1
|
Bahadoran Z, Mirmiran P, Hosseinpanah F, Kashfi K, Ghasemi A. Nitric oxide-based treatments improve wound healing associated with diabetes mellitus. Med Gas Res 2025; 15:23-35. [PMID: 39436167 PMCID: PMC11515056 DOI: 10.4103/mgr.medgasres-d-24-00020] [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: 03/12/2024] [Revised: 04/16/2024] [Accepted: 06/27/2024] [Indexed: 10/23/2024] Open
Abstract
Non-healing wounds are long-term complications of diabetes mellitus (DM) that increase mortality risk and amputation-related disability and decrease the quality of life. Nitric oxide (NO·)-based treatments (i.e., use of both systemic and topical NO· donors, NO· precursors, and NO· inducers) have received more attention as complementary approaches in treatments of DM wounds. Here, we aimed to highlight the potential benefits of NO·-based treatments on DM wounds through a literature review of experimental and clinical evidence. Various topical NO·-based treatments have been used. In rodents, topical NO·-based therapy facilitates wound healing, manifested as an increased healing rate and a decreased half-closure time. The wound healing effect of NO·-based treatments is attributed to increasing local blood flow, angiogenesis induction, collagen synthesis and deposition, re-epithelization, anti-inflammatory and anti-oxidative properties, and potent broad-spectrum antibacterial effects. The existing literature lacks human clinical evidence on the safety and efficacy of NO·-based treatments for DM wounds. Translating experimental favors of NO·-based treatments of DM wounds into human clinical practice needs conducting clinical trials with well-predefined effect sizes, i.e., wound reduction area, rate of wound healing, and hospital length of stay.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhad Hosseinpanah
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Mu Y, Zhang X, Zhang L, Luo R, Zhang Y, Wang M. MSC Exosomes Containing Valproic Acid Promote Wound Healing by Modulating Inflammation and Angiogenesis. Molecules 2024; 29:4281. [PMID: 39275128 PMCID: PMC11397650 DOI: 10.3390/molecules29174281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/16/2024] Open
Abstract
PURPOSE Chronic wounds that are difficult to heal pose a major challenge for clinicians and researchers. Currently, common treatment methods focus on isolating the wound from the outside world, relying on the tissue at the wound site to grow and heal unaided. Umbilical cord mesenchymal stem cell (MSC) exosomes can promote wound healing by enhancing new blood vessel growth at the wound site. Valproic acid (VPA) reduces the inflammatory response and acts on macrophages to accelerate wound closure. In this study, VPA was loaded into umbilical cord MSC exosomes to form a drug carrier exosome (VPA-EXO) with the aim of investigating the effect of VPA-EXO on wound healing. METHODS This study first isolated and obtained umbilical cord MSC exosomes, then added VPA to the exosomes and explored the ability of VPA-EXO to promote the proliferation and migration of human skin fibroblasts (HSFs) and human umbilical vein endothelial cells (HUVECs), as well as the ability to promote the angiogenesis of HUVECs, by using scratch, Transwell, and angiogenesis assays. An in vitro cell model was established and treated with VPA-EXO, and the expression levels of inflammation and pro-angiogenesis-related proteins and genes were examined using Western blot and qRT-PCR. The therapeutic effect of VPA-EXO on promoting wound healing in a whole skin wound model was investigated using image analysis of the wound site, H&E staining, and immunohistochemical staining experiments in a mouse wound model. RESULTS The in vitro model showed that VPA-EXO effectively promoted the proliferation and migration of human skin fibroblast cells and human umbilical vein endothelial cells; significantly inhibited the expression of MMP-9, IL-1β, IL-8, TNF-α, and PG-E2; and promoted the expression of vascular endothelial growth factors. In the mouse wound model, VPA-EXO reduced inflammation at the wound site, accelerated wound healing, and significantly increased the collagen content of tissue at the wound site. CONCLUSIONS As a complex with dual efficacy in simultaneously promoting tissue regeneration and inhibiting inflammation, VPA-EXO has potential applications in tissue wound healing and vascular regeneration. In future studies, we will further investigate the mechanism of action and application scenarios of drug-loaded exosome complexes in different types of wound healing and vascular regeneration.
Collapse
Affiliation(s)
- Yujie Mu
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Xiaona Zhang
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Linfeng Zhang
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Ruting Luo
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Yin Zhang
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Min Wang
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
3
|
Jiang Z, Chen L, Huang L, Yu S, Lin J, Li M, Gao Y, Yang L. Bioactive Materials That Promote the Homing of Endogenous Mesenchymal Stem Cells to Improve Wound Healing. Int J Nanomedicine 2024; 19:7751-7773. [PMID: 39099796 PMCID: PMC11297574 DOI: 10.2147/ijn.s455469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024] Open
Abstract
Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jiabao Lin
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Mengyao Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
4
|
Manole CG, Soare C, Ceafalan LC, Voiculescu VM. Platelet-Rich Plasma in Dermatology: New Insights on the Cellular Mechanism of Skin Repair and Regeneration. Life (Basel) 2023; 14:40. [PMID: 38255655 PMCID: PMC10817627 DOI: 10.3390/life14010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
The skin's recognised functions may undergo physiological alterations due to ageing, manifesting as varying degrees of facial wrinkles, diminished tautness, density, and volume. Additionally, these functions can be disrupted (patho)physiologically through various physical and chemical injuries, including surgical trauma, accidents, or chronic conditions like ulcers associated with diabetes mellitus, venous insufficiency, or obesity. Advancements in therapeutic interventions that boost the skin's innate regenerative abilities could significantly enhance patient care protocols. The application of Platelet-Rich Plasma (PRP) is widely recognized for its aesthetic and functional benefits to the skin. Yet, the endorsement of PRP's advantages often borders on the dogmatic, with its efficacy commonly ascribed solely to the activation of fibroblasts by the factors contained within platelet granules. PRP therapy is a cornerstone of regenerative medicine which involves the autologous delivery of conditioned plasma enriched by platelets. This is achieved by centrifugation, removing erythrocytes while retaining platelets and their granules. Despite its widespread use, the precise sequences of cellular activation, the specific cellular players, and the molecular machinery that drive PRP-facilitated healing are still enigmatic. There is still a paucity of definitive and robust studies elucidating these mechanisms. In recent years, telocytes (TCs)-a unique dermal cell population-have shown promising potential for tissue regeneration in various organs, including the dermis. TCs' participation in neo-angiogenesis, akin to that attributed to PRP, and their role in tissue remodelling and repair processes within the interstitia of several organs (including the dermis), offer intriguing insights. Their potential to contribute to, or possibly orchestrate, the skin regeneration process following PRP treatment has elicited considerable interest. Therefore, pursuing a comprehensive understanding of the cellular and molecular mechanisms at work, particularly those involving TCs, their temporal involvement in structural recovery following injury, and the interconnected biological events in skin wound healing and regeneration represents a compelling field of study.
Collapse
Affiliation(s)
- Catalin G. Manole
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M. Voiculescu
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
5
|
Asadi R, Mostafavinia A, Amini A, Ahmadi H, Ahrabi B, Omidi H, Pourhashemi E, Hajihosseintehrani M, Rezaei F, Mohsenifar Z, Chien S, Bayat M. Acceleration of a delayed healing wound repair model in diabetic rats by additive impacts of photobiomodulation plus conditioned medium of adipose-derived stem cells. J Diabetes Metab Disord 2023; 22:1551-1560. [PMID: 37975122 PMCID: PMC10638220 DOI: 10.1007/s40200-023-01285-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/17/2023] [Indexed: 11/19/2023]
Abstract
Purpose This study aimed to investigate the effects of photobiomodulation (PBM) and conditioned medium (CM) derived from human adipose-derived stem cells (h-ASCs), both individually and in combination, on the maturation stage of an ischemic infected delayed healing wound model (IIDHWM) in type I diabetic (TIDM) rats. Methods The study involved the extraction of h-ASCs from donated fat, assessment of their immunophenotypic markers, cell culture, and extraction and concentration of CM from cultured 1 × 10^6 h-ASCs. TIDM was induced in 24 male adult rats, divided into four groups: control, CM group, PBM group (80 Hz, 0.2 J/cm2, 890 nm), and rats receiving both CM and PBM. Clinical and laboratory evaluations were conducted on days 4, 8, and 16, and euthanasia was performed using CO2 on day 16. Tensiometrical and stereological examinations were carried out using two wound samples from each rat. Results Across all evaluated factors, including wound closure ratio, microbiological, tensiometrical, and stereological parameters, similar patterns were observed. The outcomes of CM + PBM, PBM, and CM treatments were significantly superior in all evaluated parameters compared to the control group (p = 0.000 for all). Both PBM and CM + PBM treatments showed better tensiometrical and stereological results than CM alone (almost all, p = 0.000), and CM + PBM outperformed PBM alone in almost all aspects (p = 0.000). Microbiologically, both CM + PBM and PBM exhibited fewer colony-forming units (CFU) than CM alone (both, p = 0.000). Conclusion PBM, CM, and CM + PBM interventions substantially enhanced the maturation stage of the wound healing process in IIDHWM of TIDM rats by mitigating the inflammatory response and reducing CFU count. Moreover, these treatments promoted new tissue formation in the wound bed and improved wound strength. Notably, the combined effects of CM + PBM surpassed the individual effects of CM and PBM. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01285-3.
