1
|
Wang H, Wu S, Bai X, Pan D, Ning Y, Wang C, Guo L, Guo J, Gu Y. Mesenchymal Stem Cell-Derived Exosomes Hold Promise in the Treatment of Diabetic Foot Ulcers. Int J Nanomedicine 2025; 20:5837-5857. [PMID: 40351704 PMCID: PMC12065540 DOI: 10.2147/ijn.s516533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic foot ulcers (DFU) represent one of the most common side effects of diabetes, significantly impacting patients' quality of life and imposing considerable financial burdens on families and society at large. Despite advancements in therapies targeting lower limb revascularization and various medications and dressings, outcomes for patients with severe lesions remain limited. A recent breakthrough in DFU treatment stems from the development of mesenchymal stem cells (MSCs). MSCs have shown promising results in treating various diseases and skin wounds due to their ability for multidirectional differentiation and immunomodulation. Recent studies highlight that MSCs primarily repair tissue through their paracrine activities, with exosomes playing a crucial role as the main biologically active components. These exosomes transport proteins, mRNA, DNA, and other substances, facilitating DFU treatment through immunomodulation, antioxidant effects, angiogenesis promotion, endothelial cell migration and proliferation, and collagen remodeling. Mesenchymal stem cell-derived exosomes (MSC-Exo) not only deliver comparable therapeutic effects to MSCs but also mitigate adverse reactions like immune rejection associated with MSCs transplantation. This article provides an overview of DFU pathophysiology and explores the mechanisms and research progress of MSC-Exo in DFU therapy.
Collapse
Affiliation(s)
- Hui Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Sensen Wu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Xinyu Bai
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Dikang Pan
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Yachan Ning
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Lianrui Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People’s Republic of China
| |
Collapse
|
2
|
Hou B, Cai W, Zhang S, Xu A, Wen Y, Wang Y, Zhu X, Wang F, Pan L, Qiu L, Sun H. Sustained-Release H 2S Nanospheres Regulate the Inflammatory Microenvironment of Wounds, Promote Angiogenesis and Collagen Deposition, and Accelerate Diabetic Wound Healing. ACS APPLIED BIO MATERIALS 2025; 8:2519-2534. [PMID: 39966083 DOI: 10.1021/acsabm.4c01955] [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: 02/20/2025]
Abstract
Diabetic wounds are blocked in the inflammatory stage, growth factors are degraded, and blood vessels are difficult to regenerate, leading to continuous necrosis and nonhealing of the wound. Hydrogen sulfide (H2S) plays an important role in the pathophysiological process of wound healing and has a long history of treating skin diseases. Although the sulfide salt solution is the preferred donor of exogenous H2S, its rapid release rate, excess production, and difficulty in accurately controlling the dose limit its use. Herein, we developed H2S sustained-release nanospheres NaHS@MS@LP for the treatment of diabetic wounds. NaHS@MS@LP nanosphere was composed of a NaHS-loaded mesoporous silicon core and a DSPE-PEG liposome outer membrane. When NaHS@MS@LP nanospheres were used to treat the wound of diabetic rats, mesoporous silicon was delivered into the cells and the loaded NaHS slowly released H2S through hydrolysis, participating in all stages of wound healing. In conclusion, NaHS@MS@LP nanospheres regulated the inflammatory microenvironment of wound skin by inducing the transformation of macrophages into M2 type and promoted angiogenesis and collagen deposition to accelerate wound healing in diabetic rats. Our findings provide strategies for the treatment of chronic wounds, including but not limited to diabetic wounds.
Collapse
Affiliation(s)
- Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Shijie Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Anjing Xu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yuanyuan Wen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yutong Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xuexue Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Fangming Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214125, China
| | - Lin Pan
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
3
|
Banerjee D, Vydiam K, Vangala V, Mukherjee S. Advancement of Nanomaterials- and Biomaterials-Based Technologies for Wound Healing and Tissue Regenerative Applications. ACS APPLIED BIO MATERIALS 2025; 8:1877-1899. [PMID: 40019109 DOI: 10.1021/acsabm.5c00075] [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: 03/01/2025]
Abstract
Patients and healthcare systems face significant social and financial challenges due to the increasing number of individuals with chronic external and internal wounds that fail to heal. The complexity of the healing process remains a serious health concern, despite the effectiveness of conventional wound dressings in promoting healing. Recent advancements in materials science and fabrication techniques have led to the development of innovative dressings that enhance wound healing. To further expedite the healing process, novel approaches such as nanoparticles, 3D-printed wound dressings, and biomolecule-infused dressings have emerged, along with cell-based methods. Additionally, gene therapy technologies are being harnessed to generate stem cell derivatives that are more functional, selective, and responsive than their natural counterparts. This review highlights the significant potential of biomaterials, nanoparticles, 3D bioprinting, and gene- and cell-based therapies in wound healing. However, it also underscores the necessity for further research to address the existing challenges and integrate these strategies into standard clinical practice.
Collapse
Affiliation(s)
- Durba Banerjee
- School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Kalyan Vydiam
- United Therapeutics, Manchester, New Hampshire 0310, United States
| | - Venugopal Vangala
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
4
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
5
|
Al Mamun A, Shao C, Geng P, Wang S, Xiao J. Recent advances in the role of neuroregulation in skin wound healing. BURNS & TRAUMA 2025; 13:tkae072. [PMID: 39872039 PMCID: PMC11770601 DOI: 10.1093/burnst/tkae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 01/29/2025]
Abstract
Neuroregulation during skin wound healing involves complex interactions between the nervous system and intricate tissue repair processes. The skin, the largest organ, depends on a complex system of nerves to manage responses to injury. Recent research has emphasized the crucial role of neuroregulation in maximizing wound healing outcomes. Recently, researchers have also explained the interactive contact between the peripheral nervous system and skin cells during the different phases of wound healing. Neurotransmitters and neuropeptides, once observed as simple signalling molecules, have since been recognized as effective regulators of inflammation, angiogenesis, and cell proliferation. The significance of skin innervation and neuromodulators is underscored by the delayed wound healing observed in patients with diabetes and the regenerative capabilities of foetal skin. Foetal skin regeneration is influenced by the neuroregulatory environment, immature immune system, abundant growth factors, and increased pluripotency of cells. Foetal skin cells exhibit greater flexibility and specialized cell types, and the extracellular matrix composition promotes regeneration. The extracellular matrix composition of foetal skin promotes regeneration, making it more capable than adult skin because neuroregulatory signals affect skin regeneration. The understanding of these systems can facilitate the development of therapeutic strategies to alter the nerve supply to the skin to enhance the process of wound healing. Neuroregulation is being explored as a potential therapeutic strategy for enhancing skin wound repair. Bioelectronic strategies and neuromodulation techniques can manipulate neural signalling, optimize the neuroimmune axis, and modulate inflammation. This review describes the function of skin innervation in wound healing, emphasizing the importance of neuropeptides released by sensory and autonomic nerve fibres. This article discusses significant discoveries related to neuroregulation and its impact on skin wound healing.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
6
|
Shah S, Mansour HM, Lucke-Wold B. Advances in Stem Cell Therapy for Huntington's Disease: A Comprehensive Literature Review. Cells 2025; 14:42. [PMID: 39791743 PMCID: PMC11719515 DOI: 10.3390/cells14010042] [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: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease characterized by uncontrolled movements, emotional disturbances, and progressive cognitive impairment. It is estimated to affect 4.3 to 10.6 per 100,000 people worldwide, and the mean prevalence rate among all published studies, reviews, and genetic HD registries is 5.7 per 100,000. A key feature of HD is the loss of striatal neurons and cortical atrophy. Although there is no cure at present, the discovery of the gene causing HD has brought us into a new DNA era and therapeutic advances for several neurological disorders. PubMed was systematically searched using three search strings: '"Huntington disease" + "stem cell"', '"Huntington disease" + Mesenchymal stromal cell', and '"Huntington disease" + "induced pluripotent stem cell"'. For each string, the search results were categorized based on cell type, and papers that included a clinical analysis were categorized as well. The data were extracted up to 2024. We did not include other databases in our search to have a comparable and systematic review of the literature on the topic. The collected data were analyzed and used for critical interpretation in the present review. Data are presented chronologically as clinical studies were published. Therapeutic strategies based on stem cells have drawn a lot of interest as possible HD therapies. Recent research indicates that NSCs have been the most often utilized stem cell type for treating HD. NSCs have been generated and extracted from a variety of sources, including HD patients' somatic cells and the brain itself. There is strong evidence supporting the transplantation of stem cells or their derivatives in HD animal models, even if stem-cell-based preclinical and clinical trials are still in their early stages. Current treatment only aims at relieving the symptoms rather than treating the pathogenesis of the disease. Although preclinical trials in HD models have shown promise in improving cognitive and motor functions, stem cell therapy still faces many challenges and disadvantages including immunosuppression and immunorejection as well as ethical, technical, and safety concerns. Further research is required for a definitive conclusion.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA (B.L.-W.)
