1
|
Huang D, Liu J, Yang J, Liang J, Zhang J, Han Q, Yu J, Yang T, Meng Q, Steinberg T, Li C, Chang Z. Restoration of Pregnancy Function Using a GT/PCL Biofilm in a Rabbit Model of Uterine Injury. Tissue Eng Part A 2025; 31:29-44. [PMID: 38526390 DOI: 10.1089/ten.tea.2023.0366] [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] [Indexed: 03/26/2024] Open
Abstract
Biomaterial scaffolds have been used successfully to promote the regenerative repair of small endometrial lesions in small rodents, providing partial restoration of gestational function. The use of rabbits in this study allowed us to investigate a larger endometrial tissue defect and myometrial injury model. A gelatin/polycaprolactone (GT/PCL) gradient-layer biofilm was sutured at the defect to guide the reconstruction of the original tissue structure. Twenty-eight days postimplantation, the uterine cavity had been restored to its original morphology, endometrial growth was accompanied by the formation of glands and blood vessels, and the fragmented myofibers of the uterine smooth muscle had begun to resemble the normal structure of the lagomorph uterine cavity, arranging in a circular luminal pattern and a longitudinal serosal pattern. In addition, the repair site supported both embryonic implantation into the placenta and normal embryonic development. Four-dimensional label-free proteomic analysis identified the cell adhesion molecules, phagosome, ferroptosis, rap1 signaling pathways, hematopoietic cell lineage, complement and coagulation cascades, tricarboxylic acid cycle, carbon metabolism, and hypoxia inducible factor (HIF)-1 signaling pathways as important in the endogenous repair process of uterine tissue injury, and acetylation of protein modification sites upregulated these signaling pathways.
Collapse
Affiliation(s)
- Di Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, P. R. China
| | - Jing Liu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Jie Yang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Junhui Liang
- Department of Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Jing Zhang
- Shandong First Medical University, Tai'an, P. R. China
| | - Qinyu Han
- Shandong First Medical University, Jinan, P. R. China
| | - Jianlong Yu
- Rizhao People's Hospital, Rizhao, P. R. China
| | - Tingting Yang
- Tai'an Maternal and Child Health Hospital, Tai'an, P. R. China
| | - Qi Meng
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, P. R. China
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, P. R. China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, P. R. China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, P. R. China
| | - Zhongle Chang
- Shandong Agriculture University, Tai'an, P. R. China
| |
Collapse
|
2
|
Shlomo Y, Gavriel M, Jaffa AJ, Grisaru D, Elad D. Arrangement into layers and mechanobiology of multi-cell co-culture models of the uterine wall. Hum Reprod 2024; 39:1767-1777. [PMID: 38876975 DOI: 10.1093/humrep/deae130] [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: 01/15/2024] [Revised: 05/18/2024] [Indexed: 06/16/2024] Open
Abstract
STUDY QUESTION Can a co-culture of three cell types mimic the in vivo layers of the uterine wall? SUMMARY ANSWER Three protocols tested for co-culture of endometrial epithelial cells (EEC), endometrial stromal cells (ESC), and myometrial smooth muscle cells (MSMC) led to formation of the distinct layers that are characteristic of the structure of the uterine wall in vivo. WHAT IS KNOWN ALREADY We previously showed that a layer-by-layer co-culture of EEC and MSMC responded to peristaltic wall shear stresses (WSS) by increasing the polymerization of F-actin in both layers. Other studies showed that WSS induced significant cellular alterations in epithelial and endothelial cells. STUDY DESIGN, SIZE, DURATION Human EEC and ESC cell lines and primary MSMC were co-cultured on a collagen-coated synthetic membrane in custom-designed wells. The co-culture model, created by seeding a mixture of all cells at once, was exposed to steady WSS of 0.5 dyne/cm2 for 10 and 30 min. PARTICIPANTS/MATERIALS, SETTING, METHODS The co-culture of the three different cells was seeded either layer-by-layer or as a mixture of all cells at once. Validation of the models was by specific immunofluorescence staining