene (PP) mesh is well-known as a gold standard of all prosthetic materials of choice for the reinforcement of soft tissues in case of hernia, organ prolapse, and urinary incontinence. The adverse effects that follow surgical mesh implantation remain an unmet medical challenge. Herein, it is outlined a new approach to allow viability and adhesion of human menstrual blood-derived mesenchymal stromal cells (MenSCs) on PP surgical meshes. A multilayered fibrin coating, based on fibrinogen and thrombin from a commercial fibrin sealant, was optimized to guarantee a homogeneous and stratified film on PP mesh. MenSCs were seeded on the optimized fibrin-coated meshes and their adhesion, viability, phenotype, gene expression, and immunomodulatory capacity were fully evaluated. This coating guaranteed MenSC viability, adhesion and did not trigger any change in their stemness and inflammatory profile. Additionally, MenSCs seeded on fibrin-coated meshes significantly decreased CD4+ and CD8+ T cell proliferation, compared to in vitro stimulated lymphocytes (p < 0.0001). Hence, the proposed fibrin coating for PP surgical meshes may allow the local administration of stromal cells and the reduction of the exacerbated inflammatory response following mesh implantation surgery. Reproducible and easy to adapt to other cell types, this method undoubtedly requires a multidisciplinary and translational approach to be improved for future clinical uses.
Collapse
Affiliation(s)
- Federica Marinaro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
| | - Joana M. Silva
- 3B’s Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (J.M.S.); (A.A.B.); (I.M.A.); (R.L.R.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Alexandre A. Barros
- 3B’s Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (J.M.S.); (A.A.B.); (I.M.A.); (R.L.R.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Ivo M. Aroso
- 3B’s Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (J.M.S.); (A.A.B.); (I.M.A.); (R.L.R.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Juan C. Gómez-Blanco
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
| | - Isaac Jardin
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10003 Cáceres, Spain; (I.J.); (J.J.L.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Cáceres, Spain;
| | - Jose J. Lopez
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, 10003 Cáceres, Spain; (I.J.); (J.J.L.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Cáceres, Spain;
| | - María Pulido
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
| | - María Ángeles de Pedro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
| | - Rui L. Reis
- 3B’s Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; (J.M.S.); (A.A.B.); (I.M.A.); (R.L.R.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Javier G. Casado
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Cáceres, Spain;
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Immunology Unit, Department of Physiology, University of Extremadura, 10003 Cáceres, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.C.G.-B.); (M.P.); (M.Á.d.P.); (E.L.)
| |
Collapse
|
23
|
Antmen E, Vrana NE, Hasirci V. The role of biomaterials and scaffolds in immune responses in regenerative medicine: macrophage phenotype modulation by biomaterial properties and scaffold architectures. Biomater Sci 2021; 9:8090-8110. [PMID: 34762077 DOI: 10.1039/d1bm00840d] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Scaffolds are an integral part of the regenerative medicine field. The contact of biomaterials with tissue, as was clearly observed over the years, induces immune reactions in a material and patient specific manner, where both surface and bulk properties of scaffolds, together with their 3D architecture, have a significant influence on the outcome. This review presents an overview of the reactions to the biomaterials with a specific focus on clinical complications with the implants in the context of immune reactions and an overview of the studies involving biomaterial properties and interactions with innate immune system cells. We emphasize the impact of these studies on scaffold selection and upscaling of microenvironments created by biomaterials from 2D to 3D using immune cell encapsulation, seeding in a 3D scaffold and co-culture with relevant tissue cells. 3D microenvironments are covered with a specific focus on innate cells since a large proportion of these studies used innate immune cells. Finally, the recent studies on the incorporation of adaptive immune cells in immunomodulatory systems are covered in this review. Biomaterial-immune cell interactions are a critical part of regenerative medicine applications. Current efforts in establishing the ground rules for such interactions following implantation can control immune response during all phases of inflammation. Thus, in the near future for complete functional recovery, tissue engineering and control over biomaterials must be considered at the first step of immune modulation and this review covers these interactions, which have remained elusive up to now.
Collapse
Affiliation(s)
- Ezgi Antmen
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey.
| | - Nihal Engin Vrana
- SPARTHA Medical, 14B Rue de la Canardiere, Strasbourg Cedex 67100, France. .,INSERM Unité 1121 Biomaterials and Bioengineering, CRBS, 1 Rue Eugène Boeckel, Strasbourg Cedex 67000, France
| | - Vasif Hasirci
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey. .,Biomaterials A&R Center, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
24
|
Abstract
Uniquely among adult tissues, the human endometrium undergoes cyclical shedding, scar-free repair and regeneration during a woman's reproductive life. Therefore, it presents an outstanding model for study of such processes. This Review examines what is known of endometrial repair and regeneration following menstruation and parturition, including comparisons with wound repair and the influence of menstrual fluid components. We also discuss the contribution of endometrial stem/progenitor cells to endometrial regeneration, including the importance of the stem cell niche and stem cell-derived extracellular vesicles. Finally, we comment on the value of endometrial epithelial organoids to extend our understanding of endometrial development and regeneration, as well as therapeutic applications.