| | | | | |
Collapse
|
7
|
Tripathi T, Mohan S, Alfaifi HA, Farasani A, R R, Sharma P, Sharma A, Koul A, Prasad GVS, Rustagi S, Anand J, Sah S, Gaidhane S, Bushi G, Jena D, Khatib MN, Shabil M, Abdelwahab SI, Bhopte K, Pant M, Mehta R, Pandey S, Brar M, Chilakam N, Balaraman AK. Efficacy and safety of stem cell therapy for fistula management: an overview of existing systematic reviews. Int J Surg 2024; 110:7573-7584. [PMID: 39468970 PMCID: PMC11634089 DOI: 10.1097/js9.0000000000002125] [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: 06/25/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Fistulas, abnormal connections between two anatomical structures, significantly impact the quality of life and can result from a variety of causes, including congenital defects, inflammatory conditions, and surgical complications. Stem cell therapy has emerged as a promising alternative due to its potential for regenerative and immunomodulatory effects. This overview of systematic reviews aimed to assess the safety and efficacy of stem cell therapy in managing fistulas, drawing on the evidence available. METHODS This umbrella review was conducted following the Joanna Briggs Institute (JBI) methodology to assess the efficacy and safety of stem cell therapy for treating various types of fistulas. A comprehensive search was performed across multiple electronic databases including PubMed, Embase, Cochrane Register, and Web of Science up to 5 May 2024. Systematic reviews focusing on stem cell therapy for fistulas were included, with data extracted on study design, stem cell types, administration methods, and outcomes. The quality of the reviews was assessed using the AMSTAR 2 tool, and meta-analyses were conducted using R software version 4.3. RESULTS Nineteen systematic reviews were included in our umbrella review. The stem cell therapy demonstrated by significant improvements in clinical remission rates, with a relative risk (RR) of 1.299 (95% CI: 1.192-1.420). Stem cell therapy enhanced fistula closure rates, both short-term (RR=1.481; 95% CI: 1.036-2.116) and long-term (RR=1.422; 95% CI: 1.091-1.854). The safety analysis revealed no significant increase in the risk of adverse events with stem cell therapy, showing a pooled RR of 0.972 (95% CI: 0.739-1.278) for general adverse events and 1.136 (95% CI: 0.821-1.572) for serious adverse events, both of which indicate a safety profile comparable to control treatments. Re-epithelialization rates also improved (RR=1.44; 95% CI: 1.322-1.572). CONCLUSION Stem cell therapy shows promise as an effective and safe treatment for fistulas, particularly in inducing remission and promoting closure of complex fistulas. The findings advocate for further high-quality research to confirm these benefits and potentially incorporate stem cell therapy into standard clinical practice for fistula management. Future studies should focus on long-term outcomes and refining stem cell treatment protocols to optimize therapeutic efficacy.
Collapse
Affiliation(s)
- Tripti Tripathi
- Department of Physiology, Integral Institute of Medical Sciences and Research, Dashauli, Uttar Pradesh, India
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Hassan A. Alfaifi
- Pharmaceutical Care Administration (Jeddah Second Health Cluster), Ministry of Health, Saudi Arabia
| | - Abdullah Farasani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, India
| | - G. V. Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Jigisha Anand
- Department of Biotechnology, Graphic Era (Deemed to be University) Clement Town Dehradun, India
- Department of Allied Sciences, Graphic Era Hill University Clement Town Dehradun, India
| | - Sanjit Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Shilpa Gaidhane
- One Health Centre, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha, India
| | - Ganesh Bushi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Diptismita Jena
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mahalaqua N. Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Muhammed Shabil
- University Center for Research and Development, Chandigarh University, Mohali, Punjab, India
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor, Malaysia
| | | | - Kiran Bhopte
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Manvi Pant
- New Delhi Institute of Management, New Delhi, India
| | - Rachana Mehta
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
- Dr Lal PathLabs - Nepal, Chandol-4, Maharajgunj, Kathmandu, Nepal
| | - Sakshi Pandey
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| | - Manvinder Brar
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
| | - Nagavalli Chilakam
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Ashok K. Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor, Malaysia
| |
Collapse
|
8
|
Jian Y, Li Y, Zhang Y, Tang M, Deng M, Liu C, Cheng M, Xiao S, Deng C, Wei Z. Lymphangiogenesis: novel strategies to promote cutaneous wound healing. BURNS & TRAUMA 2024; 12:tkae040. [PMID: 39328366 PMCID: PMC11427083 DOI: 10.1093/burnst/tkae040] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 09/28/2024]
Abstract
The cutaneous lymphatic system regulates tissue inflammation, fluid balance and immunological responses. Lymphangiogenesis or lymphatic dysfunction may lead to lymphedema, immune deficiency, chronic inflammation etc. Tissue regeneration and healing depend on angiogenesis and lymphangiogenesis during wound healing. Tissue oedema and chronic inflammation can slow wound healing due to impaired lymphangiogenesis or lymphatic dysfunction. For example, impaired lymphangiogenesis or lymphatic dysfunction has been detected in nonhealing wounds such as diabetic ulcers, venous ulcers and bedsores. This review summarizes the structure and function of the cutaneous lymphatic vessel system and lymphangiogenesis in wounds. Furthermore, we review wound lymphangiogenesis processes and remodelling, especially the influence of the inflammatory phase. Finally, we outline how to control lymphangiogenesis to promote wound healing, assess the possibility of targeting lymphangiogenesis as a novel treatment strategy for chronic wounds and provide an analysis of the possible problems that need to be addressed.
Collapse
Affiliation(s)
- Yang Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanqi Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanji Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingyuan Tang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingfu Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Chenxiaoxiao Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Maolin Cheng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| |
Collapse
|
9
|
Xie P, Xue X, Li X. Recent Progress in Mesenchymal Stem Cell-Derived Exosomes for Skin Wound Repair. Cell Biochem Biophys 2024; 82:1651-1663. [PMID: 38811472 DOI: 10.1007/s12013-024-01328-3] [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] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Exosomes are nanometer-sized, lipid bilayer membrane vesicles that are secreted by various cell types. Mesenchymal stem cells (MSCs) have been shown to exert therapeutic effects through the secretion of exosomes via a paracrine pathway. Functions: Recent studies have demonstrated that MSC-derived exosomes (MSC-Exos) can effectively transport various bioactive substances, including proteins, mRNAs, microRNAs, long non-coding RNAs, circular RNAs, and lipids, into target cells. This process regulates multiple aspects during wound repair, such as the inflammatory response, cell proliferation, migration, differentiation, angiogenesis, and matrix remodeling. POTENTIAL APPLICATIONS By promoting wound healing and inhibiting scar formation, MSC-Exos have shown great promise for clinical applications in wound repair. This review highlights the recent advances in our understanding of the role and mechanism of MSC-Exos during wound repair, providing insights into their potential use in future therapeutic strategies.
Collapse
Affiliation(s)
- Peilin Xie
- Department of Plastic Surgery, People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Xiaodong Xue
- Department of Plastic Surgery, People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Xiaodong Li
- Center for Cosmetic Surgery, General Hospital of Lanzhou Petrochemical Company (The Fourth Affiliated Hospital of Gansu University of Chinese Medicine), Lanzhou, 730060, Gansu, China.
| |
Collapse
|
10
|
Ahmadieh-Yazdi A, Karimi M, Afkhami E, Hajizadeh-Tafti F, Kuchakzadeh F, Yang P, Sheykhhasan M. Unveiling therapeutic potential: Adipose tissue-derived mesenchymal stem cells and their exosomes in the management of diabetes mellitus, wound healing, and chronic ulcers. Biochem Pharmacol 2024; 226:116399. [PMID: 38944396 DOI: 10.1016/j.bcp.2024.116399] [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: 01/31/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Diabetes mellitus (DM) is a pervasive global health issue with substantial morbidity and mortality, often resulting in secondary complications, including diabetic wounds (DWs). These wounds, arising from hyperglycemia, diabetic neuropathy, anemia, and ischemia, afflict approximately 15% of diabetic patients, with a considerable 25% at risk of lower limb amputations. The conventional approaches for chronic and diabetic wounds management involves utilizing various therapeutic substances and techniques, encompassing growth factors, skin substitutes and wound dressings. In parallel, emerging cell therapy approaches, notably involving adipose tissue-derived mesenchymal stem cells (ADMSCs), have demonstrated significant promise in addressing diabetes mellitus and its complications. ADMSCs play a pivotal role in wound repair, and their derived exosomes have garnered attention for their therapeutic potential. This review aimed to unravel the potential mechanisms and provide an updated overview of the role of ADMSCs and their exosomes in diabetes mellitus and its associated complications, with a specific focus on wound healing.
Collapse
Affiliation(s)
- Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Karimi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Afkhami
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Kuchakzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
11
|
Shi X, Li Y, Kang S, Zhao X, Liu L, Yuan F, He L, Lu H, Liu J. Dual-functional gallium/chitosan/silk/umbilical cord mesenchymal stem cell exosome sponge scaffold for diabetic wound by angiogenesis and antibacteria. Int J Biol Macromol 2024; 274:133420. [PMID: 38925194 DOI: 10.1016/j.ijbiomac.2024.133420] [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: 04/11/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
The treatment of diabetic wounds possessed significant challenges in clinical practice, which was accompanied with continuous infection, inflammation, and limited angiogenesis. Current wound dressings used for diabetic wound healing struggle to address these issues simultaneously. Therefore, Ga3+ was added to the chitosan/silk solution to confer potent antibacterial properties. Subsequently, umbilical cord mesenchymal stem cell exosomes (UCSC-Exo) were integrated into the gallium/chitosan/silk solution to enhance its angiogenesis-inducing activity. The mixture was lyophilized to prepare gallium/chitosan/silk/exosome sponge scaffolds (Ga/CSSF-Exo sponge scaffolds). The experiments of In vitro and in vivo demonstrated that Ga/CSSF-Exo sponge scaffolds exhibited sustained release of Ga3+ and bioactive exosomes, which effectively exerted continuous antibacterial effects and promoted angiogenesis. In diabetic rat wound models, Ga/CSSF-Exo sponge scaffolds facilitated angiogenesis, suppressed bacterial growth and inflammation, as well as promoted collagen deposition and re-epithelialization of wounds. Collectively, our findings suggested that Ga/CSSF-Exo held excellent potential for diabetic wound healing.
