1
|
Yang P, Chen X, Qin Y, Yu L, Ge G, Yin W, Zhang W, Li W, Li W, Xia W, Wu Z, Ding F, Bai J, Meng F, Geng D. Regulation of osteoimmune microenvironment via functional dynamic hydrogel for diabetic bone regeneration. Biomaterials 2025; 320:123273. [PMID: 40121832 DOI: 10.1016/j.biomaterials.2025.123273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Bone regeneration and repair face formidable challenges under diabetic conditions, primarily due to the disruption of macrophage polarization induced by diabetes and the inflammatory imbalance within the bone microenvironment. We have developed a novel dynamic hydrogel system (AG-CD@LINA), constructed through the coordination crosslinking of thiolated gelatin (SH-Gelatin) and gold ions (Au3+), followed by grafting with cyclodextrin to load the ligand linagliptin. This hydrogel effectively inhibits the formation of M1 macrophages and the expression of pro-inflammatory cytokines by gradually releasing linagliptin. Simultaneously, it promotes the formation of M2 macrophages and the expression of anti-inflammatory cytokines, thus improving the inflammatory microenvironment of diabetic bone defects. Consequently, it facilitates the migration of mesenchymal stem cells and angiogenic cells, augments osteogenic activity, and promotes vascularization, collectively accelerating the regeneration of diabetic bone tissue. Mechanistically, polarization occurs through the TLR3-NF-κB signaling pathway. In vivo experiments demonstrate that the in-situ injection of the hydrogel enhances the regeneration of bone tissue and the restoration of bone structure in diabetic bone defects, effectively modulating local inflammation and promoting vascular formation. This study suggests that functionalized dynamic hydrogels can improve the inflammatory microenvironment by regulating in situ macrophage polarization, thereby facilitating the reconstruction of bone microstructure. This approach represents a promising novel therapeutic strategy for diabetic bone defects.
Collapse
Affiliation(s)
- Peng Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China; Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215006, Jiangsu, China; Suzhou Key Laboratory of Orthopedic Medical Engineering, Suzhou, 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yi Qin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Weiling Yin
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zebin Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Fan Ding
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China.
| | - Fanwen Meng
- Department of Implant Dentistry, Suzhou Stomatological Hospital, Suzhou, 215005, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
2
|
Zhang M, Zhou S, Zhang T, Li J, Xue L, Liang B, Xing D. Shark skin and mussel-inspired polyurethane hydrogel sponge for wounds with infection and exudate. J Colloid Interface Sci 2025; 693:137658. [PMID: 40279845 DOI: 10.1016/j.jcis.2025.137658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Inspired by the antifouling properties of shark skin and the bioadhesion of mussels, our study presents a three-layer biomimetic wound dressing with hierarchical wettability and rapid exudate drainage capabilities. The shark skin-inspired hydrophobic modified polyurethane (PU) sponge provides antifouling properties and serves as a bacterial barrier. The mussel-inspired dopamine-functionalized carboxymethyl chitosan hydrogel (CMCS-DOP) absorbs exudates and forms an in situ hydrogel, effectively capturing and eliminating bacteria. The porous sponge layer in direct contact with the wound facilitates rapid exudate drainage, preventing excessive wound hydration. This hierarchical structure coordinates exudate transport and bacterial removal. The fabricated PCD hydrogel sponge dressing (PCD dressing) exhibits a wettability transition (contact angle: 3°-35°-101°) and a water vapor transmission rate of 1021-797-691 g/m2. It demonstrates potent bactericidal effects against Staphylococcus aureus and Escherichia coli, with survival rates of only 13 % and 14 %, respectively, and bacterial-blocking efficiencies of 89 % and 94 %. In a chronic bacterial infection wound model, the PCD dressing outperforms conventional clinical dressings, increasing the wound healing rate by 25.8 %, reducing inflammation, and enhancing angiogenesis and collagen deposition. Notably, the PCD mitigates oxidative stress at the wound site by regulating the polarization of anti-inflammatory macrophages. This exudate-draining and responsive dressing offers a promising strategy for promoting the healing of wounds with high exudate levels.
Collapse
Affiliation(s)
- Miao Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Sha Zhou
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Basic Medicine, Qingdao University, Qingdao, Shandong 266000, China
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Jiyixuan Li
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Basic Medicine, Qingdao University, Qingdao, Shandong 266000, China
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
3
|
Wang S, Cheng W, Wang X, Wu Z, Su J. Progress of microneedle targeted modulation technology in the reconstruction of immune microenvironment in diabetic wounds. Eur J Med Res 2025; 30:405. [PMID: 40394697 PMCID: PMC12090542 DOI: 10.1186/s40001-025-02667-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
Wound healing in diabetic patients is mainly hindered by a combination of long-term glycosylation, persistent inflammatory response, and immunosuppressive state. The interaction of these factors not only results in considerable prolongation of the wound healing process but also elevates the likelihood of recurrent ulcer development, profoundly affecting patients' quality of life. Traditional treatments, including surgical debridement, anti-infection, dressing application, vascular intervention, and glycaemic control, can only relieve some symptoms. However, they are often ineffective in addressing the underlying cause of impaired wound healing. It is of concern that the importance of the immune microenvironment in diabetic wound healing has not yet been fully appreciated and investigated, and the homeostasis of the immune microenvironment is crucial for promoting cell proliferation, angiogenesis, and tissue repair. However, this microenvironment is often dysregulated in the diabetic state. This paper reviews the key factors leading to dysregulation of the immune microenvironment, including immune cell dysfunction, abnormal cytokine expression, and disruption of key signalling pathways, and introduces an innovative silicone-based microneedle drug delivery method, which takes advantage of microneedle's precise targeting and highly efficient drug loading capacity to deliver drugs with immunomodulatory functions directly to the wound in a sustained manner, activate the corresponding signalling pathways, promote the polarization of M1 macrophages into the M2 phenotype, and stimulate neovascularization, providing a low inflammatory and pro-angiogenic immune microenvironment for diabetic wound healing, which provides a new therapeutic idea and means for diabetic wound healing.
Collapse
Affiliation(s)
- Shunsheng Wang
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Wei Cheng
- Jiangsu Key Laboratory of Thin Films, School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Xue Wang
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Zhuofan Wu
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Jiandong Su
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China.
| |
Collapse
|
4
|
Sharma S, Kishen A. Dysfunctional crosstalk between macrophages and fibroblasts under LPS-infected and hyperglycemic environment in diabetic wounds. Sci Rep 2025; 15:17233. [PMID: 40383800 PMCID: PMC12086240 DOI: 10.1038/s41598-025-00673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/29/2025] [Indexed: 05/20/2025] Open
Abstract
Diabetic wounds, especially diabetic foot ulcers, present a major clinical challenge due to delayed healing and prolonged inflammation. Macrophage-fibroblast interactions are essential for wound repair, yet this crosstalk is disrupted in diabetic wounds due to hyperglycemia and bacterial infection. This study investigates the dysfunctional communication between macrophages and fibroblasts, focusing on autocrine, paracrine, and juxtacrine signaling in simulated diabetic environments. Using monoculture and co-culture models of THP-1-derived macrophages and primary human dermal fibroblasts, we simulated conditions of normal glucose, LPS-induced infection, high glucose (with AGEs), and combined high glucose (with AGEs) and LPS. Macrophages in hyperglycemic and LPS-infected environments exhibited a pro-inflammatory M1 phenotype with elevated expression of CD80, and STAT1 and increased production of IL-1β, TNF-α, and MMP9. Fibroblast migration was significantly impaired under high glucose conditions, particularly in paracrine model. Secretome profiling showed heightened pro-inflammatory cytokines and proteases, with reduced anti-inflammatory markers (IL-10 and VEGF-A) under hyperglycemic conditions. Paracrine signaling exacerbated the inflammatory response, while juxtacrine signaling showed more moderate effects, conducive to healing. These findings highlight the pathological macrophage-fibroblast crosstalk in diabetic wounds, particularly under hyperglycemic and LPS-infected conditions, offering insights for potential immunomodulatory therapies aimed at restoring effective signaling and improving wound healing outcomes.
Collapse
Affiliation(s)
- Shivam Sharma
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada.
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada.
- Department of Dentistry, Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
5
|
Zhou Y, Zhou Y, Chen H, Zhang L, Bi S. Potential Role of CD99 Signaling Pathway in Schwann Cell Dysfunction in Diabetic Foot Ulcers Based on Single-Cell Transcriptome Analysis. J Diabetes Res 2025; 2025:9935400. [PMID: 40420926 PMCID: PMC12103954 DOI: 10.1155/jdr/9935400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Background: Schwann cell (SC) dysfunction contributes to the delayed healing of diabetic foot ulcers (DFUs). However, the underlying molecular mechanism regarding the unregulated SC function is poorly understood. Thus, we examined the single-cell transcriptome data from different DFU states focusing on SC characteristics. Methods: The single-cell RNA sequencing (scRNA-seq) data of DFU was obtained from the Gene Expression Omnibus (GEO) database, covering foot skin samples from nondiabetic patients, diabetic patients without DFU, DFU healers, and DFU nonhealers. After scRNA-seq data processing, downscaling, and cell cluster identification, cell communication analysis was performed by the CellChat package. Furthermore, we subclustered SC populations and ran the trajectory inference and pseudotime analysis to investigate the dynamic changes in SC. Finally, the significant pathways were validated with a db/db mouse wound model. Results: scRNA-seq analysis revealed different SC percentages and gene markers across the DFU groups. We identified that the CD99 signaling pathway was upregulated in the DFU nonhealer group. In the db/db mouse wound model, we observed that CD99 was highly expressed in the demyelinated area of the peripheral nerve fibers. Conclusion: Our study elucidated that the CD99 pathway activation may play a crucial role in SC dysfunction of DFU, providing insights into the peripheral glia regulation mechanism and potential therapeutic target of DFU.
Collapse
Affiliation(s)
- Yannan Zhou
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yaxin Zhou
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haohan Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
6
|
Valsami EA, Chu G, Guan M, Gilman J, Theocharidis G, Veves A. The Role of Omics Techniques in Diabetic Wound Healing: Recent Insights into the Application of Single-Cell RNA Sequencing, Bulk RNA Sequencing, Spatial Transcriptomics, and Proteomics. Adv Ther 2025:10.1007/s12325-025-03212-9. [PMID: 40381157 DOI: 10.1007/s12325-025-03212-9] [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] [Received: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 05/19/2025]
Abstract
Diabetic foot ulcers (DFUs) are a devastating complication of diabetes mellitus (DM) that affect millions of people worldwide every year. They have a long-term impact on patients' quality of life and pose a significant challenge for both patients and clinicians, alongside negative economic implications on affected individuals. The current therapeutic approaches are costly and, in many cases, ineffective, highlighting the urgent need to develop novel, affordable, more efficient, and personalized treatments. Recent advances in high-throughput omics technologies, including proteomics, bulk RNA sequencing (bulk RNA-seq), single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics in both preclinical animal and human clinical studies, have enhanced our understanding of the molecular function and mechanisms of DFUs, thereby offering potential for targeted therapies. Additionally, these technologies provide valuable insights behind the mechanism of action of novel wound dressings and treatments. In this review, we outline the latest application of omics technologies in DFU preclinical animal and human clinical research on diabetic wound healing, and spotlight recent findings.A graphical abstract is available with this article.
Collapse
Affiliation(s)
- Eleftheria-Angeliki Valsami
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA
| | - Guangyu Chu
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA
| | - Ming Guan
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA
| | - Jessica Gilman
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA
| | - Georgios Theocharidis
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Palmer 321A, One Deaconess Rd, Boston, MA, 02215, USA.
| |
Collapse
|
7
|
Yao X, Wang X, Zhang R, Kong L, Fan C, Qian Y. Dysregulated mast cell activation induced by diabetic milieu exacerbates the progression of diabetic peripheral neuropathy in mice. Nat Commun 2025; 16:4170. [PMID: 40325050 PMCID: PMC12052842 DOI: 10.1038/s41467-025-59562-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
Diabetic peripheral neuropathy (DPN), a common disorder in diabetes, is associated with severe microenvironment imbalance due to immunometabolic stress. However, the underlying mechanistic drivers remain unclear. Here, we generate a single-cell atlas of human peripheral nerves and identify cell-specific transcriptional changes in DPN as well as aberrant amplification of mast cells. Using streptozotocin-induced mouse diabetes models, we further find that glucose uptake mediated by GLUT3 in high-glucose (HG) diabetic milieu upregulates ERK1/2 phosphorylation in mouse mast cells. Sustained HG stimulation also induces aberrant mTOR hyperactivity, resulting in endoplasmic reticulum stress and mitochondrial oxidative stress, thereby impairing mitochondrial functions of mast cells. Dysregulated mast cells then degranulate and release histamine, tryptase and inflammatory factors into neural microenvironment to cause neuropathy in diabetic mice. Lastly, mice with mast cell deficiency are protected from the immune imbalance in nerves and progression of neuropathy. Our findings thus implicate dysregulated activation of mast cells as a potential driver in the progression of DPN.
