101
|
Hamada M, Takaya K, Wang Q, Otaki M, Imbe Y, Nakajima Y, Sakai S, Okabe K, Aramaki-Hattori N, Kishi K. Regeneration of Panniculus Carnosus Muscle in Fetal Mice Is Characterized by the Presence of Actin Cables. Biomedicines 2023; 11:3350. [PMID: 38137571 PMCID: PMC10742160 DOI: 10.3390/biomedicines11123350] [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: 10/12/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Mammalian skin, including human and mouse skin, does not regenerate completely after injury; it is repaired, leaving a scar. However, it is known that skin wounds up to a certain stage of embryonic development can regenerate. The mechanism behind the transition from regeneration to scar formation is not fully understood. Panniculus carnosus muscle (PCM) is present beneath the dermal fat layer and is a very important tissue for wound contraction. In rodents, PCM is present throughout the body. In humans, on the other hand, it disappears and becomes a shallow fascia on the trunk. Fetal cutaneous wounds, including PCM made until embryonic day 13 (E13), regenerate completely, but not beyond E14. We visualized the previously uncharacterized development of PCM in the fetus and investigated the temporal and spatial changes in PCM at different developmental stages, ranging from full regeneration to non-regeneration. Furthermore, we report that E13 epidermal closure occurs through actin cables, which are bundles of actomyosin formed at wound margins. The wound healing process of PCM suggests that actin cables may also be associated with PCM. Our findings reveal that PCM regenerates through a similar mechanism.
Collapse
Affiliation(s)
- Mariko Hamada
- Department of Plastic and Reconstructive Surgery, Tachikawa Hospital, 4-2-22 Nishiki-cho, Tachikawa-shi, Tokyo 190-8531, Japan;
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Kento Takaya
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Qi Wang
- Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-ku, Tokyo 105-8512, Japan; (Q.W.); (Y.I.)
| | - Marika Otaki
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Yuka Imbe
- Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-ku, Tokyo 105-8512, Japan; (Q.W.); (Y.I.)
| | - Yukari Nakajima
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Shigeki Sakai
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Keisuke Okabe
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Noriko Aramaki-Hattori
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.T.); (M.O.); (Y.N.); (S.S.); (K.O.); (N.A.-H.)
| |
Collapse
|
102
|
Qin W, Wang J, Hu Q, Qin R, Ma N, Zheng F, Tian W, Jiang J, Li T, Jin Y, Liao M, Qin A. Melatonin-pretreated human umbilical cord mesenchymal stem cells improved endometrium regeneration and fertility recovery through macrophage immunomodulation in rats with intrauterine adhesions†. Biol Reprod 2023; 109:918-937. [PMID: 37672216 DOI: 10.1093/biolre/ioad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/22/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Intrauterine adhesions (IUA) are a common gynecological problem. Stem cell therapy has been widely used in the treatment of IUA. However, due to the complex and harsh microenvironment of the uterine cavity, the effectiveness of such therapy is greatly inhibited. This study aimed to investigate whether melatonin pretreatment enhances the efficacy of human umbilical cord mesenchymal stem cells (HucMSCs) in IUA treatment in rats. First, we explored the effect of melatonin on the biological activity of HucMSCs in vitro through a macrophage co-culture system, Cell Counting Kit 8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU), flow cytometry, immunofluorescence staining, and qRT-PCR. Subsequently, we established the IUA rat model and tracked the distribution of HucMSCs in this model. In addition, we observed the number of M1 and M2 macrophages through immunofluorescence staining and detected the levels of inflammatory cytokines. Four weeks after cell transplantation, HE, Masson, and immunohistochemical staining were performed. In vitro experiments showed that melatonin pretreatment of HucMSCs promoted proliferation, reduced apoptosis, up-regulated the stemness gene, and regulated macrophage polarization. In vivo, melatonin pretreatment caused more HucMSCs to remain in the uterine cavity. Melatonin-pretreated HucMSCs recruited more macrophages, regulated macrophage polarization, and reduced inflammation. Melatonin-pretreated HucMSCs relieved fibrosis, increased endometrium thickness, and up-regulated CD34, vimentin, proliferating cell nuclear antigen (PCNA), and alpha small muscle antigen (α-SMA) expression. Fertility tests showed that melatonin-pretreated HucMSCs increased the number of embryos. In summary, pretreatment with melatonin was beneficial for HucMSC treatment because it enhanced the cell's ability to recruit macrophages and regulate macrophage polarization, which led to the regeneration of the endometrium and improved pregnancy outcomes.
Collapse
Affiliation(s)
- Weili Qin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiawei Wang
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Reproductive and Genetic Hospital, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qianwen Hu
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongyan Qin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Nana Ma
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fengque Zheng
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wencai Tian
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinghang Jiang
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ting Li
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yufu Jin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ming Liao
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Aiping Qin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
103
|
Kumlin M, Ungerstedt J, Cai H, Leonard E, Felländer-Tsai L, Qian H. The functional and molecular impact of triamcinolone acetonide on primary human bone marrow mesenchymal stem cells. Sci Rep 2023; 13:21787. [PMID: 38066109 PMCID: PMC10709330 DOI: 10.1038/s41598-023-48448-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Traumatic or degenerative joint pain is abundant in the population. Symptom relief by intra- and periarticular glucocorticoid administration is frequently used, however may have potentially devastating effects, changing the normal healing process of the joint. Mesenchymal stem cells (MSCs) are important for wound-healing processes due to their multipotency in regenerating osteoblasts, chondrocytes and adipocytes but also have immunomodulatory properties. The aim of this study was to investigate the impact of triamcinolone acetonide (TA) a common glucocorticoid administrated intra- and periarticularly, on human bone marrow derived MSC viability, functionality, multi-lineage differentiation and transcriptomic output. We found that TA treatment induced apoptosis and promoted adipogenesis while impairing chondrogenesis of MSCs. RNA sequencing indicated that TA modulated the inflammatory response of MSCs, which may have an impact on the immunologic environment where the inflammatory phase is a physiological part of the natural healing process. These data indicate that triamcinolone acetonide should be used with consideration bearing the patient's outcome in mind, with the intention to optimize joint recovery and homeostasis.
Collapse
Affiliation(s)
- Maritha Kumlin
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, K54, 141 86, Stockholm, Sweden.
| | - Johanna Ungerstedt
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
- ME Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Huan Cai
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| | - Elory Leonard
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| | - Li Felländer-Tsai
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, K54, 141 86, Stockholm, Sweden
| | - Hong Qian
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
104
|
Mirshekar M, Afkhami H, Razavi S, Masjedian Jazi F, Darban-Sarokhalil D, Ohadi E, Nezhad MM, Karimi R. Potential antibacterial activity and healing effect of topical administration of bone marrow and adipose mesenchymal stem cells encapsulated in collagen-fibrin hydrogel scaffold on full-thickness burn wound infection caused by Pseudomonas aeruginosa. Burns 2023; 49:1944-1957. [PMID: 38042618 DOI: 10.1016/j.burns.2023.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/20/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Burns injuries are prone to hospital-acquired infections, and Pseudomonas aeruginosa is one of the most common causes of mortality and morbidity in patients with burn injuries. Thus, this study aimed to analyze the effects of topical treatment with bone marrow (BM-MSC) and adipose mesenchymal stem cells (AD-MSC) encapsulated in collagen and fibrin scaffolds in a Balb/c model of burn wound infection. Extraction of stem cells from adipose and bone marrow tissue of rats was performed and cells were characterized using standard methods. Then, collagen, fibrin and collagen-fibrin scaffolds were constructed and the extracted cells were encapsulated in all three scaffolds. Then, 3rd degree burn was induced in mice and 1.5 × 108 (CFU/ml) of P. aeruginosa was introduced to the burn wound. Subsequently, after 24 h of inducing wound infection, encapsulated MSCs were introduced as dressings to burn wound infection and microbial load as well as rate of wound infection healing was measured. The results of this study showed that the use of BM-MSC and AD-MSC encapsulated in collagen-fibrin scaffold reduced the bacteria load down to 54 and 21 CFU/gr, respectively (P < 0.05). Moreover, BM-MSC and AD-MSC encapsulated in collagen-fibrin showed 80% and 75% wound healing, respectively (P < 0.05). Also, we found no significant between cell origin and healing. Encapsulation of MSCs into collagen-fibrin scaffolds could be effective not only against P. aeruginosa infection, but also healing and regeneration of burn wound.
Collapse
Affiliation(s)
- Maryam Mirshekar
- Microbiology Department, School of Medicine, Iran University Of Medical Science, Iran
| | - Hamed Afkhami
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Shabnam Razavi
- Microbiology Department, School of Medicine, Iran University Of Medical Science, Iran; Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| | | | | | - Elnaz Ohadi
- Microbiology Department, School of Medicine, Iran University Of Medical Science, Iran
| | - Majid Mottaghi Nezhad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Karimi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
105
|
Las Heras K, Garcia-Orue I, Aguirre JJ, de la Caba K, Guerrero P, Igartua M, Santos-Vizcaino E, Hernandez RM. Soy protein/β-chitin sponge-like scaffolds laden with human mesenchymal stromal cells from hair follicle or adipose tissue promote diabetic chronic wound healing. BIOMATERIALS ADVANCES 2023; 155:213682. [PMID: 37925826 DOI: 10.1016/j.bioadv.2023.213682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/10/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Chronic wounds are a worldwide problem that affect >40 million people every year. The constant inflammatory status accompanied by prolonged bacterial infections reduce patient's quality of life and life expectancy drastically. An important cell type involved in the wound healing process are mesenchymal stromal cells (MSCs) due to their long-term demonstrated immunomodulatory and pro-regenerative capacity. Thus, in this work, we leveraged and compared the therapeutic properties of MSCs derived from both adipose tissue and hair follicle, which we combined with sponge-like scaffolds (SLS) made of valorized soy protein and β-chitin. In this regard, the combination of these cells with biomaterials permitted us to obtain a multifunctional therapy that allowed high cell retention and growing rates while maintaining adequate cell-viability for several days. Furthermore, this combined therapy demonstrated to increase fibroblasts and keratinocytes migration, promote human umbilical vein endothelial cells angiogenesis and protect fibroblasts from highly proteolytic environments. Finally, this combined therapy demonstrated to be highly effective in reducing wound healing time in vivo with only one treatment change during all the experimental procedure, also promoting a more functional and native-like healed skin.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Jose Javier Aguirre
- Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, Pathological Anatomy Service, Vitoria-Gasteiz, Spain
| | - Koro de la Caba
- BIOMAT Research Group, University of the Basque Country (UPV/EHU), Escuela de Ingeniería de Gipuzkoa, Plaza de Europa 1, 20018 Donostia-San Sebastián, Spain; BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Pedro Guerrero
- BIOMAT Research Group, University of the Basque Country (UPV/EHU), Escuela de Ingeniería de Gipuzkoa, Plaza de Europa 1, 20018 Donostia-San Sebastián, Spain; BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; Proteinmat Materials SL, Avenida de Tolosa 72, 20018 Donostia-San Sebastián, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain.