Collapse
Affiliation(s)
- Robabeh Asadi
- Department of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences and Cognitive Neuroscience, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Omidi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | - Zhaleh Mohsenifar
- Department of Pathology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA
| | - Mohammad Bayat
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA
| |
Collapse
|
6
|
Sohrabi K, Ahmadi H, Amini A, Ahrabi B, Mostafavinia A, Omidi H, Mirzaei M, Fadaei Fathabady F, Fridoni M, Rahmannia M, Chien S, Bayat M. Promising improvement in infected Wound Healing in Type two Diabetic rats by Combined effects of conditioned medium of human adipose-derived stem cells plus Photobiomodulation. Lab Anim Res 2023; 39:29. [PMID: 37964303 PMCID: PMC10648630 DOI: 10.1186/s42826-023-00178-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND We aimed to examine the accompanying and solo impacts of conditioned medium of human adipose-derived stem cells (h-ASC-COM) and photobiomodulation (PBM) on the maturation stage of an ischemic infected delayed-healing wound model (IIDHWM) of rats with type 2 diabetes (TIIDM). RESULTS Outcomes of the wound closure ratio (WCR) results, tensiometrical microbiological, and stereological assessment followed almost identical patterns. While the outcomes of h-ASC-COM + PBM, PBM only, and h-ASC-COM only regimes were significantly better for all evaluated methods than those of group 1(all, p < 0.001), PBM alone and h-ASC-COM + PBM therapy achieved superior results than h-ASC-COM only (ranged from p = 0.05 to p < 0.001). In terms of tensiometrical and stereological examinations, the results of h-ASC-COM + PBM experienced better results than the PBM only (all, p < 0.001). CONCLUSIONS h-ASC-COM + PBM, PBM, and h-ASC-COM cures expressively accelerated the maturation stage in the wound healing process of IIDHWM with MRSA in TIIDM rats by diminishing the inflammatory reaction, and the microbial flora of MRSA; and increasing wound strength, WCR, number of fibroblasts, and new blood vessels. While the h-ASC-COM + PBM and PBM were more suitable than the effect of h-ASC-COM, the results of h-ASC-COM + PBM were superior to PBM only.
Collapse
Affiliation(s)
- Kaysan Sohrabi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - 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
| | - Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences and Cognitive Neuroscience, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Omidi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansooreh Mirzaei
- Department of Anatomy, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadjavad Fridoni
- Department of Biology and Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Rahmannia
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville and Noveratech LLC, Louisville, KY, USA
| | - Mohammad Bayat
- Price Institute of Surgical Research, University of Louisville and Noveratech LLC, Louisville, KY, USA.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Wu H, Liang W, Han M, Zhen Y, Chen L, Li H, An Y. Mechanisms regulating wound healing: Functional changes in biology mediated by lactate and histone lactylation. J Cell Physiol 2023; 238:2243-2252. [PMID: 37743554 DOI: 10.1002/jcp.31122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023]
Abstract
The high incidence, low healing rate and huge economic burden of wounds (especially chronic wounds) worldwide remain a great challenge for clinical staff and patients. The various stages of wound healing are regulated by paracrine or autocrine cytokines and growth factors, and the study of their intrinsic mechanisms is a prerequisite for better wound treatment. Lactate, the end product of glycolysis, plays a role in all stages of wound healing, and recent studies have identified lactate as an epigenetic regulator that regulates gene expression through histone lysine lactylation and stimulates posttranslational modifications to regulate related gene expression, thereby causing a series of biological functional changes. This article reviews the progress of research on lactate and lactylation during wound healing or in related diseases, including its involvement in immune regulation, inflammation control, and proliferative remodeling, and finally describes the prospects for lactate therapy regarding wound healing.
Collapse
Affiliation(s)
- Huiting Wu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
- Department of Natural Products Chemistry, School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Meng Han
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Lixia Chen
- Department of Natural Products Chemistry, School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Hua Li
- Department of Natural Products Chemistry, School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
8
|
Salvo J, Sandoval C, Schencke C, Acevedo F, del Sol M. Healing Effect of a Nano-Functionalized Medical-Grade Honey for the Treatment of Infected Wounds. Pharmaceutics 2023; 15:2187. [PMID: 37765158 PMCID: PMC10536296 DOI: 10.3390/pharmaceutics15092187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
Based on the qualities of Ulmo honey (Eucryphia cordifolia), a medical-grade honey (Ulmoplus®) has been developed. Relevant to this, the use of copper represents an emerging therapy for the treatment of wounds. Therefore, the aim of this study was to see how this medical-grade honey with copper nanoparticles (CuNPs) helped to heal infected or non-infected wounds. Twenty-four guinea pigs (Cavia porcellus) were divided into four groups for phase 1 (without and with infection, U + F1 and U + F2), and two groups for phase 2 (selected formulation, without and with infection, U + F2NI and U + F2I). Bacteriological and histopathological studies, collagen fibers content evaluation, and stereological analysis were performed. The selected formulation displayed the same antibacterial potency as Ulmoplus®, indicating that this medical-grade honey by itself can be used as an antibacterial agent. However, the evaluation of collagen content demonstrated a significant increase in fibroblast and type III collagen fibers for infected and uninfected groups, which correlated with the histopathological study. Therefore, it is correct to affirm that adding CuNPs to Ulmoplus® improved the maturation of collagen fibers. Finally, polymorphonuclear cells presented similar values between experimental groups, which would indicate that the formulation under study was able to regulate the inflammatory process despite their infectious condition.
Collapse
Affiliation(s)
- Jessica Salvo
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Temuco 4811230, Chile;
- Programa de Doctorado en Ciencias Morfológicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Cristian Sandoval
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile;
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile
| | - Carolina Schencke
- Carrera de Psicología, Facultad de Ciencias Sociales y Humanidades, Universidad Autónoma de Chile, Temuco 4810101, Chile
| | - Francisca Acevedo
- Departamento de Ciencias Básicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile;
- Núcleo Científico-Tecnológico en Biorecursos (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile
| | - Mariano del Sol
- Programa de Doctorado en Ciencias Morfológicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
- Departamento de Ciencias Básicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile;
- Centro de Excelencia en Estudios Morfológicos y Quirúrgicos (CEMyQ), Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
9
|
Salvo J, Sandoval C, Schencke C, Acevedo F, del Sol M. Healing Effect of a Nano-Functionalized Medical-Grade Honey for the Treatment of Infected Wounds. Pharmaceutics 2023; 15:2187. [DOI: https:/doi.org/10.3390/pharmaceutics15092187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Based on the qualities of Ulmo honey (Eucryphia cordifolia), a medical-grade honey (Ulmoplus®) has been developed. Relevant to this, the use of copper represents an emerging therapy for the treatment of wounds. Therefore, the aim of this study was to see how this medical-grade honey with copper nanoparticles (CuNPs) helped to heal infected or non-infected wounds. Twenty-four guinea pigs (Cavia porcellus) were divided into four groups for phase 1 (without and with infection, U + F1 and U + F2), and two groups for phase 2 (selected formulation, without and with infection, U + F2NI and U + F2I). Bacteriological and histopathological studies, collagen fibers content evaluation, and stereological analysis were performed. The selected formulation displayed the same antibacterial potency as Ulmoplus®, indicating that this medical-grade honey by itself can be used as an antibacterial agent. However, the evaluation of collagen content demonstrated a significant increase in fibroblast and type III collagen fibers for infected and uninfected groups, which correlated with the histopathological study. Therefore, it is correct to affirm that adding CuNPs to Ulmoplus® improved the maturation of collagen fibers. Finally, polymorphonuclear cells presented similar values between experimental groups, which would indicate that the formulation under study was able to regulate the inflammatory process despite their infectious condition.
Collapse
Affiliation(s)
- Jessica Salvo
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Temuco 4811230, Chile
- Programa de Doctorado en Ciencias Morfológicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Cristian Sandoval
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile
| | - Carolina Schencke
- Carrera de Psicología, Facultad de Ciencias Sociales y Humanidades, Universidad Autónoma de Chile, Temuco 4810101, Chile
| | - Francisca Acevedo
- Departamento de Ciencias Básicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile
- Núcleo Científico-Tecnológico en Biorecursos (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile
| | - Mariano del Sol
- Programa de Doctorado en Ciencias Morfológicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
- Departamento de Ciencias Básicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile
- Centro de Excelencia en Estudios Morfológicos y Quirúrgicos (CEMyQ), Facultad de Medicina, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
10
|
Zhang Y, Zhu Y, Ma P, Wu H, Xiao D, Zhang Y, Sui X, Zhang L, Dong A. Functional carbohydrate-based hydrogels for diabetic wound therapy. Carbohydr Polym 2023; 312:120823. [PMID: 37059550 DOI: 10.1016/j.carbpol.2023.120823] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Diabetes wound are grave and universal complications of diabetes. Owing to poor treatment course, high amputation rate and mortality, diabetes wound treatment and care have become a global challenge. Wound dressings have received much attention due to their ease of use, good therapeutic effect, and low costs. Among them, carbohydrate-based hydrogels with excellent biocompatibility are considered to be the best candidates for wound dressings. Based on this, we first systematically summarized the problems and healing mechanism of diabetes wounds. Next, common treatment methods and wound dressings were discussed, and the application of various carbohydrate-based hydrogels and their corresponding functionalization (antibacterial, antioxidant, autoxidation and bioactive substance delivery) in the treatment of diabetes wounds were emphatically introduced. Ultimately, the future development of carbohydrate-based hydrogel dressings was proposed. This review aims to provide a deeper understanding of wound treatment and theoretical support for the design of hydrogel dressings.
Collapse
Affiliation(s)
- Yu Zhang
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China
| | - Yingnan Zhu
- Institute of Drug Discovery and Development, Center for Drug Safety Evaluation and Research, Zhengzhou University, Zhengzhou 450001, People's Republic of China.
| | - Peirong Ma
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China
| | - Haixia Wu
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China.
| | - Douxin Xiao
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China
| | - Yanling Zhang
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China
| | - Xiaonan Sui
- College of Food Science, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| | - Lei Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin 300350, People's Republic of China.
| | - Alideertu Dong
- College of Chemistry and Chemical Engineering, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, People's Republic of China.
