1
|
Sengul T, Kirkland-Kyhn H, Gul A. Postacute Overview of Burn Injuries: Pathophysiology, Management, and Future Directions. Nurs Clin North Am 2025; 60:15-25. [PMID: 39884787 DOI: 10.1016/j.cnur.2024.08.009] [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: 02/01/2025]
Abstract
This article provides a comprehensive overview of the pathophysiology, treatment methods, outpatient rehabilitation, and future directions related to burn injuries. It details the classification of burns, treatment strategies, wound care management, and rehabilitation processes. Modern approaches, such as AI-supported models and telemedicine, are highlighted in burn treatment. The article aims to present evidence-based best practices in burn care to healthcare professionals, offering insights into future research areas. Emphasis is placed on how recent innovations and technological advancements in burn management can improve patient outcomes.
Collapse
Affiliation(s)
- Tuba Sengul
- Koç University School of Nursing, Davutpaşa Street No: 4, Topkapı 34010, Istanbul, Turkey.
| | | | - Asiye Gul
- Nursing Department, Faculty of Health Science, Istanbul Kültür University, Istanbul, Turkey
| |
Collapse
|
2
|
Razavi ZS, Aliniay Sharafshadehi S, Yousefi MH, Javaheri F, Rahimi Barghani MR, Afkhami H, Heidari F. Application of novel strategies in chronic wound management with focusing on pressure ulcers: new perspective. Arch Dermatol Res 2025; 317:320. [PMID: 39888392 DOI: 10.1007/s00403-024-03790-8] [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: 11/10/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 02/01/2025]
Abstract
Invading blood cells, extracellular tissue, and soluble mediators all play important roles in the wound-healing process. There is a substantial global burden of disease and mortality attributable to skin defects that do not heal. About 1% to 2% of the population in industrialized nations suffers from chronic wounds that don't heal, despite healthcare breakthroughs; this condition is very costly, costing about $25 billion each year in the US alone. Amputation, infection (affecting as many as 25% of chronic wounds), sepsis, and dermal replacements are all consequences of conventional therapeutic approaches like growth factor therapy and diabetic foot ulcers account for 85% of lower limb amputations. Despite these obstacles, scientists are constantly looking for new ways to speed healing and close wounds. The unique immunomodulatory capabilities and multipotency of mesenchymal stem cells (MSCs) have made them a potential therapeutic choice in tissue engineering and regenerative medicine. Animal models of wound healing have shown that MSCs can speed up the process by as much as 40% through enhancing angiogenesis, modulating inflammation, and promoting fibroblast migration. Clinical trials provide more evidence of their effectiveness; for instance, one RCT found that, after 12 weeks, patients treated with MSCs had a 72% smaller wound size than those in the control group. This review offers a thorough examination of MSCs by combining the latest research with preclinical evidence. Highlighting their potential to transform treatment paradigms, it delves into their biological properties, how they work during regeneration and healing, and therapeutic usefulness in controlling chronic wounds.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Aliniay Sharafshadehi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Fatemeh Javaheri
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | | | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
3
|
Szunerits S, Chuang EY, Yang JC, Boukherroub R, Burnouf T. Platelet extracellular vesicles-loaded hydrogel bandages for personalized wound care. Trends Biotechnol 2025:S0167-7799(24)00393-7. [PMID: 39863439 DOI: 10.1016/j.tibtech.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025]
Abstract
Autologous or allogeneic platelet-derived extracellular vesicles (pEVs) show potential in enhancing tissue recovery and healing chronic wounds. pEVs promote neovascularization and cell migration while reducing inflammation, oxidative stress, and scarring. However, their efficacy in clinical settings is challenged by their susceptibility to washout by wound exudate. Hydrogel-based bandages are effective carriers that stabilize pEVs for optimal personalized wound care. These bandages can be tailored for easy removal to minimize damage to regenerated tissue and can incorporate antibacterial or moisture-retaining properties. Furthermore, the possibility of integrating sensors in the wound bed will enable a theragnostic approach to healing. This review explores advancements in pEV-loaded hydrogels and their potential for personalized clinical applications.
Collapse
Affiliation(s)
- Sabine Szunerits
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France; Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria.
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan
| | - Jen-Chang Yang
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei City 235603, Taiwan; International PhD Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
4
|
Jain S, Bhatt J, Gupta S, Bhatia DD. Nanotechnology at the crossroads of stem cell medicine. Biomater Sci 2024; 13:161-178. [PMID: 39584588 DOI: 10.1039/d4bm01257g] [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: 11/26/2024]
Abstract
Nanotechnology in stem cell medicine is an interdisciplinary field which has gained a lot of interest recently. This domain addresses key challenges associated with stem cell medicine such as cell isolation, targeted delivery, and tracking. Nanotechnology-based approaches, including magnetic cell sorting, fluorescent tagging, and drug or biomolecule conjugation for delivery, have enhanced precision in stem cell isolation and guided cell migration, increasing the therapeutic potential. Recent studies have focused on using nanomaterials and scaffolds to drive stem cell differentiation by activating specific molecular pathways, achieved through embedding biomolecules within the scaffold or through the scaffold's material composition and structure alone. These innovations hold promise in therapeutic applications across various diseases, including cancer stem cell targeting, neurodegenerative disorders, pre-eclampsia, cardiovascular conditions, and organoid development. This review examines recent advancements in the field, explores potential applications like biosensors and nanochips, and highlights the challenges and research gaps.
Collapse
Affiliation(s)
- Sweny Jain
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Jay Bhatt
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Dhiraj Devidas Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| |
Collapse
|
5
|
Zhu Y, Mei O, Zhang H, You W, Zhong J, Collins CP, Shen G, Luo C, Wu X, Li J, Shu Y, Wen Y, Luu HH, Shi LL, Fan J, He TC, Ameer GA, Sun C, Wen L, Reid RR. Establishment and characterization of a rat model of scalp-cranial composite defect for multilayered tissue engineering. RESEARCH SQUARE 2024:rs.3.rs-4643966. [PMID: 39108474 PMCID: PMC11302684 DOI: 10.21203/rs.3.rs-4643966/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Composite cranial defects have individual functional and aesthetic ramifications, as well as societal burden, while posing significant challenges for reconstructive surgeons. Single-stage composite reconstruction of these deformities entail complex surgeries that bear many short- and long-term risks and complications. Current research on composite scalp-cranial defects is sparse and one-dimensional, often focusing solely on bone or skin. Thus, there is an unmet need for a simple, clinically relevant composite defect model in rodents, where there is a challenge in averting healing of the skin component via secondary intention. By utilizing a customizable (3D-printed) wound obturator, the scalp wound can be rendered non-healing for a long period (more than 6 weeks), with the cranial defect patent. The wound obturator shows minimal biotoxicity and will not cause severe endocranium-granulation adhesion. This composite defect model effectively slowed the scalp healing process and preserved the cranial defect, embodying the characteristics of a "chronic composite defect". In parallel, an autologous reconstruction model was established as the positive control. This positive control exhibited reproducible healing of the skin within 3 weeks with variable degrees of osseointegration, consistent with clinical practice. Both models provide a stable platform for subsequent research not only for composite tissue engineering and scaffold design but also for mechanistic studies of composite tissue healing.
Collapse
Affiliation(s)
- Yi Zhu
- The University of Chicago Medical Center
| | - Ou Mei
- The University of Chicago Medical Center
| | - Hui Zhang
- The University of Chicago Medical Center
| | - Wulin You
- The University of Chicago Medical Center
| | | | | | | | | | - Xingye Wu
- The University of Chicago Medical Center
| | | | - Yi Shu
- The University of Chicago Medical Center
| | - Ya Wen
- Capital Medical University
| | - Hue H Luu
- The University of Chicago Medical Center
| | | | | | | | | | | | - Liangyuan Wen
- Chinese Academy of Medical Sciences & Peking Union Medical College
| | | |
Collapse
|
6
|
Ramzan F, Khalid S, Ekram S, Salim A, Frazier T, Begum S, Mohiuddin OA, Khan I. 3D bio scaffold support osteogenic differentiation of mesenchymal stem cells. Cell Biol Int 2024; 48:594-609. [PMID: 38321826 DOI: 10.1002/cbin.12131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
The regeneration of osteochondral lesions by tissue engineering techniques is challenging due to the lack of physicochemical characteristics and dual-lineage (osteogenesis and chondrogenesis). A scaffold with better mechanical properties and dual lineage capability is required for the regeneration of osteochondral defects. In this study, a hydrogel prepared from decellularized human umbilical cord tissue was developed and evaluated for osteochondral regeneration. Mesenchymal stem cells (MSCs) isolated from the umbilical cord were seeded with hydrogel for 28 days, and cell-hydrogel composites were cultured in basal and osteogenic media. Alizarin red staining, quantitative polymerase chain reaction, and immunofluorescent staining were used to confirm that the hydrogel was biocompatible and capable of inducing osteogenic differentiation in umbilical cord-derived MSCs. The findings demonstrate that human MSCs differentiated into an osteogenic lineage following 28 days of cultivation in basal and osteoinductive media. The expression was higher in the cell-hydrogel composites cultured in osteoinductive media, as evidenced by increased levels of messenger RNA and protein expression of osteogenic markers as compared to basal media cultured cell-hydrogel composites. Additionally, calcium deposits were also observed, which provide additional evidence of osteogenic differentiation. The findings demonstrate that the hydrogel is biocompatible with MSCs and possesses osteoinductive capability in vitro. It may be potentially useful for osteochondral regeneration.