Collapse
Affiliation(s)
- Xiangyun Yao
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Zhang
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingchi Kong
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cunyi Fan
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yun Qian
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Bekaryssova D, Yessirkepov M, Rakisheva AU, Bakytzhan A. Diabetic foot in the context of rheumatic diseases: pathogenesis and treatment approaches. Rheumatol Int 2025; 45:132. [PMID: 40314825 DOI: 10.1007/s00296-025-05890-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Diabetic foot is a frequent and debilitating complication of diabetes mellitus that significantly impairs quality of life and increases the risk of disability and amputation. This review examines the multifactorial pathogenesis of diabetic foot, focusing on its increased incidence and severity in patients with rheumatic diseases. The development of diabetic foot is driven by diabetic neuropathy, peripheral vascular disease, and infection. In patients with rheumatic diseases, chronic systemic inflammation and vascular dysfunction further accelerate tissue damage and impair wound healing. Long-term use of pharmacologic agents such as glucocorticoids and nonsteroidal anti-inflammatory drugs also contributes to metabolic imbalance, immune suppression, and vascular complications, increasing the risk of ulceration and infection. Rheumatic disease-related joint deformities and altered foot biomechanics add mechanical stress, exacerbating the condition. Effective management of diabetic foot in patients with rheumatic diseases requires a multidisciplinary approach. This includes early diagnosis, strict glycemic control, modulation of systemic inflammation, optimization of vascular health, and preventive foot care strategies. Addressing both metabolic and rheumatologic components is essential to reduce the risk of severe outcomes such as chronic infection and limb amputation. Understanding the interplay between diabetes and rheumatic diseases is crucial for improving clinical outcomes. Targeted, integrated interventions are key to preventing complications and enhancing the quality of life for affected patients.
Collapse
Affiliation(s)
- Dana Bekaryssova
- Department of Project Management, South Kazakhstan Medical Academy, Shymkent, Kazakhstan.
| | - Marlen Yessirkepov
- Center for Life and Health Sciences, National Academy of Sciences under the President of the Republic of Kazakhstan, Almaty, Kazakhstan
- Department of Chemical Disciplines, Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | | | - Assylkhan Bakytzhan
- Department of Chemical Disciplines, Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| |
Collapse
|
9
|
Ku YC, Lee YC, Hong YK, Lo YL, Kuo CH, Wang KC, Hsu CK, Yu CH, Lin SW, Wu HL. Deciphering the Dysregulating IGF-1-SP1-CD248 Pathway in Fibroblast Functionality during Diabetic Wound Healing. J Invest Dermatol 2025; 145:1180-1195. [PMID: 39293711 DOI: 10.1016/j.jid.2024.07.035] [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: 02/26/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 09/20/2024]
Abstract
Reduced fibroblast activity is a critical factor in the progression of diabetic ulcers. CD248, a transmembrane glycoprotein prominently expressed in activated fibroblasts, plays a pivotal role in wound healing. However, the role of CD248 in diabetic wound healing and the CD248 regulatory pathway remains largely unexplored. Our study shows that CD248 expression is significantly reduced in skin wounds from both patients and mice with diabetes. Single-cell transcriptome data analyses reveal a marked reduction of CD248-enriched secretory-reticular fibroblasts in diabetic wounds. We identify IGF-1 as a key regulator of CD248 expression through the protein kinase B/mTOR signaling pathway and the SP1 transcription factor. Overexpression of CD248 enhances fibroblast motility, elucidating the under-representation of CD248-enriched fibroblasts in diabetic wounds. Immunohistochemical staining of diabetic wound samples further confirms low SP1 expression and fewer CD248-positive secretory-reticular fibroblasts. Further investigation reveals that elevated TNFα levels in diabetic environment promotes IGF-1 resistance, and inhibiting IGF-1 induced CD248 expression. In summary, our findings underscore the critical role of the IGF1-SP1-CD248 axis in activating reticular fibroblasts during wound-healing processes. Targeting this axis in fibroblasts could help develop a therapeutic regimen for diabetic ulcers.
Collapse
Affiliation(s)
- Ya-Chu Ku
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Chou Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Kai Hong
- Department of Dermatology, National Cheng Kung University Hospital, Tainan, Taiwan; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yung-Ling Lo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Hsiang Kuo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Chieh Wang
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Hung Yu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
10
|
Jia BB, Sun BK, Lee EY, Ren B. Emerging Techniques in Spatial Multiomics: Fundamental Principles and Applications to Dermatology. J Invest Dermatol 2025; 145:1017-1032. [PMID: 39503694 DOI: 10.1016/j.jid.2024.09.006] [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: 02/04/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 04/25/2025]
Abstract
Molecular pathology, such as high-throughput genomic and proteomic profiling, identifies precise disease targets from biopsies but require tissue dissociation, losing valuable histologic and spatial context. Emerging spatial multi-omic technologies now enable multiplexed visualization of genomic, proteomic, and epigenomic targets within a single tissue slice, eliminating the need for labeling multiple adjacent slices. Although early work focused on RNA (spatial transcriptomics), spatial technologies can now concurrently capture DNA, genome accessibility, histone modifications, and proteins with spatially-resolved single-cell resolution. This review outlines the principles, advantages, limitations, and potential for spatial technologies to advance dermatologic research. By jointly profiling multiple molecular channels, spatial multiomics enables novel studies of copy number variations, clonal heterogeneity, and enhancer dysregulation, replete with spatial context, illuminating the skin's complex heterogeneity.
Collapse
Affiliation(s)
- Bojing B Jia
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, California, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, California, USA
| | - Bryan K Sun
- Department of Dermatology, University of California, Irvine, Irvine, California, USA
| | - Ernest Y Lee
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Bing Ren
- Center for Epigenomics, Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, California, USA; Institute of Genomic Medicine, Moores Cancer Center, School of Medicine, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
11
|
Ni J, Chen Y, Zhang L, Wang R, Wu X, Khan NU, Xie F. Epigallocatechin gallate and vancomycin loaded poly(vinyl)-pyrrolidone-gelatine nanofibers, conceivable curative approach for wound healing. Colloids Surf B Biointerfaces 2025; 249:114506. [PMID: 39837051 DOI: 10.1016/j.colsurfb.2025.114506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/23/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
During surgical procedures, skin and soft tissue wounds are often infected by resistant strains of gram-positive bacteria and gram-negative bacteria, resulting in serious obstacles to the healing of these wounds. Commercially available dressings for such wounds are still insufficient to combat resistant infections. Here, we designed vancomycin and epigallocatechin gallate (EGCG) loaded poly(vinyl)-pyrrolidone-gelatine nanofiber's membrane dressing for potential synergistic efficiency against infected post-surgical wounds. The nanofiber's membrane was physiochemically characterized by surface morphology, chemical and physical compatibilities', thermal stability, and drug release. Disk diffusion assays, Minimum inhibitor concentrations (MICs), and fractional inhibitory concentration indexes (FICI) were measured to analyze synergistic efficiency against Escherichia coli. Furthermore, Balb/c mice were used for in vivo healing studies, and to observe the healing mechanisms, histological assessments were performed. The designed system displayed excellent physical and chemical properties. The in vitro studies unveiled controlled-release patterns of vancomycin and EGCG and, at the same time, revealed 1.5-fold higher antimicrobial synergistic efficacy (FICI 0.485) than vancomycin against E. coli. The wound healing mechanisms reflected quick and mature healing processes with the promotion of collagen and angiogenesis at wound sites. The designed electrospun nanofiber technology might be personalized, rapid wound healing remedy for scientists and healthcare providers, and may enhance patients' outcomes and quality of life.
Collapse
Affiliation(s)
- Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| | - Yanhua Chen
- Department of Pharmacy, Children's hospital of Jiangnan university (Wuxi Children's Hospital), Wuxi 214000, China
| | - Lan Zhang
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Rong Wang
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xiufeng Wu
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310012, China.
| | - Fen Xie
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China.
| |
Collapse
|
12
|
Bauer TM, Moon J, Shadiow J, Buckley S, Gallagher KA. Mechanisms of Impaired Wound Healing in Type 2 Diabetes: The Role of Epigenetic Factors. Arterioscler Thromb Vasc Biol 2025; 45:632-642. [PMID: 40109262 PMCID: PMC12018132 DOI: 10.1161/atvbaha.124.321446] [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: 03/22/2025]
Abstract
Despite decades of research, impaired extremity wound healing in type 2 diabetes remains a significant driver of patient morbidity, mortality, and health care costs. Advances in surgical and medical therapies, including the advent of endovascular interventions for peripheral artery disease and topical therapies developed to promote wound healing, have not reduced the frequency of lower leg amputations for nonhealing wounds in type 2 diabetes. This brief report is aimed at reviewing the roles of various cell types in tissue repair and summarizing the known dysfunctions of these cell types in diabetic foot ulcers. Recent advances in our understanding of the epigenetic regulation in immune cells identified to be altered in type 2 diabetes are summarized, and particular attention is paid to the developing research defining the epigenetic regulation of structural cells, including keratinocytes, fibroblasts, and endothelial cells. Gaps in knowledge are highlighted, and potential future directions are suggested based on the current state of the field.
Collapse
Affiliation(s)
- Tyler M. Bauer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jadie Moon
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - James Shadiow
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Sam Buckley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A. Gallagher
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Xiong Y, Knoedler S, Alfertshofer M, Kim BS, Jiang D, Liu G, Rinkevich Y, Mi B. Mechanisms and therapeutic opportunities in metabolic aberrations of diabetic wounds: a narrative review. Cell Death Dis 2025; 16:341. [PMID: 40280905 PMCID: PMC12032273 DOI: 10.1038/s41419-025-07583-3] [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: 01/26/2024] [Revised: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Metabolic aberrations are fundamental to the complex pathophysiology and challenges associated with diabetic wound healing. These alterations, induced by the diabetic environment, trigger a cascade of events that disrupt the normal wound-healing process. Key factors in this metabolic alternation include chronic hyperglycemia, insulin resistance, and dysregulated lipid and amino acid metabolism. In this review, we summarize the underlying mechanisms driving these metabolic changes in diabetic wounds, while emphasizing the broad implications of these disturbances. Additionally, we discuss therapeutic approaches that target these metabolic anomalies and how their integration with existing wound-healing treatments may yield synergistic effects, offering promising avenues for innovative therapies.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377, Munich, Germany
| | - Michael Alfertshofer
- Department of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University Munich, 80336, Munich, Germany
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Dongsheng Jiang
- Precision Research Centre for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377, Munich, Germany.
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Li L, Tang H, She L, Lin Z, Yu Q, Zeng Y, Chen L, Chen F, Liang G, Zhao X, Cho N, Wang Y. Dehydroabietic acid protects against cerebral ischaemia-reperfusion injury by modulating microglia-mediated neuroinflammation via targeting PKCδ. Br J Pharmacol 2025. [PMID: 40262763 DOI: 10.1111/bph.70030] [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: 10/19/2024] [Revised: 01/15/2025] [Accepted: 03/01/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Cerebral ischaemia-reperfusion injury (CIRI) is a major contributor to global morbidity and mortality, although its underlying mechanisms remain only partly understood. Emerging evidence indicates that inhibiting microglia-mediated neuroinflammation would be an effective therapeutic approach for CIRI, and pharmacological interventions targeting this pathway hold significant therapeutic promise. This study aimed to identify a potent anti-inflammatory drug from a natural compound library as a potential treatment for CIRI. EXPERIMENTAL APPROACH We used oxygen-glucose deprivation/reperfusion (OGD/R) and middle cerebral artery occlusion in male C57BL/6 mice to evaluate the efficacy of DHA in neurological deficits and the anti-inflammatory effects. Using BV2 cells and murine brain tissue, liquid chromatography-tandem mass spectrometry was used to identify potential molecular targets of DHA, followed by bio-layer interferometry, molecular docking, molecular dynamics simulations and cellular thermal shift assays to validate DHA's binding interactions with protein kinase C delta (PKCδ). KEY RESULTS DHA decreased production of pro-inflammatory cytokines following OGD/R, thereby inhibiting microglia-mediated neuroinflammation to protect neurons and reducing brain infarct size and improving neurological outcomes. Mechanistically, DHA directly bound to PKCδ, inhibiting its phosphorylation and downstream NF-κB signalling. This binding interaction involved TRP55 and LEU106 on PKCδ, as confirmed by molecular docking and other biophysical techniques. CONCLUSION AND IMPLICATIONS DHA specifically interacts with PKCδ, preventing its phosphorylation induced by ischaemia-reperfusion injury. These results suggest that DHA is a novel inhibitor of PKCδ and provide solid experimental foundations for using DHA in treating neuroinflammation-related conditions, such as CIRI.