| |
Collapse
|
11
|
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: 0.5] [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
|
12
|
Ganguly K, Espinal MM, Dutta SD, Patel DK, Patil TV, Luthfikasari R, Lim* KT. Multifunctional 3D platforms for rapid hemostasis and wound healing: Structural and functional prospects at biointerfaces. Int J Bioprint 2022; 9:648. [PMID: 36844240 PMCID: PMC9947489 DOI: 10.18063/ijb.v9i1.648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/29/2022] [Indexed: 12/05/2022] Open
Abstract
354Fabrication of multifunctional hemostats is indispensable against chronic blood loss and accelerated wound healing. Various hemostatic materials that aid wound repair or rapid tissue regeneration has been developed in the last 5 years. This review provides an overview of the three-dimensional (3D) hemostatic platforms designed through the latest technologies like electrospinning, 3D printing, and lithography, solely or in combination, for application in rapid wound healing. We critically discuss the pivotal role of micro/nano-3D topography and biomaterial properties in mediating rapid blood clots and healing at the hemostat-biointerface. We also highlight the advantages and limitations of the designed 3D hemostats. We anticipate that this review will guide the fabrication of smart hemostats of the future for tissue engineering applications.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Maria Mercedes Espinal
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Dinesh K. Patel
- Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Rachmi Luthfikasari
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ki-Taek Lim*
- Department of Biosystems Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
13
|
Bianchi E, Faccendini A, Del Favero E, Ricci C, Caliogna L, Vigani B, Pavesi FC, Perotti C, Domingues RMA, Gomes ME, Rossi S, Sandri G. Topographical and Compositional Gradient Tubular Scaffold for Bone to Tendon Interface Regeneration. Pharmaceutics 2022; 14:pharmaceutics14102153. [PMID: 36297586 PMCID: PMC9607365 DOI: 10.3390/pharmaceutics14102153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/02/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
The enthesis is an extremely specific region, localized at the tendon–bone interface (TBI) and made of a hybrid connection of fibrocartilage with minerals. The direct type of enthesis tissue is commonly subjected to full laceration, due to the stiffness gradient between the soft tissues and hard bone, and this often reoccurs after surgical reconstruction. For this purpose, the present work aimed to design and develop a tubular scaffold based on pullulan (PU) and chitosan (CH) and intended to enhance enthesis repair. The scaffold was designed with a topographical gradient of nanofibers, from random to aligned, and hydroxyapatite (HAP) nanoparticles along the tubular length. In particular, one part of the tubular scaffold was characterized by a structure similar to bone hard tissue, with a random mineralized fiber arrangement; while the other part was characterized by aligned fibers, without HAP doping. The tubular shape of the scaffold was also designed to be extemporarily loaded with chondroitin sulfate (CS), a glycosaminoglycan effective in wound healing, before the surgery. Micro CT analysis revealed that the scaffold was characterized by a continuous gradient, without interruptions from one end to the other. The gradient of the fiber arrangement was observed using SEM analysis, and it was still possible to observe the gradient when the scaffold had been hydrated for 6 days. In vitro studies demonstrated that human adipose stem cells (hASC) were able to grow and differentiate onto the scaffold, expressing the typical ECM production for tendon in the aligned zone, or bone tissue in the random mineralized part. CS resulted in a synergistic effect, favoring cell adhesion/proliferation on the scaffold surface. These results suggest that this tubular scaffold loaded with CS could be a powerful tool to support enthesis repair upon surgery.
Collapse
Affiliation(s)
- Eleonora Bianchi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Angela Faccendini
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, LITA, 20090 Segrate, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, University of Milan, LITA, 20090 Segrate, Italy
| | - Laura Caliogna
- Orthopedy, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | | | - Cesare Perotti
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| | - Rui M. A. Domingues
- 3B’s Research Group, i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics, University of Minho, 4805-017 Guimarães, Portugal
| | - Manuela E. Gomes
- 3B’s Research Group, i3Bs—Research Institute on Biomaterials Biodegradables and Biomimetics, University of Minho, 4805-017 Guimarães, Portugal
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
- Correspondence:
| |
Collapse
|
14
|
Hodge JG, Zamierowski DS, Robinson JL, Mellott AJ. Evaluating polymeric biomaterials to improve next generation wound dressing design. Biomater Res 2022; 26:50. [PMID: 36183134 PMCID: PMC9526981 DOI: 10.1186/s40824-022-00291-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 08/28/2022] [Indexed: 11/24/2022] Open
Abstract
Wound healing is a dynamic series of interconnected events with the ultimate goal of promoting neotissue formation and restoration of anatomical function. Yet, the complexity of wound healing can often result in development of complex, chronic wounds, which currently results in a significant strain and burden to our healthcare system. The advancement of new and effective wound care therapies remains a critical issue, with the current therapeutic modalities often remaining inadequate. Notably, the field of tissue engineering has grown significantly in the last several years, in part, due to the diverse properties and applications of polymeric biomaterials. The interdisciplinary cohesion of the chemical, biological, physical, and material sciences is pertinent to advancing our current understanding of biomaterials and generating new wound care modalities. However, there is still room for closing the gap between the clinical and material science realms in order to more effectively develop novel wound care therapies that aid in the treatment of complex wounds. Thus, in this review, we discuss key material science principles in the context of polymeric biomaterials, provide a clinical breadth to discuss how these properties affect wound dressing design, and the role of polymeric biomaterials in the innovation and design of the next generation of wound dressings.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.,Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - David S Zamierowski
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jennifer L Robinson
- Department of Chemical and Petroleum Engineering, University of Kansas, Mail Stop: 3051, 3901 Rainbow Blvd, Lawrence, KS, 66160, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
15
|
Martinez-Garcia FD, van Dongen JA, Burgess JK, Harmsen MC. Matrix Metalloproteases from Adipose Tissue-Derived Stromal Cells Are Spatiotemporally Regulated by Hydrogel Mechanics in a 3D Microenvironment. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9080340. [PMID: 35892753 PMCID: PMC9332414 DOI: 10.3390/bioengineering9080340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 01/16/2023]
Abstract
Adipose tissue-derived stromal cells (ASCs) are of interest in tissue engineering and regenerative medicine (TERM) due to their easy acquisition, multipotency, and secretion of a host of factors that promote regeneration. Retention of ASCs in or around lesions is poor following direct administration. Therefore, for TERM applications, ASCs can be ‘immobilized’ via their incorporation into hydrogels such as gelatine methacryloyl (GelMA). Tweaking GelMA concentration is a common approach to approximate the mechanical properties found in organs or tissues that need repair. Distinct hydrogel mechanics influence the ability of a cell to spread, migrate, proliferate, and secrete trophic factors. Mesenchymal cells such as ASCs are potent remodellers of the extracellular matrix (ECM). Not only do ASCs deposit components, they also secrete matrix metalloproteases (MMPs) which degrade ECM. In this work, we investigated if GelMA polymer concentration influenced the expression of active MMPs by ASCs. In addition, MMPs’ presence was interrogated with regard to ASCs morphology and changes in hydrogel ultrastructure. For this, immortalised ASCs were embedded in 5%, 10%, and 15% (w/v) GelMA hydrogels, photopolymerised and cultured for 14 d. Zymography in situ indicated that MMPs had a variable, hydrogel concentration-dependent influence on ASCs-secreted MMPs. In 5% GelMA, ASCs showed a high and sustained expression of MMPs, while, in 10% and 15% GelMA, such expression was almost null. ASCs morphology based on F-actin staining showed that increasing GelMA concentrations inhibit their spreading. Scanning electron microscopy (SEM) showed that hydrogel ultrastructure in terms of pore density, pore size, and percentage porosity were not consistently influenced by cells. Interestingly, changes in ultrastructural parameters were detected also in cell-free materials, albeit without a clear trend. We conclude that hydrogel concentration and its underlying mechanics influenced MMP expression by ASCs. The exact MMPs that respond to these mechanical cues should be defined in follow-up experiments.
Collapse
Affiliation(s)
- Francisco Drusso Martinez-Garcia
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Joris Anton van Dongen
- Department of Plastic Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Janette Kay Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Correspondence:
| |
Collapse
|
16
|
Salvo J, Sandoval C. Role of copper nanoparticles in wound healing for chronic wounds: literature review. BURNS & TRAUMA 2022; 10:tkab047. [PMID: 35071652 PMCID: PMC8778594 DOI: 10.1093/burnst/tkab047] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/15/2021] [Indexed: 12/12/2022]
Abstract
Chronic wounds are defined as wounds that fail to proceed through the normal phases of wound healing in an orderly and timely manner. The most common and inevitable impairment to wound healing is the installation of an infection, usually in the case of chronic wounds. Therefore, the objective of the present review was to identify the importance of copper nanoparticles in dressings for wound healing. Nanoparticles such as silver, gold and copper combat infectious processes through the inhibition of protein synthesis, peroxidation of the cell membrane and destroying the nucleic acids of bacteria and viruses. Among bioactive nanoparticles, copper plays a complex role in various cells, it modulates several cytokines and growth factor mechanisms of action and is essentially involved in all stages of the wound healing process. More importantly, copper plays a key role in skin regeneration and angiogenesis and accelerates the healing process through induction of vascular endothelial growth factor (VEGF) and angiogenesis by hypoxia-induced factor-1-alpha (HIF-1α) action where copper enhances HIF-1α expression and HIF-1α binding to the critical motifs in the promoter and putative enhancer regions of HIF-1-regulated genes.
Collapse
Affiliation(s)
- Jessica Salvo
- Escuela de Enfermería, Facultad de Ciencias, Universidad Mayor, Chile
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, 5310431, Osorno, Chile
| |
Collapse
|
17
|
Trubelja A, Kasper FK, Farach-Carson MC, Harrington DA. Bringing hydrogel-based craniofacial therapies to the clinic. Acta Biomater 2022; 138:1-20. [PMID: 34743044 PMCID: PMC9234983 DOI: 10.1016/j.actbio.2021.10.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 01/17/2023]
Abstract
This review explores the evolution of the use of hydrogels for craniofacial soft tissue engineering, ranging in complexity from acellular injectable fillers to fabricated, cell-laden constructs with complex compositions and architectures. Addressing both in situ and ex vivo approaches, tissue restoration secondary to trauma or tumor resection is discussed. Beginning with relatively simple epithelia of oral mucosa and gingiva, then moving to more functional units like vocal cords or soft tissues with multilayer branched structures, such as salivary glands, various approaches are presented toward the design of function-driven architectures, inspired by native tissue organization. Multiple tissue replacement paradigms are presented here, including the application of hydrogels as structural materials and as delivery platforms for cells and/or therapeutics. A practical hierarchy is proposed for hydrogel systems in craniofacial applications, based on their material and cellular complexity, spatial order, and biological cargo(s). This hierarchy reflects the regulatory complexity dictated by the Food and Drug Administration (FDA) in the United States prior to commercialization of these systems for use in humans. The wide array of available biofabrication methods, ranging from simple syringe extrusion of a biomaterial to light-based spatial patterning for complex architectures, is considered within the history of FDA-approved commercial therapies. Lastly, the review assesses the impact of these regulatory pathways on the translational potential of promising pre-clinical technologies for craniofacial applications. STATEMENT OF SIGNIFICANCE: While many commercially available hydrogel-based products are in use for the craniofacial region, most are simple formulations that either are applied topically or injected into tissue for aesthetic purposes. The academic literature previews many exciting applications that harness the versatility of hydrogels for craniofacial soft tissue engineering. One of the most exciting developments in the field is the emergence of advanced biofabrication methods to design complex hydrogel systems that can promote the functional or structural repair of tissues. To date, no clinically available hydrogel-based therapy takes full advantage of current pre-clinical advances. This review surveys the increasing complexity of the current landscape of available clinical therapies and presents a framework for future expanded use of hydrogels with an eye toward translatability and U.S. regulatory approval for craniofacial applications.