Collapse
Affiliation(s)
- Faiza Ramzan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shumaila Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Sobia Ekram
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Sumreen Begum
- Stem Cell Research Laboratory (SCRL), Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Omair A Mohiuddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
7
|
Zhang G, Samarawickrama PN, Gui L, Ma Y, Cao M, Zhu H, Li W, Yang H, Li K, Yang Y, Zhu E, Li W, He Y. Revolutionizing Diabetic Foot Ulcer Care: The Senotherapeutic Approach. Aging Dis 2024:AD.2024.0065. [PMID: 38739931 DOI: 10.14336/ad.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are a prevalent and profoundly debilitating complication that afflicts individuals with diabetes mellitus (DM). These ulcers are associated with substantial morbidity, recurrence rates, disability, and mortality, imposing substantial economic, psychological, and medical burdens. Timely detection and intervention can mitigate the morbidity and disparities linked to DFU. Nevertheless, current therapeutic approaches for DFU continue to grapple with multifaceted limitations. A growing body of evidence emphasizes the crucial role of cellular senescence in the pathogenesis of chronic wounds. Interventions that try to delay cellular senescence, eliminate senescent cells (SnCs), or suppress the senescence-associated secretory phenotype (SASP) have shown promise for helping chronic wounds to heal. In this context, targeting cellular senescence emerges as a novel therapeutic strategy for DFU. In this comprehensive review, we look at the pathology and treatment of DFU in a systematic way. We also explain the growing importance of investigating SnCs in DFU and highlight the great potential of senotherapeutics that target SnCs in DFU treatment. The development of efficacious and safe senotherapeutics represents a pioneering therapeutic approach aimed at enhancing the quality of life for individuals affected by DFU.
Collapse
Affiliation(s)
- Guiqin Zhang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Li Gui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yuan Ma
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Mei Cao
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Hong Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Wei Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Honglin Yang
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Kecheng Li
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Yang Yang
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Enfang Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Wen Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| |
Collapse
|
8
|
Burgos-Gutiérrez C, Álvarez-Buylla-Álvarez P, Álvarez-Viejo M, Pérez-López S, Pérez-Basterrechea M, Bea-Muñoz M, Pérez-Arias Á, De-Vicente-Rodríguez JC. Treatment of pressure ulcers in patients with spinal cord injury: Conventional surgery vs. cellular therapy. J Spinal Cord Med 2024; 47:246-254. [PMID: 34982655 PMCID: PMC10885747 DOI: 10.1080/10790268.2021.2014234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
CONTEXT Relapse and recurrence rates of pressure injuries (PIs) are very high in spinal cord injured patients. That is the reason why alternative therapies, such the stem cells derived from bone marrow, have been developed. OBJECTIVE To compare this new technique of infiltration-infusion of mononuclear cells from bone marrow with conventional surgery. DESIGN A retrospective study was carried out in patients with spinal cord injuries who had PIs, category III/IV, in the pelvic area, during a 14-year follow-up period. SETTING One group was treated with conventional surgery and, in the other group, mononuclear cells were infused. PARTICIPANTS One hundred and forty-nine patients were registered, 63 (42.3%) in the conventional surgery group and 86 (57.7%) in the mononuclear cell group. RESULTS A comparative study between these 2 groups was carried out. There were no significant differences in ulcer healing in the first 6 months, but 6 months and one-year post-treatment, they were found. At 6 months, no patient in the conventional surgery group showed dehiscence or fistulization of the wound and, one year after surgery, only 3.17% recurred in the conventional group. In addition, there was a statistically significant relationship between days of hospitalization and the type of bacterial contamination and the intervention group. CONCLUSION Bone marrow mononuclear cell infusion-infiltration is an alternative treatment for PIs and fistula during the first 6 months, instead of conventional surgery. However, in the medium-long term, conventional surgery is more effective.
Collapse
Affiliation(s)
| | | | - María Álvarez-Viejo
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Silvia Pérez-López
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marcos Pérez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Manuel Bea-Muñoz
- Servicio de Rehabilitación, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ángel Pérez-Arias
- Servicio de Cirugía Plástica, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | |
Collapse
|
9
|
Jia S, Wang X, Wang G, Wang X. Mechanism and application of β-adrenoceptor blockers in soft tissue wound healing. Med Res Rev 2024; 44:422-452. [PMID: 37470332 DOI: 10.1002/med.21984] [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: 07/03/2022] [Revised: 06/01/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
Soft tissue damage stimulates sympathetic nerves to release large amounts of catecholamine hormones which bind to β-adrenergic receptors (β-ARs) on the cell membrane surface. It activates the downstream effector molecules and impairs soft tissue wound healing. β-blockers specifically inhibit β-ARs activation in acute/chronic skin lesions and ulcerative hemangiomas. They also accelerate soft tissue wound healing by shortening the duration of inflammation, speeding keratinocyte migration and reepithelialization, promoting wound contraction and angiogenesis, and inhibiting bacterial virulence effects. In addition, β-blockers shorten wound healing periods in patients with severe thermal damage by reducing the hypermetabolic response. While β-blockers promote/inhibit corneal epithelial cell regeneration and restores limbal stem/progenitor cells function, it could well accelerate/delay corneal wound healing. Given these meaningful effects, a growing number of studies are focused on examining the efficacy and safety of β-blockers in soft tissue wound repair, including acute and chronic wounds, severe thermal damage, ulcerated infantile hemangioma, corneal wounds, and other soft tissue disorders. However, an intensive investigation on their acting mechanisms is imperatively needed. The purpose of this article is to summerize the roles of β-blockers in soft tissue wound healing and explore their clinical applications.
Collapse
Affiliation(s)
- Shasha Jia
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Xueya Wang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Guowei Wang
- Department of Stomatology, No. 971 Hospital of the Chinese Navy, Qingdao, Shandong, People's Republic of China
| | - Xiaojing Wang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
10
|
Zawrzykraj M, Deptuła M, Kondej K, Tymińska A, Pikuła M. The effect of chemotherapy and radiotherapy on stem cells and wound healing. Current perspectives and challenges for cell-based therapies. Biomed Pharmacother 2023; 168:115781. [PMID: 39491418 DOI: 10.1016/j.biopha.2023.115781] [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: 08/30/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/05/2024] Open
Abstract
Cancers are part of the group of diseases that carry a high mortality rate. According to World Health Organization in 2020 reported 10 million deaths due to cancers. Treatment of oncological patients is focused on chemotherapeutic agents, radiology, or immunology. Surgical interventions are also an important aspect of treatment. The above methods contribute to saving the patients' health and lives. However, cancer treatment possesses side effects. Commonly observed complications are hair loss, mucositis, nausea, diarrhea, or various skin damage. To improve the quality of medical care for cancer patients, new methods of reducing side effects are sought. Strategies include the use of stem cells (SCs). Due to unlimited proliferation potential and differentiating abilities, SCs are used in the treatment of many disease entities, including wounds. One of the most used types of stem cells supposed adipose-derived mesenchymal stromal cells (AD-MSCs). Clinical trials confirm the application of AD-MSCs in wound healing. Furthermore, in vivo studies considered the utilization of AD-MSCs in radiation injury. The use of stem cells in cancer treatment still involves many questions, such as the impact of treatment on SCs' condition and oncological safety. However, development in regenerative medicine research may contribute to the use of stem cells in personalized medicine, customized for the patient. This could represent a breakthrough step in preventing the side effects of cancer therapies, including chronic wounds.
Collapse
Affiliation(s)
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdansk, Poland
| | - Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland.
| |
Collapse
|
11
|
Mahmoudi M, Alizadeh P, Soltani M. Wound healing performance of electrospun PVA/70S30C bioactive glass/Ag nanoparticles mats decorated with curcumin: In vitro and in vivo investigations. BIOMATERIALS ADVANCES 2023; 153:213530. [PMID: 37356283 DOI: 10.1016/j.bioadv.2023.213530] [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: 01/27/2023] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 06/27/2023]
Abstract
Biocompatible fibrous scaffold containing polyvinyl alcohol (PVA), 70S30C bioactive glass (BG), silver (Ag) nanoparticles and curcumin (Cur) was fabricated through electrospinning method. Scanning electron microscope (SEM) and Field emission scanning electron microscopy (FESEM) were employed to investigate the morphological characteristics of the scaffolds. In addition, biodegradability, hydrophilicity, and contact angle were studied as criteria for evaluating physical properties of the scaffolds. Tensile strength was reported to be 0.971 ± 0.093 MPa. Also, the viability of fibroblasts after 7 days of cell culture was 93.58 ± 1.36 %. The antibacterial activity against Escherichia coli and Staphylococcus aureus bacteria was illustrated using inhibition zones of 13.12 ± 0.69 and 14.21 ± 1.37 mm, respectively. Histological results revealed that tissue regeneration after 14 days of surgery was much higher for the dressing group compared to the blank group. According to the obtained results, the authors introduce the PVA-BG-Ag-Cur scaffold as a promising candidate for skin tissue engineering applications.