Collapse
Affiliation(s)
- Lois A Salamonsen
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Jennifer C Hutchison
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Caroline E Gargett
- Ritchie Centre, Hudson Institute of Medical Research, 25-31 Wright St, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
25
|
Hennes DMZB, Rosamilia A, Werkmeister JA, Gargett CE, Mukherjee S. Endometrial SUSD2 + Mesenchymal Stem/Stromal Cells in Tissue Engineering: Advances in Novel Cellular Constructs for Pelvic Organ Prolapse. J Pers Med 2021; 11:jpm11090840. [PMID: 34575617 PMCID: PMC8471527 DOI: 10.3390/jpm11090840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Cellular therapy is an emerging field in clinical and personalised medicine. Many adult mesenchymal stem/progenitor cells (MSC) or pluripotent derivatives are being assessed simultaneously in preclinical trials for their potential treatment applications in chronic and degenerative human diseases. Endometrial mesenchymal stem/progenitor cells (eMSC) have been identified as clonogenic cells that exist in unique perivascular niches within the uterine endometrium. Compared with MSC isolated from other tissue sources, such as bone marrow and adipose tissue, eMSC can be extracted through less invasive methods of tissue sampling, and they exhibit improvements in potency, proliferative capacity, and control of culture-induced differentiation. In this review, we summarize the potential cell therapy and tissue engineering applications of eMSC in pelvic organ prolapse (POP), emphasising their ability to exert angiogenic and strong immunomodulatory responses that improve tissue integration of novel surgical constructs for POP and promote vaginal tissue healing.
Collapse
Affiliation(s)
- David M. Z. B. Hennes
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (A.R.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
- Pelvic Floor Disorders Unit, Monash Health, Clayton, VIC 3168, Australia
- Correspondence: (D.M.Z.B.H.); (S.M.)
| | - Anna Rosamilia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (A.R.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
- Pelvic Floor Disorders Unit, Monash Health, Clayton, VIC 3168, Australia
| | - Jerome A. Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (A.R.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (A.R.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (A.R.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
- Correspondence: (D.M.Z.B.H.); (S.M.)
| |
Collapse
|
26
|
Yang D, Zhang M, Liu K. Tissue engineering to treat pelvic organ prolapse. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:2118-2143. [PMID: 34313549 DOI: 10.1080/09205063.2021.1958184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Pelvic organ prolapse (POP) is a frequent chronic illness, which seriously affects women's living quality. In recent years, tissue engineering has made superior progress in POP treatment, and biological scaffolds have received considerable attention. Nevertheless, pelvic floor reconstruction still faces severe challenges, including the construction of ideal scaffolds, the selection of optimal seed cells, and growth factors. This paper summarizes the recent progress of pelvic floor reconstruction in tissue engineering, and discusses the problems that need to be further considered and solved to provide references for the further development of this field.
Collapse
Affiliation(s)
- Deyu Yang
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, P.R. China
| | - Min Zhang
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, P.R. China
| | - Kehai Liu
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, P.R. China
| |
Collapse
|
27
|
Chen YP, Lo TS, Lin YT, Chien YH, Lu CJ, Liu SJ. Fabrication of Drug-Eluting Polycaprolactone/poly(lactic- co-glycolic Acid) Prolapse Mats Using Solution-Extrusion 3D Printing and Coaxial Electrospinning Techniques. Polymers (Basel) 2021; 13:polym13142295. [PMID: 34301052 PMCID: PMC8309226 DOI: 10.3390/polym13142295] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
We developed biodegradable drug-eluting prolapse mats using solution-extrusion 3D printing and coaxial electrospinning techniques. The mats were composed of polycaprolactone (PCL) mesh and lidocaine-, estradiol-, metronidazole-, and connective tissue growth factor (CTGF)-incorporated poly(lactic-co-glycolic acid) (PLGA) nanofibers that mimic the structure of the natural extracellular matrix of most connective tissues. The mechanical properties of degradable prolapse membrane were assessed and compared to commercial non-degradable polypropylene knitted meshes clinically used for pelvic organ prolapse (POP) repair. The release behaviors of the drug-loaded hybrid degradable membranes were also characterized. The experimental results suggest that 3D-printed PCL meshes exhibited comparable strengths to commercial POP meshes and survived through 10,000 cycles of fatigue test without breakage. Hybrid PCL meshes/PLGA nanofibrous membranes provided a sustainable release of metronidazole, lidocaine, and estradiol for 4, 25, and 30 days, respectively, in vitro. The membranes further liberated high levels of CTGF for more than 30 days. The animal tests show that the mechanical property of PCL mesh decreased with time, mainly due to degradation of the polymers post-implantation. No adverse effect of the mesh/nanofibers was noted in the histological images. By adopting solution-extrusion 3D printing and coaxial electrospinning, degradable drug-eluting membranes can be fabricated for POP applications.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
| | - Tsia-Shu Lo
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Linkou, School of Medicine, Chang Gung University, Taoyuan 33305, Taiwan;
| | - Yu-Ting Lin
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-T.L.); (Y.-H.C.); (C.-J.L.)
| | - Yu-Han Chien
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-T.L.); (Y.-H.C.); (C.-J.L.)
| | - Chia-Jung Lu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-T.L.); (Y.-H.C.); (C.-J.L.)