and confocal microscopy. Alterations of the cytoskeletal F-actin in response to WSS were analyzed from the 2-dimensional confocal images through the Z-stacks following a previously published algorithm. MAIN RESULTS AND THE ROLE OF CHANCE We generated three multi-cell in vitro models of the uterine wall with distinct layers of EEC, ESC, and MSMC that mimic the in vivo morphology. Exposure of the mixed seeding model to WSS induced increased polymerization of F-actin in all the three layers relative to the unexposed controls. Moreover, the increased polymerization of F-actin was higher (P-value < 0.05) when the length of exposure was increased from 10 to 30 min. Furthermore, the inner layers of ESC and MSMC, which are not in direct contact with the applied shearing fluid, also increased their F-actin polymerization. LARGE SCALE DATA N/A. LIMITATIONS, RESONS FOR CAUTION The mixed seeding co-culture model was exposed to steady WSS of one magnitude, whereas the uterus is a dynamic organ with intra-uterine peristaltic fluid motions that vary in vivo with different time-dependent magnitude. Further in vitro studies may explore the response to peristaltic WSS or other physical and/or hormonal perturbations that may mimic the spectrum of pathophysiological aspects. WIDER IMPLICATIONS OF THE FINDINGS Numerous in vitro models were developed in order to mimic the human endometrium and endometrium-myometrium interface (EMI) region. The present co-culture models seem to be the first constructed from EEC, ESC, and MSMC on a collagen-coated synthetic membrane. These multi-cell in vitro models better represent the complex in vivo anatomy of the EMI region. The mixed seeding multi-cell in vitro model may easily be implemented in controlled studies of uterine function in reproduction and the pathogenesis of diseases. STUDY FINDING/COMPETING INTEREST(S) This study was supported in part by Tel Aviv University funds. All authors declare no conflict of interest.
Collapse
Affiliation(s)
- Yael Shlomo
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mark Gavriel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ariel J Jaffa
- Department of Obstetrics and Gynecology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv, Israel
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dan Grisaru
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gynecological Oncology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - David Elad
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Luo Y, Chen Y, Gu Z, Ni R, Feng P, Hu Z, Song L, Shen X, Gu C, Li J, Du T, Yang L, Zhang H, Zhu Y. Engineered muscle from micro-channeled PEG scaffold with magnetic Fe 3O 4 fixation towards accelerating esophageal muscle repair. Mater Today Bio 2023; 23:100853. [PMID: 38024845 PMCID: PMC10663962 DOI: 10.1016/j.mtbio.2023.100853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Engineered scaffolds are used for repairing damaged esophagus to allow the precise alignment and movement of smooth muscle for peristalsis. However, most of these scaffolds focus solely on inducing cell alignment through directional apparatus, often overlooking the promotion of muscle tissue formation and causing reduced esophageal muscle repair effectiveness. To address this issue, we first introduced aligned nano-ferroferric oxide (Fe3O4) assemblies on a micropatterned poly(ethylene glycol) (PEG) hydrogel to form micro-/nano-stripes. Further modification using a gold coating was found to enhance cellular adhesion, orientation and organization within these micro-/nano-stripes, which consequently prevented excessive adhesion of smooth muscle cells (SMCs) to the thin PEG ridges, thereby effectively confining the cells to the Fe3O4-laid channels. This architectural design promotes the alignment of the cytoskeleton and elongation of actin filaments, leading to the organized formation of muscle bundles and a tendency for SMCs to adopt synthetic phenotypes. Muscle patches are harvested from the micro-/nano-stripes and transplanted into a rat esophageal defect model. In vivo experiments demonstrate the exceptional viability of these muscle patches and their ability to accelerate the regeneration of esophageal tissue. Overall, this study presents an efficient strategy for constructing muscle patches with directional alignment and muscle bundle formation of SMCs, holding significant promise for muscle tissue regeneration.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yichen Chen