Collapse
Affiliation(s)
- Luyao Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
- College of Pharmacy, Chonnam National University, Gwangju, South Korea
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Hao Tang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Lingyu She
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Qin Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Yuqing Zeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Linjie Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Fan Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Guang Liang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Xia Zhao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
15
|
Wan R, Fang S, Zhang X, Zhou W, Bi X, Yuan L, Lv Q, Song Y, Tang W, Shi Y, Li T. S100A9 as a promising therapeutic target for diabetic foot ulcers. Chin Med J (Engl) 2025; 138:973-981. [PMID: 40143429 PMCID: PMC12037093 DOI: 10.1097/cm9.0000000000003543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Diabetic foot is a complex condition with high incidence, recurrence, mortality, and disability rates. Current treatments for diabetic foot ulcers are often insufficient. This study was conducted to identify potential therapeutic targets for diabetic foot. METHODS Datasets related to diabetic foot and diabetic skin were retrieved from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using R software. Enrichment analysis was conducted to screen for critical gene functions and pathways. A protein interaction network was constructed to identify node genes corresponding to key proteins. The DEGs and node genes were overlapped to pinpoint target genes. Plasma and chronic ulcer samples from diabetic and non-diabetic individuals were collected. Western blotting, immunohistochemistry, and enzyme-linked immunosorbent assays were performed to verify the S100 calcium binding protein A9 (S100A9), inflammatory cytokine, and related pathway protein levels. Hematoxylin and eosin staining was used to measure epidermal layer thickness. RESULTS In total, 283 common DEGs and 42 node genes in diabetic foot ulcers were identified. Forty-three genes were differentially expressed in the skin of diabetic and non-diabetic individuals. The overlapping of the most significant DEGs and node genes led to the identification of S100A9 as a target gene. The S100A9 level was significantly higher in diabetic than in non-diabetic plasma (178.40 ± 44.65 ng/mL vs. 40.84 ± 18.86 ng/mL) and in chronic ulcers, and the wound healing time correlated positively with the plasma S100A9 level. The levels of inflammatory cytokines (tumor necrosis factor-α, interleukin [IL]-1, and IL-6) and related pathway proteins (phospho-extracellular signal regulated kinase [ERK], phospho-p38, phospho-p65, and p-protein kinase B [Akt]) were also elevated. The epidermal layer was notably thinner in chronic diabetic ulcers than in non-diabetic skin (24.17 ± 25.60 μm vs. 412.00 ± 181.60 μm). CONCLUSIONS S100A9 was significantly upregulated in diabetic foot and was associated with prolonged wound healing. S100A9 may impair diabetic wound healing by disrupting local inflammatory responses and skin re-epithelialization.
Collapse
Affiliation(s)
- Renhui Wan
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Shuo Fang
- Department of Plastics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xingxing Zhang
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Weiyi Zhou
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xiaoyan Bi
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Le Yuan
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Qian Lv
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yan Song
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Wei Tang
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yongquan Shi
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Tuo Li
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
16
|
Zhou Q, Chi J, Yang J, Dong X, Guo J, Lian F, Mamun AA, Chen T, Zhang H, Chen J, Tao Y, Ma Y, Shi K, Xiao J. Natural, safety immunomodulatory derivatives of lactobacillus biofilms promote diabetic wound healing by metabolically regulating macrophage phenotype and alleviating local inflammation. J Adv Res 2025:S2090-1232(25)00220-6. [PMID: 40187726 DOI: 10.1016/j.jare.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/24/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
INTRODUCTION Long-term inflammatory microenvironment further impairs the healing process of diabetic wounds. Many studies have shown that Lactobacillus can regulate immune function and promote injured tissue repair. However, the immunomodulatory function and safety of Lactobacillus biofilm (LB) on wounds need further investigation. OBJECTIVES In this present research, we proposed a "bacteria-free biofilm derivative therapy" and successfully extracted Lactobacillus biofilm derivatives (LBDs) by ultrasonic separation and filtration technology for the natural and safe treatment of diabetic wounds. METHODS The study first cultured Lactobacillus anaerobically and extracted LBDs using ultrasound separation combined with filtration technology. LBDs were characterized via scanning electron microscopy, Concanavalin A fluorescence staining, and protein gel electrophoresis. In vivo diabetic wound model, wound closure rates were dynamically monitored, and tissue sections were analyzed using hematoxylin-eosin and immunofluorescence staining to evaluate LBDs' healing effects. An in vitro macrophage inflammation model was established, employing immunofluorescence, flow cytometry, and Western blotting techniques to explore the molecular mechanisms underlying LBDs' effects on macrophage phenotypes. Furthermore, whole-genome sequencing and proteomics of LBDs-treated macrophages were performed to further elucidate the intrinsic molecular mechanisms through which LBDs regulate macrophage phenotypes. RESULTS LBDs were effectively extracted utilizing ultrasonic separation coupled with filtration technology. Studies revealed that LBDs modulate the systemic metabolic reprogramming in wound-site macrophages, suppress JAK-STAT1 signaling pathway, alleviate the local inflammatory microenvironment, promote neovascularization and ultimately accelerate wound healing. CONCLUSION The LBDs retains most bioactive components of the LB. As a natural, safe and immunomodulatory agent, LBDs promote diabetic wound healing by metabolically reprogramming macrophage phenotypes and improving the local immune microenvironment, offering promising potential for regenerative applications in diabetic wound management.
Collapse
Affiliation(s)
- Qingwei Zhou
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Medicine Department, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing 311800, China
| | - Junjie Chi
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 3250035, China
| | - Jintao Yang
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 3250035, China; Medicine Department, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing 311800, China
| | - Xiaoyu Dong
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiali Guo
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Feifei Lian
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Abdullah Al Mamun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Tianling Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Haijuan Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiaojiao Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yibing Tao
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yunmiao Ma
- Medicine Department, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing 311800, China.
| | - Keqing Shi
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 3250035, China.
| | - Jian Xiao
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
17
|
Zhang Y, Fang C, Zhang L, Ma F, Sun M, Zhang N, Bai N, Wu J. Identification and validation of immune-related biomarkers and polarization types of macrophages in keloid based on bulk RNA-seq and single-cell RNA-seq analysis. Burns 2025; 51:107413. [PMID: 39923303 DOI: 10.1016/j.burns.2025.107413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Keloids are a common complication that occurs after injury. The pathogenesis of this disease remains unknown. Therefore, identifying immune-related biomarkers and macrophage polarization types in keloids can provide new insights into their treatment. METHODS In this study, keloid-related bulk RNA-seq data (GSE83286, GSE212954, GSE92566, and GSE90051) were obtained from the Gene Expression Omnibus (GEO) database. The datasets GSE83286, GSE212964, and GSE92566 were combined to form a training set, while GSE90051 was utilized as an external validation set. Differentially expressed genes (DEGs) were detected by comparing keloid and normal samples within the training set. Differentially expressed immune-related genes (DIRGs) were then determined by intersecting the DEGs with immune-related genes (IRGs). Based on the protein-protein interaction (PPI) network, the top 40 DIRGs were selected for further analyses. Weighted Gene Co-expression Network Analysis (WGCNA), in conjunction with three machine learning techniques - least absolute shrinkage and selection operator (LASSO), support vector machine-recursive feature elimination (SVM-RFE), and random forest (RF) - employed to identify biomarkers. Subsequently, a nomogram model was constructed and validated. Single-cell RNA (scRNA) analysis was used to examine the expression of biomarkers at the cell-type level. Furthermore, since keloid is a chronic inflammatory disease and the abnormal polarization of macrophages is essential for the occurrence of this kind of disease, in this study we also endeavor to elucidate the state of macrophage polarization dysregulation within keloid, with the anticipation of generating novel concepts for the treatment of keloid. Finally, western blot (WB) and immunofluorescence (IF) analyses were carried out to confirm the expression levels of the biomarkers. RESULTS A total of 740 DEGs were identified in the training set, comprising 331 up-regulated genes and 409 down-regulated genes. After intersecting with the IRGs, 73 DIRGs were obtained. Subsequently, the top 40 DIRGs were chosen for further analysis. Eventually, two biomarkers, namely BMP1 and IL1R1, were identified through WGCNA and the three machine learning methods. Their expression levels were then verified by single-cell analysis, WB, and IF analysis. Additionally, it was found that the number of M2 macrophages significantly increased, while the number of M1 macrophages decreased in keloids compared to normal samples. CONCLUSION BMP1 and IL1R1 might function as novel biomarkers and potential therapeutic targets for keloid treatment. Moreover, upregulating M1 macrophages and downregulating M2 macrophages could represent a promising approach for the treatment of keloids.
Collapse
Affiliation(s)
- Yuzhu Zhang
- Intensive care unit, Linyi People's Hospital, Linyi, Shandong, China
| | - Chenglong Fang
- Department of Rehabilitation Medicine, Lin yi People's Hospital, Linyi, Shandong, China
| | - Lizhong Zhang
- Department of pathology, Lin Yi People's Hospital, Linyi, Shandong, China
| | - Fengyu Ma
- The People's Hospital of Rizhao, Rizhao, Shandong, China
| | - Meihong Sun
- Department of Pediatric Critical Care Medicine, Lin yi People's Hospital, Linyi, Shandong, China
| | - Ning Zhang
- Emergency Department of Ning yang First Peoples Hospital, Tai an, Shandong, China
| | - Nan Bai
- Medical Cosmetology and Plastic Surgery Center, Lin Yi People's Hospital, Linyi, Shandong, China.
| | - Jun Wu
- Medical Cosmetology and Plastic Surgery Center, Lin Yi People's Hospital, Linyi, Shandong, China.
| |
Collapse
|
18
|
Liao Z, Zheng Y, Zhang M, Li X, Wang J, Xu H. Dynamic single-cell transcriptomic reveals the cellular heterogeneity and a novel fibroblast subpopulation in laryngotracheal stenosis. Biol Direct 2025; 20:40. [PMID: 40165307 PMCID: PMC11956221 DOI: 10.1186/s13062-025-00639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Laryngotracheal stenosis (LTS), a pathological narrowing of the upper airway caused by excessive extracellular matrix (ECM) deposition, often leads to dyspnea and even respiratory failure. However, systematic studies addressing the specific subpopulations and their contribution to LTS development still remain underexplored. RESULTS We collected laryngotracheal tissue at multiple time points of LTS rat model, established by injuring their laryngotracheal lining, and performed dynamic single-cell RNA sequencing (scRNA-seq) to elucidate the transcriptomic atlas of LTS development. The results showed, from the inflammatory state to the repair/fibrotic state, infiltration of immune cells such as monocyte macrophages decreased and fibroblast increased. We delineated the markers and functional status of different fibroblasts subsets and identified that fibrotic fibroblasts may originate from multiple fibroblast subpopulations, including a new subpopulation characterized by the expression of chondrogenic markers such as Ucma and Col2a1, we designated this subcluster as chondrocyte injury-related fibroblasts (CIRF). Furthermore, we categorized monocytes/macrophages into several subtypes and identified that SPP1 high macrophages represented the largest macrophage subpopulation in LTS, providing evidence to clarify the importance of SPP1 macrophages in fibrosis disease. Our findings also revealed the interactions among these cells to explore the molecular mechanism associated with LTS pathogenesis. CONCLUSIONS Our study, for the first time, conducted dynamic scRNA-seq on LTS, revealing the cellular heterogeneity and providing a valuable resource for exploring the intricate molecular landscape of LTS. We propose CIRF may represent a tissue-specific fibroblast lineage in LTS and potentially originate from cells in the perichondrium of the trachea and transform into fibrotic fibroblasts. Integration of our study with those of other respiratory fibrotic diseases will allow for a comprehensive understanding of airway remodeling in respiratory diseases and exploring potential new therapeutic targets for their treatment.
Collapse
Affiliation(s)
- Ziwei Liao
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China
| | - Yangyang Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China
| | - Mingjun Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China
| | - Xiaoyan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China.
| | - Jing Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China.
| | - Hongming Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, No. 355, Luding Road, Shanghai, 200062, People's Republic of China.
| |
Collapse
|
19
|
Wang J, Yang X, Zhou T, Ma H, Yuan X, Yan S, Wang S. Microenvironment of diabetic foot ulcers: Implications for healing and therapeutic strategies. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2025; 30:19. [PMID: 40302998 PMCID: PMC12039865 DOI: 10.4103/jrms.jrms_573_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 05/02/2025]
Abstract
Diabetic foot ulcers (DFUs) are a common yet serious complication in individuals with diabetes, often presenting as chronic, nonhealing wounds that significantly impair quality of life. The healing process of DFUs is largely influenced by the local microenvironment, which encompasses factors such as hypoxia, inflammation, and the involvement of various cell types. Poor blood circulation in the affected area results in hypoxia, compromising cellular function and restricting nutrient supply, thereby delaying wound healing. In addition, chronic inflammation disrupts immune system balance, with excessive pro-inflammatory cytokines not only failing to facilitate tissue repair but also exacerbating tissue damage. Moreover, key cell types, including fibroblasts, keratinocytes, and macrophages, play crucial roles at different stages of the healing process, contributing to collagen production and skin regeneration. A comprehensive understanding of the complex dynamics within the DFU microenvironment is essential for developing more precise therapeutic approaches, such as advanced drug delivery systems and bioactive materials, aimed at promoting wound healing and reducing the risk of recurrence.