Collapse
Affiliation(s)
- Alen Trubelja
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - F Kurtis Kasper
- Department of Orthodontics, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States; Department of BioSciences, Rice University, Houston, TX 77005, United States
| | - Daniel A Harrington
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, UTHealth Science Center at Houston, Houston, TX 77054, United States; Department of Bioengineering, Rice University, Houston, TX 77005, United States; Department of BioSciences, Rice University, Houston, TX 77005, United States.
| |
Collapse
|
18
|
Yi Y, Xie C, Liu J, Zheng Y, Wang J, Lu X. Self-adhesive hydrogels for tissue engineering. J Mater Chem B 2021; 9:8739-8767. [PMID: 34647120 DOI: 10.1039/d1tb01503f] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hydrogels consisting of a three-dimensional hydrophilic network of biocompatible polymers have been widely used in tissue engineering. Owing to their tunable mechanical properties, hydrogels have been applied in both hard and soft tissues. However, most hydrogels lack self-adhesive properties that enable integration with surrounding tissues, which may result in suture or low repair efficacy. Self-adhesive hydrogels (SAHs), an emerging class of hydrogels based on a combination of three-dimensional hydrophilic networks and self-adhesive properties, continue to garner increased attention in recent years. SAHs exhibit reliable and suitable adherence to tissues, and easily integrate into tissues to promote repair efficiency. SAHs are designed either by mimicking the adhesion mechanism of natural organisms, such as mussels and sandcastle worms, or by using supramolecular strategies. This review summarizes the design and processing strategies of SAHs, clarifies underlying adhesive mechanisms, and discusses their applications in tissue engineering, as well as future challenges.
Collapse
Affiliation(s)
- Yating Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Chaoming Xie
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| | - Jin Liu
- Lab for Aging Research and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yonghao Zheng
- School of Optoelectronic Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| |
Collapse
|
19
|
Fan F, Saha S, Hanjaya-Putra D. Biomimetic Hydrogels to Promote Wound Healing. Front Bioeng Biotechnol 2021; 9:718377. [PMID: 34616718 PMCID: PMC8488380 DOI: 10.3389/fbioe.2021.718377] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
Wound healing is a common physiological process which consists of a sequence of molecular and cellular events that occur following the onset of a tissue lesion in order to reconstitute barrier between body and external environment. The inherent properties of hydrogels allow the damaged tissue to heal by supporting a hydrated environment which has long been explored in wound management to aid in autolytic debridement. However, chronic non-healing wounds require added therapeutic features that can be achieved by incorporation of biomolecules and supporting cells to promote faster and better healing outcomes. In recent decades, numerous hydrogels have been developed and modified to match the time scale for distinct stages of wound healing. This review will discuss the effects of various types of hydrogels on wound pathophysiology, as well as the ideal characteristics of hydrogels for wound healing, crosslinking mechanism, fabrication techniques and design considerations of hydrogel engineering. Finally, several challenges related to adopting hydrogels to promote wound healing and future perspectives are discussed.
Collapse
Affiliation(s)
- Fei Fan
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Sanjoy Saha
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Donny Hanjaya-Putra
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
20
|
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: 14] [Impact Index Per Article: 4.7] [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
|
21
|
Stan D, Tanase C, Avram M, Apetrei R, Mincu NB, Mateescu AL, Stan D. Wound healing applications of creams and "smart" hydrogels. Exp Dermatol 2021; 30:1218-1232. [PMID: 34009648 PMCID: PMC8453519 DOI: 10.1111/exd.14396] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/28/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022]
Abstract
Although superficial wounds are often easy to treat for healthy individuals, there are some more severe types of wounds (burns, ulcers, diabetic wounds, etc.) that are a challenge for clinicians. A good therapeutic result is based on the delivery of a treatment at the right time, for the right patient. Our goal was to sum up useful knowledge regarding wound healing and wound treatments, based on creams and hydrogels with various active ingredients. We concluded that both preparations have application in preventing infections and promoting healing, but their efficacy is clearly conditioned by the type, depth, severity of the wound and patient profile. However, due to their superior versatility and capability of maintaining the integrity and functionality of the active ingredient, as well as it is controlled release at site, hydrogels are more suited for incorporating different active ingredients. New wound healing devices can combine smart hydrogel dressings with physical therapies to deliver a more efficient treatment to patients if the indications are appropriate.
Collapse
Affiliation(s)
- Diana Stan
- DDS Diagnostic, Bucharest, Romania.,Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Cristiana Tanase
- Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania.,Biochemistry - Proteomics Department, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | | | | | | | | |
Collapse
|
22
|
Sivaraj D, Chen K, Chattopadhyay A, Henn D, Wu W, Noishiki C, Magbual NJ, Mittal S, Mermin-Bunnell AM, Bonham CA, Trotsyuk AA, Barrera JA, Padmanabhan J, Januszyk M, Gurtner GC. Hydrogel Scaffolds to Deliver Cell Therapies for Wound Healing. Front Bioeng Biotechnol 2021; 9:660145. [PMID: 34012956 PMCID: PMC8126987 DOI: 10.3389/fbioe.2021.660145] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Cutaneous wounds are a growing global health burden as a result of an aging population coupled with increasing incidence of diabetes, obesity, and cancer. Cell-based approaches have been used to treat wounds due to their secretory, immunomodulatory, and regenerative effects, and recent studies have highlighted that delivery of stem cells may provide the most benefits. Delivering these cells to wounds with direct injection has been associated with low viability, transient retention, and overall poor efficacy. The use of bioactive scaffolds provides a promising method to improve cell therapy delivery. Specifically, hydrogels provide a physiologic microenvironment for transplanted cells, including mechanical support and protection from native immune cells, and cell-hydrogel interactions may be tailored based on specific tissue properties. In this review, we describe the current and future directions of various cell therapies and usage of hydrogels to deliver these cells for wound healing applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
23
|
Multiplex Analysis of Adipose-Derived Stem Cell (ASC) Immunophenotype Adaption to In Vitro Expansion. Cells 2021; 10:cells10020218. [PMID: 33499095 PMCID: PMC7911224 DOI: 10.3390/cells10020218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
In order to enhance the therapeutic potential, it is important that sufficient knowledge regarding the dynamic changes of adipose-derived stem cell (ASC) immunophenotypical and biological properties during in vitro growth is available. Consequently, we embarked on a study to follow the evolution of highly defined cell subsets from three unrelated donors in the course of eight passages on tissue culture polystyrene. The co-expression patterns were defined by panels encompassing seven and five cell surface markers, including CD34, CD146, CD166, CD200, CD248, CD271, and CD274 and CD29, CD31, CD36, CD201, and Stro-1, respectively. The analysis was performed using multichromatic flow cytometry. We observed a major paradigm shift, where the CD166-CD34+ combination which was found across all cell subsets early in the culture was replaced by the CD166+ phenotype as the population homogeneity increased with time. At all analysis points, the cultures were dominated by a few major clones that were highly prevalent in most of the donors. The selection process resulted in two predominant clones in the larger panel (CD166+CD34-CD146-CD271- CD274-CD248-CD200- and CD166+CD34+ CD146-CD271-CD274-CD248-CD200-) and one clone in the smaller panel (CD29+CD201+CD36- Stro-1- CD31-). The minor subsets, including CD166+CD34-CD146-CD271+CD274-CD248-CD200- and CD166+CD34+CD146+CD271-CD274-CD248-CD200-, and CD29+CD201-CD36-Stro-1-CD31-, CD29+CD201+CD36-Stro-1+CD31-, and CD29+CD201+CD36+Stro-1-CD31-, in the seven and five marker panels, respectively, were, on the other, hand highly fluctuating and donor-dependent. The results demonstrate that only a limited number of phenotypical repertoires are possible in ASC cultures. Marked differences in their relative occurrence between distinct individuals underscore the need for potency standardization of different ASC preparation to improve the clinical outcome.
Collapse
|
24
|
Landscape of transcription and expression regulated by DNA methylation related to age of donor and cell passage in adipose-derived mesenchymal stem cells. Aging (Albany NY) 2020; 12:21186-21201. [PMID: 33130636 PMCID: PMC7695361 DOI: 10.18632/aging.103809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022]
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are pluripotent stromal cells that can differentiate into a variety of cell types, including skin cells. High-throughput sequencing was performed on cells of different ages and cell passage, obtaining their methylation, mRNA expression, and protein profile data. The stemness of each sample was then calculated using the TCGAbiolinks package in R. Co-expression modules were identified using WGCNA, and a crosstalk analysis was performed on the corresponding modules. The ClusterProfile package was used for the functional annotation of module genes. Finally, the regulatory network diagram was visualized using the Cytoscape software. First, a total of 16 modules were identified, where 3 modules were screened that were most relevant to the phenotype. 29 genes were screened in combination of the RNA seq, DNA methylation seq and protein iTRAQ. Finally, a comprehensive landscape comprised of RNA expression, DNA methylation and protein profiles of age relevant ADSCs was constructed. Overall, the different omics of ADSCs were comprehensively analyzed in order to reveal mechanisms pertaining to their growth and development. The effects of age, cell passage, and stemness on the therapeutic effect of ADSCs were explored. Additionally, a theoretical basis for selecting appropriate ADSC donors for regenerative medicine was provided.