- Ningbo Women and Children's Hospital, Ningbo, 315031, China
| | - Zhaofeng Gu
- Laboratory of Infrared Materials and Devices, Advanced Technology Research Institute, Ningbo University, Ningbo, 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Peipei Feng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315010, China
| | - Zeming Hu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lei Song
- Ningbo Women and Children's Hospital, Ningbo, 315031, China
| | - Xiang Shen
- Laboratory of Infrared Materials and Devices, Advanced Technology Research Institute, Ningbo University, Ningbo, 315211, China
| | - Chenjie Gu
- Laboratory of Infrared Materials and Devices, Advanced Technology Research Institute, Ningbo University, Ningbo, 315211, China
| | - Jiajie Li
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lu Yang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Hua Zhang
- Health Science Center, Ningbo University, Ningbo, 315211, China
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310027, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
4
|
Elia E, Brownell D, Chabaud S, Bolduc S. Tissue Engineering for Gastrointestinal and Genitourinary Tracts. Int J Mol Sci 2022; 24:ijms24010009. [PMID: 36613452 PMCID: PMC9820091 DOI: 10.3390/ijms24010009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The gastrointestinal and genitourinary tracts share several similarities. Primarily, these tissues are composed of hollow structures lined by an epithelium through which materials need to flow with the help of peristalsis brought by muscle contraction. In the case of the gastrointestinal tract, solid or liquid food must circulate to be digested and absorbed and the waste products eliminated. In the case of the urinary tract, the urine produced by the kidneys must flow to the bladder, where it is stored until its elimination from the body. Finally, in the case of the vagina, it must allow the evacuation of blood during menstruation, accommodate the male sexual organ during coitus, and is the natural way to birth a child. The present review describes the anatomy, pathologies, and treatments of such organs, emphasizing tissue engineering strategies.
Collapse
Affiliation(s)
- Elissa Elia
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1J 1Z4, Canada
| | - David Brownell
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1J 1Z4, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1J 1Z4, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1J 1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-525-4444 (ext. 42282)
| |
Collapse
|
5
|
Xu R, Fang X, Wu S, Wang Y, Zhong Y, Hou R, Zhang L, Shao L, Pang Q, Zhang J, Cui X, Zuo R, Yao L, Zhu Y. Development and Prospect of Esophageal Tissue Engineering. Front Bioeng Biotechnol 2022; 10:853193. [PMID: 35252159 PMCID: PMC8892191 DOI: 10.3389/fbioe.2022.853193] [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: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Currently, patients with esophageal cancer, especially advanced patients, usually use autologous tissue for esophageal alternative therapy. However, an alternative therapy is often accompanied by serious complications such as ischemia and leakage, which seriously affect the prognosis of patients. Tissue engineering has been widely studied as one of the ideal methods for the treatment of esophageal cancer. In view of the complex multi-layer structure of the natural esophagus, how to use the tissue engineering method to design the scaffold with structure and function matching with the natural tissue is the principle that the tissue engineering method must follow. This article will analyze and summarize the construction methods, with or without cells, and repair effects of single-layer scaffold and multi-layer scaffold. Especially in the repair of full-thickness and circumferential esophageal defects, the flexible design method and the binding force between the layers of the scaffold are very important. In short, esophageal tissue engineering technology has broad prospects and plays a more and more important role in the treatment of esophageal diseases.