Collapse
Affiliation(s)
- Jixue Wang
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Xirui Yang
- Department of Ophthalmology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Tao Zhou
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Haitao Ma
- Department of Peripheral Vascular Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Xingxing Yuan
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Shuxun Yan
- Department of Endocrinology, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| | - Siqi Wang
- Department of Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
20
|
Fang S, Zhang H, Liu W, Li S, Chen Z, Min J, Dai C, An J, Zhang H, Liu D. Analysis and Validation of Mitophagy-Related Genes in Diabetic Foot Ulcers. J Inflamm Res 2025; 18:4367-4379. [PMID: 40162084 PMCID: PMC11954483 DOI: 10.2147/jir.s504001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose This study aimed to identify hub genes associated with mitophagy involved in the pathogenesis and progression of diabetic foot ulcer (DFU), and to characterize their immune cell infiltration features and single-cell expression profiles. Methods DFU-related datasets (GSE80178, GSE68183) were retrieved from the GEO database. Subsequently, differentially expressed genes (DEGs) were identified via limma analysis, followed by gene set enrichment analysis (GSEA) to assess gene function enrichment. Identified DEGs were intersected with mitophagy-related genes. Machine learning (ML) algorithms were further employed to identify hub genes. Additionally, immune cell infiltration was examined via the CIBERSORT algorithm, and the correlation between the identified genes and immune infiltration was investigated. Finally, hub genes identified were validated via the single-cell RNA sequencing dataset GSE165816, and further validated using RT-PCR and Western blot (WB) assays. Results Two hub genes, ANO6 and ALDH2, were identified and found to be significantly downregulated in the skin tissues of patients with DFU. Receiver operating characteristic (ROC) analysis demonstrated robust diagnostic potential (ANO6, AUC = 0.833, ALDH2, AUC = 0.806). Immune cell infiltration analysis demonstrated notable differences between the DFU and normal groups in naïve B cells, monocytes, resting mast cells, γδT cells, and regulatory T cells (Tregs). The findings were further validated through single-cell RNA sequencing (scRNA-seq) analysis and experimental studies, which confirmed the downregulation of ANO6 and ALDH2 in DFU tissues. Conclusion Two mitophagy-related hub genes, ANO6 and ALDH2, were identified and validated as being significantly downregulated in DFU. Both genes demonstrated diagnostic potential and showed an association with immune cell infiltration. These findings suggest that mitophagy dysfunction may contribute to the pathophysiology of DFU, potentially through the dysregulation of inflammatory pathways and immune responses. While the results provide valuable insights into DFU and its management, further studies with larger cohorts and deeper exploration of mechanistic links to inflammation are necessary to translate these findings into therapeutic strategies.
Collapse
Affiliation(s)
- Shaoyihan Fang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Huijuan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Wenjian Liu
- Department of Burns and Plastics, Jiangxi Provincial Corps Hospital of Chinese People’s Armed Police Forces, Nanchang, 330001, People’s Republic of China
| | - Shuangyan Li
- Department of Critical Care Medicine, Ezhou Central Hospital, Ezhou, Hubei, 436000, People’s Republic of China
| | - Zhenzhen Chen
- Outpatient Department, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Jingjie Min
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Chengyu Dai
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Jingwen An
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Hongxiao Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, People’s Republic of China
| |
Collapse
|
21
|
Zhao S, Yu H, Li Z, Chen W, Liu K, Dai H, Wang G, Zhang Z, Xie J, He Y, Li L. Single-cell RNA sequencing reveals a new mechanism of endothelial cell heterogeneity and healing in diabetic foot ulcers. Biol Direct 2025; 20:34. [PMID: 40121493 PMCID: PMC11929994 DOI: 10.1186/s13062-025-00628-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Diabetic foot ulcers (DFU) are a common and severe complication among diabetic patients, posing a significant burden on patients' quality of life and healthcare systems due to their high incidence, amputation rates, and mortality. This study utilized single-cell RNA sequencing technology to deeply analyze the cellular heterogeneity of the skin on the feet ofDFU patients and the transcriptomic characteristics of endothelial cells, aiming to identify key cell populations and genes associated with the healing and progression of DFU. The study found that endothelial cells from DFU patients exhibited significant transcriptomic differences under various conditions, particularly in signaling pathways related to inflammatory responses and angiogenesis. Through trajectory analysis and cell communication research, we revealed the key role of endothelial cell subsets in the development of DFU and identified multiple important gene modules associated with the progression of DFU. Notably, the promoting effect of the SH3BGRL3 gene on endothelial cell proliferation, migration, and angiogenic capabilities under high glucose conditions was experimentally verified, providing a new potential target and theoretical basis for the treatment of DFU. This study not only enhances the understanding of the pathogenesis ofDFU but also provides a scientific basis for the development ofnew therapeutic strategies.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hua Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zihao Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanying Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kaibo Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zibing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- National Key Clinical Specialty (Wound Healing), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
22
|
Li Y, Yu S, Zeng J, Zhou S, Cui X, Zhou J, Zhang P. Corilagin enhances wound healing by modulating the macrophage phenotype in diabetic mice. FASEB J 2025; 39:e70439. [PMID: 40052815 DOI: 10.1096/fj.202403085rr] [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: 12/03/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Excessive inflammation is a prominent issue in diabetic wounds, leading to delayed healing or amputation. Corilagin (Cori) is a natural polyphenolic compound with diverse pharmacological activities, particularly its anti-inflammatory properties. The aim of this study was to evaluate the anti-inflammatory effect of Cori on diabetic wounds and to explore the potential underlying mechanisms. The impact of Cori on wound healing was assessed in streptozotocin (STZ)-induced diabetic mice through morphological observation, histological staining, and gene expression analysis. Flow cytometry, qRT-PCR, western blot analysis, and RNA sequencing were conducted to elucidate the underlying mechanisms in RAW264.7 cells. The results demonstrated that Cori accelerated wound healing, inhibited excessive inflammation, and regulated macrophage polarization in diabetic mice. In Vitro, Cori decreased M1 polarization and inhibited the expression of pro-inflammatory mediators in RAW264.7 cells. Sequencing analysis revealed that Cori exerts anti-inflammatory effects on RAW 264.7 cells through multiple targeted mechanisms. Moreover, in LPS-induced macrophages, Cori dramatically decreased the activation of TLR4, MyD88, and NF-κB. Additionally, Cori enhanced M2 polarization by promoting fatty acid oxidation. In conclusion, the findings suggest that Cori modulates macrophage polarization through various targeted mechanisms, effectively suppressing inflammation and accelerating diabetic wound healing.
Collapse
Affiliation(s)
- Yun Li
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Site Yu
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Jizhang Zeng
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Situo Zhou
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Xu Cui
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Jie Zhou
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
23
|
Singh A, Bocher O, Zeggini E. Insights into the molecular underpinning of type 2 diabetes complications. Hum Mol Genet 2025; 34:469-480. [PMID: 39807636 PMCID: PMC11891870 DOI: 10.1093/hmg/ddae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Type 2 diabetes (T2D) complications pose a significant global health challenge. Omics technologies have been employed to investigate these complications and identify the biological pathways involved. In this review, we focus on four major T2D complications: diabetic kidney disease, diabetic retinopathy, diabetic neuropathy, and cardiovascular complications. We discuss advancements in omics research, summarizing findings from genetic, epigenomic, transcriptomic, proteomic, and metabolomic studies across different ancestries and disease-relevant tissues. We stress the importance of integrating multi-omics techniques to elucidate the biological mechanisms underlying T2D complications and advocate for ancestrally diverse studies. Ultimately, these insights will improve risk prediction for T2D complications and inform translation strategies.
Collapse
Affiliation(s)
- Archit Singh
- Technical University of Munich (TUM), TUM School of Medicine and Health, Graduate School of Experimental Medicine and Health Sciences, Ismaninger Straße 22, Munich 81675, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
- Munich School for Data Science (MUDS), Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Ismaninger Straße 22, Munich 81675, Germany
| |
Collapse
|
24
|
Liu Z, Bian X, Luo L, Björklund ÅK, Li L, Zhang L, Chen Y, Guo L, Gao J, Cao C, Wang J, He W, Xiao Y, Zhu L, Annusver K, Gopee NH, Basurto-Lozada D, Horsfall D, Bennett CL, Kasper M, Haniffa M, Sommar P, Li D, Landén NX. Spatiotemporal single-cell roadmap of human skin wound healing. Cell Stem Cell 2025; 32:479-498.e8. [PMID: 39729995 DOI: 10.1016/j.stem.2024.11.013] [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: 05/25/2024] [Revised: 09/24/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Wound healing is vital for human health, yet the details of cellular dynamics and coordination in human wound repair remain largely unexplored. To address this, we conducted single-cell multi-omics analyses on human skin wound tissues through inflammation, proliferation, and remodeling phases of wound repair from the same individuals, monitoring the cellular and molecular dynamics of human skin wound healing at an unprecedented spatiotemporal resolution. This singular roadmap reveals the cellular architecture of the wound margin and identifies FOSL1 as a critical driver of re-epithelialization. It shows that pro-inflammatory macrophages and fibroblasts sequentially support keratinocyte migration like a relay race across different healing stages. Comparison with single-cell data from venous and diabetic foot ulcers uncovers a link between failed keratinocyte migration and impaired inflammatory response in chronic wounds. Additionally, comparing human and mouse acute wound transcriptomes underscores the indispensable value of this roadmap in bridging basic research with clinical innovations.
Collapse
Affiliation(s)
- Zhuang Liu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Xiaowei Bian
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lihua Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Åsa K Björklund
- Department of Life Science, National Bioinformatics Infrastructure Sweden, Göteborg, Sweden; Science for Life Laboratory, Chalmers University of Technology, 41296 Göteborg, Sweden
| | - Li Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Letian Zhang
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lei Guo
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Juan Gao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Chunyan Cao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Jiating Wang
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Wenjun He
- The first affiliated hospital of Soochow University, Department of Plastic and Burn Surgery. NO.188, Shizi Street, Suzhou, Jiangsu, China
| | - Yunting Xiao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Liping Zhu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Daniela Basurto-Lozada
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - David Horsfall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Clare L Bennett
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Pehr Sommar
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China.
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden.
| |
Collapse
|
25
|
Verma SS, Sen CK, Srivastava R, Gnyawali SC, Katiyar P, Sahi AK, Kumar M, Rustagi Y, Liu S, Pandey D, Abouhashem AS, Fehme LNW, Kacar S, Mohanty SK, Faden-McCormack J, Murphy MP, Roy S, Wan J, Yoder MC, Singh K. Tissue nanotransfection-based endothelial PLCγ2-targeted epigenetic gene editing rescues perfusion and diabetic ischemic wound healing. Mol Ther 2025; 33:950-969. [PMID: 39863930 PMCID: PMC11897775 DOI: 10.1016/j.ymthe.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetic wounds are complicated by underlying peripheral vasculopathy. Reliance on vascular endothelial growth factor (VEGF) therapy to improve perfusion makes logical sense, yet clinical study outcomes on rescuing diabetic wound vascularization have yielded disappointing results. Our previous work has identified that low endothelial phospholipase Cγ2 (PLCγ2) expression hinders the therapeutic effect of VEGF on the diabetic ischemic limb. In this work, guided by single-cell RNA sequencing of human wound edge, we test the efficacy of gene-targeted therapeutic demethylation intending to improve VEGF-mediated neovascularization. PLCγ2 expression was diminished in all five identified diabetic wound-edge endothelial subclusters encompassing arterial, venous, and capillary cells. Such low expression was associated with hypermethylated PLCγ2 promoter. PLCγ2 promoter was also hypermethylated at murine diabetic ischemic wound edge. To specifically demethylate endothelial PLCγ2 promoter during VEGF therapy, a CRISPR-dCas9-based demethylation cocktail was delivered to the ischemic wound edge using tissue nanotransfection (TNT) technology. Demethylation-based upregulation of PLCγ2 during VEGF therapy improved wound tissue blood flow with an increased abundance of von Willebrand factor (vWF)+/PLCγ2+ vascular tissue elements by activating p44/p42-mitogen-activated protein kinase (MAPK) → hypoxia-inducible factor [HIF]-1α pathway. Taken together, TNT-based delivery of plasmids to demethylate the PLCγ2 gene promoter activity led to significant improvements in VEGF therapy for cutaneous diabetic wounds, resulting in better perfusion and accelerated wound closure.