Collapse
|
25
|
Analysis of Cell-seeded, Collagen-rich Hydrogel for Wound Healing. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e3049. [PMID: 32983797 PMCID: PMC7489666 DOI: 10.1097/gox.0000000000003049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/22/2020] [Indexed: 11/27/2022]
Abstract
Our laboratory has previously developed a novel collagen-rich hydrogel (cHG), which significantly increases the speed of wound healing in diabetic rats. Methods In this study, we examine the in vitro survival and migration of fibroblasts, endothelial cells, and adipose-derived stem cells in a novel cHG. Furthermore, we test the ability of adipose-derived stem cell-seeded cHG to support cell survival and accelerate healing in vivo. Results In vitro, cell survival within the cHG was retained for 25 days. We were unable to detect cellular migration into, out of, or through cHG. In the in vivo model, bioluminescence of stem cells seeded within the cHG in diabetic rat wounds was detected until day 10. Rate of wound closure was higher for cHG plus adipose-derived stem cells versus control from day 2 until day 16 and significant on days 6, 8, and 12 (P < 0.05). This significant difference was also observed on day 16 by histology (P ≤ 0.05). Conclusions We conclude that cHG is a good candidate for delivering adipose-derived stem cells, endothelial cells, and fibroblasts to wounds. Future studies will determine whether the delivery of combinations of different cell lines in cHG further enhances wound healing.
Collapse
|
26
|
Kamat P, Frueh FS, McLuckie M, Sanchez-Macedo N, Wolint P, Lindenblatt N, Plock JA, Calcagni M, Buschmann J. Adipose tissue and the vascularization of biomaterials: Stem cells, microvascular fragments and nanofat-a review. Cytotherapy 2020; 22:400-411. [PMID: 32507607 DOI: 10.1016/j.jcyt.2020.03.433] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/27/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Tissue defects in the human body after trauma and injury require precise reconstruction to regain function. Hence, there is a great demand for clinically translatable approaches with materials that are both biocompatible and biodegradable. They should also be able to adequately integrate within the tissue through sufficient vascularization. Adipose tissue is abundant and easily accessible. It is a valuable tissue source in regenerative medicine and tissue engineering, especially with regard to its angiogenic potential. Derivatives of adipose tissue, such as microfat, nanofat, microvascular fragments, stromal vascular fraction and stem cells, are commonly used in research, but also clinically to enhance the vascularization of implants and grafts at defect sites. In plastic surgery, adipose tissue is harvested via liposuction and can be manipulated in three ways (macro-, micro- and nanofat) in the operating room, depending on its ultimate use. Whereas macro- and microfat are used as a filling material for soft tissue injuries, nanofat is an injectable viscous extract that primarily induces tissue remodeling because it is rich in growth factors and stem cells. In contrast to microfat that adds volume to a defect site, nanofat has the potential to be easily combined with scaffold materials due to its liquid and homogenous consistency and is particularly attractive for blood vessel formation. The same is true for microvascular fragments that are easily isolated from adipose tissue through collagenase digestion. In preclinical animal models, it has been convincingly shown that these vascular fragments inosculate with host vessels and subsequently accelerate scaffold perfusion and host tissue integration. Adipose tissue is also an ideal source of stem cells. It yields larger quantities of cells than any other source and is easier to access for both the patient and doctor compared with other sources such as bone marrow. They are often used for tissue regeneration in combination with biomaterials. Adipose-derived stem cells can be applied unmodified or as single cell suspensions. However, certain pretreatments, such as cultivation under hypoxic conditions or three-dimensional spheroids production, may provide substantial benefit with regard to subsequent vascularization in vivo due to induced growth factor production. In this narrative review, derivatives of adipose tissue and the vascularization of biomaterials are addressed in a comprehensive approach, including several sizes of derivatives, such as whole fat flaps for soft tissue engineering, nanofat or stem cells, their secretome and exosomes. Taken together, it can be concluded that adipose tissue and its fractions down to the molecular level promote, enhance and support vascularization of biomaterials. Therefore, there is a high potential of the individual fat component to be used in regenerative medicine.
Collapse
Affiliation(s)
- Pranitha Kamat
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Florian S Frueh
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Michelle McLuckie
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nadia Sanchez-Macedo
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Petra Wolint
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nicole Lindenblatt
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jan A Plock
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
Cui Q, Zhang D, Kong D, Tang J, Liao X, Yang Q, Ren J, Gong Y, Wu G. Co-transplantation with adipose-derived cells to improve parathyroid transplantation in a mice model. Stem Cell Res Ther 2020; 11:200. [PMID: 32456711 PMCID: PMC7249357 DOI: 10.1186/s13287-020-01733-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/29/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background Accidentally removed parathyroid glands are still challenging in neck surgery, leading to hypoparathyroidism characterized with abnormally low levels of parathyroid hormone. Parathyroid auto-transplantation is usually applied in compensation. To improve the efficiency of parathyroid transplantation, we introduced a method by co-transplanting with adipose-derived cells, including stromal vascular fractions (SVFs) and adipose-derived stem cells (ADSCs), and investigated the underlying molecular mechanisms involved in parathyroid transplantation survival. Methods Rat and human parathyroid tissues were transplanted into nude mice as parathyroid transplantation model to examine the effects of SVFs and ADSCs on grafts angiogenesis and survival rates, including blood vessel assembly and parathyroid hormone levels. Several angiogenic factors, such as vascular endothelial growth factor (VEGF)-A and fibroblast growth factor (FGF) 2, were assessed in parathyroid grafts. The effects of hypoxia were investigated on ADSCs. The modulatory roles of the eyes absent homolog 1 (EYA1), which is vital in parathyroid development, was also investigated on angiogenic factor production and secretion by ADSCs. All experimental data were statistically processed. Student’s t test was used to assess significant differences between 2 groups. For multiple comparisons with additional interventions, two-way ANOVA followed by Tukey’s post hoc test was performed. P < 0.05 was considered as significant. Results SVFs improve rat parathyroid transplantation survival and blood vessel assembly, as well as FGF2 and VEGF-A expression levels in parathyroid transplantation mice. Functional human parathyroid grafts have higher microvessel density and increased VEGF-A expression. The supernatant of ADSCs induced tubule formation and migration of human endothelial cells in vitro. Hypoxia had no effect on proliferation and apoptosis of human ADSCs but induced higher angiogenic factor levels of VEGF-A and FGF2, modulated by EYA1, which was confirmed by parathyroid glands transplantation in mice. Conclusions Adipose-derived cells, including ADSCs and SVFs, improve parathyroid transplantation survival via promoting angiogenesis through EYA1-regulating angiogenetic factors in vitro and in vivo. Our studies proved an effective method to improve the parathyroid autotransplantation, which is promising for clinical patients with hypoparathyroidism when parathyroid glands were accidentally injured, removed, or devascularized.
Collapse
Affiliation(s)
- Qiuxia Cui
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Dan Zhang
- Department of Anesthesiology, Xiamen Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Deguang Kong
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Jianing Tang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Xing Liao
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Qian Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Jiangbo Ren
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China.
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, China.
| |
Collapse
|
28
|
Op 't Veld RC, Walboomers XF, Jansen JA, Wagener FADTG. Design Considerations for Hydrogel Wound Dressings: Strategic and Molecular Advances. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:230-248. [PMID: 31928151 DOI: 10.1089/ten.teb.2019.0281] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wound dressings are traditionally used to protect a wound and to facilitate healing. Currently, their function is expanding. There is an urgent need for new smart products that not only act as a protective barrier but also actively support the wound healing process. Hydrogel dressings are an example of such innovative products and typically facilitate wound healing by providing a hospitable and moist environment in which cells can thrive, while the wound can still breathe and exudate can be drained. These dressings also tend to be less painful or have a soothing effect and allow for additional drug delivery. In this review, various strategic and molecular design considerations are discussed that are relevant for developing a hydrogel into a wound dressing product. These considerations vary from material choice to ease of use and determine the dressing's final properties, application potential, and benefits for the patient. The focus of this review lies on identifying and explaining key aspects of hydrogel wound dressings and their relevance in the different phases of wound repair. Molecular targets of wound healing are discussed that are relevant when tailoring hydrogels toward specific wound healing scenarios. In addition, the potential of hydrogels is reviewed as medicine advances from a repair-based wound healing approach toward a regenerative-based one. Hydrogels can play a key role in the transition toward personal wound care and facilitating regenerative medicine strategies by acting as a scaffold for (stem) cells and carrier/source of bioactive molecules and/or drugs. Impact statement Improved wound healing will lead to a better quality of life around the globe. It can be expected that this coincides with a reduction in health care spending, as the duration of treatment decreases. To achieve this, new and modern wound care products are desired that both facilitate healing and improve comfort and outcome for the patient. It is proposed that hydrogel wound dressings can play a pivotal role in improving wound care, and to that end, this review aims to summarize the various design considerations that can be made to optimize hydrogels for the purpose of a wound dressing.
Collapse
Affiliation(s)
- Roel C Op 't Veld
- Department of Dentistry-Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.,Department of Dentistry-Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - X Frank Walboomers
- Department of Dentistry-Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - John A Jansen
- Department of Dentistry-Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry-Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| |
Collapse
|
29
|
Marusina AI, Merleev AA, Luna JI, Olney L, Haigh NE, Yoon D, Guo C, Ovadia EM, Shimoda M, Luxardi G, Boddu S, Lal NN, Takada Y, Lam KS, Liu R, Isseroff RR, Le S, Nolta JA, Kloxin AM, Maverakis E. Tunable hydrogels for mesenchymal stem cell delivery: Integrin-induced transcriptome alterations and hydrogel optimization for human wound healing. Stem Cells 2019; 38:231-245. [PMID: 31648388 DOI: 10.1002/stem.3105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/08/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Therapeutic applications for mesenchymal stem/stromal cells (MSCs) are growing; however, the successful implementation of these therapies requires the development of appropriate MSC delivery systems. Hydrogels are ideally suited to cultivate MSCs but tuning hydrogel properties to match their specific in vivo applications remains a challenge. Thus, further characterization of how hydrogel-based delivery vehicles broadly influence MSC function and fate will help lead to the next generation of more intelligently designed delivery vehicles. To date, few attempts have been made to comprehensively characterize hydrogel impact on the MSC transcriptome. Herein, we have synthesized cell-degradable hydrogels based on bio-inert poly(ethylene glycol) tethered with specific integrin-binding small molecules and have characterized their resulting effect on the MSC transcriptome when compared with 2D cultured and untethered 3D hydrogel cultured MSCs. The 3D culture systems resulted in alterations in the MSC transcriptome, as is evident by the differential expression of genes related to extracellular matrix production, glycosylation, metabolism, signal transduction, gene epigenetic regulation, and development. For example, genes important for osteogenic differentiation were upregulated in 3D hydrogel cultures, and the expression of these genes could be partially suppressed by tethering an integrin-binding RGD peptide within the hydrogel. Highlighting the utility of tunable hydrogels, when applied to ex vivo human wounds the RGD-tethered hydrogel was able to support wound re-epithelialization, possibly due to its ability to increase PDGF expression and decrease IL-6 expression. These results will aid in future hydrogel design for a broad range of applications.