Collapse
Affiliation(s)
- Xin Shi
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yabei Li
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, Chenzhou, China; The First School of Clinical Medicine, Xiangnan University, Chenzhou, China
| | - Simiao Kang
- Department of Sports Medicine and Joint Arthroplasty, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Zhao
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, Chenzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The First School of Clinical Medicine, Xiangnan University, Chenzhou, China
| | - Liang Liu
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, Chenzhou, China; The First School of Clinical Medicine, Xiangnan University, Chenzhou, China
| | - Feifei Yuan
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyun He
- Department of Health Management Center, Chenzhou No.1 People's Hospital, Chenzhou, China.
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China; Hunan Engineering Research Center of Sports and Health, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Jun Liu
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, Chenzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The First School of Clinical Medicine, Xiangnan University, Chenzhou, China.
| |
Collapse
|
12
|
Xu J, Chen X, Wang J, Zhang B, Ge W, Wang J, Yang P, Liu Y. An ADSC-loaded dermal regeneration template promotes full-thickness wound healing. Regen Ther 2024; 26:800-810. [PMID: 39309394 PMCID: PMC11415530 DOI: 10.1016/j.reth.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/02/2024] [Accepted: 08/18/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Full-thickness wounds lead to delayed wound healing and scarring. Adipose-derived stem cell (ADSC) grafting promotes wound healing and minimizes scarring, but the low efficiency of grafting has been a challenge. We hypothesized that loading ADSCs onto a clinically widely used dermal regeneration template (DRT) would improve the efficacy of ADSC grafting and promote full-thickness wound healing. Methods ADSCs from human adipose tissue were isolated, expanded, and labeled with a cell tracker. Labeled ADSCs were loaded onto the DRT. The viability, the location of ADSCs on the DRT, and the abundance of ADSCs in the wound area were confirmed using CCK8 and fluorescence microscopy. Full-thickness wounds were created on Bama minipigs, which were applied with sham, ADSC, DRT, and ADSC-DRT. Wounds from the four groups were collected at the indicated time and histological analysis was performed. RNA-seq analysis was also conducted to identify transcriptional differences among the four groups. The identified genes by RNA-seq were verified by qPCR. Immunohistochemistry and western blotting were used to assess collagen deposition. In vitro, the supernatant of ADSCs was used to culture fibroblasts to investigate the effect of ADSCs on fibroblast transformation into myofibroblasts. Results ADSCs were successfully isolated, marked, and loaded onto the DRT. The abundance of ADSCs in the wound area was significantly greater in the ADSC-DRT group than in the ADSC group. Moreover, the ADSC-DRT group exhibited better wound healing with improved re-epithelialization and denser collagen deposition than the other three groups. The RNA-seq results suggested that the application of the integrated ADSC-DRT system resulted in the differential expression of genes mainly associated with extracellular matrix remodeling. In vivo, wounds from the ADSC-DRT group exhibited an earlier increase in type III collagen deposition and alleviated scar formation. ADSCs inhibited the transformation of fibroblasts into myofibroblasts, along with increased levels of CTGF, FGF, and HGF in the supernatant of ADSCs. Wounds from the ADSC-DRT group had up-regulated expressions of CTGF, HGF, FGF, and MMP3. Conclusion The integral of ADSC-DRT increased the efficacy of ADSC grafting, and promoted full-thickness wound healing with better extracellular matrix remodeling and alleviated scar formation.
Collapse
Affiliation(s)
- Jin Xu
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Plastic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuelian Chen
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jizhuang Wang
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beibei Zhang
- Department of Plastic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjia Ge
- Department of Plastic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqiang Wang
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peilang Yang
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Burn, Ruijin Hospital, Shanghai Burn Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Yang Y, Tang L, Xiao Y, Huang W, Gao M, Xie J, Yang M, Wu Y, Fu X. miR-21-5p-loaded bone mesenchymal stem cell-derived exosomes repair ovarian function in autoimmune premature ovarian insufficiency by targeting MSX1. Reprod Biomed Online 2024; 48:103815. [PMID: 38582043 DOI: 10.1016/j.rbmo.2024.103815] [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: 08/29/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 04/08/2024]
Abstract
RESEARCH QUESTION What is the effect of micro-RNA (miR)-21-5p-loaded bone marrow mesenchymal stem cell-derived exosomes (miR-21-Exo) on autoimmune premature ovarian insufficiency (POI)? DESIGN The Cell Counting Kit 8 (CCK8) assay, fluorescence-activated cell sorting, western blotting, quantitative reverse transcriptase (qRT)-PCR and enzyme-linked immunosorbent assay (ELISA) verified the effect of miR-21-Exo on interferon-γ (IFN-γ)-induced KGN cells. qRT-PCR, western blotting and dual-luciferase reporter gene assays verified that miR-21-Exo mediated Msh homeobox 1 (MSX1) regulation of the Notch signalling pathway and that miR-21 interacted directly with MSX1. The effects of miR-21-Exo on the ovaries were verified by monitoring of the oestrous cycle, haematoxylin and eosin staining, follicle counts, ELISA, immunohistochemistry, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling (TUNEL), western blotting and qRT-PCR. RESULTS The results showed that miR-21-Exo promoted IFN-γ-induced KGN cell proliferation and hormone synthesis, and inhibited apoptosis. Using dual-luciferase reporter gene assays, miR-21 and MSX1 were shown to have direct interactions. Moreover, the findings elucidated that miR-21-Exo inhibited cell apoptosis and promoted hormone synthesis by mediating MSX1 to regulate the Notch signalling pathway. miR-21-Exo restored the ovarian structure in a mouse model of autoimmune POI, promoted endocrine function and proliferation, and inhibited apoptosis and inflammation in vivo. CONCLUSIONS This study demonstrates that miR-21-Exo regulates the MSX1-mediated Notch signalling pathway to inhibit granulosa cell apoptosis and improve hormone synthesis function, providing insight into a potential mechanism of molecular therapy for the treatment of autoimmune POI.
Collapse
Affiliation(s)
- Yutao Yang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lichao Tang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanling Xiao
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wujia Huang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meng Gao
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaxin Xie
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingxin Yang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanhong Wu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiafei Fu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Song H, Hao D, Zhou J, Farmer D, Wang A. Development of pro-angiogenic skin substitutes for wound healing. Wound Repair Regen 2024; 32:208-216. [PMID: 38308588 DOI: 10.1111/wrr.13154] [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: 08/21/2023] [Revised: 11/13/2023] [Accepted: 12/12/2023] [Indexed: 02/05/2024]
Abstract
Wounds pose significant challenges to public health, primarily due to the loss of the mechanical integrity and barrier function of the skin and impaired angiogenesis, causing physical morbidities and psychological trauma to affect patients. Reconstructing the vasculature of the wound bed is crucial for promoting wound healing, reducing scar formation and enhancing the quality of life for patients. The development of pro-angiogenic skin substitutes has emerged as a promising strategy to facilitate vascularization and expedite the healing process of burn wounds. This review provides an overview of the various types of skin substitutes employed in wound healing, explicitly emphasising those designed to enhance angiogenesis. Synthetic scaffolds, biological matrices and tissue-engineered constructs incorporating stem cells and primary cells, cell-derived extracellular vesicles (EVs), pro-angiogenic growth factors and peptides, as well as gene therapy-based skin substitutes are thoroughly examined. The review summarises the existing challenges, future directions and potential innovations in pro-angiogenic dressing for skin substitutes. It highlights the need for continued research to develop new technologies and combine multiple strategies and factors, and to overcome obstacles and advance the field, ultimately leading to improved outcomes for wound patients.
Collapse
Affiliation(s)
- Hengyue Song
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Dake Hao
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Diana Farmer
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Biomedical Engineering, UC Davis, Davis, California, USA
| |
Collapse
|
15
|
Huang C, Teng J, Liu W, Wang J, Liu A. Modulation of macrophages by a phillyrin-loaded thermosensitive hydrogel promotes skin wound healing in mice. Cytokine 2024; 177:156556. [PMID: 38417214 DOI: 10.1016/j.cyto.2024.156556] [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: 10/28/2023] [Revised: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND Impaired wound healing in traumatic skin injuries remains a severe clinical challenge due to impaired re-vascularization, harmful bacteria infection, and inflammation dysregulation. Macrophages are recognized as prominent immune cells in tissue regeneration and wound healing. Consequently, the modulation of macrophages provides a promising therapeutic target for wound healing disorders. Here, we aimed to explore whether a novel constructed combination of thermosensitive hydrogel Pluronic F-127 (PF-127) and phillyrin (PH, the main active compound of forsythia suspensa) could improve skin wound healing. METHODS Firstly, the biological effects of pH on the phenotype and inflammation of macrophages were assessed by flow cytometry and ELISA. The biocompatibility of the PF-127 plus PH combination was investigated on keratinocytes and red blood cells. The biological effect of PF-127/PH hydrogel on the migratory ability of keratinocytes in vitro was evaluated using the scratch and transwell migration assays. In addition,S. aureusandE. coliwere employed to test the antibacterial properties of the PF-127 plus PH combination. Finally, PF-127 plus PH scaffold was appliedto the full-thickness skin defect in mice. Histomorphological evaluation and immunochemistry were performed to explore the wound-healing activity of PF-127/PH hydrogel. RESULTS PH can promote the polarization of macrophages from the M1 (pro-inflammatory) phenotype to the M2 (anti-inflammatory) phenotype. The PF-127/PH hydrogel was highly biocompatible and showed a potent stimulative effect on the migration of keratinocytesin vitro. The combination of PF-127 and PH exerted a pronounced antibacterial activity onS. aureusandE. coli in vitro.PF-127/PH hydrogel potently accelerates the healing of full-thickness skin defects by promoting skin cell proliferation, accelerating angiogenesis, and inhibiting inflammation. CONCLUSIONS Our study suggests that PF-127/PH hydrogel has excellent potential for treating traumatic skin defects.