Collapse
Affiliation(s)
- Sumit S Verma
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandan K Sen
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rajneesh Srivastava
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Surya C Gnyawali
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Parul Katiyar
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ajay K Sahi
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manishekhar Kumar
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yashika Rustagi
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sheng Liu
- Center for Computational Biology and Bioinformatics (CCBB), Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Diksha Pandey
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ahmed S Abouhashem
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Leila N W Fehme
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sedat Kacar
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sujit K Mohanty
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Julie Faden-McCormack
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Michael P Murphy
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sashwati Roy
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jun Wan
- Center for Computational Biology and Bioinformatics (CCBB), Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mervin C Yoder
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kanhaiya Singh
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
26
|
Liu W, Zhao Z. Scupa: single-cell unified polarization assessment of immune cells using the single-cell foundation model. Bioinformatics 2025; 41:btaf090. [PMID: 39999031 PMCID: PMC11893155 DOI: 10.1093/bioinformatics/btaf090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/15/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025] Open
Abstract
MOTIVATION Immune cells undergo cytokine-driven polarization in response to diverse stimuli, altering their transcriptional profiles and functional states. This dynamic process is central to immune responses in health and diseases, yet a systematic approach to assess cytokine-driven polarization in single-cell RNA sequencing data has been lacking. RESULTS To address this gap, we developed single-cell unified polarization assessment (Scupa), the first computational method for comprehensive immune cell polarization assessment. Scupa leverages data from the Immune Dictionary, which characterizes cytokine-driven polarization states across 14 immune cell types. By integrating cell embeddings from the single-cell foundation model Universal Cell Embeddings, Scupa effectively identifies polarized cells across different species and experimental conditions. Applications of Scupa in independent datasets demonstrated its accuracy in classifying polarized cells and further revealed distinct polarization profiles in tumor-infiltrating myeloid cells across cancers. Scupa complements conventional single-cell data analysis by providing new insights into dynamic immune cell states, and holds potential for advancing therapeutic insights, particularly in cytokine-based therapies. AVAILABILITY AND IMPLEMENTATION The code is available at https://github.com/bsml320/Scupa.
Collapse
Affiliation(s)
- Wendao Liu
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, United States
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Zhongming Zhao
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, United States
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| |
Collapse
|
27
|
Liu Y, Wang P, Li J, Chen L, Shu B, Wang H, Liu H, Zhao S, Zhou J, Chen X, Xie J. Single-cell RNA sequencing reveals the impaired epidermal differentiation and pathological microenvironment in diabetic foot ulcer. BURNS & TRAUMA 2025; 13:tkae065. [PMID: 40040959 PMCID: PMC11879498 DOI: 10.1093/burnst/tkae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 03/06/2025]
Abstract
Background Diabetic foot ulcer (DFU) is one of the most common and complex complications of diabetes, but the underlying pathophysiology remains unclear. Single-cell RNA sequencing (scRNA-seq) has been conducted to explore novel cell types or molecular profiles of DFU from various perspectives. This study aimed to comprehensively analyze the potential mechanisms underlying impaired re-epithelization of DFU in a single-cell perspective. Methods We conducted scRNA-seq on tissues from human normal skin, acute wound, and DFU to investigate the potential mechanisms underlying impaired epidermal differentiation and the pathological microenvironment. Pseudo-time and lineage inference analyses revealed the distinct states and transition trajectories of epidermal cells under different conditions. Transcription factor analysis revealed the potential regulatory mechanism of key subtypes of keratinocytes. Cell-cell interaction analysis revealed the regulatory network between the proinflammatory microenvironment and epidermal cells. Laser-capture microscopy coupled with RNA sequencing (LCM-seq) and multiplex immunohistochemistry were used to validate the expression and location of key subtypes of keratinocytes. Results Our research provided a comprehensive map of the phenotypic and dynamic changes that occur during epidermal differentiation, alongside the corresponding regulatory networks in DFU. Importantly, we identified two subtypes of keratinocytes: basal cells (BC-2) and diabetes-associated keratinocytes (DAK) that might play crucial roles in the impairment of epidermal homeostasis. BC-2 and DAK showed a marked increase in DFU, with an inactive state and insufficient motivation for epidermal differentiation. BC-2 was involved in the cellular response and apoptosis processes, with high expression of TXNIP, IFITM1, and IL1R2. Additionally, the pro-differentiation transcription factors were downregulated in BC-2 in DFU, indicating that the differentiation process might be inhibited in BC-2 in DFU. DAK was associated with cellular glucose homeostasis. Furthermore, increased CCL2 + CXCL2+ fibroblasts, VWA1+ vascular endothelial cells, and GZMA+CD8+ T cells were detected in DFU. These changes in the wound microenvironment could regulate the fate of epidermal cells through the TNFSF12-TNFRSF12A, IFNG-IFNGR1/2, and IL-1B-IL1R2 pathways, which might result in persistent inflammation and impaired epidermal differentiation in DFU. Conclusions Our findings offer novel insights into the pathophysiology of DFU and present potential therapeutic targets that could improve wound care and treatment outcomes for DFU patients.
Collapse
Affiliation(s)
- Yiling Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Lei Chen
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Bin Shu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Hanwen Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Hengdeng Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Shixin Zhao
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Junli Zhou
- Department of Burn and Plastic Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), No. 3 Wandao Road, Dongguan 523000, China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People’s Hospital of Foshan, No. 3 Lingnan Road, Foshan 528000, China
| | - Julin Xie
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| |
Collapse
|
28
|
Jian X, Han J, Liu X, Deng Y, Gao S, Xiao S, Zhang Y, Jian S, Huang Z, Hou Y, Qi F, Deng C. Exosome-carried miR-1248 from adipose-derived stem cells improves angiogenesis in diabetes-associated wounds. Int J Biol Macromol 2025; 297:139822. [PMID: 39809401 DOI: 10.1016/j.ijbiomac.2025.139822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Chronic non-healing wounds are a common complication of diabetes, marked by impaired angiogenesis. This study explores how exosomes (Exo-miR-1248) from miR-1248-overexpressing adipose-derived stem cells enhance diabetic wound healing by modulating endothelial cell function. Adipose-derived stem cells were transfected with a lentivirus carrying miR-1248 to produce Exo-miR-1248, isolated via differential centrifugation. In vitro, Exo-miR-1248's effects on proliferation, scratch wound healing, and tube formation in human umbilical vein endothelial cells (HUVECs) were assessed. For in vivo analysis, diabetic mice were induced with streptozotocin (STZ) and full-thickness skin defects were created. The impact of Exo-miR-1248 on wound healing was evaluated through subcutaneous injections. Histological analysis included H&E staining for epithelial regeneration and wound width, Masson's staining for collagen deposition, immunofluorescence for CD31 and α-SMA expression, RT-qPCR and WB for mRNA and protein levels of pro-angiogenic genes (VEGF-A, TGF-β, and Angpt-1). Exo-miR-1248 significantly enhanced HUVEC proliferation and migration. Tube formation assays showed increased capillary-like structures. In vivo, Exo-miR-1248-treated wounds healed faster, with improved collagen deposition and blood vessel formation. RT-qPCR and WB show that the mRNA and protein levels of VEGF-A, Angpt-1, and TGF-β are upregulated. Exo-miR-1248 may enhance diabetic wound healing by upregulating pro-angiogenic factors, offering a novel therapeutic approach.
Collapse
Affiliation(s)
- Xichao Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Jiansu Han
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Xin Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Yaping Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Shaoying Gao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China; The 2011 Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Affiliated Hospital of Zunyi Medical University, PR China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, PR China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China; The 2011 Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Affiliated Hospital of Zunyi Medical University, PR China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, PR China
| | - Yan Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Shiyu Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Zhanpeng Huang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Yinchi Hou
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China
| | - Fang Qi
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China; The 2011 Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Affiliated Hospital of Zunyi Medical University, PR China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, PR China.
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, PR China; The 2011 Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Affiliated Hospital of Zunyi Medical University, PR China; The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, PR China.
| |
Collapse
|
29
|
Almet AA, Liu Y, Nie Q, Plikus MV. Integrated Single-Cell Analysis Reveals Spatially and Temporally Dynamic Heterogeneity in Fibroblast States during Wound Healing. J Invest Dermatol 2025; 145:645-659.e25. [PMID: 39019149 DOI: 10.1016/j.jid.2024.06.1281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/19/2024]
Abstract
Wound healing is a dynamic process over temporal and spatial scales. Key to repair outcomes are fibroblasts; yet, how they modulate healing across time and in different wound regions remains incompletely understood. By integrating single-cell RNA-sequencing datasets of mouse skin and wounds, we infer that fibroblasts are the most transcriptionally dynamic skin-resident cells, evolving during postnatal skin maturation and rapidly after injury toward distinct late scar states. We show that transcriptional dynamics in fibroblasts are largely driven by genes encoding extracellular matrix and signaling factors. Lineage trajectory inference and spatial gene mapping reveal that Prg4-expressing fibroblasts transiently emerge along early wound edges. Within days, they become replaced by long-lasting and likely noninterconverting fibroblast populations, including Col25a1-expressing and Pamr1-expressing fibroblasts that occupy subepidermal and deep scar regions, respectively, where they engage in reciprocal signaling with immune cells. Signaling inference shows that fibroblast-immune crosstalk repeatedly uses some signaling pathways across wound healing time, whereas use of other signaling pathways is time and space limited. Collectively, we uncovered high transcriptional plasticity by wound fibroblasts, with early states transiently forming distinct microniches along wound edges and in the fascia, followed by stable states that stratify scar tissue into molecularly dissimilar upper and lower layers.
Collapse
Affiliation(s)
- Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Yingzi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA.
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
30
|
Zhang H, Yan Z, Zhu J, Li Z, Chen L, Zheng W, Dai Z, Yang J, Yun X, Wang Y, Zhou H, Jiang Z, Yu Q, Li S, Huang W, Yang L. Extracellular Mitochondrial-Derived Vesicles Affect the Progression of Diabetic Foot Ulcer by Regulating Oxidative Stress and Mitochondrial Dysfunction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407574. [PMID: 39835574 PMCID: PMC11904950 DOI: 10.1002/advs.202407574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/23/2024] [Indexed: 01/22/2025]
Abstract
Diabetic foot ulcer (DFU) is a common and severe complication of diabetes mellitus, the etiology of which remains insufficiently understood, particularly regarding the involvement of extracellular vesicles (EVs). In this study, nanoflow cytometry to detect EVs in DFU skin tissues is used and found a significant increase in the Translocase of Outer Mitochondrial Membrane 20 (TOM20)+ mitochondrial-derived vesicles (MDVs). The role of MDVs in DFU is yet to be reported. Using single-cell datasets, it is discovered that the increase in MDVs may be regulated by Sorting Nexin 9 (SNX9). In vitro experiments revealed that MDVs secreted by fibroblasts cultured in high glucose medium exhibited similar composition and protein enrichment results to those in DFU tissues, suggesting their potential as an ideal in vitro surrogate. These MDVs promoted apoptosis and intracellular oxidative stress, disrupted mitochondrial structure, and reduced aerobic metabolism in target cells. In vivo experiments also showed that MDV drops hindered wound healing in diabetic mice; however, this effect is rescued by SNX9 inhibitors, restoring mitochondrial dynamics and balance. Under high glucose conditions, MDVs significantly upregulated oxidative stress levels and induced mitochondrial dysfunction. This study proposes targeting MDVs as a potential therapeutic strategy for DFU.
Collapse
Affiliation(s)
- Huihui Zhang
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zi Yan
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Department of Microbiology and ImmunologyCollege of Basic Medicine and Public HygieneJinan UniversityGuangzhou510632China
- Guangdong Medical Innovation Platform for Translation of 3D Printing ApplicationThe Third Affiliated Hospital of Southern Medical UniversitySouthern Medical UniversityGuangzhou510630China
| | - Junyou Zhu
- Department of BurnsFirst affiliated hospitalSun Yat‐sen UniversityGuangzhou510080China
| | - Ziyue Li
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Guangdong Medical Innovation Platform for Translation of 3D Printing ApplicationThe Third Affiliated Hospital of Southern Medical UniversitySouthern Medical UniversityGuangzhou510630China
| | - Lianglong Chen
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Weihan Zheng
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Guangdong Medical Innovation Platform for Translation of 3D Printing ApplicationThe Third Affiliated Hospital of Southern Medical UniversitySouthern Medical UniversityGuangzhou510630China
| | - Zhenning Dai
- Department of StomatologyGuangdong Provincial Key Laboratory of Research and Development in Traditional Chinese MedicineGuangdong Second Traditional Chinese Medicine HospitalGuangzhou510095China
| | - Jiaxin Yang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Xinyi Yun
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Yilin Wang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Hai Zhou
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Ziwei Jiang
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qiuyi Yu
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Shiyu Li
- Department of Microbiology and ImmunologyCollege of Basic Medicine and Public HygieneJinan UniversityGuangzhou510632China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing ApplicationGuangdong Provincial Key Laboratory of Digital Medicine and BiomechanicsNational Key Discipline of Human AnatomySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Guangdong Medical Innovation Platform for Translation of 3D Printing ApplicationThe Third Affiliated Hospital of Southern Medical UniversitySouthern Medical UniversityGuangzhou510630China
| | - Lei Yang
- Department of BurnsNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
31
|
Kumaran G, Carroll L, Muirhead N, Bottomley MJ. How Can Spatial Transcriptomic Profiling Advance Our Understanding of Skin Diseases? J Invest Dermatol 2025; 145:522-535. [PMID: 39177547 DOI: 10.1016/j.jid.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/23/2024] [Accepted: 07/04/2024] [Indexed: 08/24/2024]
Abstract
Spatial transcriptomic (ST) profiling is the mapping of gene expression within cell populations with preservation of positional context and represents an exciting new approach to develop our understanding of local and regional influences upon skin biology in health and disease. With the ability to probe from a few hundred transcripts to the entire transcriptome, multiple ST approaches are now widely available. In this paper, we review the ST field and discuss its application to dermatology. Its potential to advance our understanding of skin biology in health and disease is highlighted through the illustrative examples of 3 research areas: cutaneous aging, tumorigenesis, and psoriasis.