Collapse
Affiliation(s)
- Rui Xu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Xinnan Fang
- School of Medicine, Ningbo University, Ningbo, China
| | - Shengqian Wu
- School of Medicine, Ningbo University, Ningbo, China
| | - Yiyin Wang
- School of Medicine, Ningbo University, Ningbo, China
| | - Yi Zhong
- School of Medicine, Ningbo University, Ningbo, China
| | - Ruixia Hou
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
- *Correspondence: Ruixia Hou,
| | - Libing Zhang
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Lei Shao
- School of Medicine, Ningbo University, Ningbo, China
| | - Qian Pang
- School of Medicine, Ningbo University, Ningbo, China
| | - Jian Zhang
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Xiang Cui
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Rongyue Zuo
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Liwei Yao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Yabin Zhu
- School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
6
|
Vasanthan KS, Srinivasan V, Mathur V, Agarwal P, Negi N, Kumari S. 3D Bioprinting for esophageal tissue regeneration: A review. JOURNAL OF MATERIALS RESEARCH 2022; 37:88-113. [DOI: 10.1557/s43578-021-00409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2025]
|
7
|
Qiu S, Liang L, Zou P, Chen Q. Decellularized small intestine submucosa/polylactic-co-glycolic acid composite scaffold for potential application in hypopharyngeal and cervical esophageal tissue repair. Regen Biomater 2021; 8:rbaa061. [PMID: 33738114 PMCID: PMC7955713 DOI: 10.1093/rb/rbaa061] [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: 10/07/2020] [Revised: 11/01/2020] [Accepted: 12/18/2020] [Indexed: 11/14/2022] Open
Abstract
There has been an increase in the incidence of hypopharyngeal and cervical esophageal cancer worldwide, and hence growing needs for hypopharyngeal and cervical esophageal tissue repair. This work produced a bi-layer composite scaffold with decellularized small intestine submucosa and polylactic-co-glycolic acid, which resembled the layered architectures of its intended tissues. The decellularized small intestine submucosa contained minimal residual DNA (52.5 ± 1.2 ng/mg) and the composite scaffold exhibited satisfactory mechanical properties (a tensile modulus of 21.1 ± 4.8 MPa, an ultimate tensile strength of 14.0 ± 2.9 MPa and a failure strain of 26.9 ± 5.1%). The interactions between cells and the respective layers of the scaffold were characterized by CCK-8 assays, immunostaining and Western blotting. Desirable cell proliferation and phenotypic behaviors were observed. These results have provided an important basis for the next-step in vivo studies of the scaffold, and bode well for its future clinical applications.
Collapse
Affiliation(s)
- Shijie Qiu
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.,Department of Otorhinolaryngology-Head and Neck Surgery, Lihuili Hospital of Ningbo University, 57 Xingning Road, Ningbo, 315041, People's Republic of China
| | - Lijin Liang
- Ningbo Regen Biotech Co., Ltd, 199 East Hexiao Road, Ningbo, 315157, People's Republic of China
| | - Peng Zou
- Ningbo Regen Biotech Co., Ltd, 199 East Hexiao Road, Ningbo, 315157, People's Republic of China
| | - Qi Chen
- Ningbo Regen Biotech Co., Ltd, 199 East Hexiao Road, Ningbo, 315157, People's Republic of China
| |
Collapse
|
8
|
The effects of alignment and diameter of electrospun fibers on the cellular behaviors and osteogenesis of BMSCs. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111787. [PMID: 33545913 DOI: 10.1016/j.msec.2020.111787] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/05/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Electrospun fiber scaffolds, due to their mimicry of bone extracellular matrix (ECM), have become an important biomaterial widely applied in bone tissue engineering in recent years. While topographic cues of electrospun membranes such as alignment and diameter played vital roles in determining cellular behaviors. Yet few researches about the effects of these two significant parameters on osteogenesis have been reported. Thus, the present work explored the influence of aligned and random poly (L-lactic acid) (PLLA) fiber matrices with diameters of nanoscale (0.6 μm) and microscale (1.2 μm), respectively, on cellular responses of bone marrow mesenchymal stem cells (BMSCs), such as cell adhesion, migration, proliferation and osteogenesis. Our results revealed that aligned nanofibers (AN) could affect cell morphology and promote the migration of BMSCs after 24 h of cell culturing. Besides, AN group was observed to possess excellent biocompatibility and have significantly improved cell growth comparing with random nanofibers. More importantly, in vitro osteogenesis researches including ALP and Alizarin Red S staining, qRT-PCR and immunofluorescence staining demonstrated that BMSCs culturing on AN group exhibited higher osteogenic induction proficiency than that on aligned microfibers (AM) and random fiber substrates (RN and RM). Accordingly, aligned nanofiber scaffolds have greater application potential in bone tissue engineering.