| |
Collapse
|
106
|
Noh CH, Park S, Seong HR, Lee AY, Tsolmon KE, Geum D, Hong SC, Kim TM, Choi EK, Kim YB. An Exosome-Rich Conditioned Medium from Human Amniotic Membrane Stem Cells Facilitates Wound Healing via Increased Reepithelization, Collagen Synthesis, and Angiogenesis. Cells 2023; 12:2698. [PMID: 38067126 PMCID: PMC10705799 DOI: 10.3390/cells12232698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Tissue regeneration is an essential requirement for wound healing and recovery of organs' function. It has been demonstrated that wound healing can be facilitated by activating paracrine signaling mediated by exosomes secreted from stem cells, since exosomes deliver many functional molecules including growth factors (GFs) and neurotrophic factors (NFs) effective for tissue regeneration. In this study, an exosome-rich conditioned medium (ERCM) was collected from human amniotic membrane stem cells (AMSCs) by cultivating the cells under a low oxygen tension (2% O2 and 5% CO2). The contents of GFs and NFs including keratinocyte growth factor, epidermal growth factor, fibroblast growth factor 1, transforming growth factor-β, and vascular endothelial growth factor responsible for skin regeneration were much higher (10-30 folds) in the ERCM than in normal conditioned medium (NCM). In was found that CM-DiI-labeled exosomes readily entered keratinocytes and fibroblasts, and that ERCM not only facilitated the proliferation of keratinocytes in normal condition, but also protected against H2O2 cytotoxicity. In cell-migration assay, the scratch wound in keratinocyte culture dish was rapidly closed by treatment with ERCM. Such wound-healing effects of ERCM were confirmed in a rat whole skin-excision model: i.e., the wound closure was significantly accelerated, remaining minimal crusts, by topical application of ERCM solution (4 × 109 exosome particles/100 μL) at 4-day intervals. In the wounded skin, the deposition of collagens was enhanced by treatment with ERCM, which was supported by the increased production of collagen-1 and collagen-3. In addition, enhanced angiogenesis in ERCM-treated wounds was confirmed by increased von Willebrand factor (vWF)-positive endothelial cells. The results indicate that ERCM from AMSCs with high concentrations of GFs and NFs improves wound healing through tissue regeneration not only by facilitating keratinocyte proliferation for skin repair, but also activating fibroblasts for extracellular matrix production, in addition to the regulation of angiogenesis and scar tissue formation.
Collapse
Affiliation(s)
- Chan Ho Noh
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.H.N.)
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Sangryong Park
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Hye-Rim Seong
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Ah-young Lee
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Khan-Erdene Tsolmon
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.H.N.)
| | - Dongho Geum
- Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Soon-Cheol Hong
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Tae Myoung Kim
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Ehn-Kyoung Choi
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| | - Yun-Bae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.H.N.)
- Central Research Institute, Designed Cells Co., Ltd., Cheongju 28576, Republic of Korea (T.M.K.); (E.-K.C.)
| |
Collapse
|
107
|
Ye H, Wang F, Xu G, Shu F, Fan K, Wang D. Advancements in engineered exosomes for wound repair: current research and future perspectives. Front Bioeng Biotechnol 2023; 11:1301362. [PMID: 38033824 PMCID: PMC10682480 DOI: 10.3389/fbioe.2023.1301362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Wound healing is a complex and prolonged process that remains a significant challenge in clinical practice. Exosomes, a type of nanoscale extracellular vesicles naturally secreted by cells, are endowed with numerous advantageous attributes, including superior biocompatibility, minimal toxicity, and non-specific immunogenicity. These properties render them an exceptionally promising candidate for bioengineering applications. Recent advances have illustrated the potential of exosome therapy in promoting tissue repair. To further augment their therapeutic efficacy, the concept of engineered exosomes has been proposed. These are designed and functionally modifiable exosomes that have been tailored on the attributes of natural exosomes. This comprehensive review delineates various strategies for exosome engineering, placing specific emphasis on studies exploring the application of engineered exosomes for precision therapy in wound healing. Furthermore, this review sheds light on strategies for integrating exosomes with biomaterials to enhance delivery effectiveness. The insights presented herein provide novel perspectives and lay a robust foundation for forthcoming research in the realm of cutaneous wound repair therapies.
Collapse
Affiliation(s)
- Hailian Ye
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Wang
- Department of Burn and Plastic Surgery, Department of Wound Repair, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Guangchao Xu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Feihong Shu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kunwu Fan
- Department of Burn and Plastic Surgery, Department of Wound Repair, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Dali Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
108
|
Humenik F, Danko J, Krešáková L, Vdoviaková K, Vrabec V, Vasilová E, Giretová M, Tóth Š, Fagová Z, Babík J, Medvecký Ľ. A Chitosan-Based Biomaterial Combined with Mesenchymal Stem Cell-Conditioned Medium for Wound Healing and Skin Regeneration. Int J Mol Sci 2023; 24:16080. [PMID: 38003269 PMCID: PMC10671656 DOI: 10.3390/ijms242216080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The aim of this study was to provide a beneficial treatment effect of novel chitosan bio-polymeric material enriched with mesenchymal stem cell products derived from the canine adipose tissue (AT-MSC) on the artificial skin defect in a rabbit model. For the objectivity of the regeneration evaluation, we used histological analysis and a scoring system created by us, taking into account all the attributes of regeneration, such as inflammatory reaction, necrosis, granulation, formation of individual skin layers and hair follicles. We observed an acceleration and improvement in the healing of an artificially created skin defect after eight and ten weeks in comparison with negative control (spontaneous healing without biomaterial). Moreover, we were able to described hair follicles and epidermis layer in histological skin samples treated with a chitosan-based biomaterial on the eighth week after grafting.
Collapse
Affiliation(s)
- Filip Humenik
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Ján Danko
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Educational, Scientific and Research Institute AGEL, 811 06 Bratislava, Slovakia
| | - Lenka Krešáková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Katarína Vdoviaková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Vladimír Vrabec
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Emília Vasilová
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Mária Giretová
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Zuzana Fagová
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Ján Babík
- Clinic of Burns and Reconstructive Medicine, AGEL Hospital, 040 15 Košice-Šaca, Slovakia
| | - Ľubomír Medvecký
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| |
Collapse
|
109
|
Zawrzykraj M, Deptuła M, Kondej K, Tymińska A, Pikuła M. The effect of chemotherapy and radiotherapy on stem cells and wound healing. Current perspectives and challenges for cell-based therapies. Biomed Pharmacother 2023; 168:115781. [PMID: 39491418 DOI: 10.1016/j.biopha.2023.115781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/05/2024] Open
Abstract
Cancers are part of the group of diseases that carry a high mortality rate. According to World Health Organization in 2020 reported 10 million deaths due to cancers. Treatment of oncological patients is focused on chemotherapeutic agents, radiology, or immunology. Surgical interventions are also an important aspect of treatment. The above methods contribute to saving the patients' health and lives. However, cancer treatment possesses side effects. Commonly observed complications are hair loss, mucositis, nausea, diarrhea, or various skin damage. To improve the quality of medical care for cancer patients, new methods of reducing side effects are sought. Strategies include the use of stem cells (SCs). Due to unlimited proliferation potential and differentiating abilities, SCs are used in the treatment of many disease entities, including wounds. One of the most used types of stem cells supposed adipose-derived mesenchymal stromal cells (AD-MSCs). Clinical trials confirm the application of AD-MSCs in wound healing. Furthermore, in vivo studies considered the utilization of AD-MSCs in radiation injury. The use of stem cells in cancer treatment still involves many questions, such as the impact of treatment on SCs' condition and oncological safety. However, development in regenerative medicine research may contribute to the use of stem cells in personalized medicine, customized for the patient. This could represent a breakthrough step in preventing the side effects of cancer therapies, including chronic wounds.
Collapse
Affiliation(s)
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdansk, Poland
| | - Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Poland.
| |
Collapse
|
110
|
Anandabaskaran S, Hanna L, Iqbal N, Constable L, Tozer P, Hart A. Where Are We and Where to Next?-The Future of Perianal Crohn's Disease Management. J Clin Med 2023; 12:6379. [PMID: 37835022 PMCID: PMC10573672 DOI: 10.3390/jcm12196379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Perianal fistulizing Crohn's Disease (pCD) affects about 25% of patients with Crohn's Disease (CD). It remains a difficult entity to manage with a therapeutic ceiling of treatment success despite improving medical and surgical management. The refractory nature of the disease calls for an imminent need to better understand its immunopathogenesis and classification to better streamline our treatment options. In this article, we overview the current state of pCD management and discuss where the future of its management may lie.
Collapse
Affiliation(s)
- Sulak Anandabaskaran
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, UK
- Robin Phillip’s Fistula Research Unit, St Mark’s Hospital and Academic Institute, London HA1 3UJ, UK
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, 390 Victoria Street, Darlinghurst, NSW 2010, Australia
| | - Luke Hanna
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, UK
- Robin Phillip’s Fistula Research Unit, St Mark’s Hospital and Academic Institute, London HA1 3UJ, UK
| | - Nusrat Iqbal
- Robin Phillip’s Fistula Research Unit, St Mark’s Hospital and Academic Institute, London HA1 3UJ, UK
- Department of Surgery and Cancer, South Kensington Campus, Imperial College London, London SW7 2BX, UK
| | - Laura Constable
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, UK
| | - Phil Tozer
- Robin Phillip’s Fistula Research Unit, St Mark’s Hospital and Academic Institute, London HA1 3UJ, UK
- Department of Surgery and Cancer, South Kensington Campus, Imperial College London, London SW7 2BX, UK
| | - Ailsa Hart
- Robin Phillip’s Fistula Research Unit, St Mark’s Hospital and Academic Institute, London HA1 3UJ, UK
- Department of Surgery and Cancer, South Kensington Campus, Imperial College London, London SW7 2BX, UK
| |
Collapse
|
111
|
Andalib E, Kashfi M, Mahmoudvand G, Rezaei E, Mahjoor M, Torki A, Afkhami H. Application of hypoxia-mesenchymal stem cells in treatment of anaerobic bacterial wound infection: wound healing and infection recovery. Front Microbiol 2023; 14:1251956. [PMID: 37869672 PMCID: PMC10586055 DOI: 10.3389/fmicb.2023.1251956] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, are a type of multipotent stem cells that are typically extracted from adipose tissue and bone marrow. In the field of tissue engineering and regenerative medicine, MSCs and their exosomes have emerged as revolutionary tools. Researchers are now devoting greater attention to MSCs because of their ability to generate skin cells like fibroblasts and keratinocytes, as well as their distinctive potential to decrease inflammation and emit pro-angiogenic molecules at the site of wounds. More recent investigations revealed that MSCs can exert numerous direct and indirect antimicrobial effects that are immunologically mediated. Collectively, these antimicrobial properties can remove bacterial infections when the MSCs are delivered in a therapeutic setting. Regardless of the positive therapeutic potential of MSCs for a multitude of conditions, transplanted MSC cell retention continues to be a major challenge. Since MSCs are typically administered into naturally hypoxic tissues, understanding the impact of hypoxia on the functioning of MSCs is crucial. Hypoxia has been postulated to be among the factors determining the differentiation of MSCs, resulting in the production of inflammatory cytokines throughout the process of tissue regeneration and wound repair. This has opened new horizons in developing MSC-based systems as a potent therapeutic tool in oxygen-deprived regions, including anaerobic wound infection sites. This review sheds light on the role of hypoxia-MSCs in the treatment of anaerobic bacterial wound infection in terms of both their regenerative and antimicrobial activities.