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-T.L.); (Y.-H.C.); (C.-J.L.)
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
- Correspondence: ; Tel.: +886-3-211-8166; Fax: +886-3-211-8558
| |
Collapse
|
28
|
Identification and characterisation of maternal perivascular SUSD2 + placental mesenchymal stem/stromal cells. Cell Tissue Res 2021; 385:803-815. [PMID: 33961124 DOI: 10.1007/s00441-021-03453-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) that meet the International Society for Cellular Therapy (ISCT) criteria are obtained from placental tissue by plastic adherence. Historically, no known single marker was available for isolating placental MSCs (pMSCs) from the decidua basalis. As the decidua basalis is derived from the regenerative endometrium, we hypothesised that SUSD2, an endometrial perivascular MSC marker, would purify maternal perivascular pMSC. Perivascular pMSCs were isolated from the maternal placenta using SUSD2 magnetic bead sorting and assessed for the colony-forming unit-fibroblasts (CFU-F), surface markers, and in vitro differentiation into mesodermal lineages. Multi-colour immunofluorescence was used to colocalise SUSD2 and α-SMA, a perivascular marker in the decidua basalis. Placental stromal cell suspensions comprised 5.1%SUSD2+ cells. SUSD2 magnetic bead sorting of the placental stromal cells increased their purity approximately two-fold. SUSD2+ pMSCs displayed greater CFU-F activity than SUSD2- stromal fibroblasts (pSFs). However, both SUSD2+ pMSC and SUSD2- pSF underwent mesodermal differentiation in vitro, and both expressed the ISCT surface markers. Higher percentages of cultured SUSD2+ pMSCs expressed the perivascular markers CD146, CD140b, and SUSD2 than SUSD2- pSFs. These findings suggest that SUSD2 is a single marker that enriches maternal pMSCs, suggesting they may originate from eMSC. Placental decidua basalis can be used as an alternative source of MSC for clinical translation in situations where there is no access to endometrial tissue.
Collapse
|
29
|
Darzi S, Paul K, Leitan S, Werkmeister JA, Mukherjee S. Immunobiology and Application of Aloe Vera-Based Scaffolds in Tissue Engineering. Int J Mol Sci 2021; 22:1708. [PMID: 33567756 PMCID: PMC7915752 DOI: 10.3390/ijms22041708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Aloe vera (AV), a succulent plant belonging to the Liliaceae family, has been widely used for biomedical and pharmaceutical application. Its popularity stems from several of its bioactive components that have anti-oxidant, anti-microbial, anti-inflammatory and even immunomodulatory effects. Given such unique multi-modal biological impact, AV has been considered as a biomaterial for regenerative medicine and tissue engineering applications, where tissue repair and neo-angiogenesis are vital. This review outlines the growing scientific evidence that demonstrates the advantage of AV as tissue engineering scaffolds. We particularly highlight the recent advances in the application of AV-based scaffolds. From a tissue engineering perspective, it is pivotal that the implanted scaffolds strike an appropriate foreign body response to be well-accepted in the body without complications. Herein, we highlight the key cellular processes that regulate the foreign body response to implanted scaffolds and underline the immunomodulatory effects incurred by AV on the innate and adaptive system. Given that AV has several beneficial components, we discuss the importance of delving deeper into uncovering its action mechanism and thereby improving material design strategies for better tissue engineering constructs for biomedical applications.
Collapse
Affiliation(s)
- Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (S.D.); (K.P.); (S.L.); (J.A.W.)
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (S.D.); (K.P.); (S.L.); (J.A.W.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Shanilka Leitan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (S.D.); (K.P.); (S.L.); (J.A.W.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Jerome A. Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (S.D.); (K.P.); (S.L.); (J.A.W.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (S.D.); (K.P.); (S.L.); (J.A.W.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| |
Collapse
|
30
|
Mukherjee S, Agarwal M, Bakshi A, Sawant S, Thomas L, Fujii N, Nair P, Kode J. Chemokine SDF1 Mediated Bone Regeneration Using Biodegradable Poly(D,L-lactide- co-glycolide) 3D Scaffolds and Bone Marrow-Derived Mesenchymal Stem Cells: Implication for the Development of an "Off-the-Shelf" Pharmacologically Active Construct. Biomacromolecules 2020; 21:4888-4903. [PMID: 33136384 DOI: 10.1021/acs.biomac.0c01134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is an increasing need for bone substitutes for reconstructive orthopedic surgery following removal of bone tumors. Despite the advances in bone regeneration, the use of autologous mesenchymal stem cells (MSC) presents a significant challenge, particularly for the treatment of large bone defects in cancer patients. This study aims at developing new chemokine-based technology to generate biodegradable scaffolds that bind pharmacologically active proteins for regeneration/repair of target injured tissues in patients. Primary MSC were cultured from the uninvolved bone marrow (BM) of cancer patients and further characterized for "stemness". Their ability to differentiate into an osteogenic lineage was studied in 2D cultures as well as on 3D macroporous PLGA scaffolds incorporated with biomacromolecules bFGF and homing factor chemokine stromal-cell derived factor-1 (SDF1). MSC from the uninvolved BM of cancer patients exhibited properties similar to that reported for MSC from BM of healthy individuals. Macroporous PLGA discs were prepared and characterized for pore size, architecture, functional groups, thermostability, and cytocompatibility by ESEM, FTIR, DSC, and CCK-8 dye proliferation assay, respectively. It was observed that the MSC+PLGA+bFGF+SDF1 construct cultured for 14 days supported significant cell growth, osteo-lineage differentiation with increased osteocalcin expression, alkaline phosphatase secretion, calcium mineralization, bone volume, and soluble IL6 compared to unseeded PLGA and PLGA+MSC, as analyzed by confocal microscopy, biochemistry, ESEM, microCT imaging, flow cytometry, and EDS. Thus, chemotactic biomacromolecule SDF1-guided tissue repair/regeneration ability of MSC from cancer patients opens up the avenues for development of "off-the-shelf" pharmacologically active construct for optimal repair of the target injured tissue in postsurgery cancer patients, bone defects, damaged bladder tissue, and radiation-induced skin/mucosal lesions.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC Australia 3168