Collapse
Affiliation(s)
- Girishkumar Kumaran
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Liam Carroll
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Matthew J Bottomley
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
32
|
Xu Z, Ni T, Zhang Q, Sun X, Zhao L, Lin J, Gao W, Yi M, Zhang L, Tu L, Wu G, Yan W. Exosomes derived from fibroblasts in DFUs delay wound healing by delivering miR-93-5p to target macrophage ATG16L1. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167640. [PMID: 39761761 DOI: 10.1016/j.bbadis.2024.167640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Diabetes is an extremely costly disease, one-third of which are attributed to the management of diabetic foot disease including chronic, non-healing, diabetic foot ulcers (DFUs). Therefore, much effort is needed to understand the pathogenesis of DFUs and novel therapeutics. We utilized exosome staining to confirm the interaction between fibroblast-derived exosomes and macrophages. Subsequently, we employed public data and qPCR to screen for upregulated miRNAs in fibroblast-derived exosomes in DFUs. The relationship between was validate miR-93-5 and ATG16L1 through data prediction and dual-luciferase reporter assays. A variety of molecular biology experiments were used for subsequent pathway validation. Additionally, we established Atg16l1MKI and Nlrp3MKO mice for further validation. We identified that miR-93-5p derived from fibroblasts played an important role in M1 macrophages polarization. Predicted by database, we found that miR-93-5p can bind to ATG16L1 mRNA, thereby influencing macrophage autophagy mediated by ATG16L1 in the clearance of ROS, thus activating the NLRP3 signaling pathway. In vivo, miR-93-5p antagomir treatment accelerated diabetic wound healing and induced M2 macrophage polarization. Fibroblasts and macrophages show cell crosstalk during the development of DFUs by miR-93-5p, and that antagomir treatment may be a promising and technically advantageous alternative to DFUs therapies.
Collapse
Affiliation(s)
- Zibo Xu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China; Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Tianyi Ni
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Xiaowei Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Liping Zhao
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Jinde Lin
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Weicheng Gao
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Min Yi
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Lantian Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Liying Tu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Guoping Wu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China.
| | - Wei Yan
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China.
| |
Collapse
|
33
|
Wan X, Yao H, Wei Z, Gao D, Zheng D, Xu B, Xie M. Heterogeneous porous hypoxia-mimicking scaffolds propel urethral reconstruction by promoting angiogenesis and regulating inflammation. Biomaterials 2025; 314:122833. [PMID: 39277947 DOI: 10.1016/j.biomaterials.2024.122833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
The nasty urine microenvironment (UME) impedes neourethral regeneration by inhibiting angiogenesis and inducing an excessive inflammatory response. Cellular adaptation to hypoxia improves regeneration in numerous tissues. In this study, heterogeneous porous hypoxia-mimicking scaffolds were fabricated for urethral reconstruction via promoting angiogenesis and modulating the inflammatory response based on sustained release of dimethyloxalylglycine (DMOG) to promote HIF-1α stabilization. Such scaffolds exhibit a two-layered structure: a dense layer composed of electrospun poly (l-lactic acid) (PLLA) nanofibrous mats and a loose layer composed of a porous gelatin matrix incorporated with DMOG-loaded mesoporous silica nanoparticles (DMSNs) and coated with poly(glycerol sebacate) (PGS). The modification of PGS could significantly increase rupture elongation, making the composite scaffolds more suitable for urethral tissue regeneration. Additionally, sustained release of DMOG from the scaffold facilitates proliferation, migration, tube formation, and angiogenetic gene expression in human umbilical vein endothelial cells (HUVECs), as well as stimulates M2 macrophage polarization and its regulation of HUVECs migration and smooth muscle cell (SMCs) contractile phenotype. These effects were downstream of the stabilization of HIF-1α in HUVECs and macrophages under hypoxia-mimicking conditions. Furthermore, the scaffold achieved better urethral reconstruction in a rabbit urethral stricture model, including an unobstructed urethra with a larger urethral diameter, increased regeneration of urothelial cells, SMCs, and neovascularization. Our results indicate that heterogeneous porous hypoxia-mimicking scaffolds could promote urethral reconstruction via facilitating angiogenesis and modulating inflammatory response.
Collapse
Affiliation(s)
- Xiang Wan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Haijun Yao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ziwei Wei
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Dajun Gao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Dachao Zheng
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Minkai Xie
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
34
|
Liu Y, Andersen B, Gudjonsson JE, Plikus MV. Ancestral Diversity of Skin Reaches Single-Cell Resolution. J Invest Dermatol 2025; 145:469-472. [PMID: 39306776 DOI: 10.1016/j.jid.2024.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 02/24/2025]
Affiliation(s)
- Yingzi Liu
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Bogi Andersen
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Johann E Gudjonsson
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
35
|
Doerfler P, Schoefmann N, Cabral G, Bauer W, Berli MC, Binder B, Borst C, Botter S, French LE, Goerge T, Hafner J, Hartmann D, Høgh A, Hoetzenecker W, Holzer-Geissler JCJ, Kamolz LP, Kofler K, Luger T, Nischwitz SP, Popovits M, Rappersberger K, Restivo G, Schlager JG, Schmuth M, Stingl G, Stockinger T, Stroelin A, Stuetz A, Umlauft J, Weninger WP, Wolff-Winiski B. Development of a Cellular Assay as a Personalized Model for Testing Chronic Wound Therapeutics. J Invest Dermatol 2025; 145:631-644.e22. [PMID: 38960086 DOI: 10.1016/j.jid.2024.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 07/05/2024]
Abstract
Exudates of nonhealing wounds contain drivers of pathogenicity. We utilized >800 exudates from nonhealing and healing wounds of diverse etiologies, collected by 3 different methods, to develop a wound-specific, cell-based functional biomarker assay. Human dermal fibroblast proliferation served as readout to (i) differentiate between healing and nonhealing wounds, (ii) follow the healing process of individual patients, and (iii) assess the effects of therapeutics for chronic wounds ex vivo. We observed a strong correlation between wound chronicity and inhibitory effects of individual exudates on fibroblast proliferation, with good diagnostic sensitivity (76-90%, depending on the sample collection method). Transition of a clinically nonhealing to a healing phenotype restored fibroblast proliferation and extracellular matrix formation while reducing inflammatory cytokine production. Transcriptional analysis of fibroblasts exposed to ex vivo nonhealing wound exudates revealed an induction of inflammatory cytokine and chemokine pathways and the unfolded protein response, indicating that these changes may contribute to the pathology of nonhealing wounds. Testing the wound therapeutics, PDGF and silver sulfadiazine, yielded responses in line with clinical experience and indicates the usefulness of the assay to search for and profile new therapeutics.
Collapse
Affiliation(s)
| | | | | | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Martin C Berli
- Balgrist University Hospital, Zurich, Switzerland; Technical Orthopedics, Diabetic Foot Consultation, Wound Outpatient Clinic, Spital Limmattal, Schlieren, Switzerland
| | - Barbara Binder
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Carina Borst
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sander Botter
- Swiss Center for Musculoskeletal Biobanking, Balgrist Campus AG, Zurich, Switzerland
| | - Lars E French
- Department of Dermatology and Allergology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tobias Goerge
- Department of Dermatology, University of Münster, Muenster, Germany
| | - Juerg Hafner
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Daniela Hartmann
- Department of Dermatology and Allergology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Annette Høgh
- Department of Vascular Surgery, Regionshospitalet Viborg, Viborg, Denmark
| | | | - Judith C J Holzer-Geissler
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Lars P Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Katrin Kofler
- Department of Dermatology, Medical University of Tübingen, Tuebingen, Germany
| | - Thomas Luger
- Department of Dermatology, University of Münster, Muenster, Germany
| | - Sebastian P Nischwitz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Michael Popovits
- Department of Surgery, Barmherzige Brueder Hospital Graz, Graz, Austria; Privatklinik Graz Ragnitz, Graz, Austria
| | | | - Gaetana Restivo
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Justin G Schlager
- Department of Dermatology and Allergology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matthias Schmuth
- Department of Dermatology, Venerology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Anke Stroelin
- Department of Dermatology, Medical University of Tübingen, Tuebingen, Germany
| | | | - Julian Umlauft
- Department of Dermatology, Venerology and Allergology, Medical University of Innsbruck, Innsbruck, Austria; Dermatology, Zellmed Medalp, Zell am Ziller, Austria
| | | | | |
Collapse
|
36
|
Choi D, Bakhtiari M, Pilcher W, Huang C, Thomas BE, Mumme H, Blanco G, Rajani R, Schechter MC, Fayfman M, Santamarina G, Bhasin S, Bhasin M. Single-Cell Analysis of Debrided Diabetic Foot Ulcers Reveals Dysregulated Wound Healing Environment in Non-Hispanic Black Patients. J Invest Dermatol 2025; 145:678-690. [PMID: 39127092 DOI: 10.1016/j.jid.2024.07.017] [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: 05/09/2023] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024]
Abstract
Diabetic foot ulcer is a critical complication of diabetes, but the wound microenvironment and its healing process are not completely understood. In this study, we optimized single-cell profiling from sharp debrided ulcers. Our findings demonstrate that healing diabetic foot ulcers were significantly enriched with distinct fibroblasts-expressing genes related to inflammation (CHI3L1, IL6) and extracellular matrix remodeling (ASPN), validating our previous studies on surgically resected ulcers. The race-focused analysis depicted lower expression of key healing-associated genes such as CHIL3L1, matrix metalloproteinase 11 gene MMP11, and SFRP4 in fibroblasts of non-Hispanic Black patients than in those of White patients. In cellular communication analysis, healing-enriched fibroblasts of non-Hispanic Black patients exhibited upregulation of signaling pathways such as WNT, whereas those of White patients showed insulin-like GF and Midkine pathways upregulation. Our findings advocate race as a risk marker of diabetic foot ulcer outcomes, likely reflecting underlying disparities in environmental exposures and access to care that profoundly influence healing markers. Using sharp debrided tissues for single-cell assays, this study highlights the need for in-depth investigations into dysregulated wound healing microenvironments of under-represented racial groups.
Collapse
Affiliation(s)
- Dahim Choi
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA
| | - Mojtaba Bakhtiari
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William Pilcher
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA
| | - Chenbin Huang
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hope Mumme
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Ravi Rajani
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA; Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marcos C Schechter
- Grady Memorial Hospital, Atlanta, Georgia, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maya Fayfman
- Grady Memorial Hospital, Atlanta, Georgia, USA; Division of Endocrinology Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gabriel Santamarina
- Grady Memorial Hospital, Atlanta, Georgia, USA; Division of Vascular Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Division of Endocrinology Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Swati Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Manoj Bhasin
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA; Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| |
Collapse
|
37
|
Song P, Liang Q, Ge X, Zhou D, Yuan M, Chu W, Xu J. Adipose-Derived Stem Cell Exosomes Promote Scar-Free Healing of Diabetic Wounds via miR-204-5p/TGF- β1/Smad Pathway. Stem Cells Int 2025; 2025:6344844. [PMID: 40018015 PMCID: PMC11865461 DOI: 10.1155/sci/6344844] [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: 06/27/2024] [Accepted: 01/18/2025] [Indexed: 03/01/2025] Open
Abstract
Numerous researches have demonstrated the therapeutic potential of adipose-derived stem cell exosomes (ADSC-Exos) in promoting wound healing. In this study, we aimed to investigate the impact of ADSC-Exos on diabetic wound fibroblasts and elucidate its possible mechanisms. CCK-8, Edu, cell scratch, and Transwell tests were used to evaluate the function of ADSC-Exos on rat skin fibroblasts (RSFs) in high-glucose (HG) medium. The targeting effect of ADSC-Exo-derived microRNA (miRNA) and TGF-β1 was assessed using bioinformatic analysis and then confirmed with western blot and dual luciferase reporter assays. ADSC-Exos, miR-204-5p mimic, and anti-miR-204-5p mimic were used to stimulate RSFs, and the levels of TGF-β1/Smad pathway were analyzed by western blot. In vivo, digital photo and tissue section staining were used to evaluate the therapeutic effect of ADSC-Exos on diabetic wounds. The data showed that ADSC-Exos enhance the proliferation and migration of fibroblasts under HG conditions, reduce excessive myofibroblast differentiation and collagen deposition, and promote scarless healing of diabetic wounds. Additionally, miR-204-5p in ADSC-Exos targets TGF-β1 to inhibit p-Smad2/3, Col I, and alpha-smooth muscle actin (α-SMA), thereby reducing fibrosis. These findings suggest that ADSC-Exos have potential prospects for promoting diabetic wound healing.