Collapse
Affiliation(s)
- Elahe Andalib
- Department of Microbiology, School of Basic Sciences, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Elaheh Rezaei
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Torki
- Department of Medical Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
112
|
Cai Y, Chen K, Liu C, Qu X. Harnessing strategies for enhancing diabetic wound healing from the perspective of spatial inflammation patterns. Bioact Mater 2023; 28:243-254. [PMID: 37292231 PMCID: PMC10245071 DOI: 10.1016/j.bioactmat.2023.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 06/10/2023] Open
Abstract
Diabetic wound is a great threat to patient's health and lives. The refractory diabetic wound shows spatial inflammation patterns, in which the early-wound pattern depicts a deprived acute inflammatory response, and the long-term non-healing wound pattern delineates an excessive and persistent inflammation due to the delayed immune cell infiltration in a positive feedback loop. In this work, we give points to some strategies to normalize the dysregulated immune process based on the spatial inflammation pattern differences in diabetic wound healing. First of all, inhibiting inflammatory response to avoid subsequent persistent and excessive immune infiltration for the early diabetic wound is proposed. However, diabetic wounds are unperceptive trauma that makes patients miss the best treatment time. Therefore, we also introduce two strategies for the long-term non-healing diabetic wound. One strategy is about changing chronic wounds to acute ones, which aims to rejuvenate M1 macrophages in diabetic wounds and make spontaneous M2 polarization possible. To activate the controllable proinflammatory response, western medicine delivers proinflammatory molecules while traditional Chinese medicine develops "wound-pus promoting granulation tissue growth theory". Another strategy to solve long-term non-healing wounds is seeking switches that target M1/M2 transition directly. These investigations draw a map that delineates strategies for enhancing diabetic wound healing from the perspective of spatial inflammation patterns systematically.
Collapse
Affiliation(s)
- Yixin Cai
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, China
| | - Kangli Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, China
| | - Xue Qu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai, 200237, China
| |
Collapse
|
113
|
Zhang W, Zhang J, Cui Y, Zhao Y, Lei X. Stem Cells and Exosome Applications for Cutaneous Wound Healing: From Ground to Microgravity Environment. Stem Cell Rev Rep 2023; 19:2094-2108. [PMID: 37458936 DOI: 10.1007/s12015-023-10571-9] [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] [Accepted: 05/27/2023] [Indexed: 10/17/2023]
Abstract
The increasing number of astronauts entering microgravity environments for long-term space missions has resulted in serious health problems, including accidental injury and trauma. Skin, as the largest organ and outermost layer of the human body, has the ability to self-renew and withstand a variety of harmful biological and environmental influences. Recent spaceflight experiments and simulated studies have begun to concern the effects of microgravity on the growth of skin cells and the process of cutaneous wound healing. However, the mechanisms of the adverse effects of microgravity on skin cells and potential intervention measures are still limited. Stem cells and their exosomes provide unique opportunities for the cutaneous wound healing as they have been used to improve skin repair. This review discusses the effects of microgravity on wound healing, from cell morphological changes to molecular level alterations. Furthermore, the current research on wound healing treatment utilizing stem cells and their exosomes on the ground is summarized. Finally, this review proposes promising therapeutic strategies using stem cells or exosomes for wound healing in the microgravity environment. Graphical Abstract.
Collapse
Affiliation(s)
- Wenya Zhang
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jinhua Zhang
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Yongxiang Zhao
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China.
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
114
|
Wen Z, Chen Y, Liao P, Wang F, Zeng W, Liu S, Wu H, Wang N, Moroni L, Zhang M, Duan Y, Chen H. In Situ Precision Cell Electrospinning as an Efficient Stem Cell Delivery Approach for Cutaneous Wound Healing. Adv Healthc Mater 2023; 12:e2300970. [PMID: 37379527 DOI: 10.1002/adhm.202300970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/10/2023] [Indexed: 06/30/2023]
Abstract
Mesenchymal stem cell (MSC) therapies have been brought forward as a promising treatment modality for cutaneous wound healing. However, current approaches for stem cell delivery have many drawbacks, such as lack of targetability and cell loss, leading to poor efficacy of stem cell therapy. To overcome these problems, in the present study, an in situ cell electrospinning system is developed as an attractive approach for stem cell delivery. MSCs have a high cell viability of over 90% even with a high applied voltage of 15 kV post-cell electrospinning process. In addition, cell electrospinning does not show any negative effect on the surface marker expression and differentiation capacity of MSCs. In vivo studies demonstrate that in situ cell electrospinning treatment can promote cutaneous wound healing through direct deposition of bioactive fish gelatin fibers and MSCs onto wound sites, leading to a synergic therapeutic effect. The approach enhances extracellular matrix remodeling by increasing collagen deposition, promotes angiogenesis by increasing the expression of vascular endothelial growth factor (VEGF) and forming small blood vessels, and dramatically reduces the expression of interleukin-6 (IL-6) during wound healing. The use of in situ cell electrospinning system potentially provides a rapid, no touch, personalized treatment for cutaneous wound healing.
Collapse
Affiliation(s)
- Zhengbo Wen
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yuxin Chen
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Peilin Liao
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Fengyu Wang
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Weiping Zeng
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Haibing Wu
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Minmin Zhang
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510006, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
115
|
Song Y, You Y, Xu X, Lu J, Huang X, Zhang J, Zhu L, Hu J, Wu X, Xu X, Tan W, Du Y. Adipose-Derived Mesenchymal Stem Cell-Derived Exosomes Biopotentiated Extracellular Matrix Hydrogels Accelerate Diabetic Wound Healing and Skin Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304023. [PMID: 37712174 PMCID: PMC10602544 DOI: 10.1002/advs.202304023] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/30/2023] [Indexed: 09/16/2023]
Abstract
Wound healing is an urgent clinical challenge, particularly in the case of chronic wounds. Traditional approaches to wound healing have limited therapeutic efficacy due to lengthy healing times, risk of immune rejection, and susceptibility to infection. Recently, adipose-derived mesenchymal stem cell-derived exosomes (ADSC-exos) have emerged as a promising modality for tissue regeneration and wound repair. In this study, the development of a novel extracellular matrix hydrogel@exosomes (ECM@exo) is reported, which entails incorporation of ADSC-exos into an extracellular matrix hydrogel (ECM hydrogel). This solution forms a hydrogel at physiological temperature (≈37 °C) upon local injection into the wound site. ECM@exo enables sustained release of ADSC-exos from the ECM hydrogel, which maintains high local concentrations at the wound site. The ECM hydrogel displays good biocompatibility and biodegradability. The in vivo and in vitro results demonstrate that ECM@exo treatment effectively reduces inflammation and promotes angiogenesis, collagen deposition, cell proliferation, and migration, thereby accelerating the wound healing process. Overall, this innovative therapeutic approach offers a new avenue for wound healing via a biological hydrogel with controlled exosome release.
Collapse
Affiliation(s)
- Yanling Song
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Yuchan You
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xinyi Xu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jingyi Lu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiajie Huang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jucong Zhang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Luwen Zhu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jiahao Hu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiaochuan Wu
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiaoling Xu
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouZhejiang310015P. R. China
| | - Weiqiang Tan
- Department of Plastic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
| | - Yongzhong Du
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
- Department of Plastic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
- Department of PharmacySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016P. R. China
- Innovation Center of Translational PharmacyJinhua Institute of Zhejiang UniversityJinhua321299P. R. China
| |
Collapse
|
116
|
He X, Wang D, Yi Y, Tan Y, Wu M, Wang H, Hu W, Chen H, Zhang Q, Wu Y. δ-Tocotrienol preconditioning improves the capability of bone marrow-derived mesenchymal stem cells in promoting wound healing by inhibiting BACH1-related ferroptosis. Cell Death Discov 2023; 9:349. [PMID: 37739949 PMCID: PMC10516898 DOI: 10.1038/s41420-023-01653-1] [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: 07/06/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023] Open
Abstract
Wound healing is a complex physiological process for maintaining skin integrity after a wound. Bone marrow-derived mesenchymal stem cells (BMSCs) are excellent cellular candidates for wound healing, which could be enhanced by exogenous stimulation. We aimed to explore the role of δ-Tocotrienol (δ-TT) in BMSC ability of wound healing. Firstly, transcriptome and single-cell analysis were used to explore the genes and pathways related to ferroptosis in wound tissues. In vitro, cell proliferation, migration, and angiogenesis of δ-TT-BMSCs were detected. In addition, qRT-PCR and immunofluorescence (IF) were applied for observing the promoting wound healing ability of δ-TT-BMSC conditioned medium (CM) on NIH-3T3 and PAM-212 cells. The level of ferroptosis was determined by the mitochondrial membrane potential and total/lipid reactive oxygen species (ROS) in the cells and the morphological changes of mitochondria were observed by transmission electron microscope. The BTB and CNC homology 1 (BACH1) expression and activation of the PI3K/AKT signaling pathway were detected by IF and western blot (WB). The effect of δ-TT-BMSCs on wound healing was observed in vivo. The regulatory mechanism of δ-TT-BMSCs on ferroptosis was verified by IHC and IF staining. In vitro, δ-TT-BMSCs declined the level of lipid ROS in NIH-3T3 and PAM-212 cells and enhanced mitochondrial membrane potential. In vivo, δ-TT-BMSCs promoted wound healing in mice by decreasing ferroptosis. In terms of mechanism, δ-TT-BMSCs inhibited the expression of BACH1 and activated PI3K/AKT signaling pathway. This study demonstrated the ability of δ-TT-BMSCs to promote wound healing by inhibiting BACH1-related ferroptosis. In addition, PI3K/AKT signaling pathway was activated by δ-TT-BMSCs and could be involved in wound healing. δ-TT-BMSCs might be a promising strategy for treating wounds.
Collapse
Affiliation(s)
- Xiao He
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dawei Wang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yi Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yufang Tan
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Haiping Wang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Weijie Hu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Hongbo Chen
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
117
|
Zhu M, Cao L, Melino S, Candi E, Wang Y, Shao C, Melino G, Shi Y, Chen X. Orchestration of Mesenchymal Stem/Stromal Cells and Inflammation During Wound Healing. Stem Cells Transl Med 2023; 12:576-587. [PMID: 37487541 PMCID: PMC10502569 DOI: 10.1093/stcltm/szad043] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/13/2023] [Indexed: 07/26/2023] Open
Abstract
Wound healing is a complex process and encompasses a number of overlapping phases, during which coordinated inflammatory responses following tissue injury play dominant roles in triggering evolutionarily highly conserved principals governing tissue repair and regeneration. Among all nonimmune cells involved in the process, mesenchymal stem/stromal cells (MSCs) are most intensely investigated and have been shown to play fundamental roles in orchestrating wound healing and regeneration through interaction with the ordered inflammatory processes. Despite recent progress and encouraging results, an informed view of the scope of this evolutionarily conserved biological process requires a clear understanding of the dynamic interplay between MSCs and the immune systems in the process of wound healing. In this review, we outline current insights into the ways in which MSCs sense and modulate inflammation undergoing the process of wound healing, highlighting the central role of neutrophils, macrophages, and T cells during the interaction. We also draw attention to the specific effects of MSC-based therapy on different pathological wound healing. Finally, we discuss how ongoing scientific advances in MSCs could be efficiently translated into clinical strategies, focusing on the current limitations and gaps that remain to be overcome for achieving preferred functional tissue regeneration.