Collapse
Affiliation(s)
- Masoud Mahmoudi
- Department of Materials Science and Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran
| | - Parvin Alizadeh
- Department of Materials Science and Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran.
| | - Mohammad Soltani
- Department of Materials Science and Engineering, Faculty of Engineering & Technology, Tarbiat Modares University, P. O. Box: 14115-143, Tehran, Iran
| |
Collapse
|
12
|
Ozel C, Apaydin E, Sariboyaci AE, Tamayol A, Avci H. A multifunctional sateen woven dressings for treatment of skin injuries. Colloids Surf B Biointerfaces 2023; 224:113197. [PMID: 36822118 DOI: 10.1016/j.colsurfb.2023.113197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cutaneous wounds with impaired healing such as diabetic ulcers and burns constitute major and rapidly growing threat to healthcare systems worldwide. Accelerating wound healing requires the delivery of biological factors that induce angiogenesis, support cellular proliferation, and modulate inflammation while minimizing infection. In this study, we engineered a dressing made by weaving of composite fibers (CFs) carrying mesenchymal stem cells (MSCs) and a model antibiotic using a scalable sateen textile technique. In this regard, two different sets of CFs carrying MSCs or an antimicrobial agent were used to generate a multifunctional dressing. According to cell viability and metabolic activity as CCK-8 and live/dead with qRT-PCR results, more than %90 the encapsulated MSCs remain viable for 28 days and their expression levels of the wound repair factors including ECM remodeling, angiogenesis and immunomodulatory maintained in MSCs post dressing manufacturing for 14 days. Post 10 days culture of the dressing, MSCs within CFs had 10-fold higher collagen synthesis (p < 0.0001) determined by hydroxyproline assay which indicates the enhanced healing properties. According to in vitro antimicrobial activity results determined by disk diffusion and broth microdilution tests, the first day and the total amount of release gentamicin loaded dressing samples during the 28 days were higher than determined minimal inhibition concentration (MIC) values for S. aureus and K. pneumonia without negatively impacting the viability and functionality of encapsulated MSCs within the dressing. The dressing is also flexible and can conform to skin curvatures making the dressing suitable for the treatment of different skin injuries such as burns and diabetic ulcers.
Collapse
Affiliation(s)
- Ceren Ozel
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Elif Apaydin
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskişehir 26470, Turkey
| | - Ayla Eker Sariboyaci
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06269, USA.
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey; Translational Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey.
| |
Collapse
|
13
|
Microbiological Aspects of Pharmaceutical Manufacturing of Adipose-Derived Stem Cell-Based Medicinal Products. Cells 2023; 12:cells12050680. [PMID: 36899816 PMCID: PMC10000438 DOI: 10.3390/cells12050680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Subcutaneous adipose tissue is an excellent source of mesenchymal stem cells (ADSCs), which can be used in cell therapies as an active substance in advanced therapy medicinal products (ATMPs). Because of the short shelf-life of ATMPs and the time needed to obtain the results of microbiological analysis, the final product is often administered to the patient before sterility is confirmed. Because the tissue used for cell isolation is not sterilized to maintain cell viability, controlling and ensuring microbiological purity at all stages of production is crucial. This study presents the results of monitoring the contamination incidence during ADSC-based ATMP manufacturing over two years. It was found that more than 40% of lipoaspirates were contaminated with thirteen different microorganisms, which were identified as being physiological flora from human skin. Such contamination was successfully eliminated from the final ATMPs through the implementation of additional microbiological monitoring and decontamination steps at various stages of production. Environmental monitoring revealed incidental bacterial or fungal growth, which did not result in any product contamination and was reduced thanks to an effective quality assurance system. To conclude, the tissue used for ADSC-based ATMP manufacturing should be considered contaminated; therefore, good manufacturing practices specific to this type of product must be elaborated and implemented by the manufacturer and the clinic in order to obtain a sterile product.
Collapse
|
14
|
Wang J, Fu S, Li H, Wu Y. A CS-based composite scaffold with excellent photothermal effect and its application in full-thickness skin wound healing. Regen Biomater 2023; 10:rbad028. [PMID: 37091498 PMCID: PMC10118997 DOI: 10.1093/rb/rbad028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/29/2023] [Accepted: 02/19/2023] [Indexed: 04/25/2023] Open
Abstract
The development of natural polymer-based scaffolds with excellent biocompatibility, antibacterial activity, and blood compatibility, able to facilitate full-thickness skin wound healing, remains challenging. In this study, we have developed three chitosan (CS)-based porous scaffolds, including CS, CS/CNT (carbon nanotubes) and CS/CNT/HA (nano-hydroxyapatite, n-HA) using a freeze-drying method. All three scaffolds have a high swelling ratio, excellent antibacterial activity, outstanding cytocompatibility and blood compatibility in vitro. The introduction of CNTs exhibited an obvious increase in mechanical properties and exerts excellent photothermal response, which displays excellent healing performance as a wound dressing in mouse full-thickness skin wound model when compared to CS scaffolds. CS/CNT/HA composite scaffolds present the strongest ability to promote full-thickness cutaneous wound closure and skin regeneration, which might be ascribed to the synergistic effect of photothermal response from CNT and excellent bioactivity from n-HA. Overall, the present study indicated that CNT and n-HA can be engineered as effective constituents in wound dressings to facilitate full-thickness skin regeneration.
Collapse
Affiliation(s)
| | - Shijia Fu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Huishan Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yue Wu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
15
|
Sharma P, Kumar A, Dey AD. Cellular Therapeutics for Chronic Wound Healing: Future for Regenerative Medicine. Curr Drug Targets 2022; 23:1489-1504. [PMID: 35748548 DOI: 10.2174/138945012309220623144620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/10/2021] [Accepted: 02/01/2022] [Indexed: 01/25/2023]
Abstract
Chronic wounds are associated with significant morbidity and mortality, which demand long-term effective treatment and represent a tremendous financial strain on the global healthcare systems. Regenerative medicines using stem cells have recently become apparent as a promising approach and are an active zone of investigation. They hold the potential to differentiate into specific types of cells and thus possess self-renewable, regenerative, and immune-modulatory effects. Furthermore, with the rise of technology, various cell therapies and cell types such as Bone Marrow and Adipose-derived Mesenchymal Cell (ADMSC), Endothelial Progenitor Cells (EPCs), Embryonic Stem Cells (ESCs), Mesenchymal Stem Cell (MSCs), and Pluripotent Stem Cells (PSCs) are studied for their therapeutic impact on reparative processes and tissue regeneration. Cell therapy has proven to have substantial control over enhancing the quality and rate of skin regeneration and wound restoration. The literature review brings to light the mechanics of wound healing, abnormalities resulting in chronic wounds, and the obstacles wound care researchers face, thus exploring the multitude of opportunities for potential improvement. Also, the review is focused on providing particulars on the possible cell-derived therapeutic choices and their associated challenges in healing, in the context of clinical trials, as solutions to these challenges will provide fresh and better future opportunities for improved study design and therefore yield a substantial amount of data for the development of more specialized treatments.
Collapse
Affiliation(s)
- Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.,Government Pharmacy College Kangra, Nagrota Bhagwan, Himachal Pradesh, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
16
|
PLGA/Gelatin/Hyaluronic Acid Fibrous Membrane Scaffold for Therapeutic Delivery of Adipose-Derived Stem Cells to Promote Wound Healing. Biomedicines 2022; 10:biomedicines10112902. [PMID: 36428471 PMCID: PMC9687264 DOI: 10.3390/biomedicines10112902] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Hyaluronic acid (HA) has been suggested to be a preferential material for the delivery of adipose-derived stem cells (ASCs) in wound healing. By incorporating HA in electrospun poly (lactide-co-glycolide) (PLGA)/gelatin (PG) fibrous membrane scaffolds (FMS), we aim to fabricate PLGA/gelatin/HA (PGH) FMS to provide a milieu for 3D culture and delivery of ASCs. The prepared FMS shows adequate cytocompatibility and is suitable for attachment and growth of ASCs. Compared with PG, the PGH offers an enhanced proliferation rate of ASCs, shows higher cell viability, and better maintains an ASC-like phenotype during in vitro cell culture. The ASCs in PGH also show upregulated expression of genes associated with angiogenesis and wound healing. From a rat full-thickness wound healing model, a wound treated with PGH/ASCs can accelerate the wound closure rate compared with wounds treated with PGH, alginate wound dressing, and gauze. From H&E and Masson's trichrome staining, the PGH/ASC treatment can promote wound healing by increasing the epithelialization rate and forming well-organized dermis. This is supported by immunohistochemical staining of macrophages and α-smooth muscle actin, where early recruitment of macrophages, macrophage polarization, and angiogenesis was found due to the delivered ASCs. The content of type III collagen is also higher than type I collagen within the newly formed skin tissue, implying scarless wound healing. Taken together, using PGH FMS as a topical wound dressing material for the therapeutic delivery of ASCs, a wound treated with PGH/ASCs was shown to accelerate wound healing significantly in rats, through modulating immunoreaction, promoting angiogenesis, and reducing scar formation at the wound sites.