| | - Manish Agarwal
- Department of Orthopaedic Oncology, Tata Memorial Hospital, TMC, Parel, Mumbai 400012, India
- Department of Orthopedic Oncology, P.D. Hinduja National Hospital & Medical Research Centre, Mumbai, India
| | - Ashish Bakshi
- Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- Department of Bone Marrow Transplantation, Department of Medical Oncology, Hiranandani Hospital, Powai, Mumbai 400076, India
| | - Sharada Sawant
- Electron Microscopy Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
| | - Lynda Thomas
- Laboratory for Polymer Analysis, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology Poojappura, Trivandrum, India
| | - Nobutaka Fujii
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Prabha Nair
- Laboratory for Polymer Analysis, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology Poojappura, Trivandrum, India
| | - Jyoti Kode
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
31
|
Aghaei-Ghareh-Bolagh B, Mukherjee S, Lockley KM, Mithieux SM, Wang Z, Emmerson S, Darzi S, Gargett CE, Weiss AS. A novel tropoelastin-based resorbable surgical mesh for pelvic organ prolapse repair. Mater Today Bio 2020; 8:100081. [PMID: 33210083 PMCID: PMC7658716 DOI: 10.1016/j.mtbio.2020.100081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 11/03/2022] Open
Abstract
Pelvic organ prolapse is a common condition that affects 1 in 4 women across all age groups. It is mainly caused by vaginal birth injury and can be exacerbated by obesity and increased age. Until recently, treatment strategies often used non-degradable synthetic meshes for reconstructive surgery. However, owing to their frequent, unacceptable rate of adverse events such as mesh erosion, transvaginal meshes have been banned in many countries. Recent reports have highlighted the urgent need for biocompatible design of meshes for a safe and effective treatment in the long term. This study reports the design and evaluation of a novel, elastin based degradable mesh using an ovine model of POP as a potential surgical treatment. Elastin is a protein component of the ECM and provides elasticity to tissues throughout the body. Tropoelastin, the monomer subunit of elastin, has been used with success in electrospun constructs as it is a naturally cell interactive polymer. Biomaterials that incorporate tropoelastin support cell attachment and proliferation, and have been proven to encourage elastogenesis and angiogenesis in vitro and in vivo. The biological properties of tropoelastin were combined with the physical properties of PCL, a degradable synthetic polymer, with the aim of producing, characterizing and assessing the performance of continuous tropoelastin:PCL electrospun yarns. Using a modified spinneret electrospinning system and adjusting settings based on relative humidity, four blends of tropoelastin:PCL yarns were fabricated with concentration ratios of 75:25, 50:50, 25:75 and 0:100. Yarns were assessed for ease of manufacture, fibrous architecture, protein/polymer content, yarn stability - including initial tropoelastin release, mechanical strength, and ability to support cell growth. Based on overall favorable properties, a mesh woven from the 50:50 tropoelastin:PCL yarn was implanted into the vagina of a parous ewe with vaginal wall weakness as a model of pelvic organ prolapse. This mesh showed excellent integration with new collagen deposition by SEM and a predominant M2 macrophage response with few pro-inflammatory M1 macrophages after 30 days. The woven tropoelastin:PCL electrospun mesh shows potential as an alternative to non-degradable, synthetic pelvic organ prolapse mesh products.
Collapse
Affiliation(s)
- B Aghaei-Ghareh-Bolagh
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - K M Lockley
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S M Mithieux
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - Z Wang
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia
| | - S Emmerson
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - S Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia
| | - C E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Victoria, 3168, Australia
| | - A S Weiss
- Charles Perkins Centre, University of Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia.,The University of Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| |
Collapse
|
32
|
Cun X, Hosta-Rigau L. Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2070. [PMID: 33092104 PMCID: PMC7590059 DOI: 10.3390/nano10102070] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising strategy to treat tissue and organ loss or damage caused by injury or disease. During the past two decades, mesenchymal stem cells (MSCs) have attracted a tremendous amount of interest in tissue engineering due to their multipotency and self-renewal ability. MSCs are also the most multipotent stem cells in the human adult body. However, the application of MSCs in tissue engineering is relatively limited because it is difficult to guide their differentiation toward a specific cell lineage by using traditional biochemical factors. Besides biochemical factors, the differentiation of MSCs also influenced by biophysical cues. To this end, much effort has been devoted to directing the cell lineage decisions of MSCs through adjusting the biophysical properties of biomaterials. The surface topography of the biomaterial-based scaffold can modulate the proliferation and differentiation of MSCs. Presently, the development of micro- and nano-fabrication techniques has made it possible to control the surface topography of the scaffold precisely. In this review, we highlight and discuss how the main topographical features (i.e., roughness, patterns, and porosity) are an efficient approach to control the fate of MSCs and the application of topography in tissue engineering.