Collapse
Affiliation(s)
- Mengting Zhu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,”Rome, Italy
| | - Lijuan Cao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,”Rome, Italy
| | - Sonia Melino
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,”Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,”Rome, Italy
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Shanghai, People’s Republic of China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China
| | - Gerry Melino
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,”Rome, Italy
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China
| | - Xiaodong Chen
- Wuxi Sinotide New Drug Discovery Institutes, Huishan Economic and Technological Development Zone, Wuxi, Jiangsu, People’s Republic of China
| |
Collapse
|
118
|
Chen J, Yu W, Xiao C, Su N, Han Y, Zhai L, Hou C. Exosome from adipose-derived mesenchymal stem cells attenuates scar formation through microRNA-181a/SIRT1 axis. Arch Biochem Biophys 2023; 746:109733. [PMID: 37652148 DOI: 10.1016/j.abb.2023.109733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/17/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Pathological scarring is the greatest challenge after injury. Exosome from adipose-derived mesenchymal stem cells has been reported effective to improve hypertrophic scar. This study focused on the possible mechanisms during this process. Exosomes from adipose-derived mesenchymal stem cells were extracted first. Hypertrophic scar tissue and paired normal skin tissue were collected from patients. Mice skin incision model and fibroblasts model were established. TGF-β1 was used to stimulate fibroblasts to myofibroblasts transdifferentiation. It was found that exosomes injection could decrease collagen sediment after wound healing. During which, the expression of microRNA-181a decreased. Further, we found that expression of microRNA-181a in scar tissue was higher than in normal skin. Then hypertrophic scar-derived fibroblasts were used for in vitro study. It was found that similar to the use of exosomes, microRNA-181a inhibitor decreased the expression of collagen and α-SMA. While microRNA-181a mimics suppressed the effects of exosomes. During fibroblast to myofibroblast trans-differentiation, level of microRNA-181a well as levels of scar-related molecules also decreased with the use of exosomes and vice versa. SIRT1 was confirmed one of the downstream targets of microRNA-181a. Suppression of SIRT1 led to diminished effects of exosomes in hypertrophic scar derived fibroblasts. In mice skin incision model, injection of SIRT1 inhibitor led to increased collagen synthesis. In conclusion, exosomes from Adipose-derived mesenchymal stem cells are promising to antagonize scarring through the regulation of microRNA-181a/SIRT1 axis.
Collapse
Affiliation(s)
- Jie Chen
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China; Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, China
| | - Wenjuan Yu
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China
| | - Chao Xiao
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China
| | - Na Su
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China
| | - Yubo Han
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China
| | - Liang Zhai
- Department of Plastic Surgery, The Second Affiliated Hospital of Xi'an Medical College, China
| | - Chen Hou
- Shaanxi Provincial People's Hospital, China.
| |
Collapse
|
119
|
Daneste H, Mohammadzadeh Boukani L, Ramezani N, Asadi F, Zaidan HK, Sadeghzade A, Ehsannia M, Azarashk A, Gholizadeh N. Combination therapy along with mesenchymal stem cells in wound healing; the state of the art. Adv Med Sci 2023; 68:441-449. [PMID: 37924749 DOI: 10.1016/j.advms.2023.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/23/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are being increasingly used in various therapeutic applications including skin tissue repair and wound healing. The positive effects of the MSCs therapy are largely elicited by immunomodulation, increasing angiogenesis, supporting extracellular matrix (ECM) and thus favoring skin structure. However, the therapeutic competences of MSC-based therapies are somewhat hindered by their apparent modest clinical merits, conferring the need for methods that would rise the efficacy of such therapies. A plethora of reports have shown that therapeutic properties of MSCs could be enhanced with other strategies and compounds like biomaterial and platelet-rich plasma (PRP) to target key possessions of MSCs and properties of adjacent tissues concurrently. Manipulation of cellular stress-response mechanisms to improve cell resistance to oxidative stress prior to or during MSC injection could also improve therapeutic efficacy of MSCs. In the current review, we shed light on the recent advances in MSCs combination therapy with other ingredients and procedures to sustain MSCs-mediated effects in wound healing.
Collapse
Affiliation(s)
- Hossein Daneste
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Narges Ramezani
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Fatemeh Asadi
- Department of Genetics, Izeh Branch, Islamic Azad University, Izeh, Iran
| | - Haider Kamil Zaidan
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Azita Sadeghzade
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Ehsannia
- Faculty of Basic Sciences, Islamic Azad University, Tehran East Branch, Tehran, Iran
| | - Ali Azarashk
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
120
|
Huerta CT, Ortiz YY, Li Y, Ribieras AJ, Voza F, Le N, Dodson C, Wang G, Vazquez-Padron RI, Liu ZJ, Velazquez OC. Novel Gene-Modified Mesenchymal Stem Cell Therapy Reverses Impaired Wound Healing in Ischemic Limbs. Ann Surg 2023; 278:383-395. [PMID: 37334717 PMCID: PMC10414148 DOI: 10.1097/sla.0000000000005949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
OBJECTIVE Here, we report a new method to increase the therapeutic potential of mesenchymal stem/stromal cells (MSCs) for ischemic wound healing. We tested biological effects of MSCs modified with E-selectin, a cell adhesion molecule capable of inducing postnatal neovascularization, on a translational murine model. BACKGROUND Tissue loss significantly worsens the risk of extremity amputation for patients with chronic limb-threatening ischemia. MSC-based therapeutics hold major promise for wound healing and therapeutic angiogenesis, but unmodified MSCs demonstrate only modest benefits. METHODS Bone marrow cells harvested from FVB/ROSA26Sor mTmG donor mice were transduced with E-selectin-green fluorescent protein (GFP)/AAV-DJ or GFP/AAV-DJ (control). Ischemic wounds were created via a 4 mm punch biopsy in the ipsilateral limb after femoral artery ligation in recipient FVB mice and subsequently injected with phosphate-buffered saline or 1×10 6 donor MSC GFP or MSC E-selectin-GFP . Wound closure was monitored daily for 7 postoperative days, and tissues were harvested for molecular and histologic analysis and immunofluorescence. Whole-body DiI perfusion and confocal microscopy were utilized to evaluate wound angiogenesis. RESULTS Unmodified MSCs do not express E-selectin, and MSC E-selectin-GFP gain stronger MSC phenotype yet maintain trilineage differentiation and colony-forming capability. MSC E-selectin-GFP therapy accelerates wound healing compared with MSC GFP and phosphate-buffered saline treatment. Engrafted MSC E-selectin-GFP manifest stronger survival and viability in wounds at postoperative day 7. Ischemic wounds treated with MSC E-selectin-GFP exhibit more abundant collagen deposition and enhanced angiogenic response. CONCLUSIONS We establish a novel method to potentiate regenerative and proangiogenic capability of MSCs by modification with E-selectin/adeno-associated virus. This innovative therapy carries the potential as a platform worthy of future clinical studies.
Collapse
Affiliation(s)
- Carlos Theodore Huerta
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Yulexi Y. Ortiz
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Yan Li
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Antoine J. Ribieras
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Francesca Voza
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Nga Le
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Caroline Dodson
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Zhao-Jun Liu
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Omaida C. Velazquez
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
121
|
Liu Q, Li J, Chang J, Guo Y, Wen D. The characteristics and medical applications of antler stem cells. Stem Cell Res Ther 2023; 14:225. [PMID: 37649124 PMCID: PMC10468909 DOI: 10.1186/s13287-023-03456-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023] Open
Abstract
Antlers are the only fully regenerable mammalian appendages whose annual renewal is initiated by antler stem cells (ASCs), defined as a specialized type of mesenchymal stem cells (MSCs) with embryonic stem cell properties. ASCs possess the same biological features as MSCs, including the capacity for self-renewal and multidirectional differentiation, immunomodulatory functions, and the maintenance of stem cell characteristics after multiple passages. Several preclinical studies have shown that ASCs exhibit promising potential in wound healing, bone repair, osteoarthritis, anti-tissue fibrosis, anti-aging, and hair regeneration. Medical applications based on ASCs and ASC-derived molecules provide a new source of stem cells and therapeutic modalities for regenerative medicine. This review begins with a brief description of antler regeneration and the role of ASCs. Then, the properties and advantages of ASCs are described. Finally, medical research advances regarding ASCs are summarized, and the prospects and challenges of ASCs are highlighted.
Collapse
Affiliation(s)
- Qi Liu
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiannan Li
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinghui Chang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Guo
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Dacheng Wen
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
122
|
Zhuang X, Jiang Y, Yang X, Fu L, Luo L, Dong Z, Zhao J, Hei F. Advances of mesenchymal stem cells and their derived extracellular vesicles as a promising therapy for acute respiratory distress syndrome: from bench to clinic. Front Immunol 2023; 14:1244930. [PMID: 37711624 PMCID: PMC10497773 DOI: 10.3389/fimmu.2023.1244930] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory lung injury characterized by diffuse alveolar damage. The period prevalence of ARDS was 10.4% of ICU admissions in 50 countries. Although great progress has been made in supportive care, the hospital mortality rate of severe ARDS is still up to 46.1%. Moreover, up to now, there is no effective pharmacotherapy for ARDS and most clinical trials focusing on consistently effective drugs have met disappointing results. Mesenchymal stem cells (MSCs) and their derived extracellular vesicles (EVs) have spawned intense interest of a wide range of researchers and clinicians due to their robust anti-inflammatory, anti-apoptotic and tissue regeneration properties. A growing body of evidence from preclinical studies confirmed the promising therapeutic potential of MSCs and their EVs in the treatment of ARDS. Based on the inspiring experimental results, clinical trials have been designed to evaluate safety and efficacy of MSCs and their EVs in ARDS patients. Moreover, trials exploring their optimal time window and regimen of drug administration are ongoing. Therefore, this review aims to present an overview of the characteristics of mesenchymal stem cells and their derived EVs, therapeutic mechanisms for ARDS and research progress that has been made over the past 5 years.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Feilong Hei
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
123
|
Al-Sharabi N, Gruber R, Sanz M, Mohamed-Ahmed S, Kristoffersen EK, Mustafa K, Shanbhag S. Proteomic Analysis of Mesenchymal Stromal Cells Secretome in Comparison to Leukocyte- and Platelet-Rich Fibrin. Int J Mol Sci 2023; 24:13057. [PMID: 37685865 PMCID: PMC10487446 DOI: 10.3390/ijms241713057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Secretomes of mesenchymal stromal cells (MSCs) are emerging as a novel growth factor (GF)-based strategy for periodontal and bone regeneration. The objective of this study was to compare the secretome of human bone marrow MSC (BMSC) to that of leukocyte- and platelet-rich fibrin (L-PRF), an established GF-based therapy, in the context of wound healing and regeneration. Conditioned media from human BMSCs (BMSC-CM) and L-PRF (LPRF-CM) were subjected to quantitative proteomic analysis using liquid chromatography with tandem mass spectrometry. Global profiles, gene ontology (GO) categories, differentially expressed proteins (DEPs), and gene set enrichment (GSEA) were identified using bioinformatic methods. Concentrations of selected proteins were determined using a multiplex immunoassay. Among the proteins identified in BMSC-CM (2157 proteins) and LPRF-CM (1420 proteins), 1283 proteins were common. GO analysis revealed similarities between the groups in terms of biological processes (cellular organization, protein metabolism) and molecular functions (cellular/protein-binding). Notably, more DEPs were identified in BMSC-CM (n = 550) compared to LPRF-CM (n = 118); these included several key GF, cytokines, and extracellular matrix (ECM) proteins involved in wound healing. GSEA revealed enrichment of ECM (especially bone ECM)-related processes in BMSC-CM and immune-related processes in LPRF-CM. Similar trends for intergroup differences in protein detection were observed in the multiplex analysis. Thus, the secretome of BMSC is enriched for proteins/processes relevant for periodontal and bone regeneration. The in vivo efficacy of this therapy should be evaluated in future studies.