Collapse
|
17
|
Derakhshan R, Ahmadi H, Bayat M, Mehboudi L, Pourhashemi E, Amini A, Vatandoust D, Aghamiri S, Asadi R, Sabet B. The Combined Effects of a Methacrylate Powder Dressing (Altrazeal Powder) and Photobiomodulation Therapy on the Healing of a Severe Diabetic Foot Ulcer in a Diabetic Patient: A Case Report. J Lasers Med Sci 2022; 13:e38. [PMID: 36743151 PMCID: PMC9841389 DOI: 10.34172/jlms.2022.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/14/2022] [Indexed: 01/27/2023]
Abstract
Weakened wound healing is a popular, severe complication of patients with diabetes which poses a risk for foot infection and amputation. Researchers have searched for new treatments for treating diabetic foot ulcers (DFUs) in recent years. In this case report, for the first time, we applied photobiomodulation therapy (PBMT) and Altrazeal powder together to treat a severe case of DFU in a 47-year-old woman who was suffering from type 1 diabetes. Along with the progress of combination therapy, we observed that the ulcer area was significantly reduced, and the wound healed within 16 weeks. Furthermore, dermatitis and purulent secretion were treated, and the pain was reduced. This reported case study indicated the beneficial effect of the combination of PBMT and Altrazeal powder for the healing of a severe DFU in a patient with type one diabetes. The combined application of PBMT plus Altrazeal powder demonstrated an additive effect. Further clinical trials in the clinical setting are suggested to validate the results further. Besides, more studies in preclinical models are suggested to find the mechanism of the action of combination therapy.
Collapse
Affiliation(s)
- Roya Derakhshan
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran,Price Institute of Surgical Research, University of Louisville, and Noveratech LLC, Louisville, KY, USA
| | - Leila Mehboudi
- Department of Anesthesia, Faculty of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Erfan Pourhashemi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Dorsa Vatandoust
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Robabeh Asadi
- Department of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Sabet
- Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran,Correspondence to Babak Sabet,
| |
Collapse
|
18
|
Patton D, Avsar P, Wilson P, Mairghani M, O'Connor T, Nugent L, Moore Z. Treatment of diabetic foot ulcers: review of the literature with regard to the TIME clinical decision support tool. J Wound Care 2022; 31:771-779. [PMID: 36113541 DOI: 10.12968/jowc.2022.31.9.771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The aim of this clinically orientated paper is to offer an overview of diabetic foot ulcer (DFU) dressings generally, and more specifically, their use in the treatment of DFUs. METHOD The TIME clinical decision support tool (CDST) has been used as a clinical tool that can help clinicians bring together the different aspects of dressings for DFU treatment into a holistic approach to patient care. RESULTS DFUs are often difficult to heal, are painful and impact negatively on the individual's quality of life. Most DFU dressings are designed to support the healing of hard-to-heal wounds and represent one part of the management of DFUs. Apart from providing a moist environment, absorbing increased exudate, enhancing granulation and assisting in autolysis, the dressings need to be cost-effective. Wound dressing selection is based on clinical knowledge that ensures the dressing is most appropriate for the individual and the wound, taking into account the comorbidities that the individual may have. CONCLUSION This paper has highlighted how the use of the TIME CDST model can enhance clinical care and is a further tool clinicians should consider when developing and executing DFU treatment plans. Future research needs to focus on large multicentre studies using robust methodologies, given the current gaps in the evidence, to determine the effectiveness of dressing products for DFUs.
Collapse
Affiliation(s)
- Declan Patton
- School of Nursing & Midwifery, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin.,Skin, Wounds and Trauma Research Centre, School of Nursing and Midwifery. RCSI University of Medicine and Health Sciences, Dublin.,Adjunct Associate Professor, Fakeeh College of Health Sciences, Jeddah, Saudi Arabia.,Honorary Senior Fellow, Faculty of Science, Medicine and Health, University of Wollongong, Australia.,Adjunct Professor, Griffith University, Australia
| | - Pinar Avsar
- Skin, Wounds and Trauma Research Centre, School of Nursing and Midwifery. RCSI University of Medicine and Health Sciences, Dublin
| | - Pauline Wilson
- Skin, Wounds and Trauma Research Centre, School of Nursing and Midwifery. RCSI University of Medicine and Health Sciences, Dublin
| | - Maisoon Mairghani
- Public Health and Epidemiology, RCSI University of Medicine and Health Sciences
| | - Tom O'Connor
- School of Nursing & Midwifery, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin.,Skin, Wounds and Trauma Research Centre, School of Nursing and Midwifery. RCSI University of Medicine and Health Sciences, Dublin.,Adjunct Professor, Griffith University, Australia.,Honorary Professor, Lida Institute, Shanghai, China.,Professor, Fakeeh College of Health Sciences
| | - Linda Nugent
- School of Nursing & Midwifery, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin.,Adjunct Assistant Professor, Fakeeh College of Health Sciences, Jeddah, Saudi Arabia
| | - Zena Moore
- School of Nursing & Midwifery, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin.,Skin, Wounds and Trauma Research Centre, School of Nursing and Midwifery. RCSI University of Medicine and Health Sciences, Dublin.,Honorary Professor, Lida Institute, Shanghai, China.,Professor, Fakeeh College of Health Sciences.,Professor, Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium.,Visiting Professor, University of Wales, Cardiff, UK
| |
Collapse
|
19
|
El-Serafi AT, El-Serafi I, Steinvall I, Sjöberg F, Elmasry M. A Systematic Review of Keratinocyte Secretions: A Regenerative Perspective. Int J Mol Sci 2022; 23:ijms23147934. [PMID: 35887279 PMCID: PMC9323141 DOI: 10.3390/ijms23147934] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Cell regenerative therapy is a modern solution for difficult-to-heal wounds. Keratinocytes, the most common cell type in the skin, are difficult to obtain without the creation of another wound. Stem cell differentiation towards keratinocytes is a challenging process, and it is difficult to reproduce in chemically defined media. Nevertheless, a co-culture of keratinocytes with stem cells usually achieves efficient differentiation. This systematic review aims to identify the secretions of normal human keratinocytes reported in the literature and correlate them with the differentiation process. An online search revealed 338 references, of which 100 met the selection criteria. A total of 80 different keratinocyte secretions were reported, which can be grouped mainly into cytokines, growth factors, and antimicrobial peptides. The growth-factor group mostly affects stem cell differentiation into keratinocytes, especially epidermal growth factor and members of the transforming growth factor family. Nevertheless, the reported secretions reflected the nature of the involved studies, as most of them focused on keratinocyte interaction with inflammation. This review highlights the secretory function of keratinocytes, as well as the need for intense investigation to characterize these secretions and evaluate their regenerative capacities.
Collapse
Affiliation(s)
- Ahmed T. El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
- Correspondence:
| | - Ibrahim El-Serafi
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
- Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Ingrid Steinvall
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| | - Moustafa Elmasry
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| |
Collapse
|
20
|
Acellular Dermal Matrix Favorably Modulates the Healing Response after Surgery. Plast Reconstr Surg 2022; 150:290e-299e. [PMID: 35653544 DOI: 10.1097/prs.0000000000009338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
SUMMARY When first described for breast reconstruction, the presence of acellular dermal matrices (ADM) was associated with increased seroma formation and infection. However, clinical safety data has gradually improved with surgeon experience to an acceptable outcomes profile of ADM-assisted reconstruction when compared to submuscular implant coverage. In fact, ADM use potentially decreases capsular contracture rates and facilitates expansion for staged pre-pectoral breast reconstruction. Due to new regulatory requirements, the collection of unbiased, well-powered pre-market approval data summarizing long-term clinical outcomes will be essential over the coming years to understand the clinical performance of ADM usage in breast reconstruction.Currently, we can highlight the physiologic benefits of ADM use in breast reconstruction by evaluating the components of surgical wound healing that are favorably augmented by the implanted collagen substrate. ADM takes advantage of the wound healing cascade to incorporate into the patient's tissues - a process that requires a coordinated inflammatory infiltrate and angiogenesis. The presence of ADM augments and modulates the wound healing process to its advantage by simultaneously increasing the invasion of appropriate cellular constituents to facilitate expeditious healing and accelerate angiogenesis. Herein, we summarize the wound healing literature to demonstrate the mechanisms ADM use to biointegrate and the literature in which cellular constituents and soluble growth factors are upregulated in the presence of ADM. Lastly, we use our experimental observations of ADM incorporation to corroborate the literature.