Collapse
Affiliation(s)
- Chenggang Huang
- Department of Proctology, The Central Hospital of Yueyang, Yueyang 414020, PR China
| | - Jiajia Teng
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, PR China
| | - Wei Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, PR China
| | - Junzhe Wang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, PR China
| | - An Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, PR China.
| |
Collapse
|
16
|
Kulus M, Farzaneh M, Bryja A, Zehtabi M, Azizidoost S, Abouali Gale Dari M, Golcar-Narenji A, Ziemak H, Chwarzyński M, Piotrowska-Kempisty H, Dzięgiel P, Zabel M, Mozdziak P, Bukowska D, Kempisty B, Antosik P. Phenotypic Transitions the Processes Involved in Regulation of Growth and Proangiogenic Properties of Stem Cells, Cancer Stem Cells and Circulating Tumor Cells. Stem Cell Rev Rep 2024; 20:967-979. [PMID: 38372877 PMCID: PMC11087301 DOI: 10.1007/s12015-024-10691-w] [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] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial process with significance in the metastasis of malignant tumors. It is through the acquisition of plasticity that cancer cells become more mobile and gain the ability to metastasize to other tissues. The mesenchymal-epithelial transition (MET) is the return to an epithelial state, which allows for the formation of secondary tumors. Both processes, EMT and MET, are regulated by different pathways and different mediators, which affects the sophistication of the overall tumorigenesis process. Not insignificant are also cancer stem cells and their participation in the angiogenesis, which occur very intensively within tumors. Difficulties in effectively treating cancer are primarily dependent on the potential of cancer cells to rapidly expand and occupy secondarily vital organs. Due to the ability of these cells to spread, the concept of the circulating tumor cell (CTC) has emerged. Interestingly, CTCs exhibit molecular diversity and stem-like and mesenchymal features, even when derived from primary tumor tissue from a single patient. While EMT is necessary for metastasis, MET is required for CTCs to establish a secondary site. A thorough understanding of the processes that govern the balance between EMT and MET in malignancy is crucial.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Artur Bryja
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afsaneh Golcar-Narenji
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Hanna Ziemak
- Veterinary Clinic of the Nicolaus Copernicus University in Torun, Torun, Poland
| | - Mikołaj Chwarzyński
- Veterinary Clinic of the Nicolaus Copernicus University in Torun, Torun, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, Zielona Góra, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, USA
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland.
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland.
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, USA.
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic.
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| |
Collapse
|
17
|
Shang S, Zhuang K, Chen J, Zhang M, Jiang S, Li W. A bioactive composite hydrogel dressing that promotes healing of both acute and chronic diabetic skin wounds. Bioact Mater 2024; 34:298-310. [PMID: 38261910 PMCID: PMC10796815 DOI: 10.1016/j.bioactmat.2023.12.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024] Open
Abstract
Mesenchymal stem cell derived exosomes (MSC-Exos) demonstrate beneficial effects on wound healing via anti-inflammatory and angiogenic properties. Chitosan (CS) exhibits excellent biocompatibility and accelerates cellular migration, adhesion, and proliferation. The ions released from bioactive glass (BG) and titanium dioxide (TiO2) nanoparticles exhibit sustained angiogenic and antibacterial potency. In this study, CMCS-CEBT hydrogel was synthesized from exosomes encapsulated carboxymethyl chitosan (CMCS), chitosan nanoparticles (CS-NPs), BG, and TiO2 nanoparticles for a preliminary evaluation of its impacts on the treatment of full-thickness skin defects, diabetic wounds, and burn skin injury due to burns. In vitro analysis indicated that the hydrogel exhibits excellent cell compatibility, stimulates endothelial cell adhesion and proliferation, and presents anti-inflammatory, angiogenic, and antibacterial activities. In vivo, the composite hydrogel dressing accelerated a wound healing acceleration effect, stimulated angiogenesis, and increased collagen deposition and the expression of anti-inflammatory factors. This innovative composite hydrogel dressing as a potential clinical therapy, utilizing bioactive materials, holds promise as a potential clinical therapy that aims to facilitate the regeneration of acute and chronically damaged skin tissue.
Collapse
Affiliation(s)
- Shunlai Shang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Kaiting Zhuang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Jianwen Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Ming Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, 100020, China
| | - Shimin Jiang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
18
|
Shahin H, Belcastro L, Das J, Perdiki Grigoriadi M, Saager RB, Steinvall I, Sjöberg F, Olofsson P, Elmasry M, El-Serafi AT. MicroRNA-155 mediates multiple gene regulations pertinent to the role of human adipose-derived mesenchymal stem cells in skin regeneration. Front Bioeng Biotechnol 2024; 12:1328504. [PMID: 38562669 PMCID: PMC10982420 DOI: 10.3389/fbioe.2024.1328504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: The role of Adipose-derived mesenchymal stem cells (AD-MSCs) in skin wound healing remains to be fully characterized. This study aims to evaluate the regenerative potential of autologous AD-MSCs in a non-healing porcine wound model, in addition to elucidate key miRNA-mediated epigenetic regulations that underlie the regenerative potential of AD-MSCs in wounds. Methods: The regenerative potential of autologous AD-MSCs was evaluated in porcine model using histopathology and spatial frequency domain imaging. Then, the correlations between miRNAs and proteins of AD-MSCs were evaluated using an integration analysis in primary human AD-MSCs in comparison to primary human keratinocytes. Transfection study of AD-MSCs was conducted to validate the bioinformatics data. Results: Autologous porcine AD-MSCs improved wound epithelialization and skin properties in comparison to control wounds. We identified 26 proteins upregulated in human AD-MSCs, including growth and angiogenic factors, chemokines and inflammatory cytokines. Pathway enrichment analysis highlighted cell signalling-associated pathways and immunomodulatory pathways. miRNA-target modelling revealed regulations related to genes encoding for 16 upregulated proteins. miR-155-5p was predicted to regulate Fibroblast growth factor 2 and 7, C-C motif chemokine ligand 2 and Vascular cell adhesion molecule 1. Transfecting human AD-MSCs cell line with anti-miR-155 showed transient gene silencing of the four proteins at 24 h post-transfection. Discussion: This study proposes a positive miR-155-mediated gene regulation of key factors involved in wound healing. The study represents a promising approach for miRNA-based and cell-free regenerative treatment for difficult-to-heal wounds. The therapeutic potential of miR-155 and its identified targets should be further explored in-vivo.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
- Faculty of Biotechnology, Modern Sciences and Arts University, October City, Cairo, Egypt
| | - Luigi Belcastro
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Jyotirmoy Das
- Bioinformatics Unit, Core Facility (KEF), Faculty of Medicine and Health Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, SciLife Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Rolf B. Saager
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| | - Pia Olofsson
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Moustafa Elmasry
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Ahmed T. El-Serafi
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| |
Collapse
|
19
|
Wein S, Schemmer C, Al Enezy-Ulbrich MA, Jung SA, Rütten S, Kühnel M, Jonigk D, Jahnen-Dechent W, Pich A, Neuss S. Fibrin-Based Hydrogels with Reactive Amphiphilic Copolymers for Mechanical Adjustments Allow for Capillary Formation in 2D and 3D Environments. Gels 2024; 10:182. [PMID: 38534600 DOI: 10.3390/gels10030182] [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: 02/15/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
This study focuses on enhancing controllable fibrin-based hydrogels for tissue engineering, addressing existing weaknesses. By integrating a novel copolymer, we improved the foundation for cell-based angiogenesis with adaptable structural features. Tissue engineering often faces challenges like waste disposal and nutrient supply beyond the 200 µm diffusion limit. Angiogenesis breaks through this limitation, allowing the construction of larger constructs. Our innovative scaffold combination significantly boosts angiogenesis, resulting in longer branches and more capillary network junctions. The copolymer attached to fibrin fibers enables precise adjustment of hydrogel mechanical dynamic properties for specific applications. Our material proves effective for angiogenesis, even under suppression factors like suramin. In our study, we prepared fibrin-based hydrogels with and without the copolymer PVP12400-co-GMA10mol%. Using a co-culture system of human umbilical vein endothelial cells (HUVEC) and mesenchymal stem cells (MSC), we analyzed angiogenetic behavior on and within the modified hydrogels. Capillary-like structures were reproducibly formed on different surfaces, demonstrating the general feasibility of three-dimensional endothelial cell networks in fibrin-based hydrogels. This highlights the biomaterial's suitability for in vitro pre-vascularization of biohybrid implants.