Collapse
Affiliation(s)
- Niyaz Al-Sharabi
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway; (N.A.-S.); (S.M.-A.); (K.M.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria;
- Austrian Cluster for Tissue Regeneration, 1090 Vienna, Austria
- Department of Periodontology, School of Dental Medicine, University of Bern, 3012 Bern, Switzerland
| | - Mariano Sanz
- ETEP Research Group, Faculty of Odontology, University Complutense of Madrid, 28040 Madrid, Spain;
| | - Samih Mohamed-Ahmed
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway; (N.A.-S.); (S.M.-A.); (K.M.)
| | - Einar K. Kristoffersen
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5021 Bergen, Norway;
- Department of Clinical Science, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway; (N.A.-S.); (S.M.-A.); (K.M.)
| | - Siddharth Shanbhag
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway; (N.A.-S.); (S.M.-A.); (K.M.)
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5021 Bergen, Norway;
| |
Collapse
|
124
|
Du CL, Zhang X, Yang H. Clinical value of combined serum MMP-2, MMP-9 and TIMP-1 for the prognosis of perianal fistula patients who received minimally invasive surgery. Eur J Gastroenterol Hepatol 2023; 35:843-847. [PMID: 37395236 DOI: 10.1097/meg.0000000000002586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
OBJECTIVE This study aimed to investigate the clinical value of combined serum matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) for the prognosis of perianal fistula patients. METHODS Patients diagnosed and treated for perianal fistula by minimally invasive surgery (MIS) were enrolled. The concentrations of serum MMP-2, MMP-9 and TIMP-1 were measured at 24 h after surgery. Different levels of wound secretion, growth of granulation tissue and wound pain were used as criteria to evaluate surgical incision healing. The receiver operating characteristic curve was used to analyze the predicted assessment value. RESULTS The concentrations of serum MMP-2 and MMP-9 were significantly higher, while the concentrations of serum TIMP-1 at 24 h after surgery were significantly lower in the poor healing group than in the good healing group. It was further found that high levels of serum MMP-2 and MMP-9 were risk factors for poor healing, while high concentrations of serum TIMP-1 at 24 h after surgery were protective factors for poor healing. CONCLUSION High concentrations of serum MMP-2 and MMP-9 and low concentrations of serum TIMP at 24 h after surgery are risk factors for poor healing in perianal fistula patients who receive MIS, and the combined test has a higher predictive value.
Collapse
Affiliation(s)
- Chang-Liang Du
- Department of Anorectal Diseases, Daqing Oilfield General Hospital, Saertu District, Daqing, Heilongjiang, China
| | | | | |
Collapse
|
125
|
Jin Y, Li S, Yu Q, Chen T, Liu D. Application of stem cells in regeneration medicine. MedComm (Beijing) 2023; 4:e291. [PMID: 37337579 PMCID: PMC10276889 DOI: 10.1002/mco2.291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/21/2023] Open
Abstract
Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self-replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.
Collapse
Affiliation(s)
- Ye Jin
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Shuangyang Li
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Qixuan Yu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Tianli Chen
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Da Liu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| |
Collapse
|
126
|
Sukmana BI, Margiana R, Almajidi YQ, Almalki SG, Hjazi A, Shahab S, Romero-Parra RM, Alazbjee AAA, Alkhayyat A, John V. Supporting wound healing by mesenchymal stem cells (MSCs) therapy in combination with scaffold, hydrogel, and matrix; State of the art. Pathol Res Pract 2023; 248:154575. [PMID: 37285734 DOI: 10.1016/j.prp.2023.154575] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
Non-healing wounds impose a huge annual cost on the survival of different countries and large populations in the world. Wound healing is a complex and multi-step process, the speed and quality of which can be changed by various factors. To promote wound healing, compounds such as platelet-rich plasma, growth factors, platelet lysate, scaffolds, matrix, hydrogel, and cell therapy, in particular, with mesenchymal stem cells (MSCs) are suggested. Nowadays, the use of MSCs has attracted a lot of attention. These cells can induce their effect by direct effect and secretion of exosomes. On the other hand, scaffolds, matrix, and hydrogels provide suitable conditions for wound healing and the growth, proliferation, differentiation, and secretion of cells. In addition to generating suitable conditions for wound healing, the combination of biomaterials and MSCs increases the function of these cells at the site of injury by favoring their survival, proliferation, differentiation, and paracrine activity. In addition, other compounds such as glycol, sodium alginate/collagen hydrogel, chitosan, peptide, timolol, and poly(vinyl) alcohol can be used along with these treatments to increase the effectiveness of treatments in wound healing. In this review article, we take a glimpse into the merging scaffolds, hydrogels, and matrix application with MSCs therapy to favor wound healing.
Collapse
Affiliation(s)
- Bayu Indra Sukmana
- Oral Biology Department, Lambung Mangkurat University, Banjarmasin, Indonesia
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Yasir Qasim Almajidi
- Department of Pharmacy (Pharmaceutics), Baghdad College of Medical Sciences, Baghdad, Iraq
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Sana Shahab
- Department of Business Administration, College of Business Administration, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | | | | | - Afa Alkhayyat
- College of Pharmacy, the Islamic University, 54001 Najaf, Iraq
| | - Vivek John
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
127
|
Iacopetti I, Perazzi A, Patruno M, Contiero B, Carolo A, Martinello T, Melotti L. Assessment of the quality of the healing process in experimentally induced skin lesions treated with autologous platelet concentrate associated or unassociated with allogeneic mesenchymal stem cells: preliminary results in a large animal model. Front Vet Sci 2023; 10:1219833. [PMID: 37559892 PMCID: PMC10407250 DOI: 10.3389/fvets.2023.1219833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/13/2023] [Indexed: 08/11/2023] Open
Abstract
Regenerative medicine for the treatment of skin lesions is an innovative and rapidly developing field that aims to promote wound healing and restore the skin to its original condition before injury. Over the years, different topical treatments have been evaluated to improve skin wound healing and, among them, mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP) have shown promising results for this purpose. This study sought to evaluate the quality of the healing process in experimentally induced full-thickness skin lesions treated with PRP associated or unassociated with MSCs in a sheep second intention wound healing model. After having surgically created full-thickness wounds on the back of three sheep, the wound healing process was assessed by performing clinical evaluations, histopathological examinations, and molecular analysis. Treated wounds showed a reduction of inflammation and contraction along with an increased re-epithelialization rate and better maturation of the granulation tissue compared to untreated lesions. In particular, the combined treatment regulated the expression of collagen types I and III resulting in a proper resolution of the granulation tissue contrary to what was observed in untreated wounds; moreover, it led to a better maturation and organization of skin adnexa and collagen fibers in the repaired skin compared to untreated and PRP-treated wounds. Overall, both treatments improved the wound healing process compared to untreated wounds. Wounds treated with PRP and MSCs showed a healing progression that qualitatively resembles a restitutio ad integrum of the repaired skin, showing features typical of a mature healthy dermis.
Collapse
Affiliation(s)
- Ilaria Iacopetti
- Department of Animal Medicine, Production and Health, University of Padua, Padova, Italy
| | - Anna Perazzi
- Department of Animal Medicine, Production and Health, University of Padua, Padova, Italy
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padua, Padova, Italy
| | - Barbara Contiero
- Department of Animal Medicine, Production and Health, University of Padua, Padova, Italy
| | - Anna Carolo
- Department of Comparative Biomedicine and Food Science, University of Padua, Padova, Italy
| | | | - Luca Melotti
- Department of Comparative Biomedicine and Food Science, University of Padua, Padova, Italy
| |
Collapse
|
128
|
Eom Y, Eom SY, Lee J, Hwang S, Won J, Kim H, Chung S, Kim HJ, Lee MY. Therapeutic Effects and Underlying Mechanism of SOCS-com Gene-Transfected ADMSCs in Pressure Ulcer Mouse Models. Cells 2023; 12:1840. [PMID: 37508509 PMCID: PMC10378383 DOI: 10.3390/cells12141840] [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: 05/24/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Although the proportion of ulcer patients with medical problems among the elderly has increased with the extension of human life expectancy, treatment efficiency is drastically low, incurring substantial social costs. MSCs have independent regeneration potential, making them useful in clinical trials of difficult-to-treat diseases. In particular, ADMSCs are promising in the stem cell therapy industry as they can be obtained in vast amounts using non-invasive methods. Furthermore, studies are underway to enhance the regeneration potential of ADMSCs using cytokines, growth factors, and gene delivery to generate highly functional ADMSCs. In this study, key regulators of wound healing, SOCS-1, -3, and -5, were combined to maximize the regenerative potential of ADMSCs in pressure ulcer treatments. After transfecting SOCS-1, -3, -5, and SOCS-com into ADMSCs using a non-viral method, the expression of the inflammatory factors TNF-alpha, INF-gamma, and IL-10 was confirmed. ADMSCs transfected with SOCS-com showed decreased overall expression of inflammatory factors and increased expression of anti-inflammatory factors. Based on these results, we implanted ADMSCs transfected with SOCS-com into a pressure ulcer mouse model to observe their subsequent wound-healing effects. Notably, SOCS-com improved wound closure in ulcers, and reconstruction of the epidermis and dermis was observed. The healing mechanism of ADMSCs transfected with SOCS-com was examined by RNA sequencing. Gene analysis results confirmed that expression changes occurred in genes of key regulators of wound healing, such as chemokines, MMP-1, 9, CSF-2, and IL-33, and that such genetic changes enhanced wound healing in ulcers. Based on these results, we demonstrate the potential of ADMSCs transfected with SOCS-com as an ulcer treatment tool.
Collapse
Affiliation(s)
- Youngsic Eom
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - So Young Eom
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jeonghwa Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Saeyeon Hwang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 34943, Republic of Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyunsoo Kim
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hye Joung Kim
- Institute of Chemical Engineering Convergence System, Korea University, Seoul 02841, Republic of Korea
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
129
|
Zhao W, Zhang H, Liu R, Cui R. Advances in Immunomodulatory Mechanisms of Mesenchymal Stem Cells-Derived Exosome on Immune Cells in Scar Formation. Int J Nanomedicine 2023; 18:3643-3662. [PMID: 37427367 PMCID: PMC10327916 DOI: 10.2147/ijn.s412717] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023] Open
Abstract
Pathological scars are the result of over-repair and excessive tissue proliferation of the skin injury. It may cause serious dysfunction, resulting in psychological and physiological burdens on the patients. Currently, mesenchymal stem cells-derived exosomes (MSC-Exo) displayed a promising therapeutic effect on wound repair and scar attenuation. But the regulatory mechanisms are opinions vary. In view of inflammation has long been proven as the initial factor of wound healing and scarring, and the unique immunomodulation mechanism of MSC-Exo, the utilization of MSC-Exo may be promising therapeutic for pathological scars. However, different immune cells function differently during wound repair and scar formation. The immunoregulatory mechanism of MSC-Exo would differ among different immune cells and molecules. Herein, this review gave a comprehensive summary of MSC-Exo immunomodulating different immune cells in wound healing and scar formation to provide basic theoretical references and therapeutic exploration of inflammatory wound healing and pathological scars.