Collapse
|
21
|
Leibrock LB, Hofmann DM, Fuchs B, Birt A, Reinholz M, Guertler A, Frank K, Giunta RE, Egaña JT, Nickelsen J, Schenck TL, Moellhoff N. In vitro and in vivo detection of microbial gene expression in bioactivated scaffolds seeded with cyanobacteria. Lett Appl Microbiol 2022; 75:401-409. [PMID: 35587396 DOI: 10.1111/lam.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022]
Abstract
Dermal replacement materials bioactivated with cyanobacteria have shown promising potential for wound regeneration. To date, extraction of cyanobacteria RNA from seeded scaffolds has not been described. Aim of this study was to develop a method to isolate total RNA from bioactivated scaffolds and to propose a new approach in determining living bacteria based on real-time PCR. Transgenic synechococcus sp. PCC 7002 (tSyn7002) were seeded in liquid cultures or in scaffolds for dermal regeneration in vitro and in vivo for 7 days. RNA was extracted with a 260/280 ratio of ≥ 2. The small subunit of the 30S ribosome in prokaryotes (16S) and RNAse P protein (rnpA) were validated as reference transcripts for PCR analysis. Gene expression patterns differed in vitro and in vivo. Expression of 16S was significantly upregulated in scaffolds in vitro, as compared to liquid cultures, while rnpA expression was comparable. In vivo, both 16S and rnpA showed reduced expression compared to in vitro (16S: in vivo Ct value 13.21±0.32, in vitro 12.44±0.42; rnpA in vivo Ct value 19.87±0.41, in vitro 17.75±1.41). Overall, the results demonstrate rnpA and 16S expression after 7 days of implantation in vitro and in vivo, proving presence of living bacteria embedded in scaffolds using qPCR.
Collapse
Affiliation(s)
- Lars B Leibrock
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - Daniel M Hofmann
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - Benedikt Fuchs
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - Alexandra Birt
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - Markus Reinholz
- Department of Dermatology and Allergy, University Hospital of Munich, LMU, Germany
| | - Anne Guertler
- Department of Dermatology and Allergy, University Hospital of Munich, LMU, Germany
| | - Konstantin Frank
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - Riccardo E Giunta
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| | - José T Egaña
- Institute for Biological and Medical Engineering, Schools of Engineering, Biological Sciences and Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Joerg Nickelsen
- Molecular Plant Science, Department Biology I, LMU Munich, Munich, Germany
| | - Thilo L Schenck
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany.,Frauenklinik Dr. Geisenhofer GmbH, 80538, Munich, Germany
| | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Germany
| |
Collapse
|
22
|
Aulanni’am A, Raissa R, Riawan W, Wuragil DK, Permata FS, Beltran MAG. Epidermal Stem Cell in Wound Healing of Gliricidia sepium Leaves from Indonesia and the Philippines in Rats (Rattus norvegicus). Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: This study intended to investigate the regenerate wound, due to the ointment therapy containing Gliricidia sepium leaves that has potential-induced epidermal stem cells producing. It determined its effect on the expression of transforming growth factor-β1 (TGF-β1), Smad-3, β-catenin, LGR-6.
MATERIALS AND METHODS: About 16 Wistar male rats aged approximately 2 months (150–200g) were used and were divided into four treatment groups (T1, positive control; T2, negative control; T3, wounds treated with G. sepium from Indonesia; and T4, wounds treated with G. sepium from the Philippines). The treatment of ointment was applied to the wound for 3 days. The expression of TGF-β1, Smad-3, β-catenin, and LGR-6 was observed by immunohistochemistry staining.
RESULTS: G. sepium leaves significantly (p < 0.05) upregulated the expression of TGF-β1, Smad-3, β-catenin, and LGR-6 in the group treated with Indonesian G. sepium leaves were higher than that in the group treated with G. sepium leaves from the Philippines.
CONCLUSIONS: Both leaves Varian contain flavonoids, saponins, and tannins, which act as producing epidermal stem cell agents to enhance the wound healing process. It can be concluded that both Gl. sepium Varian Indonesia and the Philippines have a potential effect on wound healing.
Collapse
|
23
|
Li Y, Li Y, Li L, Wang H, Wang B, Feng L, Lin S, Li G. The emerging translational potential of GDF11 in chronic wound healing. J Orthop Translat 2022; 34:113-120. [PMID: 35891714 PMCID: PMC9283991 DOI: 10.1016/j.jot.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 11/28/2022] Open
Abstract
Chronic skin wounds impose immense suffers and economic burdens. Current research mainly focuses on acute wound management which exhibits less effective in chronic wound healing. Growth differentiation factor 11 (GDF11) has profound effects on several important physiological processes related to chronic wound healing, such as inflammation, cell proliferation, migration, angiogenesis, and neurogenesis. This review summarizes recent advances in biology of chronic wounds and the potential role of GDF11 on wound healing with its regenerative effects, as well as the potential delivery methods of GDF11. The challenges and future perspectives of GDF11-based therapy for chronic wound care are also discussed. The Translational Potential of this Article: This review summarized the significance of GDF11 in the modulation of inflammation, vascularization, cell proliferation, and remodeling, which are important physiological processes of chronic wound healing. The potential delivery methods of GDF11 in the management of chronic wound healing is also summarized. This review may provide potential therapeutic approaches based on GDF11 for chronic wound healing.
Collapse
|
24
|
Zhong J, Wang H, Yang K, Wang H, Duan C, Ni N, An L, Luo Y, Zhao P, Gou Y, Sheng S, Shi D, Chen C, Wagstaff W, Hendren-Santiago B, Haydon RC, Luu HH, Reid RR, Ho SH, Ameer GA, Shen L, He TC, Fan J. Reversibly immortalized keratinocytes (iKera) facilitate re-epithelization and skin wound healing: Potential applications in cell-based skin tissue engineering. Bioact Mater 2022; 9:523-540. [PMID: 34820586 PMCID: PMC8581279 DOI: 10.1016/j.bioactmat.2021.07.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Skin injury is repaired through a multi-phase wound healing process of tissue granulation and re-epithelialization. Any failure in the healing process may lead to chronic non-healing wounds or abnormal scar formation. Although significant progress has been made in developing novel scaffolds and/or cell-based therapeutic strategies to promote wound healing, effective management of large chronic skin wounds remains a clinical challenge. Keratinocytes are critical to re-epithelialization and wound healing. Here, we investigated whether exogenous keratinocytes, in combination with a citrate-based scaffold, enhanced skin wound healing. We first established reversibly immortalized mouse keratinocytes (iKera), and confirmed that the iKera cells expressed keratinocyte markers, and were responsive to UVB treatment, and were non-tumorigenic. In a proof-of-principle experiment, we demonstrated that iKera cells embedded in citrate-based scaffold PPCN provided more effective re-epithelialization and cutaneous wound healing than that of either PPCN or iKera cells alone, in a mouse skin wound model. Thus, these results demonstrate that iKera cells may serve as a valuable skin epithelial source when, combining with appropriate biocompatible scaffolds, to investigate cutaneous wound healing and skin regeneration.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Ke Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- The Pediatric Research Institute, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Huifeng Wang
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
| | - Chongwen Duan
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
| | - Na Ni
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Liqin An
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yetao Luo
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Piao Zhao
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shiyan Sheng
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Bryce Hendren-Santiago
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
| | - Sherwin H. Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Guillermo A. Ameer
- Biomedical Engineering Department, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60616, USA
| | - Le Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL, 60208, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, And Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| |
Collapse
|
25
|
Burn Wound Healing: Clinical Complications, Medical Care, Treatment, and Dressing Types: The Current State of Knowledge for Clinical Practice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031338. [PMID: 35162360 PMCID: PMC8834952 DOI: 10.3390/ijerph19031338] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023]
Abstract
According to the World Health Organization (WHO), it is estimated that each year approximately 11 million people suffer from burn wounds, 180,000 of whom die because of such injuries. Regardless of the factors causing burns, these are complicated wounds that are difficult to heal and are associated with high mortality rates. Medical care of a burn patient requires a lot of commitment, experience, and multidirectional management, including surgical activities and widely understood pharmacological approaches. This paper aims to comprehensively review the current literature concerning burn wounds, including classification of burns, complications, medical care, and pharmacological treatment. We also overviewed the dressings (with an emphasis on the newest innovations in this field) that are currently used in medical practice to heal wounds.
Collapse
|
26
|
Karkanitsa M, Fathi P, Ngo T, Sadtler K. Mobilizing Endogenous Repair Through Understanding Immune Reaction With Biomaterials. Front Bioeng Biotechnol 2021; 9:730938. [PMID: 34917594 PMCID: PMC8670074 DOI: 10.3389/fbioe.2021.730938] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022] Open
Abstract
With few exceptions, humans are incapable of fully recovering from severe physical trauma. Due to these limitations, the field of regenerative medicine seeks to find clinically viable ways to repair permanently damaged tissue. There are two main approaches to regenerative medicine: promoting endogenous repair of the wound, or transplanting a material to replace the injured tissue. In recent years, these two methods have fused with the development of biomaterials that act as a scaffold and mobilize the body's natural healing capabilities. This process involves not only promoting stem cell behavior, but by also inducing activity of the immune system. Through understanding the immune interactions with biomaterials, we can understand how the immune system participates in regeneration and wound healing. In this review, we will focus on biomaterials that promote endogenous tissue repair, with discussion on their interactions with the immune system.