Collapse
Affiliation(s)
- Peijun Song
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Qiu Liang
- Department of Plastic Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou City 225000, Jiangsu Province, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Xiuyu Ge
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Danlian Zhou
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Mei Yuan
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Weiwei Chu
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| | - Jing Xu
- Department of Plastic Surgery and Burn, The First Affiliated Hospital of Bengbu Medical University, Bengbu City 233000, Anhui Province, China
| |
Collapse
|
38
|
Mahmoudi N, Sharifi S, Leshchiner D, Horibata S, Lin Z, Ghazali N, Shahbazi MA, Priyam A, Williams RJ, Pastar I, Gould L, Matoori S, Nisbet DR, Mahmoudi M. Tailored bioengineering and nanomedicine strategies for sex-specific healing of chronic wounds. Br J Dermatol 2025; 192:390-401. [PMID: 39565404 DOI: 10.1093/bjd/ljae457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024]
Abstract
Chronic wounds, defined by their prolonged healing process, significantly impair patients' quality of life and impose a hefty financial burden on healthcare systems worldwide. Sex- and gender-specific mechanisms regulate inflammation and infection, angiogenesis, matrix synthesis and cell recruitment. All of these processes contribute to cutaneous wound healing but remain largely understudied. This review aims to spotlight the innovative realm of bioengineering and nanomedicine, which is at the helm of revolutionizing complex chronic wound care. It underscores the significance of integrating patient sex into the development and (pre)clinical testing of these avant-garde treatment modalities, in order to enhance healing prospects for all patients regardless of sex. Moreover, we explore the representation of male and female patients in clinical trials of bioengineered and nanomedicine products. Finally, we examine the primary reasons for the historical neglect in translating sex-specific wound healing research into clinical practice and propose strategic solutions. By tackling these issues, the article advocates advanced treatment frameworks that could significantly improve healing outcomes for individuals of all sexes, thereby optimizing both efficacy and inclusivity in chronic wound management.
Collapse
Affiliation(s)
- Negar Mahmoudi
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Shahriar Sharifi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Dmitry Leshchiner
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Sachi Horibata
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Noor Ghazali
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ayushi Priyam
- IMPACT, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- IMPACT, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Irena Pastar
- Dr Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lisa Gould
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- South Shore Health Center for Wound Healing, Weymouth, MA, USA
| | - Simon Matoori
- Faculté de Pharmacie, Université de Montréal, Montreal, QC, Canada
| | - David R Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
- Connors Center for Women's Health & Gender Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Jabbari P, Kim JH, Le BH, Zhang W, Zhang H, Martins-Green M. Chronic Wound Initiation: Single-Cell RNAseq of Cutaneous Wound Tissue and Contributions of Oxidative Stress to Initiation of Chronicity. Antioxidants (Basel) 2025; 14:214. [PMID: 40002400 PMCID: PMC11852160 DOI: 10.3390/antiox14020214] [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: 12/14/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic wounds (CWs) in humans affect millions of people in the US alone, cost billions of dollars, cause much suffering, and still there are no effective treatments. Patients seek medical care when wound chronicity is already established, making it impossible to investigate factors that initiate chronicity. In this study, we used a diabetic mouse model of CWs that mimics many aspects of chronicity in humans. We performed scRNAseq to compare the cell composition and function during the first 72 h post-injury and profiled 102,737 cells into clusters of all major cell types involved in healing. We found two types of fibroblasts. Fib 1 (pro-healing) was enriched in non-CWs (NCWs) whereas Fib 2 (non-healing) was in CWs. Both showed disrupted proliferation and migration, and extracellular matrix (ECM) deposition in CWs. We identified several subtypes of keratinocytes, all of which were more abundant in NCWs, except for Channel-related keratinocytes, and showed altered migration, apoptosis, and response to oxidative stress (OS) in CWs. Vascular and lymphatic endothelial cells were both less abundant in CWs and both had impaired migration affecting the development of endothelial and lymphatic microvessels. Study of immune cells showed that neutrophils and mast cells are less abundant in CWs and that NCWs contained more proinflammatory macrophages (M1) whereas CWs were enriched in anti-inflammatory macrophages (M2). Also, several genes involved in mitochondrial function were abnormally expressed in CWs, suggesting impaired mitochondrial function and/or higher OS. Heat shock proteins needed for response to OS were downregulated in CWs, potentially leading to higher cellular damage. In conclusion, the initiation of chronicity is multifactorial and involves various cell types and cellular functions, indicating that one type of treatment will not fix all problems, unless the root cause is fundamental to the cell and molecular mechanisms of healing. We propose that such a fundamental process is high OS and its association with wound infection/biofilm.
Collapse
Affiliation(s)
- Parnian Jabbari
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, CA 92521, USA; (P.J.); (J.H.K.); (H.Z.)
| | - Jane H. Kim
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, CA 92521, USA; (P.J.); (J.H.K.); (H.Z.)
| | - Brandon H. Le
- Institute of Integrative Genome Biology, University of California, Riverside, CA 92521, USA; (B.H.L.); (W.Z.)
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92521, USA
| | - Wei Zhang
- Institute of Integrative Genome Biology, University of California, Riverside, CA 92521, USA; (B.H.L.); (W.Z.)
- Department of Botany and Plant Sciences, University of California, Riverside, CA 92521, USA
| | - Huimin Zhang
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, CA 92521, USA; (P.J.); (J.H.K.); (H.Z.)
| | - Manuela Martins-Green
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, CA 92521, USA; (P.J.); (J.H.K.); (H.Z.)
| |
Collapse
|
40
|
Balukoff NC, Houk G, Gonzalez T, Berton Y, Ronfard V, Pastar I, Tomic-Canic M. Out of this World: Wound Healing on Earth and in Space. J Invest Dermatol 2025:S0022-202X(25)00027-2. [PMID: 39955658 DOI: 10.1016/j.jid.2024.12.024] [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: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/17/2025]
Abstract
Impaired wound healing is a significant concern for humans in space, where the unique microgravity environment poses challenges to the natural healing processes of the body. Similar to chronic wounds on earth, such as diabetic foot ulcers and venous leg ulcers, wounds inflicted in space exhibit delayed or impaired healing responses. These wounds share common features, including dysregulated cellular signaling, altered cytokine profiles, and impaired tissue regeneration. Little is known about the mechanisms underlying wound healing under microgravity. In this review, we focused on exploring the parallels between wound healing in space and chronic wounds on earth as a fundamental approach for developing effective countermeasures to promote healing and mitigate associated health risks during long-space missions.
Collapse
Affiliation(s)
- Nathan C Balukoff
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Garrett Houk
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Tammy Gonzalez
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
41
|
Gomes MLNP, Krijnen PAJ, Middelkoop E, Niessen HWM, Boekema BKHL. Fetal Skin Wound Healing: Key Extracellular Matrix Components and Regulators in Scarless Healing. J Invest Dermatol 2025; 145:280-302. [PMID: 39152955 DOI: 10.1016/j.jid.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 08/19/2024]
Abstract
Fetal skin at early gestational stage is able to regenerate and heal rapidly after wounding. The exact mechanisms and molecular pathways involved in this process are however still largely unknown. The numerous differences in the skin of the early fetus versus skin in later developmental stages might provide clues for the mechanisms of scarless healing. This review summarizes the differences between mammalian fetal skin and the skin at later developmental phases in healthy and wounded conditions, focusing on extracellular matrix components, which are crucial factors in the microenvironment that direct cells and tissue functions and hence the wound healing process.
Collapse
Affiliation(s)
- Madalena Lopes Natário Pinto Gomes
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Esther Middelkoop
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Burn Centre, Red Cross Hospital, Beverwijk, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands; Department of Cardio-thoracic Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Bouke K H L Boekema
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands.
| |
Collapse
|
42
|
Li J, Meng H, Guo W, Zhou L, Wu S, Gao G, Liu Q, You D, Qu W. In-Situ Electrospinning Dressings Loaded with Kaempferol for Reducing MMP9 to Promote Diabetic Ulcer Healing. Int J Nanomedicine 2025; 20:1101-1117. [PMID: 39895983 PMCID: PMC11786600 DOI: 10.2147/ijn.s501370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Background Diabetic foot ulcers (DFUs) are often associated with persistent inflammatory response, impaired macrophage polarization, and slow vascular regeneration. Existing treatments cannot be adapted to wounds and do not achieve the desired therapeutic effects. The high porosity of biomaterials induces more M2 macrophages, while the natural compound kaempferol inhibits the expression of matrix metalloproteinase 9 (MMP9) and thus inhibits the associated inflammatory and immunological responses. Methods portable electrospinning dressings (PEDs) were prepared from the spinning solution using a portable electrospinning device. The material properties of PEDs were examined by scanning electron microscope, contact angle tester and WVTR-C3. Then, the in vitro biocompatibility of the dressings was evaluated using NIH3T3 cells. The in vivo wound healing efficacy of the dressings was analyzed in the diabetic wound model rats. Histological and immunofluorescence staining were performed to determine the status of epithelization, collagen deposition, MMP9 expression, macrophage polarization, inflammation response and angiogenesis. Results Material science experiments have shown that the dressing has optimal fiber micromorphology and good water vapor transport properties (WVTR: 4.88 kg m-2 24h-1); in vivo, diabetic wound experiments have shown that the high porosity and pharmacological effects of PED4 can mutually promote the rapid healing of diabetic wounds (healed 95.9% on day 15), facilitate the transformation of macrophages from M1-type to M2-type and regulate the expression of MMP9. Conclusion Portable electrospinning dressings equipped with kaempferol not only better fit irregular wounds, but also promote wound healing through MMP9 and macrophage polarization. Thus, PEDs show great promise for advancing research of personalized diabetic wound healing.
Collapse
Affiliation(s)
- Jianwen Li
- Gastroenteric Medicine and Digestive Endoscopy Center, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
- Joint International Research Laboratory of Ageing Active Strategy and Bionic Health in Northeast Asia of Ministry of Education, Changchun, 130041, People’s Republic of China
| | - Hongqi Meng
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Wenlai Guo
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Lubin Zhou
- Polymeric and Soft Materials Laboratory, Advanced Institute of Materials Science, School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, People’s Republic of China
| | - Siyu Wu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Guanghui Gao
- Polymeric and Soft Materials Laboratory, Advanced Institute of Materials Science, School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, People’s Republic of China
| | - Quanzhe Liu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Di You
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, 130012, People’s Republic of China
| | - Wenrui Qu
- Joint International Research Laboratory of Ageing Active Strategy and Bionic Health in Northeast Asia of Ministry of Education, Changchun, 130041, People’s Republic of China
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| |
Collapse
|
43
|
Ishikawa K, Hoshino Y, Osawa M, Funayama E, Miura T, Hojo M, Sasaki Y, Sasaki S, Yamamoto Y, Maeda T. Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat Accelerates Wound Healing in a Mouse Hind limb Lymphedema Model. Adv Wound Care (New Rochelle) 2025. [PMID: 39853221 DOI: 10.1089/wound.2024.0237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
Objective: Drugs regulating hypoxia-inducible factor (HIF)-1α have not been investigated for wound healing in lymphedema. Therefore, we examined the effects of drug modulation of HIF-1α activity for wound healing in our previously developed mouse model of nonirradiated hind limb lymphedema. Approach: Mouse hind limb lymphedema models (n = 17) and a sham group (n = 6) were created using 8- to 10-week-old male C57BL/6N mice. Mice with hind limb lymphedema were randomized into experimental groups receiving roxadustat, 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1), or dimethyl sulfoxide and were given intraperitoneal injections every 2 days for up to 2 weeks. Four days after the surgery, an 8-mm diameter full-thickness skin wound was created in the hind limb. The number of days required for wound closure and the percentage of wounds closed were measured. Skin samples taken at wound creation were evaluated by histological and molecular analysis. Results: Administration of roxadustat accelerated wound healing, whereas YC-1 delayed it, with a significant decrease and increase in skin thickness, respectively. The relative mRNA expression of Hif1α, matrix metalloproteinase-3, and interleukin-6 was significantly higher in the roxadustat group and that of metalloproteinase-9 was significantly lower in the roxadustat group compared with the control group. Innovation: This study is the first to demonstrate delayed wound healing in a mouse model of hind limb lymphedema and the first to demonstrate the promotion of significant wound healing through the use of roxadustat. Conclusion: Roxadustat exerts wound-healing effects and may promote the regulation of extracellular matrix remodeling via gene expression in hind limb lymphedema wound models.