Collapse
Affiliation(s)
- Wen Zhao
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Huimin Zhang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Rui Liu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Rongtao Cui
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
130
|
Shu F, Lu J, Zhang W, Huang H, Lin J, Jiang L, Liu W, Liu T, Xiao S, Zheng Y, Xia Z. JAM-A Overexpression in Human Umbilical Cord-Derived Mesenchymal Stem Cells Accelerated the Angiogenesis of Diabetic Wound By Enhancing Both Paracrine Function and Survival of Mesenchymal Stem Cells. Stem Cell Rev Rep 2023; 19:1554-1575. [PMID: 37060532 DOI: 10.1007/s12015-023-10518-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 04/16/2023]
Abstract
Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.
Collapse
Affiliation(s)
- Futing Shu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jianyu Lu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hongchao Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jiezhi Lin
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Wenzhang Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Tianyi Liu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China.
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
131
|
Go YY, Lee CM, Chae SW, Song JJ. Regenerative capacity of trophoblast stem cell-derived extracellular vesicles on mesenchymal stem cells. Biomater Res 2023; 27:62. [PMID: 37370189 DOI: 10.1186/s40824-023-00396-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Human mesenchymal stem cells (MSCs) are therapeutic for clinical applications because of their excellent immunomodulatory and multiple lineage differentiation abilities at tissue injury sites. However, insufficient number of cells and lack of regenerative properties during in vitro expansion still limit the clinical applicability of MSC therapies. Here, we demonstrated a preconditioning strategy with trophoblast stem cell-derived extracellular vesicles (TSC-EVs) to boost the proliferation and regenerative capacity of MSCs. METHODS We employed cell proliferation analyses such as CCK8 and BrdU assays to determine the proliferation-promoting role of TSC-EVs on MSCs. Osteogenic effects of TSC-EVs on MSCs were assessed by alkaline phosphatase (ALP) activity, calcium assays, and calvarial bone defect animal models. For skin regenerative effects, skin wound mice model was exploited to analyze wound-healing rate in this study, as well as immunofluorescence and histological staining evaluates. We also performed the small RNA profiling and RNA-sequencing analyzes to understand the cellular mechanism of TSC-EVs on MSCs. RESULTS TSC-EVs significantly promoted MSC proliferation under xeno-free conditions and facilitated the therapeutic effects of MSCs, including osteogenesis, anti-senescence, and wound healing. Transcriptomic analysis also provided evidence that specific microRNAs in TSC-EVs and differentially expressed genes (DEGs) in TSC-EV-treated MSCs showed the possibility of TSC-EVs triggering the regenerative abilities of MSCs with cytokine interaction. Hence, we found that NGF/Akt signaling mediated the regenerative effects of TSC-EVs on MSCs as a particular cellular signaling pathway. CONCLUSION The results of this study demonstrated the functional properties of TSC-EVs on MSCs for MSC-based therapeutic applications, suggesting that TSC-EVs may serve as a potential preconditioning source for MSC therapy in the clinical field of regenerative medicine.
Collapse
Affiliation(s)
- Yoon-Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
| | - Chan-Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea.
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
132
|
Devi A, Pahuja I, Singh SP, Verma A, Bhattacharya D, Bhaskar A, Dwivedi VP, Das G. Revisiting the role of mesenchymal stem cells in tuberculosis and other infectious diseases. Cell Mol Immunol 2023; 20:600-612. [PMID: 37173422 PMCID: PMC10176304 DOI: 10.1038/s41423-023-01028-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/29/2023] [Indexed: 05/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play diverse roles ranging from regeneration and wound healing to immune signaling. Recent investigations have indicated the crucial role of these multipotent stem cells in regulating various aspects of the immune system. MSCs express unique signaling molecules and secrete various soluble factors that play critical roles in modulating and shaping immune responses, and in some other cases, MSCs can also exert direct antimicrobial effects, thereby helping with the eradication of invading organisms. Recently, it has been demonstrated that MSCs are recruited at the periphery of the granuloma containing Mycobacterium tuberculosis and exert "Janus"-like functions by harboring pathogens and mediating host protective immune responses. This leads to the establishment of a dynamic balance between the host and the pathogen. MSCs function through various immunomodulatory factors such as nitric oxide (NO), IDO, and immunosuppressive cytokines. Recently, our group has shown that M.tb uses MSCs as a niche to evade host protective immune surveillance mechanisms and establish dormancy. MSCs also express a large number of ABC efflux pumps; therefore, dormant M.tb residing in MSCs are exposed to a suboptimal dose of drugs. Therefore, it is highly likely that drug resistance is coupled with dormancy and originates within MSCs. In this review, we discussed various immunomodulatory properties of MSCs, their interactions with important immune cells, and soluble factors. We also discussed the possible roles of MSCs in the outcome of multiple infections and in shaping the immune system, which may provide insight into therapeutic approaches using these cells in different infection models.
Collapse
Affiliation(s)
- Annu Devi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Isha Pahuja
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Molecular Medicine, Jamia Hamdard University, New Delhi, India
| | - Shashi Prakash Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Akanksha Verma
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Ashima Bhaskar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
133
|
Zhou Z, Xun J, Wu C, Ji C, Ji S, Shu F, Wang Y, Chen H, Zheng Y, Xiao S. Acceleration of burn wound healing by micronized amniotic membrane seeded with umbilical cord-derived mesenchymal stem cells. Mater Today Bio 2023; 20:100686. [PMID: 37334186 PMCID: PMC10276167 DOI: 10.1016/j.mtbio.2023.100686] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/11/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Umbilical cord-derived mesenchymal stem cells (UC-MSC) are promising candidates for wound healing. However, the low amplification efficiency of MSC in vitro and their low survival rates after transplantation have limited their medical application. In this study, we fabricated a micronized amniotic membrane (mAM) as a microcarrier to amplify MSC in vitro and used mAM and MSC (mAM-MSC) complexes to repair burn wounds. Results showed that MSC could live and proliferate on mAM in a 3D culture system, exhibiting higher cell activity than in 2D culture. Transcriptome sequencing of MSC showed that the expression of growth factor-related, angiogenesis-related, and wound healing-related genes was significantly upregulated in mAM-MSC compared to traditional 2D-cultured MSC, which was verified via RT-qPCR. Gene ontology (GO) analysis of differentially expressed genes (DEGs) showed significant enrichment of terms related to cell proliferation, angiogenesis, cytokine activity, and wound healing in mAM-MSC. In a burn wound model of C57BL/6J mice, topical application of mAM-MSC significantly accelerated wound healing compared to MSC injection alone and was accompanied by longer survival of MSC and greater neovascularization in the wound.
Collapse
Affiliation(s)
- Zixuan Zhou
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jingnan Xun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Chenghao Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Chao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Futing Shu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hao Chen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
134
|
Doshi A, Erickson P, Teryek M, Parekkadan B. Dynamics of Ex Vivo Mesenchymal Stromal Cell Potency under Continuous Perfusion. Int J Mol Sci 2023; 24:ijms24119602. [PMID: 37298556 DOI: 10.3390/ijms24119602] [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: 04/29/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a candidate for cell immunotherapy due to potent immunomodulatory activity found in their secretome. Though studies on their secreted substances have been reported, the time dynamics of MSC potency remain unclear. Herein, we report on the dynamics of MSC secretome potency in an ex vivo hollow fiber bioreactor using a continuous perfusion cell culture system that fractionated MSC-secreted factors over time. Time-resolved fractions of MSC-conditioned media were evaluated for potency by incubation with activated immune cells. Three studies were designed to characterize MSC potency under: (1) basal conditions, (2) in situ activation, and (3) pre-licensing. Results indicate that the MSC secretome is most potent in suppressing lymphocyte proliferation during the first 24 h and is further stabilized when MSCs are prelicensed with a cocktail of pro-inflammatory cytokines, IFNγ, TNFα, and IL-1β. The evaluation of temporal cell potency using this integrated bioreactor system can be useful in informing strategies to maximize MSC potency, minimize side effects, and allow greater control for the duration of ex vivo administration approaches.
Collapse
Affiliation(s)
- Aneesha Doshi
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Patrick Erickson
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Matthew Teryek
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
135
|
Dou Z, Qiu T, Ren Y, Wang X, Wen Q, Shen Y, Wu L, Han L, Jiang T, Xia X. Bilayer Silk Fibroin/Sodium Alginate Scaffold Delivered hUC-MSCs to Enhance Skin Scarless Healing and Hair Follicle Regeneration with the IRE1/XBP1 Pathway Inhibition. ACS Biomater Sci Eng 2023. [PMID: 37256923 DOI: 10.1021/acsbiomaterials.3c00059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Efficient local delivery of mesenchymal stem cells (MSCs) is a decisive factor for their application in regeneration processes. Here, we prepared a biomimetic bilayer silk fibroin/sodium alginate (SF/SA) scaffold to deliver human umbilical mesenchymal stem cells (hUC-MSCs) for wound healing. An SA membrane was prepared by the casting method on the upper layer of the scaffold to simulate the dense epidermal structure. On the lower layer, porous materials simulating the loose structure of the dermis were formed by the freeze-drying method. In vitro, the scaffold was proven to have a high-density pore structure, good swelling property, and suitable degradation rate. The hUC-MSCs could survive on the scaffold for up to 14 days and maintain cell stemness for at least 7 days. In vivo, SF/SA scaffolds loaded with hUC-MSCs (M-SF/SA) were applied to full-thickness defect wounds and compared with the local injection of hUC-MSCs. The M-SF/SA group showed excellent therapeutic efficacy, characterized by induction of macrophage polarization, regulation of TGF-β expression and collagen components, and enhancement of vascular regeneration, thereby preventing scar formation and promoting hair follicle regeneration. Furthermore, the expression of endoplasmic reticulum stress markers IRE1, XBP1, and CHOP was inhibited significantly in M-SF/SA treatment. In conclusion, the bilayer SF/SA scaffold is an ideal delivery platform for hUC-MSCs, and the M-SF/SA system could locally promote scarless skin healing and hair follicle regeneration by alleviating the IRE1/XBP1 signal pathway.
Collapse
Affiliation(s)
- Zhaona Dou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Yimeng Ren
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Quan Wen
- Yinfeng Academy of Life Science, Yichang Laboratory of Hematopoietic Stem Cells, 1109 Gangxing Sanlu, Jinan, Shandong Province 250100, China
| | - Ying Shen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Lin Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Lei Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Tao Jiang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Xinke Xia
- Institute of Life Science, Yinfeng Biological Group, High-tech Zone, Jinan, Shandong Province 250100, China
| |
Collapse
|
136
|
Ma H, Siu WS, Leung PC. The Potential of MSC-Based Cell-Free Therapy in Wound Healing-A Thorough Literature Review. Int J Mol Sci 2023; 24:ijms24119356. [PMID: 37298306 DOI: 10.3390/ijms24119356] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
A wound is an interruption of the normal anatomic structure and function of the skin, which is critical in protecting against foreign pathogens, regulating body temperature and water balance. Wound healing is a complex process involving various phases, including coagulation, inflammation, angiogenesis, re-epithelialization, and re-modeling. Factors such as infection, ischemia, and chronic diseases such as diabetes can compromise wound healing, leading to chronic and refractory ulcers. Mesenchymal stem cells (MSCs) have been used to treat various wound models due to their paracrine activity (secretome) and extracellular vehicles (exosomes) that contain several molecules, including long non-coding RNAs (lncRNAs), micro-RNAs (miRNAs), proteins, and lipids. Studies have shown that MSCs-based cell-free therapy using secretome and exosomes has great potential in regenerative medicine compared to MSCs, as there are fewer safety concerns. This review provides an overview of the pathophysiology of cutaneous wounds and the potential of MSCs-based cell-free therapy in each phase of wound healing. It also discusses clinical studies of MSCs-based cell-free therapies.