Collapse
Affiliation(s)
| | | | | | - Kaitlyn Sadtler
- Section on Immuno-Engineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
27
|
Aleksandrowicz H, Owczarczyk-Saczonek A, Placek W. Venous Leg Ulcers: Advanced Therapies and New Technologies. Biomedicines 2021; 9:1569. [PMID: 34829797 PMCID: PMC8615583 DOI: 10.3390/biomedicines9111569] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/08/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
The prevalence of venous leg ulcers (VLUs) differs between 1.5% and 3% in the general population. The challenge in treating VLUs is common recurrence. Moreover, VLUs can be resistant to healing, despite appropriate treatment. In these cases, advanced wound therapies should be considered. The number of new technologies, applied in VLUs treatment, has increased in the last years. These therapies include biophysical interventions such as ultrasound therapy, electrical stimulations, electromagnetic therapy, or phototherapy. Furthermore, stem cell therapies, biologic skin equivalents, platelet-rich plasma therapy, oxygen therapies, anti-TNF therapy, or negative pressure wound therapy are advanced venous ulcer therapeutic methods that may support the standard of care. Medical devices, such as a muscle pump activator, or intermittent pneumatic compression device, may be especially useful for specific subgroups of patients suffering from VLUs. Some of the above-mentioned technologies require broader evidence of clinical efficacy and are still considered experimental therapies in dermatology.
Collapse
Affiliation(s)
- Hubert Aleksandrowicz
- Department and Clinic of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, University of Warmia and Mazury, Al. Wojska Polskiego 30, 10-959 Olsztyn, Poland; (A.O.-S.); (W.P.)
| | | | | |
Collapse
|
28
|
Azam M, Ghufran H, Butt H, Mehmood A, Ashfaq R, Ilyas AM, Ahmad MR, Riazuddin S. Curcumin preconditioning enhances the efficacy of adipose-derived mesenchymal stem cells to accelerate healing of burn wounds. BURNS & TRAUMA 2021; 9:tkab021. [PMID: 34514007 PMCID: PMC8430278 DOI: 10.1093/burnst/tkab021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/26/2021] [Indexed: 01/09/2023]
Abstract
Background Following recent findings from our group that curcumin preconditioning augments the therapeutic efficacy of adipose-derived stem cells in the healing of diabetic wounds in rats, we aimed to investigate the regenerative effects of curcumin preconditioned adipose-derived mesenchymal stem cells (ASCs) for better recovery of acid inflicted burns in this study. Methods ASCs were preconditioned with 5 μM curcumin for 24 hours and assessed for proliferation, migration, paracrine release potential and gene expression comparative to naïve ASCs. Subsequently, the healing capacity of curcumin preconditioned ASCs (Cur-ASCs) versus naïve ASCs was examined using acidic wounds in rats. For this, acid inflicted burns of 20 mm in diameter were made on the back of male Wistar rats. Then, 2 × 106 cells of Cur-ASCs and naïve ASCs were intradermally injected in the wound periphery (n = 6) for comparison with an untreated saline control. Post-transplantation, wounds were macroscopically analysed and photographed to evaluate the percentage of wound closure and period of re-epithelization. Healed wound biopsies were excised and used for histological evaluation and expression analysis of wound healing markers at molecular level by quantitative PCR and western blotting. Results We found that Cur-ASCs exhibited greater proliferation, migration and paracrine potential in vitro. Further, Cur-ASCs showed more effective recovery than naïve ASCs as exhibited by gross morphology, faster wound closure and earlier re-epithelialization. Masson’s trichrome and hematoxylin and eosin staining demonstrated the improved architecture of the healing burns, as evidenced by reduced infiltration of inflammatory cells, compact collagen and marked granulation in Cur-ASC treated rats. Corroborating these findings, molecular assessment showed significantly reduced expressions of pro-inflammatory factors (interleukin-1 beta, interleukin-6, tumor necrosis factor alpha) a with striking upsurge of an oxidative marker (superoxide dismutase 1), pro-angiogenic factors (vascular endothelial growth factor, hepatocyte growth factor, hypoxia-inducible factor-1 alpha) and collagen markers (transforming growth factor beta 1, fibroblast growth factor-2, collagen type 1 alpha 1), verifying that Cur-ASCs modulate the regulation of pro-inflammatory and healing markers at burn sites. Conclusions Treatment with Cur-ASCs resulted in faster re-epithelization of acid inflicted burns compared to the treatment with naïve ASCs. Based on observed findings, we suggest the transplantation of Cur-ASCs is a valuable therapy for the potent clinical management of acidic burns.
Collapse
Affiliation(s)
- Maryam Azam
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Hafiz Ghufran
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Hira Butt
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Ramla Ashfaq
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Asad M Ilyas
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Muhammad R Ahmad
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
29
|
Ajit A, Ambika Gopalankutty I. Adipose-derived stem cell secretome as a cell-free product for cutaneous wound healing. 3 Biotech 2021; 11:413. [PMID: 34476171 DOI: 10.1007/s13205-021-02958-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds continue to be a substantial public health concern contributing to both humanistic and economic burden worldwide. The magnitude of chronic wounds as a global healthcare crisis is likely to increase due to the rising geriatric and diabetic population, demanding novel therapeutic approaches that can restore the functionality of the skin at a reduced cost. Stem cell therapy has been widely acknowledged as a promising strategy for the repair of damaged tissues due to its regenerative potential. This potential attributes to a concoction of bioactive molecules secreted by the stem cells, collectively called the secretome, that mediates paracrine and autocrine functions. Among the stem cell types, adipose tissue-derived mesenchymal stem cells (ADMSCs) have been receiving increased attention for its ease of isolation, abundance in tissue and notable impact on improving chronic wound healing. Owing to the reported advantages of cell-free preparations like the secretome over cellular products, developing secretome as a ready-to-use product for wound healing applications seems promising. In this review, we discuss the functional benefits of adipose stem cell secretome in wound healing, the techniques to enrich the secretome and the recommendations for the scale-up and commercialization of secretome products.
Collapse
Affiliation(s)
- Amita Ajit
- Scientific Consultant and Life Member, Kerala Academy of Sciences, Sasthra Bhavan, Pattom, Thiruvananthapuram, 695004 Kerala India
| | | |
Collapse
|
30
|
Lima TDPDL, Passos MF. Skin wounds, the healing process, and hydrogel-based wound dressings: a short review. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1910-1925. [PMID: 34156314 DOI: 10.1080/09205063.2021.1946461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skin wounds are damage to the epithelial layer and the integrity of living tissue. The healing mechanism is dynamic and complex, and often treatments with wound dressings help in tissue regeneration, reducing the risk of infections. Polymeric hydrogels become good candidates for wet curing process. These materials prevent dehydration of the tissue and avoid discomfort to the patient when changing the dressing. In this short review, we demonstrate the importance of the healing process, the types of skin wounds, and the hydrogels that are potentially attractive as wound dressings.
Collapse
|
31
|
Lu Q, Yin Z, Shen X, Li J, Su P, Feng M, Xu X, Li W, He C, Shen Y. Clinical effects of high-intensity laser therapy on patients with chronic refractory wounds: a randomised controlled trial. BMJ Open 2021; 11:e045866. [PMID: 34253665 PMCID: PMC8276284 DOI: 10.1136/bmjopen-2020-045866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE To investigate the effectiveness of high-intensity laser therapy (HILT) on chronic refractory wounds.DesignRandomised controlled trial. SETTING The outpatient wound care department of the Affiliated Jiangsu Shengze Hospital of Nanjing Medical University from August 2019 to June 2020. PARTICIPANTS Sixty patients were enrolled in this study and were randomised into control (n=30) and treatment (n=30) groups. INTERVENTIONS AND OUTCOME MEASURES The control group was treated only with conventional wound dressing, whereas the treatment group received irradiation with HILT in addition to standard wound care, such as debridement, wound irrigation with normal saline solution and application of dressing and sterile gauze. Patient scores on the Bates-Jensen Wound Assessment Tool (BWAT) and Pressure Ulcer Scale for Healing (PUSH) were evaluated before and after 1, 2 and 3 weeks of treatment. RESULTS One patient was excluded from the control group, and a total of 59 subjects completed the trial. The BWAT scores significantly decreased in the treatment group compared with the control group at the end of 3-week treatment (difference=-3.6; 95% CI -6.3 to-0.8; p<0.01). Similarly, patients in treatment group showed a significant reduction of PUSH scores compared with the control group (difference=-5.3; 95% CI -8.1 to -2.6; p<0.01). CONCLUSIONS The therapeutic effects of HILT on chronic refractory wounds are significant and far more superior to those of conventional wound dressing. TRIAL REGISTRATION NUMBER Chinese Clinical Trial Registry; ChiCTR1900023157. URL: http://www.chictr.org.cn/showproj.aspx?proj=38866.