Collapse
Affiliation(s)
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Nils Koppels Allé, Building 423, 2800 Kgs. Lyngby, Denmark;
| |
Collapse
|
33
|
Abstract
Mesenchymal stem cells (MSCs), also referred to as multipotent stromal cells or mesenchymal stromal cells, are present in multiple tissues and capable of differentiating into diverse cell lineages, holding a great promise in developing cell-based therapy for a wide range of conditions. Pelvic floor disorders (PFDs) is a common degenerative disease in women and may diminish a woman's quality of life at any age. Since the treatments for this disease are limited by the high rates of recurrence and surgical complications, seeking an ideal therapy in the restoration of pelvic floor function is an urgent issue at present. Herein, we summarize the cell sources of MSCs used for PFDs and discuss the potential mechanisms of MSCs in treating PFDs. Specifically, we also provide a comprehensive review of current preclinical and clinical trials dedicated to investigating MSC-based therapy for PFDs. The novel therapy has presented promising therapeutic effects which include relieving the symptoms of urinary or fecal incontinence, improving the biological properties of implanted meshes and promoting the injured tissue repair. Nevertheless, MSC-based therapies for PFDs are still experimental and the unstated issues on their safety and efficacy should be carefully addressed before their clinical applications.
Collapse
|
34
|
Lucciola R, Vrljicak P, Gurung S, Filby C, Darzi S, Muter J, Ott S, Brosens JJ, Gargett CE. Impact of Sustained Transforming Growth Factor-β Receptor Inhibition on Chromatin Accessibility and Gene Expression in Cultured Human Endometrial MSC. Front Cell Dev Biol 2020; 8:567610. [PMID: 32984350 PMCID: PMC7490520 DOI: 10.3389/fcell.2020.567610] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Endometrial mesenchymal stem cells (eMSC) drive the extraordinary regenerative capacity of the human endometrium. Clinical application of eMSC for therapeutic purposes is hampered by spontaneous differentiation and cellular senescence upon large-scale expansion in vitro. A83-01, a selective transforming growth factor-β receptor (TGFβ-R) inhibitor, promotes expansion of eMSC in culture by blocking differentiation and senescence, but the underlying mechanisms are incompletely understood. In this study, we combined RNA-seq and ATAC-seq to study the impact of sustained TGFβ-R inhibition on gene expression and chromatin architecture of eMSC. Treatment of primary eMSC with A83-01 for 5 weeks resulted in differential expression of 1,463 genes. Gene ontology analysis showed enrichment of genes implicated in cell growth whereas extracellular matrix genes and genes involved in cell fate commitment were downregulated. ATAC-seq analysis demonstrated that sustained TGFβ-R inhibition results in opening and closure of 3,555 and 2,412 chromatin loci, respectively. Motif analysis revealed marked enrichment of retinoic acid receptor (RAR) binding sites, which was paralleled by the induction of RARB, encoding retinoic acid receptor beta (RARβ). Selective RARβ inhibition attenuated proliferation and clonogenicity of A83-01 treated eMSC. Taken together, our study provides new insights into the gene networks and genome-wide chromatin changes that underpin maintenance of an undifferentiated phenotype of eMSC in prolonged culture.
Collapse
Affiliation(s)
- Raffaella Lucciola
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Pavle Vrljicak
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Caitlin Filby
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Joanne Muter
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Sascha Ott
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Jan J. Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Warwick Medical School, University Hospitals Coventry and Warwickshire National Health Service Trust, Coventry, United Kingdom
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, Gargett CE. Endometrial and Menstrual Blood Mesenchymal Stem/Stromal Cells: Biological Properties and Clinical Application. Front Cell Dev Biol 2020; 8:497. [PMID: 32742977 PMCID: PMC7364758 DOI: 10.3389/fcell.2020.00497] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFβ receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.
Collapse
Affiliation(s)
- Mahmood Bozorgmehr
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Shohreh Nikoo
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobitechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
36
|
Wu X, Jia Y, Sun X, Wang J. Tissue engineering in female pelvic floor reconstruction. Eng Life Sci 2020; 20:275-286. [PMID: 32647506 PMCID: PMC7336160 DOI: 10.1002/elsc.202000003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/16/2022] Open
Abstract
Pelvic organ prolapse is a common and frequently occurring disease in middle-aged and elderly women. Mesh implantation is an ideal surgical treatment. The polypropylene mesh commonly used in clinical practice has good mechanical properties, but there are long-term complications. The application of tissue engineering technology in the treatment of pelvic organ prolapse disease can not only meet the mechanical requirements of pelvic floor support, but also be more biocompatible than traditional polypropylene mesh, and can promote tissue repair to a certain extent. In this paper, the progress of tissue engineering was summarized to understand the application of tissue engineering in the treatment of pelvic organ prolapse disease and will help in research.