Collapse
Affiliation(s)
- Svenja Wein
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Carina Schemmer
- Chair for Laser Technology LLT, RWTH Aachen University, Steinbachstraße 15, 52074 Aachen, Germany
| | - Miriam Aischa Al Enezy-Ulbrich
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Shannon Anna Jung
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Stephan Rütten
- Electron Microscopic Facility, University Clinics, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Mark Kühnel
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Danny Jonigk
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Wilhelm Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Andrij Pich
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
20
|
Gheytasvand A, Bagheri H, Pourbeyranvand S, Salehnia M. Enhancement of Wound Healing and Angiogenesis Using Mouse Embryo Fibroblasts Loaded in Decellularized Skin Scaffold. IRANIAN BIOMEDICAL JOURNAL 2024; 28:90-101. [PMID: 38770915 PMCID: PMC11186609 DOI: 10.61186/ibj.3971] [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: 07/03/2024] [Accepted: 04/03/2024] [Indexed: 05/22/2024]
Abstract
Background Synthetic and natural polymer scaffolds can be used to design wound dressing for repairing the damaged skin tissue. This study investigated acute wound healing process using a decellularized skin scaffold and mouse embryo fibroblast (MEF). Methods Mouse skin fragments were decellularized and evaluated by DNA content, toxicity, H&E staining, Raman confocal microscopy, Masson’s trichrome staining, SEM, and biodegradation assays. The fragments were recellularized by the MEFs, and cell attachment and penetration were studied. De- and decellularized scaffolds were used wound dressings in mouse acute wound models as two experimental groups. Using morphological and immunohistochemical assessments, wound healing was evaluated and compared among the experimental and control groups. Results DNA content of the decellularized tissue significantly reduced compared to the native control group (7% vs. 100%; p < 0.05). extracellular matrix components, e.g. collagen types I, III, and IV, elastin, and glycosaminoglycan, were well preserved in the decellularized group. The porosity and fiber arrangement in the stroma had a structure similar to normal skin tissue. A significant reduction in healing time was observed in the group treated with a decellularized scaffold. A thicker epidermis layer was observed in the recovered tissue in both experimental groups compared to the control group. Immunostaining showed a positive reaction for CD31 as an endothelial marker in both experimental groups, confirming new vascularization in these groups. Conclusion Using MEFs with decellularized skin as a wound dressing increases the rate of wound healing and also the formation of new capillaries. This system could be beneficial for clinical applications in the field of tissue engineering.
Collapse
Affiliation(s)
- Armaghan Gheytasvand
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Hamed Bagheri
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Shahram Pourbeyranvand
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojdeh Salehnia
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
21
|
Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci 2024; 25:2356. [PMID: 38397032 PMCID: PMC10889096 DOI: 10.3390/ijms25042356] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Adipose tissue (AT) is a large and important energy storage organ as well as an endocrine organ with a critical role in many processes. Additionally, AT is an enormous and easily accessible source of multipotent cell types used in our day for all types of tissue regeneration. The ability of adipose-derived stem cells (ADSCs) to differentiate into other types of cells, such as endothelial cells (ECs), vascular smooth muscle cells, or cardiomyocytes, is used in tissue engineering in order to promote/stimulate the process of angiogenesis. Being a key for future successful clinical applications, functional vascular networks in engineered tissue are targeted by numerous in vivo and ex vivo studies. The article reviews the angiogenic potential of ADSCs and explores their capacity in the field of tissue engineering (TE).
Collapse
Affiliation(s)
- Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Toronto, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada
| |
Collapse
|
22
|
Andleeb A, Khan H, Andleeb A, Khan M, Tariq M. Advances in Chronic Wound Management: From Conventional Treatment to Novel Therapies and Biological Dressings. Crit Rev Biomed Eng 2024; 52:29-62. [PMID: 38884212 DOI: 10.1615/critrevbiomedeng.2024053066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Chronic wounds can be classified as diabetic foot ulcers, pressure ulcers, or venous leg ulcers. Chronic wound management has become a threat to clinicians and constitutes a major healthcare burden. The healing process of chronic wounds requires many factors to work in concert to achieve optimal healing. Various treatment options, ranging from hypoxia to infection, have evolved considerably to address the challenges associated with chronic wound healing. The conventional and accelerating treatments for chronic wounds still represent an unmet medical need due to the complex pathophysiology of the chronic wound microenvironment. In clinical settings, traditional chronic wound care practices rely on nonspecific topical treatment, which can reduce pain and alleviate disease progression with varying levels of success but fail to completely cure the wounds. Conventional wound dressings, such as hydrocolloids, gauze, foams, and films, have also shown limited success for the treatment of chronic wounds and only act as a physical barrier and absorb wound exudates. Emerging advances in treatment approaches, including novel therapies (stem cells, microRNAs, and nanocarrier-based delivery systems) and multifunctional biological dressings, have been reported for chronic wound repair. This review summarizes the challenges offered by chronic wounds and discusses recent advancements in chronic wound treatment.
Collapse
Affiliation(s)
- Anisa Andleeb
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan
| | - Hamza Khan
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Aneeta Andleeb
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Centre for Biotechnology and Microbiology, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Tariq
- Department of Biotechnology, Mirpur University of Science and Technology, Mirpur, Azad Jammu and Kashmir, Pakistan
| |
Collapse
|
23
|
Mandakhbayar N, Ji Y, El-Fiqi A, Patel KD, Yoon DS, Dashnyam K, Bayaraa O, Jin G, Tsogtbaatar K, Kim TH, Lee JH, Kim HW. Double hits with bioactive nanozyme based on cobalt-doped nanoglass for acute and diabetic wound therapies through anti-inflammatory and pro-angiogenic functions. Bioact Mater 2024; 31:298-311. [PMID: 37637079 PMCID: PMC10458956 DOI: 10.1016/j.bioactmat.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/27/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023] Open
Abstract
Regeneration of pathological wounds, such as diabetic ulcers, poses a significant challenge in clinical settings, despite the widespread use of drugs. To overcome clinical side effects and complications, drug-free therapeutics need to be developed to promote angiogenesis while overcoming inflammation to restore regenerative events. This study presents a novel bioactive nanozyme based on cobalt-doped nanoglass (namely, CoNZ), which exhibits high enzymatic/catalytic activity while releasing therapeutic ions. Cobalt oxide "Co3O4" tiny crystallites produced in situ through a chemical reaction with H2O2 within CoNZ nanoparticles play a crucial role in scavenging ROS. Results showed that CoNZ-treatment to full-thickness skin wounds in mice significantly accelerated the healing process, promoting neovascularization, matrix deposition, and epithelial lining while reducing pro-inflammatory signs. Notably, CoNZ was highly effective in treating pathological wounds (streptozotocin-induced diabetic wounds). Rapid scavenging of ROS by CoNZ and down-regulation of pro-inflammatory markers while up-regulating tissue healing signs with proliferative cells and activated angiogenic factors contributed to the observed healing events. In vitro experiments involving CoNZ-cultures with macrophages and endothelial cells exposed to high glucose and ROS-generating conditions further confirmed the effectiveness of CoNZ. CoNZ-promoted angiogenesis was attributed to the release of cobalt ions, as evidenced by the comparable effects of CoNZ-extracted ionic medium in enhancing endothelial migration and tubule formation via activated HIF-1α. Finally, we compared the in vivo efficacy of CoNZ with the clinically-available drug deferoxamine. Results demonstrated that CoNZ was as effective as the drug in closing the diabetic wound, indicating the potential of CoNZ as a novel drug-free therapeutic approach.
Collapse
Affiliation(s)
- Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - YunSeong Ji
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ahmed El-Fiqi
- Glass Research Department, National Research Centre, Cairo, 12622, Egypt
| | - Kapil D. Patel
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- School of Cellular and Molecular Medicine (CMM), University of Bristol, Bristol, BS8 1TD United Kingdom
| | - Dong Suk Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomedical Science, Hwasung Medi-Science University, Hwaseong‑Si 18274, Gyeonggi‑Do, Republic of Korea
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Drug Research Institute, Mongolian University of Pharmaceutical Sciences, Ulaanbaatar 14250, Mongolia
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Drug Research Institute, Mongolian University of Pharmaceutical Sciences, Ulaanbaatar 14250, Mongolia
| | - Gangshi Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Khaliunsarnai Tsogtbaatar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Drug Research Institute, Mongolian University of Pharmaceutical Sciences, Ulaanbaatar 14250, Mongolia
| | - Tae-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- R&D Center, TE Bios, Osong, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
24
|
Xu CB, Xu Z, Yu C, Jiang Z. Hydrogel loaded with exosomes from Wharton 's Jelly-derived mesenchymal stem cells enhances wound healing in mice. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:766-776. [PMID: 38171508 PMCID: PMC10764182 DOI: 10.3724/zdxbyxb-2023-0316] [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: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVES To explore the effect of hydrogel loaded with exosomes from Wharton's Jelly-derived mesenchymal stem cell (WJMSC) on wound healing. METHODS Exosomes were extracted from WJMSC, and the morphology and size of WJMSC-derived exosomes (WEX) were analyzed by transmission electron microscopy and nanoparticle size analyzer, respectively. The surface markers CD9, CD81, and Calnexin of WEX were detected by Western blotting. Exosome-loaded alginate hydrogel (WEX-gel) was prepared; its morphology was studied by scanning electron microscope, and its rheological behavior was examined by a rheometer. The in vitro drug release performance of WEX-gel was investigated by BCA method. RAW264.7 cells were treated with alginate hydrogel, WEX and WEX-gel, respectively; and the expression of CD86 and CD206 in macrophages was detected by flow cytometry. A full-thickness skin wound model was established in mice; the model mice were randomly divided into blank control group, WEX control group and WEX-gel group, and PBS, WEX and WEX-gel were applied to the wound area of mice, respectively. On day 3, the skin tissue of mice was excised, and the antibacterial effect of WEX hydrogel was evaluated by plate counting. On day 15, the mice were euthanized and the percentage of residual wounds was calculated. The histological changes of the skin wound were observed after hematoxylin and eosin (HE) and Masson stainings. The expression of CD86, CD206, CD31 and vascular endothelial growth factor (VEGF) in the skin wound tissue was detected by immunohistochemistry. RESULTS Exosomes were successfully extracted from WJMSC. WEX-gel presented a regular three-dimensional network structure, good rheology and controlled drug release performance. WEX-gel promoted the polarization of RAW264.7 cells from the M1 phenotype to M2 phenotype in vitro. The residual wound percentage in blank control group, WEX control group and WEX-gel group were (27.5±3.4)%, (15.3±1.2)% and (7.6±1.1)%, respectively (P<0.05). The antibacterial property of WEX-gel is better than that of WEX (P<0.05). The dermis thickness, the number of new hair follicles, and the rate of collagen deposition in the WEX-gel group were significantly higher than those in the other two groups (all P<0.05). The expression of CD206, CD31 and VEGF in skin wound tissue was higher and the expression of CD86 was lower in WEX-gel group than those in other two groups (all P<0.05). CONCLUSIONS WEX-gel can significantly promote wound healing in mice by regulating the polarization of macrophages.