Collapse
|
9
|
Mondésert H, Bossard F, Favier D. Anisotropic electrospun honeycomb polycaprolactone scaffolds: Elaboration, morphological and mechanical properties. J Mech Behav Biomed Mater 2020; 113:104124. [PMID: 33091720 DOI: 10.1016/j.jmbbm.2020.104124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022]
Abstract
Tissue engineering technology requires porous scaffolds, based on biomaterials, which have to mimic as closely as possible the morphological and anisotropic mechanical properties of the native tissue to substitute. Anisotropic fibrous scaffolds fabricated by template-assisted electrospinning are investigated in this study. Fibers of electrospun Polycaprolactone (PCL) were successfully arranged spatially into honeycomb structures by using well-shaped 3D micro-architected metal collectors. Fibrous scaffolds present 2 × 4 mm2 wide elementary patterns with low and high fiber density areas. Distinct regions of the honeycomb patterns were analyzed through SEM images revealing different fiber diameters with specific fiber orientation depending on the regions of interest. Tensile test experiments were carried out with an optical observation of the local deformation at the pattern scale, allowing the determination and analysis, at small and large deformation, of the axial and transverse local strains. The honeycomb patterned mats showed significantly different mechanical properties along the two orthogonal directions probing an anisotropic ratio of 4.2. Stress relaxation test was performed on scaffolds at 15% of strain. This measurement pointed out the low contribution of the viscosity of about 20% in the mechanical response of the scaffold. An orthotropic linear elastic model was consequently proposed to characterize the anisotropic behavior of the produced patterned membranes. This new versatile method to produce architected porous materials, adjustable to several polymers and structures, will provide appealing benefits for soft regenerative medicine application and the development of custom-made scaffolds.
Collapse
Affiliation(s)
- Hugues Mondésert
- Univ. Grenoble Alpes, CNRS, Grenoble INP(1), LRP, Grenoble, 38000, France; Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP(1), TIMC-IMAG, Grenoble, 38000, France
| | - Frédéric Bossard
- Univ. Grenoble Alpes, CNRS, Grenoble INP(1), LRP, Grenoble, 38000, France.
| | - Denis Favier
- Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP(1), TIMC-IMAG, Grenoble, 38000, France
| |
Collapse
|
10
|
Tissue-engineered multi-cellular models of the uterine wall. Biomech Model Mechanobiol 2020; 19:1629-1639. [PMID: 31997029 DOI: 10.1007/s10237-020-01296-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
The human uterus is composed of three layers: endometrium, myometrium and perimetrium. It remodels during the monthly menstrual cycle and more significantly during the complex stages of reproduction. In vivo studies of the human uterine wall are yet incomplete due to ethical and technical limitations. The objective of this study was to develop in vitro uterine wall models that mimic the in vivo structure in humans. We co-cultured multiple cellular models of endometrial epithelial cells, endometrial stromal cells and smooth muscle cells on a synthetic membrane mounted in multi-purpose custom-designed wells. Immunofluorescence staining and confocal imaging confirmed that the new model represents the in vivo anatomical architecture of the inner uterine wall. Hormonal treatment with progesterone and β-estradiol demonstrated increased expression of progestogen-associated endometrial protein, which is associated with the in vivo receptive uterus. The new tissue-engineered in vitro models of the uterine wall will enable deeper investigation of molecular and biomechanical aspects of the blastocyst-uterus interaction during the window of implantation.
Collapse
|
11
|
Huyan Y, Lian Q, Zhao T, Li D, He J. Pilot Study of the Biological Properties and Vascularization of 3D Printed Bilayer Skin Grafts. Int J Bioprint 2020; 6:246. [PMID: 32596551 PMCID: PMC7294694 DOI: 10.18063/ijb.v6i1.246] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
Abstract
The skin is the largest human organ, and defects in the skin with a diameter greater than 4 cm do not heal without treatment. Allogeneic skin transplantation has been used to allow wound healing, but many grafts do not survive after implantation, due to multiple complications in the procedure. In the present study, the vascularization of three-dimensional (3D) printed full-thickness skin grafts was investigated. Dermal-epithelial grafts were transplanted into a nude mouse model to evaluate integration with the host tissue and the extent of wound healing. To create microvessels in the skin grafts, a bilayer structure consisting of human dermal fibroblasts, keratinocytes, and microvascular endothelial cells was designed and fabricated using an extruded 3D printer. Human dermal fibroblasts and human microvascular endothelial cells were mixed with gelatin-sodium alginate composite hydrogel as the dermis, and human keratinocytes were mixed with gel as the epithelium. Confocal imaging allowed visualization of the location of the cells in the double-layer skin grafts. A full-thickness wound was created on the backs of nude mice and then covered with a double-layer skin graft. Various groups of mice were tested. Animals were euthanized and tissue samples collected after specified time points. Compared with the control group, wound contraction improved by approximately 10%. Histological analysis demonstrated that the new skin had an appearance similar to that of normal skin and with a significant degree of angiogenesis. The results of the immunohistochemical analysis demonstrated that the transplanted cells survived and participated in the healing process.