Collapse
Affiliation(s)
- Hui Ma
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Wing-Sum Siu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
137
|
Valsecchi C, Croce S, Lenta E, Acquafredda G, Comoli P, Avanzini MA. TITLE: New therapeutic approaches in pediatric diseases: Mesenchymal stromal cell and mesenchymal stromal cell-derived extracellular vesicles as new drugs. Pharmacol Res 2023; 192:106796. [PMID: 37207738 DOI: 10.1016/j.phrs.2023.106796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Mesenchymal Stromal Cell (MSC) clinical applications have been widely reported and their therapeutic potential has been documented in several diseases. MSCs can be isolated from several human tissues and easily expanded in vitro, they are able to differentiate in a variety of cell lineages, and they are known to interact with most immunological cells, showing immunosuppressive and tissue repair properties. Their therapeutic efficacy is closely associated with the release of bioactive molecules, namely Extracellular Vesicles (EVs), effective as their parental cells. EVs isolated from MSCs act by fusing with target cell membrane and releasing their content, showing a great potential for the treatment of injured tissues and organs, and for the modulation of the host immune system. EV-based therapies provide, as major advantages, the possibility to cross the epithelium and blood barrier and their activity is not influenced by the surrounding environment. In the present review, we deal with pre-clinical reports and clinical trials to provide data in support of MSC and EV clinical efficacy with particular focus on neonatal and pediatric diseases. Considering pre-clinical and clinical data so far available, it is likely that cell-based and cell-free therapies could become an important therapeutic approach for the treatment of several pediatric diseases.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Stefania Croce
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Elisa Lenta
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Gloria Acquafredda
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Patrizia Comoli
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| |
Collapse
|
138
|
Parascandolo A, Di Tolla MF, Liguoro D, Lecce M, Misso S, Micieli F, Ambrosio MR, Cabaro S, Beguinot F, Pelagalli A, D'Esposito V, Formisano P. Human Platelet-Rich Plasma Regulates Canine Mesenchymal Stem Cell Migration through Aquaporins. Stem Cells Int 2023; 2023:8344259. [PMID: 37223543 PMCID: PMC10202607 DOI: 10.1155/2023/8344259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
Platelet products are commonly used in regenerative medicine due to their effects on the acceleration and promotion of wound healing, reduction of bleeding, synthesis of new connective tissue, and revascularization. Furthermore, a novel approach for the treatment of damaged tissues, following trauma or other pathological damages, is represented by the use of mesenchymal stem cells (MSCs). In dogs, both platelet-rich plasma (PRP) and MSCs have been suggested to be promising options for subacute skin wounds. However, the collection of canine PRP is not always feasible. In this study, we investigated the effect of human PRP (hPRP) on canine MSCs (cMSCs). We isolated cMSCs and observed that hPRP did not modify the expression levels of the primary class of major histocompatibility complex genes. However, hPRP was able to increase cMSC viability and migration by at least 1.5-fold. hPRP treatment enhanced both Aquaporin (AQP) 1 and AQP5 protein levels, and their inhibition by tetraethylammonium chloride led to a reduction of PRP-induced migration of cMSCs. In conclusion, we have provided evidence that hPRP supports cMSC survival and may promote cell migration, at least through AQP activation. Thus, hPRP may be useful in canine tissue regeneration and repair, placing as a promising tool for veterinary therapeutic approaches.
Collapse
Affiliation(s)
- Alessia Parascandolo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Michele Francesco Di Tolla
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Domenico Liguoro
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Manuela Lecce
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Saverio Misso
- Unit of Transfusion Medicine, Azienda Sanitaria Locale Caserta, Caserta, Italy
| | - Fabiana Micieli
- Department of Veterinary Medicine and Animal Productions, University of Napoli Federico II, 80137 Naples, Italy
| | - Maria Rosaria Ambrosio
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Napoli Federico II, 80131 Naples, Italy
- Institute of Biostructures and Bioimages, National Research Council, 80145 Naples, Italy
| | - Vittoria D'Esposito
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
139
|
Liu Y, Graves DT, Wang S. Development and clinical application of human mesenchymal stem cell drugs. Sci Bull (Beijing) 2023; 68:860-863. [PMID: 37045665 PMCID: PMC10784989 DOI: 10.1016/j.scib.2023.03.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing 100050, China; Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
140
|
An S, Anwar K, Ashraf M, Lee H, Jung R, Koganti R, Ghassemi M, Djalilian AR. Wound-Healing Effects of Mesenchymal Stromal Cell Secretome in the Cornea and the Role of Exosomes. Pharmaceutics 2023; 15:1486. [PMID: 37242728 PMCID: PMC10221647 DOI: 10.3390/pharmaceutics15051486] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) and their secreted factors have been shown to have immunomodulatory and regenerative effects. In this study, we investigated human bone-marrow-derived MSC secretome (MSC-S) for the treatment of corneal epithelial wounds. Specifically, we evaluated the role of MSC extracellular vesicles (EV)/exosomes in mediating the wound-healing effects of the MSC-S. In vitro studies using human corneal epithelial cells showed that MSC-CM increased cell proliferation in HCEC and HCLE cells, while EV-depleted MSC-CM showed lower cell proliferation in both cell lines compared to the MSC-CM group. In vitro and in vivo experiments revealed that 1X MSC-S consistently promoted wound healing more effectively than 0.5X MSC-S, and MSC-CM promoted wound healing in a dose-dependent manner, while exosome deprivation delayed wound healing. We further evaluated the incubation period of MSC-CM on corneal wound healing and showed that MSC-S collected for 72 h is more effective than MSC-S collected for 48 h. Finally, we evaluated the stability of MSC-S under different storage conditions and found that after one cycle of freeze-thawing, MSC-S is stable at 4 °C for up to 4 weeks. Collectively, we identified the following: (i) MSC-EV/Exo as the active ingredient in MSC-S that mediates the wound-healing effects in the corneal epithelium, providing a measure to optimize its dosing for a potential clinical product; (ii) Treatment with EV/Exo-containing MSC-S resulted in an improved corneal barrier and decreased corneal haze/edema relative to EV/Exo-depleted MSC-S; (iii) The stability of MSC-CM for up to 4 weeks showed that the regular storage condition did not significantly impact its stability and therapeutic functions.
Collapse
Affiliation(s)
- Seungwon An
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Khandaker Anwar
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Mohammadjavad Ashraf
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Hyungjo Lee
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Rebecca Jung
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Mahmood Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (K.A.); (M.A.); (H.L.); (R.J.); (R.K.); (M.G.)
| |
Collapse
|
141
|
Humenik F, Maloveská M, Hudáková N, Petroušková P, Šufliarska Z, Horňáková Ľ, Valenčáková A, Kožár M, Šišková B, Mudroňová D, Bartkovský M, Čížková D. Impact of Canine Amniotic Mesenchymal Stem Cell Conditioned Media on the Wound Healing Process: In Vitro and In Vivo Study. Int J Mol Sci 2023; 24:ijms24098214. [PMID: 37175924 PMCID: PMC10179513 DOI: 10.3390/ijms24098214] [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: 03/15/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of this study was to provide a beneficial treatment effect of mesenchymal stem cell products derived from the canine amniotic membrane (AM-MSC) on the complicated wound healing process in dogs. AM-MSCs were characterized in terms of morphology, phenotypic profile, and multilineage differentiation potential. The in vitro study of the effect of canine amniotic mesenchymal stem cell conditioned media (AMMSC-CM) on a primary skin fibroblast cell culture scratch assay showed a decrease in the measured scratch area of about 66.39% against the negative control (Dulbecco's Modified Eagle's Medium-32.55%) and the positive control (Dulbecco's Modified Eagle's Medium supplemented with FGF2, N2, B27, and EGF-82.077%) after 72 h treatment. In the experimental study, seven dogs with complicated nonhealing wounds were treated with a combination of antibiotics, NSAIDs, and local AMMSC-CM application. After 15 days of therapy, we observed a 98.47% reduction in the wound surface area as opposed to 57.135% in the control group treated by conventional therapy based on debridement of necrotic tissue, antibiotic therapy, pain management, and change of wound dressing.
Collapse
Affiliation(s)
- Filip Humenik
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Marcela Maloveská
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Nikola Hudáková
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Patrícia Petroušková
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Zuzana Šufliarska
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Ľubica Horňáková
- Small Animal Clinic, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Alexandra Valenčáková
- Small Animal Clinic, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Martin Kožár
- Small Animal Clinic, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Barbora Šišková
- Small Animal Clinic, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Dagmar Mudroňová
- Institute of Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Martin Bartkovský
- Department of Food Hygiene, Technology and Safety, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| | - Daša Čížková
- Centre of Experimental and Clinical Regenerative Medicine, The University of Veterinary Medicine and Pharmacy in Kosice, 040 01 Kosice, Slovakia
| |
Collapse
|
142
|
Ren H, Liu M, Jihu Y, Zeng H, Yao C, Yan H. Hypoxia activates the PI3K/AKT/HIF-1α pathway to promote the anti-inflammatory effect of adipose mesenchymal stem cells. Acta Histochem 2023; 125:152042. [PMID: 37137202 DOI: 10.1016/j.acthis.2023.152042] [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: 03/04/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/05/2023]
Abstract
This study aimed to investigate the effect of hypoxia on the anti-inflammatory effect of adipose-derived mesenchymal stem cells (AMSCs) in vitro and its possible mechanism. AMSCs were cultured in vitro in a hypoxic environment with 3% O2, and a normoxic (21% O2) environment was used as the control. The cells were identified by in vitro adipogenic and osteogenic differentiation and cell surface antigen detection, and the cell viability were detected. The effect of hypoxic AMSCs on macrophage inflammation was analyzed by co-culture. The results showed that under hypoxia, AMSCs had better viability, significantly downregulated the expression of inflammatory factors, alleviated macrophage inflammation, and activated the PI3K/AKT/HIF-1α pathway.