Collapse
Affiliation(s)
- Xiaotong Wu
- Department of Obstetrics and GynecologyPeking University People's HospitalBeijingP. R. China
- Beijing Key Laboratory of Female Pelvic Floor DisordersBeijingP. R. China
| | - YuanYuan Jia
- Department of Obstetrics and GynecologyPeking University People's HospitalBeijingP. R. China
- Beijing Key Laboratory of Female Pelvic Floor DisordersBeijingP. R. China
| | - Xiuli Sun
- Department of Obstetrics and GynecologyPeking University People's HospitalBeijingP. R. China
- Beijing Key Laboratory of Female Pelvic Floor DisordersBeijingP. R. China
| | - Jianliu Wang
- Department of Obstetrics and GynecologyPeking University People's HospitalBeijingP. R. China
- Beijing Key Laboratory of Female Pelvic Floor DisordersBeijingP. R. China
| |
Collapse
|
37
|
Design of Novel Perovskite-Based Polymeric Poly(l-Lactide-Co-Glycolide) Nanofibers with Anti-Microbial Properties for Tissue Engineering. NANOMATERIALS 2020; 10:nano10061127. [PMID: 32517379 PMCID: PMC7353416 DOI: 10.3390/nano10061127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/19/2023]
Abstract
There is a growing need for anti-microbial materials in several biomedical application areas, such are hernia, skin grafts as well as gynecological products, owing to the complications caused by infection due to surgical biomaterials. The anti-microbial effects of silver in the form of nanoparticles, although effective, can be toxic to surrounding cells. In this study, we report, for the first time, a novel biomedical application of Ag0.3Na1.7La2Ti3O10-layered perovskite particles, blended with poly(L-lactide-co-glycolide) (PLGA), aimed at designing anti-microbial and tissue engineering scaffolds. The perovskite was incorporated in three concentrations of 1, 5, 10 and 15 w/w% and electrospun using dimethylformamide (DMF) and chloroform. The morphology of the resultant nanofibers revealed fiber diameters in the range of 408 to 610 nm by scanning electron microscopy. Mechanical properties of perovskite-based nanofibers also matched similar mechanical properties to human skin. We observed impressive anti-microbial activity, against Gram-negative, Gram-positive bacteria and even fungi, to Ag0.3Na1.7La2Ti3O10 in powder as well as nanofiber-incorporated forms. Furthermore, cytotoxicity assay and immunocytochemistry revealed that perovskite-based nanofibers promoted the proliferation of human dermal fibroblasts whist maintaining normal cellular protein expression. Our study shows that perovskite-nanofibers have potential as scaffolds for biomedical applications with anti-microbial needs.
Collapse
|
38
|
Paul K, Darzi S, Werkmeister JA, Gargett CE, Mukherjee S. Emerging Nano/Micro-Structured Degradable Polymeric Meshes for Pelvic Floor Reconstruction. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1120. [PMID: 32517067 PMCID: PMC7353440 DOI: 10.3390/nano10061120] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Pelvic organ prolapse (POP) is a hidden women's health disorder that impacts 1 in 4 women across all age groups. Surgical intervention has been the only treatment option, often involving non-degradable meshes, with variable results. However, recent reports have highlighted the adverse effects of meshes in the long term, which involve unacceptable rates of erosion, chronic infection and severe pain related to mesh shrinkage. Therefore, there is an urgent unmet need to fabricate of new class of biocompatible meshes for the treatment of POP. This review focuses on the causes for the downfall of commercial meshes, and discusses the use of emerging technologies such as electrospinning and 3D printing to design new meshes. Furthermore, we discuss the impact and advantage of nano-/microstructured alternative meshes over commercial meshes with respect to their tissue integration performance. Considering the key challenges of current meshes, we discuss the potential of cell-based tissue engineering strategies to augment the new class of meshes to improve biocompatibility and immunomodulation. Finally, this review highlights the future direction in designing the new class of mesh to overcome the hurdles of foreign body rejection faced by the traditional meshes, in order to have safe and effective treatment for women in the long term.
Collapse
Affiliation(s)
- Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (K.P.); (S.D.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (K.P.); (S.D.); (J.A.W.); (C.E.G.)