Collapse
Affiliation(s)
- Cui Bocheng Xu
- Department of Anorectal Surgery, Taizhou Central Hospital & Affiliated Hospital of Taizhou College, Taizhou 318000, Zhejiang Province, China.
| | - Zhengbao Xu
- Department of Anorectal Surgery, Taizhou Central Hospital & Affiliated Hospital of Taizhou College, Taizhou 318000, Zhejiang Province, China
| | - Chengyang Yu
- Department of Anorectal Surgery, Taizhou Central Hospital & Affiliated Hospital of Taizhou College, Taizhou 318000, Zhejiang Province, China
| | - Zufu Jiang
- Department of Anorectal Surgery, Taizhou Central Hospital & Affiliated Hospital of Taizhou College, Taizhou 318000, Zhejiang Province, China.
| |
Collapse
|
25
|
Zhang HM, Yang ML, Xi JZ, Yang GY, Wu QN. Mesenchymal stem cells-based drug delivery systems for diabetic foot ulcer: A review. World J Diabetes 2023; 14:1585-1602. [DOI: 10.4239/wjd.v14.i11.1585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/16/2023] [Accepted: 09/11/2023] [Indexed: 11/14/2023] Open
Abstract
The complication of diabetes, which is known as diabetic foot ulcer (DFU), is a significant concern due to its association with high rates of disability and mortality. It not only severely affects patients’ quality of life, but also imposes a substantial burden on the healthcare system. In spite of efforts made in clinical practice, treating DFU remains a challenging task. While mesenchymal stem cell (MSC) therapy has been extensively studied in treating DFU, the current efficacy of DFU healing using this method is still inadequate. However, in recent years, several MSCs-based drug delivery systems have emerged, which have shown to increase the efficacy of MSC therapy, especially in treating DFU. This review summarized the application of diverse MSCs-based drug delivery systems in treating DFU and suggested potential prospects for the future research.
Collapse
Affiliation(s)
- Hong-Min Zhang
- Department of Endocrinology, People’s Hospital of Chongqing Liangjiang New Area, Chongqing 400030, China
| | - Meng-Liu Yang
- Department of Endocrinology, The Second Affiliated Hospital of The Chongqing Medical University, Chongqing 400030, China
| | - Jia-Zhuang Xi
- Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, Chongqing 406230, China
| | - Gang-Yi Yang
- Department of Endocrinology, The Second Affiliated Hospital of The Chongqing Medical University, Chongqing 400030, China
| | - Qi-Nan Wu
- Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, Chongqing 406230, China
| |
Collapse
|
26
|
Ghassemi K, Inouye K, Takhmazyan T, Bonavida V, Yang JW, de Barros NR, Thankam FG. Engineered Vesicles and Hydrogel Technologies for Myocardial Regeneration. Gels 2023; 9:824. [PMID: 37888397 PMCID: PMC10606880 DOI: 10.3390/gels9100824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Increased prevalence of cardiovascular disease and potentially life-threatening complications of myocardial infarction (MI) has led to emerging therapeutic approaches focusing on myocardial regeneration and restoration of physiologic function following infarction. Extracellular vesicle (EV) technology has gained attention owing to the biological potential to modulate cellular immune responses and promote the repair of damaged tissue. Also, EVs are involved in local and distant cellular communication following damage and play an important role in initiating the repair process. Vesicles derived from stem cells and cardiomyocytes (CM) are of particular interest due to their ability to promote cell growth, proliferation, and angiogenesis following MI. Although a promising candidate for myocardial repair, EV technology is limited by the short retention time of vesicles and rapid elimination by the body. There have been several successful attempts to address this shortcoming, which includes hydrogel technology for the sustained bioavailability of EVs. This review discusses and summarizes current understanding regarding EV technology in the context of myocardial repair.
Collapse
Affiliation(s)
- Kaitlyn Ghassemi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Keiko Inouye
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Tatevik Takhmazyan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Victor Bonavida
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Finosh G. Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| |
Collapse
|
27
|
Andalib E, Kashfi M, Mahmoudvand G, Rezaei E, Mahjoor M, Torki A, Afkhami H. Application of hypoxia-mesenchymal stem cells in treatment of anaerobic bacterial wound infection: wound healing and infection recovery. Front Microbiol 2023; 14:1251956. [PMID: 37869672 PMCID: PMC10586055 DOI: 10.3389/fmicb.2023.1251956] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, are a type of multipotent stem cells that are typically extracted from adipose tissue and bone marrow. In the field of tissue engineering and regenerative medicine, MSCs and their exosomes have emerged as revolutionary tools. Researchers are now devoting greater attention to MSCs because of their ability to generate skin cells like fibroblasts and keratinocytes, as well as their distinctive potential to decrease inflammation and emit pro-angiogenic molecules at the site of wounds. More recent investigations revealed that MSCs can exert numerous direct and indirect antimicrobial effects that are immunologically mediated. Collectively, these antimicrobial properties can remove bacterial infections when the MSCs are delivered in a therapeutic setting. Regardless of the positive therapeutic potential of MSCs for a multitude of conditions, transplanted MSC cell retention continues to be a major challenge. Since MSCs are typically administered into naturally hypoxic tissues, understanding the impact of hypoxia on the functioning of MSCs is crucial. Hypoxia has been postulated to be among the factors determining the differentiation of MSCs, resulting in the production of inflammatory cytokines throughout the process of tissue regeneration and wound repair. This has opened new horizons in developing MSC-based systems as a potent therapeutic tool in oxygen-deprived regions, including anaerobic wound infection sites. This review sheds light on the role of hypoxia-MSCs in the treatment of anaerobic bacterial wound infection in terms of both their regenerative and antimicrobial activities.
Collapse
Affiliation(s)
- Elahe Andalib
- Department of Microbiology, School of Basic Sciences, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Elaheh Rezaei
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Torki
- Department of Medical Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
28
|
Vasalou V, Kotidis E, Tatsis D, Boulogeorgou K, Grivas I, Koliakos G, Cheva A, Ioannidis O, Tsingotjidou A, Angelopoulos S. The Effects of Tissue Healing Factors in Wound Repair Involving Absorbable Meshes: A Narrative Review. J Clin Med 2023; 12:5683. [PMID: 37685753 PMCID: PMC10488606 DOI: 10.3390/jcm12175683] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Wound healing is a complex and meticulously orchestrated process involving multiple phases and cellular interactions. This narrative review explores the intricate mechanisms behind wound healing, emphasizing the significance of cellular processes and molecular factors. The phases of wound healing are discussed, focusing on the roles of immune cells, growth factors, and extracellular matrix components. Cellular shape alterations driven by cytoskeletal modulation and the influence of the 'Formin' protein family are highlighted for their impact on wound healing processes. This review delves into the use of absorbable meshes in wound repair, discussing their categories and applications in different surgical scenarios. Interleukins (IL-2 and IL-6), CD31, CD34, platelet rich plasma (PRP), and adipose tissue-derived mesenchymal stem cells (ADSCs) are discussed in their respective roles in wound healing. The interactions between these factors and their potential synergies with absorbable meshes are explored, shedding light on how these combinations might enhance the healing process. Recent advances and challenges in the field are also presented, including insights into mesh integration, biocompatibility, infection prevention, and postoperative complications. This review underscores the importance of patient-specific factors and surgical techniques in optimizing mesh placement and healing outcomes. As wound healing remains a dynamic field, this narrative review provides a comprehensive overview of the current understanding and potential avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Varvara Vasalou
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
- Andreas Syggros Hospital, 11528 Athens, Greece
| | - Efstathios Kotidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Dimitris Tatsis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
- Oral and Maxillofacial Surgery Department, School of Dentistry, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Kassiani Boulogeorgou
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.B.)
| | - Ioannis Grivas
- Laboratory of Anatomy, Histology & Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Koliakos
- Department of Biochemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Angeliki Cheva
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.B.)