Collapse
Affiliation(s)
- Hongjing Ren
- Southwest Medical University, NO.1 Section 1, Xianglin Road, Luzhou City, Sichuan Province 646000, China
| | - Mengchang Liu
- Southwest Medical University, NO.1 Section 1, Xianglin Road, Luzhou City, Sichuan Province 646000, China
| | - Yueda Jihu
- Southwest Medical University, NO.1 Section 1, Xianglin Road, Luzhou City, Sichuan Province 646000, China
| | - Huizhen Zeng
- Southwest Medical University, NO.1 Section 1, Xianglin Road, Luzhou City, Sichuan Province 646000, China
| | - Chong Yao
- Southwest Medical University, NO.1 Section 1, Xianglin Road, Luzhou City, Sichuan Province 646000, China
| | - Hong Yan
- Department of Plastic and Burn Surgery, Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, NO.25 Taiping Street, Jiangyang District, Luzhou 646000 Sichuan Province, China.
| |
Collapse
|
143
|
Min Q, Yang L, Tian H, Tang L, Xiao Z, Shen J. Immunomodulatory Mechanism and Potential Application of Dental Pulp-Derived Stem Cells in Immune-Mediated Diseases. Int J Mol Sci 2023; 24:ijms24098068. [PMID: 37175774 PMCID: PMC10178746 DOI: 10.3390/ijms24098068] [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: 03/03/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are mesenchymal stem cells (MSCs) derived from dental pulp tissue, which have high self-renewal ability and multi-lineage differentiation potential. With the discovery of the immunoregulatory ability of stem cells, DPSCs have attracted much attention because they have similar or even better immunomodulatory effects than MSCs from other sources. DPSCs and their exosomes can exert an immunomodulatory ability by acting on target immune cells to regulate cytokines. DPSCs can also migrate to the lesion site to differentiate into target cells to repair the injured tissue, and play an important role in tissue regeneration. The aim of this review is to summarize the molecular mechanism and target cells of the immunomodulatory effects of DPSCs, and the latest advances in preclinical research in the treatment of various immune-mediated diseases, providing new reflections for their clinical application. DPSCs may be a promising source of stem cells for the treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Qi Min
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Liqiong Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Hua Tian
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Lu Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| |
Collapse
|
144
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
145
|
Saadh MJ, Ramírez-Coronel AA, Saini RS, Arias-Gonzáles JL, Amin AH, Gavilán JCO, Sârbu I. Advances in mesenchymal stem/stromal cell-based therapy and their extracellular vesicles for skin wound healing. Hum Cell 2023:10.1007/s13577-023-00904-8. [PMID: 37067766 DOI: 10.1007/s13577-023-00904-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023]
Abstract
Wound healing is a dynamic and complicated process containing overlapping phases. Presently, definitive therapy is not available, and the investigation into optimal wound care is influenced by the efficacy and cost-effectiveness of developing therapies. Accumulating evidence demonstrated the potential role of mesenchymal stem/stromal cell (MSC) therapy in several tissue injuries and diseases due to their high proliferation and differentiation abilities along with an easy collection procedure, low tumorigenesis, and immuno-privileged status. MSCs have also accelerated wound repair in all phases through their advantageous properties, such as accelerating wound closure, improving re-epithelialization, elevating angiogenesis, suppressing inflammation, and modulating extracellular matrix (ECM) remodeling. In addition, the beneficial therapeutic impacts of MSCs are largely associated with their paracrine functions, including extracellular vesicles (EVs). Exosomes and microvesicles are the two main subgroups of EVs. These vesicles are heterogeneous bilayer membrane structures that contain several proteins, lipids, and nucleic acids. EVs have emerged as a promising alternative to stem cell-based therapies because of their lower immunogenicity, tumorigenicity, and ease of management. MSCs from various sources have been widely investigated in skin wound healing and regeneration. Considering these features, in this review, we highlighted recent studies that the investigated therapeutic potential of various MSCs and MSC-EVs in skin damages and wounds.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
| | | | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, Pontifical University of Peru, San Miguel, Peru
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | | | - Ioan Sârbu
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, "Grigore T. Popa", University of Medicine and Pharmacy, 700115, Iași, Romania.
| |
Collapse
|
146
|
Ozel C, Apaydin E, Sariboyaci AE, Tamayol A, Avci H. A multifunctional sateen woven dressings for treatment of skin injuries. Colloids Surf B Biointerfaces 2023; 224:113197. [PMID: 36822118 DOI: 10.1016/j.colsurfb.2023.113197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cutaneous wounds with impaired healing such as diabetic ulcers and burns constitute major and rapidly growing threat to healthcare systems worldwide. Accelerating wound healing requires the delivery of biological factors that induce angiogenesis, support cellular proliferation, and modulate inflammation while minimizing infection. In this study, we engineered a dressing made by weaving of composite fibers (CFs) carrying mesenchymal stem cells (MSCs) and a model antibiotic using a scalable sateen textile technique. In this regard, two different sets of CFs carrying MSCs or an antimicrobial agent were used to generate a multifunctional dressing. According to cell viability and metabolic activity as CCK-8 and live/dead with qRT-PCR results, more than %90 the encapsulated MSCs remain viable for 28 days and their expression levels of the wound repair factors including ECM remodeling, angiogenesis and immunomodulatory maintained in MSCs post dressing manufacturing for 14 days. Post 10 days culture of the dressing, MSCs within CFs had 10-fold higher collagen synthesis (p < 0.0001) determined by hydroxyproline assay which indicates the enhanced healing properties. According to in vitro antimicrobial activity results determined by disk diffusion and broth microdilution tests, the first day and the total amount of release gentamicin loaded dressing samples during the 28 days were higher than determined minimal inhibition concentration (MIC) values for S. aureus and K. pneumonia without negatively impacting the viability and functionality of encapsulated MSCs within the dressing. The dressing is also flexible and can conform to skin curvatures making the dressing suitable for the treatment of different skin injuries such as burns and diabetic ulcers.
Collapse
Affiliation(s)
- Ceren Ozel
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Elif Apaydin
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskişehir 26470, Turkey
| | - Ayla Eker Sariboyaci
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06269, USA.
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir 26040, Turkey; Department of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey; Translational Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey.
| |
Collapse
|
147
|
Yang Q, Qin B, Hou W, Qin H, Yin F. Pathogenesis and therapy of radiation enteritis with gut microbiota. Front Pharmacol 2023; 14:1116558. [PMID: 37063268 PMCID: PMC10102376 DOI: 10.3389/fphar.2023.1116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
Radiotherapy is widely used in clinic due to its good effect for cancer treatment. But radiotherapy of malignant tumors in the abdomen and pelvis is easy to cause radiation enteritis complications. Gastrointestinal tract contains numerous microbes, most of which are mutualistic relationship with the host. Abdominal radiation results in gut microbiota dysbiosis. Microbial therapy can directly target gut microbiota to reverse microbiota dysbiosis, hence relieving intestinal inflammation. In this review, we mainly summarized pathogenesis and novel therapy of the radiation-induced intestinal injury with gut microbiota dysbiosis and envision the opportunities and challenges of radiation enteritis therapy.
Collapse
Affiliation(s)
- Qilin Yang
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- School of Clinical Medicine of Nanjing Medical University, Nanjing, China
| | - Bingzhi Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| |
Collapse
|
148
|
Brembilla NC, Modarressi A, André-Lévigne D, Brioudes E, Lanza F, Vuagnat H, Durual S, Marger L, Boehncke WH, Krause KH, Preynat-Seauve O. Adipose-Derived Stromal Cells within a Gelatin Matrix Acquire Enhanced Regenerative and Angiogenic Properties: A Pre-Clinical Study for Application to Chronic Wounds. Biomedicines 2023; 11:biomedicines11030987. [PMID: 36979966 PMCID: PMC10046849 DOI: 10.3390/biomedicines11030987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
This study evaluates the influence of a gelatin sponge on adipose-derived stromal cells (ASC). Transcriptomic data revealed that, compared to ASC in a monolayer, a cross-linked porcine gelatin sponge strongly influences the transcriptome of ASC. Wound healing genes were massively regulated, notably with the inflammatory and angiogenic factors. Proteomics on conditioned media showed that gelatin also acted as a concentrator and reservoir of the regenerative ASC secretome. This secretome promoted fibroblast survival and epithelialization, and significantly increased the migration and tubular assembly of endothelial cells within fibronectin. ASC in gelatin on a chick chorioallantoic membrane were more connected to vessels than an empty sponge, confirming an increased angiogenesis in vivo. No tumor formation was observed in immunodeficient nude mice to which an ASC gelatin sponge was transplanted subcutaneously. Finally, ASC in a gelatin sponge prepared from outbred rats accelerated closure and re-vascularization of ischemic wounds in the footpads of rats. In conclusion, we provide here preclinical evidence that a cross-linked porcine gelatin sponge is an optimal carrier to concentrate and increase the regenerative activity of ASC, notably angiogenic. This formulation of ASC represents an optimal, convenient and clinically compliant option for the delivery of ASC on ischemic wounds.
Collapse
Affiliation(s)
- Nicolo Costantino Brembilla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Division of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Ali Modarressi
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Dominik André-Lévigne
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Estelle Brioudes
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Florian Lanza
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Hubert Vuagnat
- Program for Wounds and Wound Healing, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Stéphane Durual
- Laboratory of Biomaterials, Faculty of Dental Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Laurine Marger
- Laboratory of Biomaterials, Faculty of Dental Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Division of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Laboratory of Therapy and Stem Cells, Geneva University Hospitals, 1205 Geneva, Switzerland
- Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
149
|
Peng Y, Meng H, Li PX, Jiang YF, Fu XB. [Research advances of stem cell-based tissue engineering repair materials in promoting the healing of chronic refractory wounds on the body surface]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2023; 39:290-295. [PMID: 37805728 DOI: 10.3760/cma.j.cn501225-20220407-00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
Repairing chronic refractory wounds on the body surface is a complex medical problem involving all stages of wound healing. In recent years, stem cells (SCs) and tissue engineering (TE) have brought hope for repairing chronic refractory wounds. SCs have excellent regenerative and paracrine effects; various TE strategies have the potential to repair chronic refractory wounds on the body surface and also improve the delivery efficiency of SCs. This article reviews the pathological characteristics of chronic refractory wounds, SCs used to repair chronic refractory wounds, and SC-based TE wound repair strategies.
Collapse
Affiliation(s)
- Y Peng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, the PLA General Hospital, Beijing 100048, China
| | - H Meng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, the PLA General Hospital, Beijing 100048, China
| | - P X Li
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Y F Jiang
- Department for Wound Repair and Plastic Surgery, PLA Strategic Support Force Characteristic Medical Center, Beijing 100005, China
| | - X B Fu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, the PLA General Hospital, Beijing 100048, China School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
150
|
Yu X, Liu P, Li Z, Zhang Z. Function and mechanism of mesenchymal stem cells in the healing of diabetic foot wounds. Front Endocrinol (Lausanne) 2023; 14:1099310. [PMID: 37008908 PMCID: PMC10061144 DOI: 10.3389/fendo.2023.1099310] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Diabetes has become a global public health problem. Diabetic foot is one of the most severe complications of diabetes, which often places a heavy economic burden on patients and seriously affects their quality of life. The current conventional treatment for the diabetic foot can only relieve the symptoms or delay the progression of the disease but cannot repair damaged blood vessels and nerves. An increasing number of studies have shown that mesenchymal stem cells (MSCs) can promote angiogenesis and re-epithelialization, participate in immune regulation, reduce inflammation, and finally repair diabetic foot ulcer (DFU), rendering it an effective means of treating diabetic foot disease. Currently, stem cells used in the treatment of diabetic foot are divided into two categories: autologous and allogeneic. They are mainly derived from the bone marrow, umbilical cord, adipose tissue, and placenta. MSCs from different sources have similar characteristics and subtle differences. Mastering their features to better select and use MSCs is the premise of improving the therapeutic effect of DFU. This article reviews the types and characteristics of MSCs and their molecular mechanisms and functions in treating DFU to provide innovative ideas for using MSCs to treat diabetic foot and promote wound healing.
Collapse
Affiliation(s)
- Xiaoping Yu
- School of Medicine and Nursing, Chengdu University, Chengdu, Sichuan, China
| | - Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zheng Li
- People’s Hospital of Jiulongpo District, Chongqing, China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|