Collapse
Affiliation(s)
- Qian Lu
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhifei Yin
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Shen
- Department of Outpatient Wound Care, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jinhua Li
- Department of Outpatient Wound Care, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Panpan Su
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Min Feng
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xingjun Xu
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Li
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Chuan He
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ying Shen
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
32
|
Zhao X, Hu DA, Wu D, He F, Wang H, Huang L, Shi D, Liu Q, Ni N, Pakvasa M, Zhang Y, Fu K, Qin KH, Li AJ, Hagag O, Wang EJ, Sabharwal M, Wagstaff W, Reid RR, Lee MJ, Wolf JM, El Dafrawy M, Hynes K, Strelzow J, Ho SH, He TC, Athiviraham A. Applications of Biocompatible Scaffold Materials in Stem Cell-Based Cartilage Tissue Engineering. Front Bioeng Biotechnol 2021; 9:603444. [PMID: 33842441 PMCID: PMC8026885 DOI: 10.3389/fbioe.2021.603444] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Cartilage, especially articular cartilage, is a unique connective tissue consisting of chondrocytes and cartilage matrix that covers the surface of joints. It plays a critical role in maintaining joint durability and mobility by providing nearly frictionless articulation for mechanical load transmission between joints. Damage to the articular cartilage frequently results from sport-related injuries, systemic diseases, degeneration, trauma, or tumors. Failure to treat impaired cartilage may lead to osteoarthritis, affecting more than 25% of the adult population globally. Articular cartilage has a very low intrinsic self-repair capacity due to the limited proliferative ability of adult chondrocytes, lack of vascularization and innervation, slow matrix turnover, and low supply of progenitor cells. Furthermore, articular chondrocytes are encapsulated in low-nutrient, low-oxygen environment. While cartilage restoration techniques such as osteochondral transplantation, autologous chondrocyte implantation (ACI), and microfracture have been used to repair certain cartilage defects, the clinical outcomes are often mixed and undesirable. Cartilage tissue engineering (CTE) may hold promise to facilitate cartilage repair. Ideally, the prerequisites for successful CTE should include the use of effective chondrogenic factors, an ample supply of chondrogenic progenitors, and the employment of cell-friendly, biocompatible scaffold materials. Significant progress has been made on the above three fronts in past decade, which has been further facilitated by the advent of 3D bio-printing. In this review, we briefly discuss potential sources of chondrogenic progenitors. We then primarily focus on currently available chondrocyte-friendly scaffold materials, along with 3D bioprinting techniques, for their potential roles in effective CTE. It is hoped that this review will serve as a primer to bring cartilage biologists, synthetic chemists, biomechanical engineers, and 3D-bioprinting technologists together to expedite CTE process for eventual clinical applications.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Ofir Hagag
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin H. Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
33
|
Zhang Y, Wu D, Zhao X, Pakvasa M, Tucker AB, Luo H, Qin KH, Hu DA, Wang EJ, Li AJ, Zhang M, Mao Y, Sabharwal M, He F, Niu C, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Chen C, Wagstaff W, Reid RR, Athiviraham A, Ho S, Lee MJ, Hynes K, Strelzow J, He TC, El Dafrawy M. Stem Cell-Friendly Scaffold Biomaterials: Applications for Bone Tissue Engineering and Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:598607. [PMID: 33381499 PMCID: PMC7767872 DOI: 10.3389/fbioe.2020.598607] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic organ with high regenerative potential and provides essential biological functions in the body, such as providing body mobility and protection of internal organs, regulating hematopoietic cell homeostasis, and serving as important mineral reservoir. Bone defects, which can be caused by trauma, cancer and bone disorders, pose formidable public health burdens. Even though autologous bone grafts, allografts, or xenografts have been used clinically, repairing large bone defects remains as a significant clinical challenge. Bone tissue engineering (BTE) emerged as a promising solution to overcome the limitations of autografts and allografts. Ideal bone tissue engineering is to induce bone regeneration through the synergistic integration of biomaterial scaffolds, bone progenitor cells, and bone-forming factors. Successful stem cell-based BTE requires a combination of abundant mesenchymal progenitors with osteogenic potential, suitable biofactors to drive osteogenic differentiation, and cell-friendly scaffold biomaterials. Thus, the crux of BTE lies within the use of cell-friendly biomaterials as scaffolds to overcome extensive bone defects. In this review, we focus on the biocompatibility and cell-friendly features of commonly used scaffold materials, including inorganic compound-based ceramics, natural polymers, synthetic polymers, decellularized extracellular matrix, and in many cases, composite scaffolds using the above existing biomaterials. It is conceivable that combinations of bioactive materials, progenitor cells, growth factors, functionalization techniques, and biomimetic scaffold designs, along with 3D bioprinting technology, will unleash a new era of complex BTE scaffolds tailored to patient-specific applications.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Andrew Blake Tucker
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Laboratory Diagnostic Medicine, The Affiliated Hospital of the University of Chinese Academy of Sciences, Chongqing General Hospital, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery Section of Plastic and Reconstructive Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
34
|
Weigelt MA, Sivamani R, Lev-Tov H. The therapeutic potential of cannabinoids for integumentary wound management. Exp Dermatol 2020; 30:201-211. [PMID: 33205468 DOI: 10.1111/exd.14241] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022]
Abstract
The increasing legalization of Cannabis for recreational and medicinal purposes in the United States has spurred renewed interest in the therapeutic potential of cannabinoids (CBs) for human disease. The skin has its own endocannabinoid system (eCS) which is a key regulator of various homeostatic processes, including those necessary for normal physiologic wound healing. Data on the use of CBs for wound healing are scarce. Compelling pre-clinical evidence supporting the therapeutic potential of CBs to improve wound healing by modulating key molecular pathways is herein reviewed. These findings merit further exploration in basic science, translational and clinical studies.
Collapse
Affiliation(s)
- Maximillian A Weigelt
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Raja Sivamani
- Department of Dermatology, University of California-Davis, Sacramento, CA, USA.,Department of Biological Sciences, California State University, Sacramento, CA, USA.,School of Medicine, California Northstate University, Elk Grove, CA, USA.,Pacific Skin Institute, Sacramento, CA, USA.,Zen Dermatology, Sacramento, CA, USA
| | - Hadar Lev-Tov
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
35
|
Ahmadi H, Amini A, Fadaei Fathabady F, Mostafavinia A, Zare F, Ebrahimpour-malekshah R, Ghalibaf MN, Abrisham M, Rezaei F, Albright R, Ghoreishi SK, Chien S, Bayat M. Transplantation of photobiomodulation-preconditioned diabetic stem cells accelerates ischemic wound healing in diabetic rats. Stem Cell Res Ther 2020; 11:494. [PMID: 33239072 PMCID: PMC7688005 DOI: 10.1186/s13287-020-01967-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diabetic foot ulcer is the most costly and complex challenge for patients with diabetes. We hereby assessed the effectiveness of different preconditioned adipose-derived mesenchymal stem cells (AD-MSCs) and photobiomodulation protocols on treating an infected ischemic wound in type 1 diabetic rats. METHODS There were five groups of rats: (1) control, (2) control AD-MSCs [diabetic AD-MSCs were transplanted (grafted) into the wound bed], (3) AD-MSC + photobiomodulation in vivo (diabetic AD-MSCs were grafted into the wound, followed by in vivo PBM treatment), (4) AD-MSCs + photobiomodulation in vitro, and (5) AD-MSCs + photobiomodulation in vitro + in vivo. RESULTS Diabetic AD-MSCs preconditioned with photobiomodulation had significantly risen cell function compared to diabetic AD-MSC. Groups 3 and 5 had significantly decreased microbial flora correlated to groups 1 and 2 (all, p = 0.000). Groups 2, 3, 4, and 5 had significantly improved wound closure rate (0.4, 0.4, 0.4, and 0.8, respectively) compared to group 1 (0.2). Groups 2-5 had significantly increased wound strength compared to group 1 (all p = 0.000). In most cases, group 5 had significantly better results than groups 2, 3, and 4. CONCLUSIONS Preconditioning diabetic AD-MSCs with photobiomodulation in vitro plus photobiomodulation in vivo significantly hastened healing in the diabetic rat model of an ischemic infected delayed healing wound.