Collapse
Affiliation(s)
- Alina I Marusina
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Alexander A Merleev
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Jesus I Luna
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Laura Olney
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Nathan E Haigh
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Daniel Yoon
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Chen Guo
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Elisa M Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Michiko Shimoda
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Guillaume Luxardi
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Sucharita Boddu
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Nelvish N Lal
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Yoshikazu Takada
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California
| | - R Rivkah Isseroff
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Stephanie Le
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis, Sacramento, California
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware.,Department of Materials Science and Engineering, University of Delaware, Newark, Delaware
| | - Emanual Maverakis
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
30
|
Giatsidis G, Succar J, Waters TD, Liu W, Rhodius P, Wang C, Nilsen TJ, Chnari E, Orgill DP. Tissue-Engineered Soft-Tissue Reconstruction Using Noninvasive Mechanical Preconditioning and a Shelf-Ready Allograft Adipose Matrix. Plast Reconstr Surg 2019; 144:884-895. [PMID: 31568297 DOI: 10.1097/prs.0000000000006085] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Adipose tissue defects leading to severe functional (disability) and morphologic (disfigurement) morbidity are often treated in plastic surgery with fat grafting, which can be limited by resorption, necrosis, and cyst formation. This study aimed to assess whether adipose scaffolds could provide an environment for in situ autologous fat grafting, and to study whether adipose cell migration and proliferation (adipogenesis) within scaffolds could be enhanced by preliminarily increasing the vascularity (preconditioning) of the surrounding tissue receiving the scaffolds. METHODS Using an established rodent model of subcutaneous tissue/scaffold grafting, the authors tested the potential of a human-derived, shelf-ready, injectable, decellularized allograft adipose matrix to reconstruct soft-tissue defects when used in combination with noninvasive mechanical (suction-induced) skin preconditioning. RESULTS Combined use of the allograft adipose matrix and noninvasive skin preconditioning significantly improved long-term volume retention (50 to 80 percent higher at a 12-week follow-up) and histologic quality of reconstructed tissues compared with standard of care (autologous adipose grafts). The components of the allograft adipose matrix supported adipogenesis and angiogenesis. Combining the allograft adipose matrix with living adipose grafts mitigated negative outcomes (lower long-term volume retention, higher presence of cystic-like areas). CONCLUSIONS This study suggests that the synergistic use of the allograft adipose matrix and noninvasive tissue preconditioning provides an effective solution for improving fat grafting. These strategies can easily be tested in clinical trials and could establish the basis for a novel therapeutic paradigm in reconstructive surgery.
Collapse
Affiliation(s)
- Giorgio Giatsidis
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Julien Succar
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Trevon D Waters
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Wenyue Liu
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Patrick Rhodius
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Chenglong Wang
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Todd J Nilsen
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Evangelia Chnari
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| | - Dennis P Orgill
- From the Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School; Department of Molecular Medicine, University of Padova; Preventive Medicine, University of New Mexico; Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; and the Musculoskeletal Transplant Foundation
| |
Collapse
|
31
|
da Silva LP, Reis RL, Correlo VM, Marques AP. Hydrogel-Based Strategies to Advance Therapies for Chronic Skin Wounds. Annu Rev Biomed Eng 2019; 21:145-169. [DOI: 10.1146/annurev-bioeng-060418-052422] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chronic skin wounds are the leading cause of nontraumatic foot amputations worldwide and present a significant risk of morbidity and mortality due to the lack of efficient therapies. The intrinsic characteristics of hydrogels allow them to benefit cutaneous healing essentially by supporting a moist environment. This property has long been explored in wound management to aid in autolytic debridement. However, chronic wounds require additional therapeutic features that can be provided by a combination of hydrogels with biochemical mediators or cells, promoting faster and better healing. We survey hydrogel-based approaches with potential to improve the healing of chronic wounds by reviewing their effects as observed in preclinical models. Topics covered include strategies to ablate infection and resolve inflammation, the delivery of bioactive agents to accelerate healing, and tissue engineering approaches for skin regeneration. The article concludes by considering the relevance of treating chronic skin wounds using hydrogel-based strategies.
Collapse
Affiliation(s)
- Lucília P. da Silva
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Vitor M. Correlo
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Alexandra P. Marques
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| |
Collapse
|
32
|
Browne S, Healy KE. Matrix-assisted cell transplantation for tissue vascularization. Adv Drug Deliv Rev 2019; 146:155-169. [PMID: 30605738 DOI: 10.1016/j.addr.2018.12.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/30/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022]
Abstract
Cell therapy offers much promise for the treatment of ischemic diseases by augmenting tissue vasculogenesis. Matrix-assisted cell transplantation (MACT) has been proposed as a solution to enhance cell survival and integration with host tissue following transplantation. By designing semi synthetic matrices (sECM) with the correct physical and biochemical signals, encapsulated cells are directed towards a more angiogenic phenotype. In this review, we describe the choice of cells suitable for pro-angiogenic therapies, the properties that should be considered when designing sECM for transplantation and their relative importance. Pre-clinical models where MACT has been successfully applied to promote angiogenesis are reviewed to show the great potential of this strategy to treat ischemic conditions.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA; Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
33
|
Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 2019; 146:97-125. [PMID: 30267742 DOI: 10.1016/j.addr.2018.09.010] [Citation(s) in RCA: 449] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/15/2018] [Accepted: 09/24/2018] [Indexed: 12/19/2022]
Abstract
The enhancement of wound healing has been a goal of medical practitioners for thousands of years. The development of chronic, non-healing wounds is a persistent medical problem that drives patient morbidity and increases healthcare costs. A key aspect of many non-healing wounds is the reduced presence of vessel growth through the process of angiogenesis. This review surveys the creation of new treatments for healing cutaneous wounds through therapeutic angiogenesis. In particular, we discuss the challenges and advancement that have been made in delivering biologic, pharmaceutical and cell-based therapies as enhancers of wound vascularity and healing.
Collapse
|
34
|
Green LJ, Zhou H, Padmanabhan V, Shikanov A. Adipose-derived stem cells promote survival, growth, and maturation of early-stage murine follicles. Stem Cell Res Ther 2019; 10:102. [PMID: 30898159 PMCID: PMC6427888 DOI: 10.1186/s13287-019-1199-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/14/2019] [Accepted: 03/01/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency is a common complication of anticancer treatments in young women and girls. The ovary is a complex, highly regulated reproductive organ, whose proper function is contingent upon the bidirectional endocrine, paracrine, and autocrine signaling. These factors facilitate the development of the follicles, the functional units of the ovary, to progress from the gonadotropin-independent, paracrine-controlled early stage to the gonadotropin-dependent, endocrine-controlled later stage. We hypothesized that the low survival rate of individually cultured early-stage follicles could be improved with co-culture of adipose-derived stem cells (ADSCs) that secrete survival- and growth-promoting factors. MATERIALS AND METHODS Ovarian follicles ranging from 85 to 115 μm in diameter, from 10- to 12-day-old B6CBAF1 mice were mechanically isolated and co-encapsulated with ADSCs within alginate-based 3D culture system. The follicles were cultured for 14 days, imaged using light microscopy every 2 days, and matured at the end. Follicle media were changed every 2 days and collected for hormone measurements. Follicle diameter, morphology, number of transzonal projections, and survival and maturation rates were recorded. Statistical analyses using one- and two-way ANOVA were performed to compare hormone levels, survival of the follicles and ADSCs, oocyte maturation rates, and follicle growth. RESULTS The co-encapsulation of the follicles with ADSCs increased follicle survival, ranging from 42.4% for the 86-95 μm to 86.2% for the 106-115-μm follicle size group. Co-culture also improved the follicle growth, the rate of antrum formation and oocyte maturation compared to the follicles cultured alone. The levels of androstenedione, estradiol, and progesterone of co-encapsulated follicles increased progressively with time in culture. CONCLUSIONS To our knowledge, this is the first report of an in vitro system utilizing mouse adipose-derived stem cells to support the development of the mouse follicles. Our findings suggest that co-encapsulation of ADSCs with early-stage follicles supports follicular development, through secretion of cytokines that promote follicular survival, antrum formation, and meiotic competence. The unique 3D culture system that supports the survival of both cell types has translational implications, as ADSCs could be used as an autologous source for in vitro maturation of early-stage human follicles.