| | - Jerome A. Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (K.P.); (S.D.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (K.P.); (S.D.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton 3168, Australia; (K.P.); (S.D.); (J.A.W.); (C.E.G.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| |
Collapse
|
39
|
Mukherjee S, Darzi S, Paul K, Cousins FL, Werkmeister JA, Gargett CE. Electrospun Nanofiber Meshes With Endometrial MSCs Modulate Foreign Body Response by Increased Angiogenesis, Matrix Synthesis, and Anti-Inflammatory Gene Expression in Mice: Implication in Pelvic Floor. Front Pharmacol 2020; 11:353. [PMID: 32265721 PMCID: PMC7107042 DOI: 10.3389/fphar.2020.00353] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/09/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Transvaginal meshes for the treatment of Pelvic Organ Prolapse (POP) have been associated with severe adverse events and have been banned for clinical use in many countries. We recently reported the design of degradable poly L-lactic acid-co-poly ε-caprolactone nanofibrous mesh (P nanomesh) bioengineered with endometrial mesenchymal stem/stromal cells (eMSC) for POP repair. We showed that such bioengineered meshes had high tissue integration as well as immunomodulatory effects in vivo. This study aimed to determine the key molecular players enabling eMSC-based foreign body response modulation. Methods SUSD2+ eMSC were purified from single cell suspensions obtained from endometrial biopsies from cycling women by magnetic bead sorting. Electrospun P nanomeshes with and without eMSC were implanted in a NSG mouse skin wound repair model for 1 and 6 weeks. Quantitative PCR was used to assess the expression of extracellular matrix (ECM), cell adhesion, angiogenesis and inflammation genes as log2 fold changes compared to sham controls. Histology and immunostaining were used to visualize the ECM, blood vessels, and multinucleated foreign body giant cells around implants. Results Bioengineered P nanomesh/eMSC constructs explanted after 6 weeks showed significant increase in 35 genes associated with ECM, ECM regulation, cell adhesion angiogenesis, and immune response in comparison to P nanomesh alone. In the absence of eMSC, acute inflammatory genes were significantly elevated at 1 week. However, in the presence of eMSC, there was an increased expression of anti-inflammatory genes including Mrc1 and Arg1 by 6 weeks. There was formation of multinucleated foreign body giant cells around both implants at 6 weeks that expressed CD206, a M2 macrophage marker. Conclusion This study reveals that eMSC modulate the foreign body response to degradable P nanomeshes in vivo by altering the expression profile of mouse genes. eMSC reduce acute inflammatory and increase ECM synthesis, angiogenesis and anti-inflammatory gene expression at 6 weeks while forming newly synthesized collagen within the nanomeshes and neo-vasculature in close proximity. From a tissue engineering perspective, this is a hallmark of a highly successful implant, suggesting significant potential as alternative surgical constructs for the treatment of POP.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Fiona L Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
40
|
Gargett CE, Gurung S, Darzi S, Werkmeister JA, Mukherjee S. Tissue engineering approaches for treating pelvic organ prolapse using a novel source of stem/stromal cells and new materials. Curr Opin Urol 2020; 29:450-457. [PMID: 31008783 DOI: 10.1097/mou.0000000000000634] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Nondegradable transvaginal polypropylene meshes for treating pelvic organ prolapse (POP) are now generally unavailable or banned. In this review, we summarize recent developments using tissue engineering approaches combining alternate degradable scaffolds with a novel source of mesenchymal stem/stromal cells from human endometrium (eMSC). RECENT FINDINGS Tissue engineering constructs comprising immunomodulatory, reparative eMSC and biomimetic materials with nanoarchitecture are a promising approach for vaginal repair and improving outcomes of POP surgery. Culture expansion of eMSC that maintains them (and other MSC) in the undifferentiated state has been achieved using a small molecule transforming growth factor-β receptor inhibitor, A83-01. The mechanism of action of A83-01 has been determined and its suitability for translation into the clinic explored. Novel blends of electrospun synthetic and natural polymers combined with eMSC shows this approach promotes host cell infiltration and slows biomaterial degradation that has potential to strengthen the vaginal wall during healing. Improving the preclinical ovine transvaginal surgical model by adapting the human clinical POP-Quantification system for selection of multiparous ewes with vaginal wall weakness enables assessment of this autologous eMSC/nanobiomaterial construct. SUMMARY A tissue engineering approach using autologous eMSC with degradable nanobiomaterials offers a new approach for treating women with POP.
Collapse
Affiliation(s)
- Caroline E Gargett
- The Ritchie Centre.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson institute of Medical Research
| | - Saeedeh Darzi
- The Ritchie Centre.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
41
|
Yang C, Zhao C, Wang X, Shi M, Zhu Y, Jing L, Wu C, Chang J. Stimulation of osteogenesis and angiogenesis by micro/nano hierarchical hydroxyapatite via macrophage immunomodulation. NANOSCALE 2019; 11:17699-17708. [PMID: 31545331 DOI: 10.1039/c9nr05730g] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Biomaterial topography-based strategies are regarded as an effective way to regulate the osteoimmune environment which plays an indispensable role in the bone regeneration process. The rapid development of manufacture techniques makes it possible to investigate the cell-topography interactions by preparing various micro and nano-topographical surfaces on biomaterials. Still, it is a challenge to prepare well-defined micro/nano hierarchical structures of bioceramics due to the inherent brittleness of ceramic materials. Also, the correlation between osteoimmunomodulation initiated by micro/nano hierarchical topographies and the tissue regeneration outcomes is unclear. In this study, we prepared well-defined micro/nano hierarchical structures on hydroxyapatite (HA) bioceramics through the combination of the photolithography and hydrothermal techniques. Three different microscale circular patterns (4 μm, 12 μm and 36 μm) and nanotopographies (nanoneedle, nanosheet and nanorod) were fabricated by changing the size of the mask and the condition of the hydrothermal reaction. The macrophage responses on the nanoneedle structures with different micropatterns were investigated and the micro/nano hierarchical structures with appropriate pattern sizes could either promote or alleviate the macrophage polarization, which further affected the outcomes of the osteogenic differentiation of human bone marrow stromal cells (hBMSCs) and angiogenic activity of human umbilical vein endothelial cells (HUVECs). Our study demonstrated that osteoimmunomodulation could be manipulated via tuning the micro/nano hierarchical structures, which could lead to a new strategy for the development of bone biomaterials with favorable osteoimmunomodulatory properties.