Collapse
Affiliation(s)
- Yige Huyan
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Qin Lian
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Tingze Zhao
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
12
|
Yisireyili M, Wulamu W, Aili A, Li Y, Alimujiang A, Aipire A, Aizezi M, Zhang W, Cao Z, Mijiti A, Abudureyimu K. Chronic restraint stress induces esophageal fibrosis with enhanced oxidative stress in a murine model. Exp Ther Med 2019; 18:1375-1383. [PMID: 31316626 DOI: 10.3892/etm.2019.7669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 04/11/2019] [Indexed: 12/29/2022] Open
Abstract
Although the underlying mechanism of stress remains unknown, it has been associated with the pathophysiology of gastroesophageal reflux diseases, the development of which appear to be accelerated by oxidative stress and fibrosis. The aim of the current study was to investigate the effect of chronic restraint stress on esophageal oxidative stress and fibrosis, as well as the impact of oxidative stress in a murine model whereby 8-week old C57BL/6J male mice were subjected to intermittent chronic restraint stress for a two-week period. The current study demonstrated that chronic restraint stress significantly reduced the body weight of mice compared with the control group. Although chronic restraint stress did not significantly alter the levels of triglycerides or cholesterol, free fatty acid concentration was significantly increased compared with the control group. Furthermore, chronic restraint stress significantly upregulated the expression levels of several fibrotic biomarkers including collagen type I, transforming growth factor β-1, α-smooth muscle actin and SMAD-3 compared with the control group. In addition, the expression levels of the reactive oxygen species (ROS) NADPH oxidase-4 and malondialdehyde were significantly increased, while the expression levels of nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 were significantly decreased in esophageal tissue from mice in the chronic restraint stress group compared with the control group. In conclusion, chronic restraint stress may induce esophageal fibrosis by accumulating ROS and increasing fibrotic gene expression in a murine model.
Collapse
Affiliation(s)
- Maimaiti Yisireyili
- Research Institute of General and Minimally Invasive Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China.,Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Wubulikasimu Wulamu
- Research Institute of General and Minimally Invasive Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China.,Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Aikebaier Aili
- Research Institute of General and Minimally Invasive Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China.,Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Yiliang Li
- Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Aziguli Alimujiang
- Department of Obstetrics and Gynecology Clinic, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Aliyeguli Aipire
- Research Institute of General and Minimally Invasive Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Maimaitiaili Aizezi
- Department of Cardiac Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Weimin Zhang
- Department of Cardiac Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Zhengyi Cao
- Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Abulajiang Mijiti
- Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Kelimu Abudureyimu
- Research Institute of General and Minimally Invasive Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China.,Department of Minimally Invasive Surgery, Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| |
Collapse
|
13
|
Liu P, Fu K, Zeng X, Chen N, Wen X. Fabrication and Characterization of Composite Meshes Loaded with Antimicrobial Peptides. ACS APPLIED MATERIALS & INTERFACES 2019; 11:24609-24617. [PMID: 31199612 DOI: 10.1021/acsami.9b07246] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Biomaterials-centered infection or implant-associated infection plays critical roles in all areas of medicine with implantable devices. The widespread over use of antibiotics has caused severe bacterial resistance and even super bugs. Therefore, the development of anti-infection implantable devices with non-antibiotic-based new antimicrobial agents is indeed a priority for all of us. In this study, antimicrobial composite meshes were fabricated with broad-spectrum antimicrobial peptides (AMPs). Macroporous polypropylene meshes with poly-caprolactone electrospun nanosheets were utilized as a substrate to load AMPs and gellan gum presented as a media to gel with AMPs. Different amounts of AMPs were loaded onto gellan gum to determine the appropriate dose. The surface morphologies, Fourier-transform infrared spectroscopy spectra, in vitro release profiles, mechanical performances, in vitro antimicrobial properties, and cytocompatibility of composite scaffolds were evaluated. Results showed that AMPs were loaded into the meshes successfully, the in vitro release of AMPs in phosphate-buffered saline was prolonged, and less than 60% peptides were released in 10 days. The mechanical properties of composite meshes were also within the scope of several commercial surgical meshes. Composite meshes with the AMP loading amount of over 3 mg/cm2 showed inhibition against both Gram-negative and Gram-positive bacteria effectively, while they presented no toxicity to mammalian cells even at a loading amount of 10 mg/cm2. These results demonstrate a new simple and practicable method to offer antimicrobial properties to medical devices for hernia repair.