Collapse
Affiliation(s)
- Lisa J. Green
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI USA
- Present Address: Department of Obstetrics and Gynecology, University of South Carolina School of Medicine, Greenville, SC USA
| | - Hong Zhou
- Department of Biomedical Engineering, University of Michigan, 2126 Lurie Biomedical Engineering Building, 1101 Beal Ave., Ann Arbor, MI 48109 USA
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI USA
- Department of Pediatrics, University of Michigan, Ann Arbor, MI USA
- Department Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, 2126 Lurie Biomedical Engineering Building, 1101 Beal Ave., Ann Arbor, MI 48109 USA
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
35
|
Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: A Cellular Perspective. Physiol Rev 2019; 99:665-706. [PMID: 30475656 PMCID: PMC6442927 DOI: 10.1152/physrev.00067.2017] [Citation(s) in RCA: 1301] [Impact Index Per Article: 260.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 02/08/2023] Open
Abstract
Wound healing is one of the most complex processes in the human body. It involves the spatial and temporal synchronization of a variety of cell types with distinct roles in the phases of hemostasis, inflammation, growth, re-epithelialization, and remodeling. With the evolution of single cell technologies, it has been possible to uncover phenotypic and functional heterogeneity within several of these cell types. There have also been discoveries of rare, stem cell subsets within the skin, which are unipotent in the uninjured state, but become multipotent following skin injury. Unraveling the roles of each of these cell types and their interactions with each other is important in understanding the mechanisms of normal wound closure. Changes in the microenvironment including alterations in mechanical forces, oxygen levels, chemokines, extracellular matrix and growth factor synthesis directly impact cellular recruitment and activation, leading to impaired states of wound healing. Single cell technologies can be used to decipher these cellular alterations in diseased states such as in chronic wounds and hypertrophic scarring so that effective therapeutic solutions for healing wounds can be developed.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Nina Kosaric
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Clark A Bonham
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
36
|
Fibrin Glue Enhances Adipose-Derived Stromal Cell Cytokine Secretion and Survival Conferring Accelerated Diabetic Wound Healing. Stem Cells Int 2018; 2018:1353085. [PMID: 30662467 PMCID: PMC6313983 DOI: 10.1155/2018/1353085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/13/2018] [Indexed: 02/08/2023] Open
Abstract
Introduction Although chronic wounds are a major personal and economic burden, treatment options are still limited. Among those options, adipose-derived stromal cell- (ASC-) based therapies rank as a promising approach but are restricted by the harsh wound environment. Here we use a commercially available fibrin glue to provide a deliverable niche for ASCs in chronic wounds. Material and Methods To investigate the in vitro effect of fibrin glue, cultivation experiments were performed and key cytokines for regeneration were quantified. By using an established murine chronic diabetic wound-healing model, we evaluated the influence of fibrin glue spray seeding on cell survival (In Vivo Imaging System, IVIS), wound healing (wound closure kinetics), and neovascularization of healed wounds (CD31 immunohistochemistry). Results Fibrin glue seeding leads to a significantly enhanced secretion of key cytokines (SDF-1, bFGF, and MMP-2) of human ASCs in vitro. IVIS imaging showed a significantly prolonged murine ASC survival in diabetic wounds and significantly accelerated complete wound closure in the fibrin glue seeded group. CD31 immunohistochemistry revealed significantly more neovascularization in healed wounds treated with ASCs spray seeded in fibrin glue vs. ASC injected into the wound bed. Conclusion Although several vehicles have shown to successfully act as cell carrier systems in preclinical trials, regulatory issues have prohibited clinical usage for chronic wounds. By demonstrating the ability of fibrin glue to act as a carrier vehicle for ASCs, while simultaneously enhancing cellular regenerative function and viability, this study is a proponent of clinical translation for ASC-based therapies.
Collapse
|
37
|
Single-Cell Gene Expression Analysis and Evaluation of the Therapeutic Function of Murine Adipose-Derived Stromal Cells (ASCs) from the Subcutaneous and Visceral Compartment. Stem Cells Int 2018; 2018:2183736. [PMID: 30651733 PMCID: PMC6311719 DOI: 10.1155/2018/2183736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022] Open
Abstract
Introduction Adipose-derived stromal cells (ASCs) are a promising resource for wound healing and tissue regeneration because of their multipotent properties and cytokine secretion. ASCs are typically isolated from the subcutaneous fat compartment, but can also be obtained from visceral adipose tissue. The data on their equivalence diverges. The present study analyzes the cell-specific gene expression profiles and functional differences of ASCs derived from the subcutaneous (S-ASCs) and the visceral (V-ASCs) compartment. Material and Methods Subcutaneous and visceral ASCs were obtained from mouse inguinal fat and omentum. The transcriptional profiles of the ASCs were compared on single-cell level. S-ASCs and V-ASCs were then compared in a murine wound healing model to evaluate their regenerative functionality. Results On a single-cell level, S-ASCs and V-ASCs displayed distinct transcriptional profiles. Specifically, significant differences were detected in genes associated with neoangiogenesis and tissue remodeling (for example, Ccl2, Hif1α, Fgf7, and Igf). In addition, a different subpopulation ecology could be identified employing a cluster model. Nevertheless, both S-ASCs and V-ASCs induced accelerated healing rates and neoangiogenesis in a mouse wound healing model. Conclusion With similar therapeutic potential in vivo, the significantly different gene expression patterns of ASCs from the subcutaneous and visceral compartments suggest different signaling pathways underlying their efficacy. This study clearly demonstrates that review of transcriptional results in vivo is advisable to confirm the tentative effect of cell therapies.
Collapse
|
38
|
Adipose-Derived Stem Cells Can Contribute to Vascular Network Formation in Poly(ethylene Glycol) Hydrogel Scaffolds. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0075-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Stone Ii R, Natesan S, Kowalczewski CJ, Mangum LH, Clay NE, Clohessy RM, Carlsson AH, Tassin DH, Chan RK, Rizzo JA, Christy RJ. Advancements in Regenerative Strategies Through the Continuum of Burn Care. Front Pharmacol 2018; 9:672. [PMID: 30038569 PMCID: PMC6046385 DOI: 10.3389/fphar.2018.00672] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/05/2018] [Indexed: 01/09/2023] Open
Abstract
Burns are caused by several mechanisms including flame, scald, chemical, electrical, and ionizing and non-ionizing radiation. Approximately half a million burn cases are registered annually, of which 40 thousand patients are hospitalized and receive definitive treatment. Burn care is very resource intensive as the treatment regimens and length of hospitalization are substantial. Burn wounds are classified based on depth as superficial (first degree), partial-thickness (second degree), or full-thickness (third degree), which determines the treatment necessary for successful healing. The goal of burn wound care is to fully restore the barrier function of the tissue as quickly as possible while minimizing infection, scarring, and contracture. The aim of this review is to highlight how tissue engineering and regenerative medicine strategies are being used to address the unique challenges of burn wound healing and define the current gaps in care for both partial- and full-thickness burn injuries. This review will present the current standard of care (SOC) and provide information on various treatment options that have been tested pre-clinically or are currently in clinical trials. Due to the complexity of burn wound healing compared to other skin injuries, burn specific treatment regimens must be developed. Recently, tissue engineering and regenerative medicine strategies have been developed to improve skin regeneration that can restore normal skin physiology and limit adverse outcomes, such as infection, delayed re-epithelialization, and scarring. Our emphasis will be centered on how current clinical and pre-clinical research of pharmacological agents, biomaterials, and cellular-based therapies can be applied throughout the continuum of burn care by targeting the stages of wound healing: hemostasis, inflammation, cell proliferation, and matrix remodeling.
Collapse
Affiliation(s)
- Randolph Stone Ii
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Shanmugasundaram Natesan
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Christine J Kowalczewski
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Lauren H Mangum
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States.,Extremity Trauma and Regenerative Medicine, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Nicholas E Clay
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Ryan M Clohessy
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Anders H Carlsson
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - David H Tassin
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Rodney K Chan
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Julie A Rizzo
- Burn Flight Team, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Robert J Christy
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| |
Collapse
|
40
|
Abstract
The normal wound healing process involves a well-organized cascade of biological pathways and any failure in this process leads to wounds becoming chronic. Non-healing wounds are a burden on healthcare systems and set to increase with aging population and growing incidences of obesity and diabetes. Stem cell-based therapies have the potential to heal chronic wounds but have so far seen little success in the clinic. Current research has been focused on using polymeric biomaterial systems that can act as a niche for these stem cells to improve their survival and paracrine activity that would eventually promote wound healing. Furthermore, different modification strategies have been developed to improve stem cell survival and differentiation, ultimately promoting regenerative wound healing. This review focuses on advanced polymeric scaffolds that have been used to deliver stem cells and have been tested for their efficiency in preclinical animal models of wounds.
Collapse
|
41
|
|
42
|
Promotion of Cell-Based Therapy: Special Focus on the Cooperation of Mesenchymal Stem Cell Therapy and Gene Therapy for Clinical Trial Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:103-118. [PMID: 30155859 DOI: 10.1007/5584_2018_256] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regenerative medicine (RM) is a promising new field of medicine that has mobilized several new tools to repair or replace lost or damaged cells or tissues by stimulating natural regenerative mechanisms nearby cell and tissue-based therapy approaches. However, mesenchymal stem cell (MSC) based therapy has been shown to be safe and effective to a certain degree in multiple clinical trial studies (CTSs) of several diseases, in most MSC CTSs the efficacy of treatment has been reported low. Therefore, researchers have focused on efficacy enhancing of MSC to improve migratory and homing, survival, stemness, differentiation and other therapeutic applicable properties by using different approaches. Gene therapy is one of the experimental technique tools that uses genes to change cells for therapeutic and investigation purposes. In this study has been focused on genetically modified MSCs for use in RM with an emphasis on CTSs. We highlight the basic concept of genetic modifications and also discuss recent clinical studies aspects. Recently reviewed studies show that MSC therapy with assistant gene therapy can be used in cancer therapy, heart diseases, Fanconi anemia and several other diseases.
Collapse
|
43
|
Rodrigues M, Gurtner G. Black, White, and Gray: Macrophages in Skin Repair and Disease. CURRENT PATHOBIOLOGY REPORTS 2017; 5:333-342. [PMID: 30288366 PMCID: PMC6166434 DOI: 10.1007/s40139-017-0152-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Macrophages alter their responses during the temporal stages of wound healing. During the inflammatory phase macrophages perform phagocytosis. During neovascularization macrophages activate angiogenesis. In the proliferation phase of wound healing, macrophages deposit extracellular matrix and during wound resolution macrophages phagocytize excessive cellular components. This review addresses how these changing phenotypes affect skin repair and disease. RECENT FINDINGS Macrophages can determine the outcome of repair and can shift the normal wound healing response into fibrosis or chronic wounds. Emerging single cell technologies for the first time provide us with tools to uncover macrophage origin, heterogeneity and function. SUMMARY Macrophages may exist as one population where all cells alter their phenotype in response to signals from the microenvironment. Alternatively, macrophages may exist as distinct subsets that can control wound outcomes. A clarified understanding will strengthen our knowledge of skin biology and aid in the development of wound healing therapies.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, USA
| | - Geoffrey Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, USA
| |
Collapse
|
44
|
Oh S, Kim DY, Baek MK, Byun K, Woo JH. The Effect of Human Adipose Tissue-Derived Mesenchymal Stem Cells in Rat's Subglottic Stenosis Model. Ann Otol Rhinol Laryngol 2017; 127:5-12. [PMID: 29185355 DOI: 10.1177/0003489417739920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Subglottic stenosis remains a clinical challenge. The aim of this study was to evaluate the effect of human adipose tissue-derived mesenchymal stem cells (hAMSCs) in rat model of subglottic stenosis. SUBJECTS AND METHODS Ninety-six 13-week-old male rats were enrolled in this study. They were divided into 3 groups as normal control (NC) group, a subglottic injury and media injection (SM) group, and a subglottic injury and media-stem cell injection (SMSC) group. The hAMSCs were immediately injected into subglottis after injury. Histologic characteristics of subglottis; the mRNA expressions of interleukin-1β, cyclooxygenase-2, tumor growth factor-β and basic fibroblast growth factor; and hAMSCs' survival were evaluated. RESULTS The hAMSCs survived in the subglottis of the rat until 10 days after implantation. The NC and SMSC groups had a significantly wider subglottic lumen and thinner lamina propria than the SM group at 56 days after injury. Collagen intensity of subglottis was significantly higher in the SM group than in the NC and SMSC groups at 28 days after injury. Gene expression didn't show significant difference between the SM group and the SMSC group. CONCLUSIONS The hAMSCs injection was found to be helpful for preventing subglottic stenosis in a rat model.