Collapse
Affiliation(s)
- Houssein Ahmadi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atarodsadat Mostafavinia
- Department of Anatomy, Faculty of Medicine, Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Zare
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mustafa Neshat Ghalibaf
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Matin Abrisham
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemehalsadat Rezaei
- University of Kentucky, College of Pharmacy, 789 South Limestone, Lexington, Kentucky 40536 USA
| | | | | | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC, Louisville, KY USA
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC, Louisville, KY USA
| |
Collapse
|
36
|
Beccia E, Carbone A, Cecchino LR, Pedicillo MC, Annacontini L, Lembo F, Di Gioia S, Parisi D, Angiolillo A, Pannone G, Portincasa A, Conese M. Adipose Stem Cells and Platelet-Rich Plasma Induce Vascular-Like Structures in a Dermal Regeneration Template. Tissue Eng Part A 2020; 27:631-641. [PMID: 32907520 DOI: 10.1089/ten.tea.2020.0175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In the context of biointeractive dressings used for enhancing wound healing, the use of stromal vascular fraction (SVF) or adipose-derived stem cells (ASCs) hereof derived has not been fully exploited yet. Noncultured SVF, a heterogeneous mesenchymal population of cells, is attractive in the field of dermal regeneration because it can be instantaneously obtained, avoids genomic alterations, and is comparatively safer than cultured ASCs. Integra® Dermal Regeneration Template (DRT) was sprinkled with ASCs in complete medium supplemented with 10% fetal bovine serum (FBS), or SVF, obtained from emulsified or nonemulsified fat, in medium supplemented with 2% platelet-rich plasma (PRP). The presence and differentiation of cells were evaluated by standard histochemistry and immunohistochemistry, whereas conditioned media were analyzed for vascular endothelial growth factors (VEGF) by ELISA. In vitro experiments were conducted to analyze ASC proliferation in the presence of either FBS or PRP. Deposition of ASCs in medium supplemented with FBS caused their integration into Integra DRT as early as 1 h. ASCs were found as aggregates until 6-10 days without forming organized structures. When seeded onto Integra DRT, SVF cells in medium supplemented with PRP formed aggregates at early times, which at 7 and 10 days organized into vascular-like structures, lined by CD31+ and smooth muscle actin-positive cells. With nonemulsified fat, the lacunar structures did not show an organized distribution of SVF cells. PRP induced ASC proliferation although at lower level than FBS. VEGF secretion was enhanced when fat emulsification was introduced into the protocol. In conclusion, the combination of SVF cells obtained from emulsified fat, PRP, and Integra DRT exhibit synergistic effect on the formation of vessel-like structures indicating a step forward aimed at regenerative surgery for chronic wound healing.
Collapse
Affiliation(s)
- Elisa Beccia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.,Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Foggia, Italy
| | | | | | - Luigi Annacontini
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Fedele Lembo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Domenico Parisi
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences "V. Tiberio," University of Molise, Campobasso, Italy
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Aurelio Portincasa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
37
|
Frazier T, Alarcon A, Wu X, Mohiuddin OA, Motherwell JM, Carlsson AH, Christy RJ, Edwards JV, Mackin RT, Prevost N, Gloster E, Zhang Q, Wang G, Hayes DJ, Gimble JM. Clinical Translational Potential in Skin Wound Regeneration for Adipose-Derived, Blood-Derived, and Cellulose Materials: Cells, Exosomes, and Hydrogels. Biomolecules 2020; 10:E1373. [PMID: 32992554 PMCID: PMC7650547 DOI: 10.3390/biom10101373] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Acute and chronic skin wounds due to burns, pressure injuries, and trauma represent a substantial challenge to healthcare delivery with particular impacts on geriatric, paraplegic, and quadriplegic demographics worldwide. Nevertheless, the current standard of care relies extensively on preventive measures to mitigate pressure injury, surgical debridement, skin flap procedures, and negative pressure wound vacuum measures. This article highlights the potential of adipose-, blood-, and cellulose-derived products (cells, decellularized matrices and scaffolds, and exosome and secretome factors) as a means to address this unmet medical need. The current status of this research area is evaluated and discussed in the context of promising avenues for future discovery.
Collapse
Affiliation(s)
- Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Andrea Alarcon
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| | - Omair A. Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi 75270, Pakistan;
| | | | - Anders H. Carlsson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Robert J. Christy
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Judson V. Edwards
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Robert T. Mackin
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Nicolette Prevost
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Elena Gloster
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Qiang Zhang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Daniel J. Hayes
- Department of Biomedical Engineering, State College, Pennsylvania State University, Centre County, PA 16802, USA;
| | - Jeffrey M. Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| |
Collapse
|
38
|
Raghuram AC, Yu RP, Lo AY, Sung CJ, Bircan M, Thompson HJ, Wong AK. Role of stem cell therapies in treating chronic wounds: A systematic review. World J Stem Cells 2020; 12:659-675. [PMID: 32843920 PMCID: PMC7415243 DOI: 10.4252/wjsc.v12.i7.659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/03/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The impairment of cutaneous wound healing results in chronic, non-healing wounds that are caused by altered wound environment oxygenation, tissue injury, and permissive microbial growth. Current modalities for the treatment of these wounds inadequately address the complex changes involved in chronic wound pathogenesis. Consequently, stem cell therapies have emerged as a potential therapeutic modality to promote cutaneous regeneration through trophic and paracrine activity.
AIM To investigate current literature regarding use of stem cell therapies for the clinical treatment of chronic, non-healing wounds.
METHODS PubMed, EMBASE, Cochrane Library, Web of Science, and Scopus were queried with combinations of the search terms “mesenchymal stem cells,” “adult stem cells,” “embryonic stem cells,” “erythroid precursor cells,” “stem cell therapies,” and “chronic wounds” in order to find relevant articles published between the years of 2000 and 2019 to review a 20-year experience. Reference lists from the articles were reviewed to identify additional pertinent articles. Retrieved manuscripts (reviews, case reports/series, retrospective/prospective studies, and clinical trials) were evaluated by the authors for their depiction of clinical stem cell therapy use. Data were extracted from the articles using a standardized collection tool.
RESULTS A total of 43 articles describing the use of stem cell therapies for the treatment of chronic wounds were included in this review. While stem cell therapies have been explored in in vitro and in vivo applications in the past, recent efforts are geared towards assessing their clinical role. A review of the literature revealed that adipose-derived stem cells, bone marrow-derived stem cells, bone marrow-derived mononuclear cells, epidermally-derived mesenchymal stem cells, fibroblast stem cells, keratinocyte stem cells, placental mesenchymal stem cells, and umbilical cord mesenchymal stem cells have all been employed in the treatment of chronic wounds of various etiologies. Most recently, embryonic stem cells have emerged as a novel stem cell therapy with the capacity for multifaceted germ cell layer differentiation. With the capacity for self-renewal and differentiation, stem cells can enrich existing cell populations in chronic wounds in order to overcome barriers impeding the progression of wound healing. Further, stem cell therapies can be utilized to augment cell engraftment, signaling and activity, and resultant patient outcomes.
CONCLUSION Assessing observed clinical outcomes, potential for stem cell use, and relevant therapeutic challenges allows wound care stakeholders to make informed decisions regarding optimal treatment approaches for their patients’ chronic wounds.
Collapse
Affiliation(s)
- Anjali C Raghuram
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Roy P Yu
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Andrea Y Lo
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Cynthia J Sung
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Melissa Bircan
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Holly J Thompson
- Wilson Dental Library, Herman Ostrow School of Dentistry of USC, Los Angeles, CA 90089, United States
| | - Alex K Wong
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| |
Collapse
|
39
|
Przekora A. A Concise Review on Tissue Engineered Artificial Skin Grafts for Chronic Wound Treatment: Can We Reconstruct Functional Skin Tissue In Vitro? Cells 2020; 9:cells9071622. [PMID: 32640572 PMCID: PMC7407512 DOI: 10.3390/cells9071622] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic wounds occur as a consequence of a prolonged inflammatory phase during the healing process, which precludes skin regeneration. Typical treatment for chronic wounds includes application of autografts, allografts collected from cadaver, and topical delivery of antioxidant, anti-inflammatory, and antibacterial agents. Nevertheless, the mentioned therapies are not sufficient for extensive or deep wounds. Moreover, application of allogeneic skin grafts carries high risk of rejection and treatment failure. Advanced therapies for chronic wounds involve application of bioengineered artificial skin substitutes to overcome graft rejection as well as topical delivery of mesenchymal stem cells to reduce inflammation and accelerate the healing process. This review focuses on the concept of skin tissue engineering, which is a modern approach to chronic wound treatment. The aim of the article is to summarize common therapies for chronic wounds and recent achievements in the development of bioengineered artificial skin constructs, including analysis of biomaterials and cells widely used for skin graft production. This review also presents attempts to reconstruct nerves, pigmentation, and skin appendages (hair follicles, sweat glands) using artificial skin grafts as well as recent trends in the engineering of biomaterials, aiming to produce nanocomposite skin substitutes (nanofilled polymer composites) with controlled antibacterial activity. Finally, the article describes the composition, advantages, and limitations of both newly developed and commercially available bioengineered skin substitutes.
Collapse
Affiliation(s)
- Agata Przekora
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| |
Collapse
|
40
|
Wang Z, Shi C. Cellular senescence is a promising target for chronic wounds: a comprehensive review. BURNS & TRAUMA 2020; 8:tkaa021. [PMID: 32607375 PMCID: PMC7309580 DOI: 10.1093/burnst/tkaa021] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/07/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic wounds include, but are not limited to, radiation ulcers, pressure ulcers, vascular ulcers and diabetic foot ulcers. These chronic wounds can persist for years without healing and severe ulcers may lead to amputation. Unfortunately, the underlying pathologies of refractory chronic wounds are not fully characterized, and new treatments are urgently needed. Recently, increasing evidence has indicated that cell senescence plays an important role in the development of chronic wounds, and preventing cell senescence or removing senescent cells holds promise as a new therapeutic strategy. In this review, we aim to probe these latest findings to promote the understanding of cellular senescence in the pathological process and potential management of chronic wounds.
Collapse
Affiliation(s)
- Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Chongqing, 400038, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Chongqing, 400038, China
| |
Collapse
|