Collapse
Affiliation(s)
- Pengbi Liu
- College of Textiles , Donghua University , Shanghai 201620 , P. R. China
- Department of Chemical and Life Science Engineering, School of Engineering , Virginia Commonwealth University , Richmond , Virginia 23284 , United States
| | - Kun Fu
- Department of Chemical and Life Science Engineering, School of Engineering , Virginia Commonwealth University , Richmond , Virginia 23284 , United States
- Department of Stomatology , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan 450052 , P. R. China
| | - Xiaomei Zeng
- Department of Chemical and Life Science Engineering, School of Engineering , Virginia Commonwealth University , Richmond , Virginia 23284 , United States
| | - Nanliang Chen
- College of Textiles , Donghua University , Shanghai 201620 , P. R. China
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, School of Engineering , Virginia Commonwealth University , Richmond , Virginia 23284 , United States
- Beijing Ditan Hospital , Capital Medical University , Beijing 100015 , P. R. China
| |
Collapse
|
14
|
Biological properties of a bionic scaffold for esophageal tissue engineering research. Colloids Surf B Biointerfaces 2019; 179:208-217. [PMID: 30959233 DOI: 10.1016/j.colsurfb.2019.03.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/24/2019] [Accepted: 03/31/2019] [Indexed: 12/26/2022]
Abstract
Polyurethane is a good matrix material with wide application prospects in tissue engineering because of its adjustable and mechanical properties. A novel biodegradable crosslinked poly(ester urethane) (CPU) with flexible poly(caprolactone) (PCL) and hydrophilic poly(ethylene glycol) (PEG) components has been synthesized using a ferric iron catalyst in our laboratory. In the present study, to promote the interaction between the CPU material and cells, the material was superficially modified by silk fibroin (SF) grafting using an aminolysis and glutaraldehyde crosslinking method to achieve a biocompatible material, CPU-SF. Considering the esophageal-specific architecture, three types of scaffolds were fabricated. S1 was a CPU-SF channel (200 μm in diameter and 30 μm in depth with 30 μm of wall thickness) to support muscle regeneration; S2 was the decellularized matrix of the esophageal mucosa/submucosa obtained by enzyme treatment; and S3 was a combination of S1 and S2, aiming to promote esophageal regeneration with histological structure and function. The biological properties and functions of the materials and scaffolds were investigated by qualitative and quantitative analyses using scanning electron microscopy, immunofluorescence staining, cell adhesion and proliferation measurements, and western blotting technology. The results showed that esophageal smooth muscle cells (SMCs) and epithelial cells (ECs) were very well supported by the scaffolds. In particular, SMCs exhibited guided directional growth and ECs infiltrated the acellular mucosa with retained biological functions when co-cultured on the composite scaffold S3. These findings suggest that the composite bionic scaffold will be a good alternative for esophageal replacement.