| | - Orestis Ioannidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology & Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Stamatis Angelopoulos
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| |
Collapse
|
29
|
Liu L, Yao S, Mao X, Fang Z, Yang C, Zhang Y. Thermosensitive hydrogel coupled with sodium ascorbyl phosphate promotes human umbilical cord-derived mesenchymal stem cell-mediated skin wound healing in mice. Sci Rep 2023; 13:11909. [PMID: 37488143 PMCID: PMC10366115 DOI: 10.1038/s41598-023-38666-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Poor survival and restricted function of transplanted stem cells are regarded as limiting their efficacy in wound recovery greatly. Consequently, it is necessary to identify innovative therapeutic strategies to solve these issues. Firstly, the biological effect of PF-127 hydrogel alone and in combination with SAP on the survival, and migration of cultured HUCMSCs was assessed by cell viability, apoptosis, and scratch wound assays. S. aureus and E. coli were used to evaluate the antibacterial activity of PF-127 plus SAP combination. Further, the ability of HUCMSCs-conditioned medium (HUCMSCs-CM) to promote the angiogenesis and migration of human umbilical vein endothelial cells (HUVECs) in vitro was evaluated using tube formation and transwell migration assays. Finally, the HUCMSCs embedded in PF-127 plus SAP scaffold were administered onto mice's excisional cutaneous wound bed. Histological and immunohistochemical analyses were employed to investigate the wound healing capacity as well as cellular responses of PF-127/HUCMSCs/SAP hydrogel. PF-127 showed cytotoxicity on HUCMSCs, whereas the addition of SAP significantly promoted cell viability and alleviated apoptosis of HUCMSCs encapsulated in PF-127 hydrogel in vitro. SAP supplementation substantially abrogated the inhibiting effect of PF-127 on the migration of HUCMSCs in vitro. The combination of PF-127 and SAP exerted an obvious bacteriostatic function on S. aureus and E. coli. Moreover, the co-treatment with SAP could remarkably enhance the stimulative effect of HUCMSCs-CM on the angiogenesis and migration of HUVECs in vitro. PF-127 combined SAP-embedded HUCMSCs transplantation resulted in a potently accelerated wound healing process, promoted the number of proliferating cells and newly formed blood vessels, as well as enhanced expression of vascular endothelial growth factor. PF-127 coupled with SAP contributes to HUCMSCs-mediated traumatic wound closure in mice by promoting cell survival, antibacterial action, and angiogenesis. Our results offered a theoretical foundation for the clinical treatment of traumatic skin defects.
Collapse
Affiliation(s)
- Liji Liu
- Department of Bone and Joint, The Central Hospital of Yueyang, Yueyang, 414020, China
| | - Sheng Yao
- Huarong County People's Hospital, Yueyang, 414207, China
| | - Xianhua Mao
- Yueyang Vocational and Technical College, Yueyang, 414000, China
| | - Zheng Fang
- Department of Bone and Joint, The Central Hospital of Yueyang, Yueyang, 414020, China
| | - Cheng Yang
- Department of Bone and Joint, The Central Hospital of Yueyang, Yueyang, 414020, China
| | - Yan Zhang
- Department of Bone and Joint, The Central Hospital of Yueyang, Yueyang, 414020, China.
| |
Collapse
|
30
|
Bashiri Z, Rajabi Fomeshi M, Ghasemi Hamidabadi H, Jafari D, Alizadeh S, Nazm Bojnordi M, Orive G, Dolatshahi-Pirouz A, Zahiri M, Reis RL, Kundu SC, Gholipourmalekabadi M. 3D-printed placental-derived bioinks for skin tissue regeneration with improved angiogenesis and wound healing properties. Mater Today Bio 2023; 20:100666. [PMID: 37273796 PMCID: PMC10239019 DOI: 10.1016/j.mtbio.2023.100666] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 06/06/2023] Open
Abstract
Extracellular matrix (ECM)-based bioinks has attracted much attention in recent years for 3D printing of native-like tissue constructs. Due to organ unavailability, human placental ECM can be an alternative source for the construction of 3D print composite scaffolds for the treatment of deep wounds. In this study, we use different concentrations (1.5%, 3% and 5%w/v) of ECM derived from the placenta, sodium-alginate and gelatin to prepare a printable bioink biomimicking natural skin. The printed hydrogels' morphology, physical structure, mechanical behavior, biocompatibility, and angiogenic property are investigated. The optimized ECM (5%w/v) 3D printed scaffold is applied on full-thickness wounds created in a mouse model. Due to their unique native-like structure, the ECM-based scaffolds provide a non-cytotoxic microenvironment for cell adhesion, infiltration, angiogenesis, and proliferation. In contrast, they do not show any sign of immune response to the host. Notably, the biodegradation, swelling rate, mechanical property, cell adhesion and angiogenesis properties increase with the increase of ECM concentrations in the construct. The ECM 3D printed scaffold implanted into deep wounds increases granulation tissue formation, angiogenesis, and re-epithelialization due to the presence of ECM components in the construct, when compared with printed scaffold with no ECM and no treatment wound. Overall, our findings demonstrate that the 5% ECM 3D scaffold supports the best deep wound regeneration in vivo, produces a skin replacement with a cellular structure comparable to native skin.
Collapse
Affiliation(s)
- Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Motahareh Rajabi Fomeshi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sanaz Alizadeh
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Nazm Bojnordi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01009, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029, Madrid, Spain
- University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007, Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore, 169856, Singapore
| | | | - Maria Zahiri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Department of Anatomical Sciences, School of Medical Sciences, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradable and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, Portugal
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Ma S, Yang Y, Mu Y, Peng H, Wei P, Jing W, Peng C, Liu X, Zhao B, Cai M, Liu Z, Yu X, Deng J. Modification of the small intestinal submucosa membrane with oligopeptides screened from intrinsically disordered regions to promote angiogenesis and accelerate wound healing. BIOMATERIALS ADVANCES 2023; 148:213360. [PMID: 36905827 DOI: 10.1016/j.bioadv.2023.213360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
A slow vascularization rate is considered one of the major disadvantages of biomaterials used for accelerating wound healing. Several efforts, including cellular and acellular technologies, have been made to facilitate biomaterial-induced angiogenesis. However, no well-established techniques for promoting angiogenesis have been reported. In this study, a small intestinal submucosa (SIS) membrane modified by an angiogenesis-promoting oligopeptide (QSHGPS) screened from intrinsically disordered regions (IDRs) of MHC class II was used to promote angiogenesis and accelerate wound healing. Because the main component of SIS membranes is collagen, the collagen-binding peptide sequence TKKTLRT and the pro-angiogenic oligopeptide sequence QSHGPS were used to construct chimeric peptides to obtain specific oligopeptide-loaded SIS membranes. The resulting chimeric peptide-modified SIS membranes (SIS-L-CP) significantly promoted the expression of angiogenesis-related factors in umbilical vein endothelial cells. Furthermore, SIS-L-CP exhibited excellent angiogenic and wound-healing abilities in a mouse hindlimb ischaemia model and a rat dorsal skin defect model. The high biocompatibility and angiogenic capacity of the SIS-L-CP membrane make it promising in angiogenesis- and wound healing-related regenerative medicine.
Collapse
Affiliation(s)
- Shiqing Ma
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yilin Yang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yuzhu Mu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Huizhen Peng
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing 102600, China
| | - Wei Jing
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing 102600, China; Foshan (Southern China) Institute for New Materials, Foshan 528220, China
| | - Cheng Peng
- Department of Stomatology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing 102600, China
| | - Mingxiang Cai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou 510630, China.
| | - Zihao Liu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Xueqiao Yu
- Beijing Biosis Healing Biological Technology Co., Ltd, Beijing 102600, China.
| | - Jiayin Deng
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
32
|
Chang SY, Lee JH, Oh SC, Lee MY, Lim NK. Human Fibroblast Growth Factor-Treated Adipose-Derived Stem Cells Facilitate Wound Healing and Revascularization in Rats with Streptozotocin-Induced Diabetes Mellitus. Cells 2023; 12:cells12081146. [PMID: 37190055 DOI: 10.3390/cells12081146] [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: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetes mellitus contributes to 15-25% of all chronic foot ulcers. Peripheral vascular disease is a cause of ischemic ulcers and exacerbates diabetic foot disease. Cell-based therapies are viable options to restore damaged vessels and induce the formation of new vessels. Adipose-derived stem cells (ADSCs) have the potential for angiogenesis and regeneration because of their greater paracrine effect. Preclinical studies are currently using other forced enhancement techniques (e.g., genetic modification or biomaterials) to increase the efficacy of human ADSC (hADSC) autotransplantation. Unlike genetic modifications and biomaterials, many growth factors have been approved by the equivalent regulatory authorities. This study confirmed the effect of enhanced human ADSC (ehADSC)s with a cocktail of FGF and other pharmacological agents to promote wound healing in diabetic foot disease. In vitro, ehADSCs exhibited a long and slender spindle-shaped morphology and showed significantly increased proliferation. In addition, it was shown that ehADSCs have more functionalities in oxidative stress toleration, stem cell stemness, and mobility. In vivo, the local transplantation of 1.2 × 106 hADSCs or ehADSCs was performed in animals with diabetes induced by STZ. The ehADSC group showed a statistically decreased wound size and increased blood flow compared with the hADSC group and the sham group. Human Nucleus Antigen (HNA) positive cells were observed in some ADSC-transplanted animals. The ehADSC group showed a relatively higher portion of HNA-positive animals than the hADSC group. The blood glucose levels showed no significant difference among the groups. In conclusion, the ehADSCs showed a better performance in vitro, compared with conventional hADSCs. Additionally, a topical injection of ehADSCs into diabetic wounds enhanced wound healing and blood flow, while improving histological markers suggesting revascularization.