Collapse
Affiliation(s)
- Chen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Paul K, Darzi S, McPhee G, Del Borgo MP, Werkmeister JA, Gargett CE, Mukherjee S. 3D bioprinted endometrial stem cells on melt electrospun poly ε-caprolactone mesh for pelvic floor application promote anti-inflammatory responses in mice. Acta Biomater 2019; 97:162-176. [PMID: 31386931 DOI: 10.1016/j.actbio.2019.08.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/18/2022]
Abstract
Endometrial mesenchymal stem/stromal cells (eMSCs) exhibit excellent regenerative capacity in the endometrial lining of the uterus following menstruation and high proliferative capacity in vitro. Bioprinting eMSCs onto a mesh could be a potential therapy for Pelvic Organ Prolapse (POP). This study reports an alternative treatment strategy targeting vaginal wall repair using bioprinting of eMSCs encapsulated in a hydrogel and 3D melt electrospun mesh to generate a tissue engineering construct. Following a CAD, 3D printed poly ε-caprolactone (PCL) meshes were fabricated using melt electrospinning (MES) at different temperatures using a GMP clinical grade GESIM Bioscaffolder. Electron and atomic force microscopies revealed that MES meshes fabricated at 100 °C and with a speed 20 mm/s had the largest open pore diameter (47.2 ± 11.4 μm) and the lowest strand thickness (121.4 ± 46 μm) that promoted optimal eMSC attachment. An Aloe Vera-Sodium Alginate (AV-ALG) composite based hydrogel was optimised to a 1:1 mixture (1%AV-1%ALG) and eMSCs, purified from human endometrial biopsies, were then bioprinted in this hydrogel onto the MES printed meshes. Acute in vivo foreign body response assessment in NSG mice revealed that eMSC printed on MES constructs promoted tissue integration, eMSC retention and an anti-inflammatory M2 macrophage phenotype characterised by F4/80+CD206+ colocalization. Our results address an unmet medical need highlighting the potential of 3D bioprinted eMSC-MES meshes as an alternative approach to overcome the current challenges with non-degradable knitted meshes in POP treatment. STATEMENT OF SIGNIFICANCE: This study presents the first report of bioprinting mesenchymal stem cells derived from woman endometrium (eMSCs) to boost Pelvic Organ Prolapse (POP) treatment. It impacts over 50% of elderly women with no optimal treatment at present. The overall study is conducted in three stages as fabricating a melt electrospun (MES) mesh, bioprinting eMSCs into a Ca2+ free Aloe Vera-Alginate (AV-Alg) based hydrogel and in vivo study. Our data showed that AV-ALG hydrogel potentially suppresses the foreign body response and further addition of eMSCs triggered a high influx of anti-inflammatory CD206+ M2 macrophages. Our final construct demonstrates a favourable foreign body response to predict expected tissue integration, therefore, provides a potential for developing an alternative treatment for POP.
Collapse
Affiliation(s)
- Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Gordon McPhee
- Monash Health Translation Precinct, Cell Therapies and Regenerative Medicine Platform, Australia
| | - Mark P Del Borgo
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia.
| |
Collapse
|
43
|
Mukherjee S, Darzi S, Paul K, Werkmeister JA, Gargett CE. Mesenchymal stem cell-based bioengineered constructs: foreign body response, cross-talk with macrophages and impact of biomaterial design strategies for pelvic floor disorders. Interface Focus 2019; 9:20180089. [PMID: 31263531 PMCID: PMC6597526 DOI: 10.1098/rsfs.2018.0089] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
An excessive foreign body response (FBR) has contributed to the adverse events associated with polypropylene mesh usage for augmenting pelvic organ prolapse surgery. Consequently, current biomaterial research considers the critical role of the FBR and now focuses on developing better biocompatible biomaterials rather than using inert implants to improve the clinical outcomes of their use. Tissue engineering approaches using mesenchymal stem cells (MSCs) have improved outcomes over traditional implants in other biological systems through their interaction with macrophages, the main cellular player in the FBR. The unique angiogenic, immunomodulatory and regenerative properties of MSCs have a direct impact on the FBR following biomaterial implantation. In this review, we focus on key aspects of the FBR to tissue-engineered MSC-based implants for supporting pelvic organs and beyond. We also discuss the immunomodulatory effects of the recently discovered endometrial MSCs on the macrophage response to new biomaterials designed for use in pelvic floor reconstructive surgery. We conclude with a focus on considerations in biomaterial design that take into account the FBR and will likely influence the development of the next generation of biomaterials for gynaecological applications.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia.,CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia.,CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|