Collapse
|
15
|
Liu P, Chen N, Jiang J, Wen X. New surgical meshes with patterned nanofiber mats. RSC Adv 2019; 9:17679-17690. [PMID: 35520597 PMCID: PMC9064676 DOI: 10.1039/c9ra01917k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/26/2019] [Indexed: 01/21/2023] Open
Abstract
Abdominal wall hernia repair is one of the most common general surgeries nowadays. Surgical meshes used in hernia repair indeed improved the outcomes, but complications like chronic pain or hernia recurrence partly caused by mechanical mismatch cannot be ignored. This work designed six warp-knitted polypropylene (PP) meshes and found the properties of surgical meshes could be improved to better mimic the performances of human abdominal wall by designing meshes with appropriate textile structures. Poly-caprolactone was electrospun onto newly designed PP meshes and formed a thin layer of patterned nanofiber mat. The pattern of nanofiber mats was affected by the structure of meshes. Diverse nanofiber morphology (straight aligned, straight random or spiral random pattern) and fiber diameters (50–70 nm ultra-thin nanofibers or from 330 nm to 700 nm nanofibers) were observed in different regions of a single patterned nanofiber scaffold. The addition of electrospinning nanofibers enhanced cell adherence and proliferation as compared with naked PP meshes. Cell actin filaments spread along the nanofibers and formed a morphology exactly similar with the patterned mats on day 7. Furthermore, cells on thin and aligned patterned nanofibers showed much more elongation and better orientation than that of the spiral random fibers, suggesting that cell morphology can be altered by changing the patterns of scaffolds. This study helps us in further understanding the properties of hernia repair meshes with their textile structures and the biological interactions of cells with different substrates in order to develop new biomedical scaffolds with desired properties. Newly designed warp-knitted meshes with different textile parameters and the interactions between cell and patterned nanofiber mats and different meshes.![]()
Collapse
Affiliation(s)
- Pengbi Liu
- College of Textiles
- Donghua University
- Shanghai 201620
- P. R. China
- Department of Chemical and Life Science Engineering
| | - Nanliang Chen
- College of Textiles
- Donghua University
- Shanghai 201620
- P. R. China
| | - Jinhua Jiang
- College of Textiles
- Donghua University
- Shanghai 201620
- P. R. China
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering
- School of Engineering
- Virginia Commonwealth University
- Richmond
- USA
| |
Collapse
|
16
|
Zhang Z, Jiang F, Zeng L, Wang X, Tu S. PHACTR1 regulates oxidative stress and inflammation to coronary artery endothelial cells via interaction with NF-κB/p65. Atherosclerosis 2018; 278:180-189. [PMID: 30293016 DOI: 10.1016/j.atherosclerosis.2018.08.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 08/06/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Genome-wide association studies have showed that genetic variants in phosphatase and actin regulator 1 (PHACTR1) are associated with coronary artery disease and myocardial infarction. However, the underlying mechanism of PHACTR1 in atherosclerosis remains unknown. METHODS Immunoblots were performed to evaluate the expression of PHACTR1 and phosphorylation of NF-κB signaling. Reactive oxygen species (ROS) labeled with DCFH-DA were assessed by flow cytometry. Fluorescence microscope was used to detect the translocation of p65 in human coronary artery endothelial cells (HACECs). Co-immunoprecipitation was performed to determine the interaction of PHACTR1 with MRTF-A. RESULTS The mRNA and protein levels of PHACTR1 were markedly increased in carotid plaquescompared with normal carotid arteries. Immunofluorescence staining indicated that PHACTR1 was constitutively expressed in endothelial cells in carotid plaques. Knockdown of PHACTR1 reduced excessive ICAM-1, VCAM-1 and VE-cadherin expression induced by oxidized low density lipoprotein (ox-LDL) in HCAECs. Additionally, silencing PHACTR1 alleviated p47phox phosphorylation and intracellular oxidative stress reflected by the reduction of ROS. Molecular experiments revealed that knockdown of PHACTR1 attenuated NF-κB activity without affecting IκBα and IKKα/β phosphorylation. In contrast, nuclear translation of p65 was blocked by depletion of PHACTR1. Furthermore, co-immunoprecipitation showed that PHACTR1 interacted with MRTF-A and p65 in HCAECs. Knockdown of MRTF-A suppressed the interaction of PHACTR1 with p65, subsequently blocking the nuclear translocation of p65. CONCLUSIONS Our finding suggest that silencing PHACTR1 alleviates the nuclear accumulation of p65 and NF-κB via interaction with MRTF-A, ensuing attenuating oxidative stress and inflammation in HCAECs.
Collapse
Affiliation(s)
- Zhihui Zhang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Fenglin Jiang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Lixiong Zeng
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Xiaoyan Wang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China
| | - Shan Tu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, China.
| |
Collapse
|