Collapse
Affiliation(s)
- So-Young Chang
- Beckman Laser Institute Korea, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
| | - Se Cheol Oh
- Stem Cell R&D Center, N-BIOTEK Inc., Bucheon 14449, Republic of Korea
| | - Min Young Lee
- Beckman Laser Institute Korea, Dankook University, Cheonan 31116, Republic of Korea
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Nam Kyu Lim
- Department of Plastic and Reconstructive surgery, College of medicine, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
33
|
Fu S, Zhang H, Li X, Zhang Q, Guo C, Qiu K, Feng J, Liu X, Liu D. Exosomes Derived from Human Amniotic Mesenchymal Stem Cells Facilitate Diabetic Wound Healing by Angiogenesis and Enrich Multiple lncRNAs. Tissue Eng Regen Med 2023; 20:295-308. [PMID: 36696086 PMCID: PMC10070558 DOI: 10.1007/s13770-022-00513-w] [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: 11/10/2022] [Revised: 11/29/2022] [Accepted: 12/18/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Diabetic wound healing remains a major challenge due to the impaired functionality of angiogenesis by persistent hyperglycemia. Mesenchymal stem cell exosomes are appropriate candidates for regulating the formation of angiogenesis in tissue repair and regeneration. Here, we explored the effects of exosomes derived from human amniotic mesenchymal stem cell (hAMSC-Exos) on the biological activities of human umbilical vein endothelial cells (HUVECs) treated with high glucose and on diabetic wound healing and investigate lncRNAs related to angiogenesis in hAMSC-Exos. METHODS hAMSCs and hAMSC-Exos were isolated and identified by flow cytometry or western blot. A series of functional assays such as cell counting kit-8, scratching, transwell and tube formation assays were performed to evaluate the potential effect of hAMSC-Exos on high glucose-treated HUVECs. The effect of hAMSC-Exos on diabetic wound healing were tested by measuring wound closure rates and immunohistochemical staining of CD31. Subsequently, the lncRNAs profiles in hAMSC-Exos and hAMSCs were examined to screen the lncRNAs related to angiogenesis. RESULTS The isolated hAMSC-Exos had a size range of 30-150 nm and were positive for CD9, CD63 and CD81. The hAMSC-Exos facilitate the functional properties of high glucose-treated HUVECs including the proliferation, migration and the angiogenic activities as well as wound closure and angiogenesis in diabetic wound. hAMSC-Exos were enriched lncRNAs that related to angiogenesis, including PANTR1, H19, OIP5-AS1 and NR2F1-AS1. CONCLUSION Our findings demonstrated hAMSC-Exos facilitate diabetic wound healing by angiogenesis and contain several exosomal lncRNAs related to angiogenesis, which may represent a promising strategy for diabetic wound healing.
Collapse
Affiliation(s)
- Shangfeng Fu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Hongyan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xiancai Li
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Qiling Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Chunyan Guo
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Keqing Qiu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Junyun Feng
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xiaoxiao Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
34
|
Li M, Cheng X, Feng S, Zhu H, Lu P, Zhang P, Cai X, Qiao P, Gu X, Wang G, Xue C, Wang H. Skin precursor‐derived Schwann cells accelerate in vivo prevascularization of tissue‐engineered nerves to promote peripheral nerve regeneration. Glia 2023; 71:1755-1769. [PMID: 36971489 DOI: 10.1002/glia.24367] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Prevascularization strategies have become a hot spot in tissue engineering. As one of the potential candidates for seed cells, skin precursor-derived Schwann cells (SKP-SCs) were endowed with a new role to more efficiently construct prevascularized tissue-engineered peripheral nerves. The silk fibroin scaffolds seeded with SKP-SCs were prevascularized through subcutaneously implantation, which was further assembled with the SKP-SC-containing chitosan conduit. SKP-SCs expressed pro-angiogenic factors in vitro and in vivo. SKP-SCs significantly accelerated the satisfied prevascularization in vivo of silk fibroin scaffolds compared with VEGF. Moreover, the NGF expression revealed that pregenerated blood vessels adapted to the nerve regeneration microenvironment through reeducation. The short-term nerve regeneration of SKP-SCs-prevascularization was obviously superior to that of non-prevascularization. At 12 weeks postinjury, both SKP-SCs-prevascularization and VEGF-prevascularization significantly improved nerve regeneration with a comparable degree. Our figures provide a new enlightenment for the optimization of prevascularization strategies and how to further utilize tissue engineering for better repair.
Collapse
Affiliation(s)
- Meiyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiyang Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Shuyue Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Hui Zhu
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Panjian Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Ping Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaodong Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Pingping Qiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Gang Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Chengbin Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
35
|
Tiwari R, Pathak K. Local Drug Delivery Strategies towards Wound Healing. Pharmaceutics 2023; 15:pharmaceutics15020634. [PMID: 36839956 PMCID: PMC9964694 DOI: 10.3390/pharmaceutics15020634] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/30/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
A particular biological process known as wound healing is connected to the overall phenomena of growth and tissue regeneration. Several cellular and matrix elements work together to restore the integrity of injured tissue. The goal of the present review paper focused on the physiology of wound healing, medications used to treat wound healing, and local drug delivery systems for possible skin wound therapy. The capacity of the skin to heal a wound is the result of a highly intricate process that involves several different processes, such as vascular response, blood coagulation, fibrin network creation, re-epithelialisation, collagen maturation, and connective tissue remodelling. Wound healing may be controlled with topical antiseptics, topical antibiotics, herbal remedies, and cellular initiators. In order to effectively eradicate infections and shorten the healing process, contemporary antimicrobial treatments that include antibiotics or antiseptics must be investigated. A variety of delivery systems were described, including innovative delivery systems, hydrogels, microspheres, gold and silver nanoparticles, vesicles, emulsifying systems, nanofibres, artificial dressings, three-dimensional printed skin replacements, dendrimers and carbon nanotubes. It may be inferred that enhanced local delivery methods might be used to provide wound healing agents for faster healing of skin wounds.
Collapse
Affiliation(s)
- Ruchi Tiwari
- Pranveer Singh Institute of Technology (Pharmacy), Kanpur 208020, Uttar Pradesh, India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Etawah 206130, Uttar Pradesh, India
- Correspondence:
| |
Collapse
|
36
|
Wound healing approach based on excretory-secretory product and lysate of liver flukes. Sci Rep 2022; 12:21639. [PMID: 36517588 PMCID: PMC9751068 DOI: 10.1038/s41598-022-26275-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Exogenous bioactive peptides are considered promising for the wound healing therapy in humans. In this regard, parasitic trematodes proteins may potentially become a new perspective agents. Foodborne trematode Opisthorchis felineus is widespread in Europe and has the ability to stimulate proliferation of bile duct epithelium. In this study, we investigated skin wound healing potential of O. felineus proteins in mouse model. C57Bl/6 mice were inflicted with superficial wounds with 8 mm diameter. Experimental groups included several non-specific controls and specific treatment groups (excretory-secretory product and lysate). After 10 days of the experiment, the percentage of wound healing in the specific treatment groups significantly exceeded the control values. We also found that wound treatment with excretory-secretory product and worm lysate resulted in: (i) inflammation reducing, (ii) vascular response modulating, (iii) type 1 collagen deposition promoting dermal ECM remodeling. An additional proteomic analysis of excretory-secretory product and worm lysate samples was revealed 111 common proteins. The obtained data indicate a high wound-healing potential of liver fluke proteins and open prospects for further research as new therapeutic approaches.
Collapse
|
37
|
Liu H, Wu B, Shi X, Cao Y, Zhao X, Liang D, Qin Q, Liang X, Lu W, Wang D, Liu J. Aerobic exercise-induced circulating extracellular vesicle combined decellularized dermal matrix hydrogel facilitates diabetic wound healing by promoting angiogenesis. Front Bioeng Biotechnol 2022; 10:903779. [PMID: 36082169 PMCID: PMC9445842 DOI: 10.3389/fbioe.2022.903779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Insufficient blood supply results in unsatisfactory wound healing, especially for challenging wound repair such as diabetic wound defects. Regular exercise training brings a lot of benefits to cardiovascular fitness and metabolic health including attenuation of T2DM progression. Circulating extracellular vesicles (EVs) are postulated to carry a variety of signals involved in tissue crosstalk by their modified cargoes, representing novel mechanisms for the effects of exercise. Prominently, both acute and chronic aerobic exercise training can promote the release of exercise-induced cytokines and enhance the angiogenic function of circulating angiogenic cell–derived EVs.Methods: We investigated the possible angiogenesis potential of aerobic exercise-induced circulating EVs (EXE-EVs) on diabetic wound healing. Circulating EVs were isolated from the plasma of rats subjected to 4 weeks of moderate aerobic exercise or sedentariness 24 h after the last training session. The therapeutic effect of circulating EVs was evaluated in vitro by proliferation, migration, and tube formation assays of human umbilical vein endothelial cells (HUVECs), as well as in vivo by quantification of angiogenesis and cutaneous wound healing in diabetic rats.Results: The number of circulating EVs did not change significantly in exercised rats 24 h post-exercise in comparison with the sedentary rats. Nevertheless, EXE-EVs showed remarkable pro-angiogenic effect by augmenting proliferation, migration, and tube formation of HUVECs. Furthermore, the findings of animal experiments revealed that the EXE-EVs delivered by decellularized dermal matrix hydrogel (DDMH) could significantly promote the repair of skin defects through stimulating the regeneration of vascularized skin.Discussion: The present study is the first attempt to demonstrate that aerobic exercise-induced circulating EVs could be utilized as a cell-free therapy to activate angiogenesis and promote diabetic wound healing. Our findings suggest that EXE-EVs may stand for a potential strategy for diabetic soft tissue wound repair.
Collapse
Affiliation(s)
- Haifeng Liu
- Guangzhou Medical University, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Bing Wu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xin Shi
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yanpeng Cao
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Xin Zhao
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Daqiang Liang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Qihuang Qin
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xinzhi Liang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Wei Lu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Daping Wang
- Guangzhou Medical University, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- *Correspondence: Daping Wang, ; Jun Liu,
| | - Jun Liu
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
- *Correspondence: Daping Wang, ; Jun Liu,
| |
Collapse
|