Collapse
Affiliation(s)
- Seyeon Oh
- 1 Center for Genomics and Proteomics & Center for Regenerative Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Dong Young Kim
- 2 Department of Otolaryngology-Head and Neck Surgery, Gachon University, Gil Medical Center, Incheon, Republic of Korea
| | - Min Kwan Baek
- 2 Department of Otolaryngology-Head and Neck Surgery, Gachon University, Gil Medical Center, Incheon, Republic of Korea
| | - Kyunghee Byun
- 1 Center for Genomics and Proteomics & Center for Regenerative Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.,3 Department of Anatomy and Cell Biology, Gachon University, Incheon, Republic of Korea
| | - Joo Hyun Woo
- 2 Department of Otolaryngology-Head and Neck Surgery, Gachon University, Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
45
|
Zheng Y, Ji S, Wu H, Tian S, Zhang Y, Wang L, Fang H, Luo P, Wang X, Hu X, Xiao S, Xia Z. Topical administration of cryopreserved living micronized amnion accelerates wound healing in diabetic mice by modulating local microenvironment. Biomaterials 2016; 113:56-67. [PMID: 27810642 DOI: 10.1016/j.biomaterials.2016.10.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 01/09/2023]
Abstract
Approximately 25% of diabetic patients suffer from diabetic lower-extremity ulcer throughout their lives and 7%-20% of patients will eventually need an amputation despite standard care treatment. The development of new therapies to treat diabetic wounds is urgent. In this study, we used cryopreserved living micronized amnion (300-600 μm) to treat wounds in diabetic mice. Post-thaw micronized amnion retained high cell viability, as well as intact cell morphology and membrane structure. When transplanted onto the wounds of db/db mice, the cryopreserved living micronized amnion greatly promoted wound healing in diabetic mice mainly by secreting growth, inflammation, and chemotaxis-related factors that regulated macrophage migration and phenotype switch, recruited CD34+ progenitor cells, and increased neovascularization. In addition, the micronized amnion matrix can exist in the dermis and serve as a long-term dermal scaffold. These results demonstrated the potential of the cryopreserved living micronized amnion as a ready-to-use living dermal substitute that addresses multiple defective physiological processes of impaired wounds to treat diabetic ulcers and other chronic wounds in clinics.
Collapse
Affiliation(s)
- Yongjun Zheng
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shizhao Ji
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Haibin Wu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Song Tian
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yunqing Zhang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Li Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - He Fang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Pengfei Luo
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xingtong Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoyan Hu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shichu Xiao
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Zhaofan Xia
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
46
|
Clevenger TN, Luna G, Boctor D, Fisher SK, Clegg DO. Cell-mediated remodeling of biomimetic encapsulating hydrogels triggered by adipogenic differentiation of adipose stem cells. J Tissue Eng 2016; 7:2041731416670482. [PMID: 27733898 PMCID: PMC5040235 DOI: 10.1177/2041731416670482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/31/2016] [Indexed: 01/18/2023] Open
Abstract
One of the most common regenerative therapies is autologous fat grafting, which frequently suffers from unexpected volume loss. One approach is to deliver adipose stem cells encapsulated in the engineered hydrogels supportive of cell survival, differentiation, and integration after transplant. We describe an encapsulating, biomimetic poly(ethylene)-glycol hydrogel, with embedded peptides for attachment and biodegradation. Poly(ethylene)-glycol hydrogels containing an Arg–Gly–Asp attachment sequence and a matrix metalloprotease 3/10 cleavage site supported adipose stem cell survival and showed remodeling initiated by adipogenic differentiation. Arg–Gly–Asp–matrix metalloprotease 3/10 cleavage site hydrogels showed an increased number and area of lacunae or holes after adipose stem cell differentiation. Image analysis of adipose stem cells in Arg–Gly–Asp–matrix metalloprotease 3/10 cleavage site hydrogels showed larger Voronoi domains, while cell density remained unchanged. The differentiated adipocytes residing within these newly remodeled spaces express proteins and messenger RNAs indicative of adipocytic differentiation. These engineered scaffolds may provide niches for stem cell differentiation and could prove useful in soft tissue regeneration.
Collapse
Affiliation(s)
- Tracy N Clevenger
- Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Gabriel Luna
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Center for Bio-Image Informatics, University of California, Santa Barbara, CA, USA
| | - Daniel Boctor
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Steven K Fisher
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Center for Bio-Image Informatics, University of California, Santa Barbara, CA, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| |
Collapse
|
47
|
Duscher D, Luan A, Rennert RC, Atashroo D, Maan ZN, Brett EA, Whittam AJ, Ho N, Lin M, Hu MS, Walmsley GG, Wenny R, Schmidt M, Schilling AF, Machens HG, Huemer GM, Wan DC, Longaker MT, Gurtner GC. Suction assisted liposuction does not impair the regenerative potential of adipose derived stem cells. J Transl Med 2016; 14:126. [PMID: 27153799 PMCID: PMC4859988 DOI: 10.1186/s12967-016-0881-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/27/2016] [Indexed: 01/22/2023] Open
Abstract
Background Adipose-derived stem cells (ASCs) have been identified as a population of multipotent cells with promising applications in tissue engineering and regenerative medicine. ASCs are abundant in fat tissue, which can be safely harvested through the minimally invasive procedure of liposuction. However, there exist a variety of different harvesting methods, with unclear impact on ASC regenerative potential. The aim of this study was thus to compare the functionality of ASCs derived from the common technique of suction-assisted lipoaspiration (SAL) versus resection. Methods Human adipose tissue was obtained from paired abdominoplasty and SAL samples from three female donors, and was processed to isolate the stromal vascular fraction. Fluorescence-activated cell sorting was used to determine ASC yield, and cell viability was assayed. Adipogenic and osteogenic differentiation capacity were assessed in vitro using phenotypic staining and quantification of gene expression. Finally, ASCs were applied in an in vivo model of tissue repair to evaluate their regenerative potential. Results SAL specimens provided significantly fewer ASCs when compared to excised fat tissue, however, with equivalent viability. SAL-derived ASCs demonstrated greater expression of the adipogenic markers FABP-4 and LPL, although this did not result in a difference in adipogenic differentiation. There were no differences detected in osteogenic differentiation capacity as measured by alkaline phosphatase, mineralization or osteogenic gene expression. Both SAL- and resection-derived ASCs enhanced significantly cutaneous healing and vascularization in vivo, with no significant difference between the two groups. Conclusion SAL provides viable ASCs with full capacity for multi-lineage differentiation and tissue regeneration, and is an effective method of obtaining ASCs for cell-based therapies.
Collapse
Affiliation(s)
- Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria. .,Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany.
| | - Anna Luan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David Atashroo
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth A Brett
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander J Whittam
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Natalie Ho
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Lin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Raphael Wenny
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Manfred Schmidt
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Arndt F Schilling
- Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany
| | - Hans-Günther Machens
- Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany
| | - Georg M Huemer
- Section of Plastic, Aesthetic and Reconstructive Surgery, Johannes Kepler University, Linz, Austria
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
48
|
Jinno C, Morimoto N, Ito R, Sakamoto M, Ogino S, Taira T, Suzuki S. A Comparison of Conventional Collagen Sponge and Collagen-Gelatin Sponge in Wound Healing. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4567146. [PMID: 27218103 PMCID: PMC4863089 DOI: 10.1155/2016/4567146] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 01/13/2023]
Abstract
The objective of this study was to compare the effectiveness of the collagen-gelatin sponge (CGS) with that of the collagen sponge (CS) in dermis-like tissue regeneration. CGS, which achieves the sustained release of basic fibroblast growth factor (bFGF), is a promising material in wound healing. In the present study, we evaluated and compared CGSs and conventional CSs. We prepared 8 mm full-thickness skin defects on the backs of rats. Either CGSs or CSs were impregnated with normal saline solution (NSS) or 7 μg/cm(2) of bFGF solution and implanted into the defects. At 1 and 2 weeks after implantation, tissue specimens were obtained from the rats of each group (n = 3, total n = 24). The wound area, neoepithelial length, dermis-like tissue area, and the number and area of capillaries were evaluated at 1 and 2 weeks after implantation. There were no significant differences in the CGS without bFGF and CS groups. Significant improvements were observed in the neoepithelial length, the dermis-like tissue area, and the number of newly formed capillaries in the group of rats that received CGSs impregnated with bFGF. The effects on epithelialization, granulation, and vascularization of wound healing demonstrated that, as a scaffold, CGSs are equal or superior to conventional CSs.
Collapse
Affiliation(s)
- Chizuru Jinno
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-machi, Syougoin, Sakyou-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata City, Osaka 573-1010, Japan
| | - Ran Ito
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-machi, Syougoin, Sakyou-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Michiharu Sakamoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-machi, Syougoin, Sakyou-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Shuichi Ogino
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-machi, Syougoin, Sakyou-ku, Kyoto City, Kyoto 606-8507, Japan
| | - Tsuguyoshi Taira
- GUNZE Ltd. QOL Research Center Laboratory, 1 Ishiburo, Inokura Shinmachi, Ayabe, Kyoto 623-8512, Japan
| | - Shigehiko Suzuki
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-machi, Syougoin, Sakyou-ku, Kyoto City, Kyoto 606-8507, Japan
| |
Collapse
|