Collapse
Affiliation(s)
- Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshitada Hoshino
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masayuki Osawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Emi Funayama
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Hojo
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Sasaki
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Sasaki
- Department of Plastic and Reconstructive Surgery, Center for Vascular Anomalies, Tonan Hospital, Sapporo, Japan
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
44
|
Chen C, Yang J, Shang R, Tang Y, Cai X, Chen Y, Liu Z, Hu W, Zhang W, Zhang X, Huang Y, Hu X, Yin W, Lu Q, Sheng H, Fan D, Ju Z, Luo G, He W. Orchestration of Macrophage Polarization Dynamics by Fibroblast-Secreted Exosomes during Skin Wound Healing. J Invest Dermatol 2025; 145:171-184.e6. [PMID: 38838771 DOI: 10.1016/j.jid.2024.05.007] [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: 02/12/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 06/07/2024]
Abstract
Macrophages undertake pivotal yet dichotomous functions during skin wound healing, mediating both early proinflammatory immune activation and late anti-inflammatory tissue remodeling processes. The timely phenotypic transition of macrophages from inflammatory M1 to proresolving M2 activation states is essential for efficient healing. However, the endogenous mechanisms calibrating macrophage polarization in accordance with the evolving tissue milieu remain undefined. In this study, we reveal an indispensable immunomodulatory role for fibroblast-secreted exosomes in directing macrophage activation dynamics. Fibroblast-derived exosomes permitted spatiotemporal coordination of macrophage phenotypes independent of direct intercellular contact. Exosomes enhanced macrophage sensitivity to both M1 and M2 polarizing stimuli, yet they also accelerated timely switching from M1 to M2 phenotypes. Exosome inhibition dysregulated macrophage responses, resulting in aberrant inflammation and impaired healing, whereas provision of exogenous fibroblast-derived exosomes corrected defects. Topical application of fibroblast-derived exosomes onto chronic diabetic wounds normalized dysregulated macrophage activation to resolve inflammation and restore productive healing. Our findings elucidate fibroblast-secreted exosomes as remote programmers of macrophage polarization that calibrate immunological transitions essential for tissue repair. Harnessing exosomes represents a previously unreported approach to steer productive macrophage activation states with immense therapeutic potential for promoting healing in chronic inflammatory disorders.
Collapse
Affiliation(s)
- Cheng Chen
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Jiacai Yang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Ruoyu Shang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yuanyang Tang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Cai
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yunxia Chen
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Zhihui Liu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Wengang Hu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Weiguang Zhang
- Department of Intensive Care, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaorong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yong Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Xiaohong Hu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Wenjing Yin
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China; Academy of Biological Engineering, Chongqing University, Chongqing, China
| | - Qudong Lu
- Department of Urology, Army 73rd Group Military Hospital, Xiamen, China
| | - Hao Sheng
- Department of Urology, The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dejiang Fan
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China.
| | - Weifeng He
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China.
| |
Collapse
|
45
|
Cho C, Haddadi NS, Kidacki M, Woodard GA, Shakiba S, Yıldız-Altay Ü, Richmond JM, Vesely MD. Spatial Transcriptomics in Inflammatory Skin Diseases Using GeoMx Digital Spatial Profiling: A Practical Guide for Applications in Dermatology. JID INNOVATIONS 2025; 5:100317. [PMID: 39559817 PMCID: PMC11570843 DOI: 10.1016/j.xjidi.2024.100317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 07/05/2024] [Accepted: 08/29/2024] [Indexed: 11/20/2024] Open
Abstract
The spatial organization of the skin is critical for its function. In particular, the skin immune microenvironment is arranged spatially and temporally, such that imbalances in the immune milieu are indicative of disease. Spatial transcriptomic platforms are helping to provide additional insights into aberrant inflammation in tissues that are not captured by tissue processing required for single-cell RNA sequencing. In this paper, we discuss a technical and user experience overview of NanoString's GeoMx Digital Spatial Profiler to perform in-depth spatial analysis of the transcriptome in inflammatory skin diseases. Our objective is to provide potential pitfalls and methods to optimize RNA capture that are not readily available in the manufacturer's guidelines. We use concrete examples from our experiments to demonstrate these strategies in inflammatory skin diseases, including psoriasis, lichen planus, and discoid lupus erythematosus. Overall, we hope to illustrate the potential of digital spatial profiling to dissect skin disease pathogenesis in a spatially resolved manner and provide a framework for other skin biology investigators using digital spatial profiling.
Collapse
Affiliation(s)
- Christina Cho
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nazgol-Sadat Haddadi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Michal Kidacki
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Gavitt A. Woodard
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Saeed Shakiba
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ümmügülsüm Yıldız-Altay
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jillian M. Richmond
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Matthew D. Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
46
|
Kim SE, Noda R, Liu YC, Nakajima Y, Kameoka S, Motooka D, Mizuno S, Takahashi S, Takaya K, Murase T, Ikematsu K, Tratsiakova K, Motoyama T, Nakashima M, Kishi K, Martin P, Seno S, Okuzaki D, Mori R. Novel integrated multiomics analysis reveals a key role for integrin beta-like 1 in wound scarring. EMBO Rep 2025; 26:122-152. [PMID: 39558136 PMCID: PMC11724056 DOI: 10.1038/s44319-024-00322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Exacerbation of scarring can originate from a minority fibroblast population that has undergone inflammatory-mediated genetic changes within the wound microenvironment. The fundamental relationship between molecular and spatial organization of the repair process at the single-cell level remains unclear. We have developed a novel, high-resolution spatial multiomics method that integrates spatial transcriptomics with scRNA-Seq; we identified new characteristic features of cell-cell communication and signaling during the repair process. Data from PU.1-/- mice, which lack an inflammatory response, combined with scRNA-Seq and Visium transcriptomics, led to the identification of nine genes potentially involved in inflammation-related scarring, including integrin beta-like 1 (Itgbl1). Transgenic mouse experiments confirmed that Itgbl1-expressing fibroblasts are required for granulation tissue formation and drive fibrogenesis during skin repair. Additionally, we detected a minority population of Acta2high-expressing myofibroblasts with apparent involvement in scarring, in conjunction with Itgbl1 expression. IL1β signaling inhibited Itgbl1 expression in TGFβ1-treated primary fibroblasts from humans and mice. Our novel methodology reveal molecular mechanisms underlying fibroblast-inflammatory cell interactions that initiate wound scarring.
Collapse
Affiliation(s)
- Sang-Eun Kim
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Ryota Noda
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Yu-Chen Liu
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yukari Nakajima
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shoichiro Kameoka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kento Takaya
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takehiko Murase
- Department of Forensic Pathology and Science, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kita, Kagawa, 761-0793, Japan
| | - Kazuya Ikematsu
- Department of Forensic Pathology and Science, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Katsiaryna Tratsiakova
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Takahiro Motoyama
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Paul Martin
- Department of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Ryoichi Mori
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan.
- Department of Tissue Repair and Regenerative Medical Science, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan.
- Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan.
| |
Collapse
|
47
|
Liao Y, Peng Z, Fu S, Hua Y, Luo W, Liu R, Chen Y, Gu W, Zhao P, Zhao J, Wang Y, Wang H. Elevated EBF2 in mouse but not pig drives the progressive brown fat lineage specification via chromatin activation. J Adv Res 2024:S2090-1232(24)00624-6. [PMID: 39736442 DOI: 10.1016/j.jare.2024.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/01/2025] Open
Abstract
Brown adipose tissue (BAT) is responsible for non-shivering thermogenesis, but it is absent in some mammals, including pigs. During development, BAT progenitors are derived from paired box 7 (Pax7)-expressing somitic mesodermal stem cells, which also give rise to skeletal muscle. However, the intrinsic mechanisms underlying the fate decisions between brown fat and muscle progenitors remain elusive across species. In this study, we analyzed the dynamics of chromatin landscape during the segregation and specification of brown fat and muscle lineages from Pax7+ multipotent mesodermal stem cells, aiming to uncover epigenetic factors that drive de novo BAT formation. Notably, myogenic progenitors were specified at embryonic day (E) 12.5, exhibiting high levels of H3K4me3 and low H3K27me3 at muscle-related genes. In contrast, the specification of the BAT lineage occurred much later, with coordinated step-wise depositions of histone modifications at BAT-associated genes from E10.5 to E14.5. We identified the transcription factor early B-cell factor 2 (EBF2) as a key driver of the progressive specification of brown fat lineage and the simultaneous deviation away from the muscle lineage. Mechanistically, EBF2 interacts with transcriptional co-activators CREB binding protein/ E1A-binding protein p300 (CBP/P300) to induce H3K27ac deposition and chromatin activation at BAT-associated genes to promote brown adipogenesis. Both mouse and pig EBF2 could potently stimulate adipogenesis in unspecified multipotent mesodermal stem cells. However, in pigs, EBF2 expression was depleted during the critical lineage specification time window, thus preventing the embryonic formation and development of porcine BAT. Hence, the elevation of EBF2 in mice, but not in pigs, promote chromatin activation to drive the progressive specification of brown fat lineage.
Collapse
Affiliation(s)
- Yinlong Liao
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; Yazhouwan National Laboratory, Sanya, China
| | - Zhelun Peng
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shanshan Fu
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Yao Hua
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Wenzhe Luo
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Ruige Liu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yingjin Chen
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Gu
- Shandong Provincial Key Laboratory of Animal Microecologics and Efficient Breeding of Livestock and Poultry, Shandong Baolai-Leelai Bio-Tech Co., Ltd, Taian, China
| | - Pengxiang Zhao
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Jianguo Zhao
- Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Yanfang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Heng Wang
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
48
|
Liu S, Chen H, Yang X, Wen Y, Chen L. Identification and validation of up-regulated TNFAIP6 in osteoarthritis with type 2 diabetes mellitus. Sci Rep 2024; 14:31450. [PMID: 39733138 PMCID: PMC11682049 DOI: 10.1038/s41598-024-82985-5] [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: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Lines of evidence have indicated that type 2 diabetes mellitus (T2DM) is an independent risk factor for osteoarthritis (OA) progression. However, the study focused on the relationship between T2DM and OA at the transcriptional level remains empty. We downloaded OA- and T2DM-related bulk RNA-sequencing and single-cell RNA sequencing data from the Gene Expression Omnibus (GEO) dataset. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed to screen out hub genes between OA and T2DM, and functional enrichment was done. Single-cell sequencing analysis was further used to screen key genes on OA and T2DM datasets. Rat chondrocytes and human articular cartilage were used to validate biomarkers among OA and T2DM. Sixty-eight hub genes were obtained, which were mainly enriched in the inflammatory response. We found that the hub gene TNFAIP6 is not only closely related to OA and T2DM but also a marker of prehypertrophic chondrocytes, which are closely related to the progression of OA. TNFAIP6 was found to be significantly elevated in CD14 + monocytes in T2DM patients, and this group of cells can promote inflammation. Validation on rat chondrocytes and human cartilage showed that TNFAIP6 was highly expressed in OA and further increased in the presence of T2DM or high glucose. Our study identified several characteristic modules and hub genes in the pathogenesis of T2DM-induced OA, which may facilitate further investigation of its molecular mechanisms. Up-regulated TNFAIP6 may contribute to OA in patients with T2DM by the recruitment of pro-inflammatory CD14 + monocytes in the OA synovium, which provides a potential target for the diagnosis and treatment of T2DM-associated OA.
Collapse
Affiliation(s)
- Siyi Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Joint Disease Research Center of Wuhan University, Wuhan, 430071, China
| | - Haitao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Joint Disease Research Center of Wuhan University, Wuhan, 430071, China
| | - Xu Yang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Joint Disease Research Center of Wuhan University, Wuhan, 430071, China
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Joint Disease Research Center of Wuhan University, Wuhan, 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Joint Disease Research Center of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
49
|
Zhang Z, Ma X, La Y, Guo X, Chu M, Bao P, Yan P, Wu X, Liang C. Advancements in the Application of scRNA-Seq in Breast Research: A Review. Int J Mol Sci 2024; 25:13706. [PMID: 39769466 PMCID: PMC11677372 DOI: 10.3390/ijms252413706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Single-cell sequencing technology provides apparent advantages in cell population heterogeneity, allowing individuals to better comprehend tissues and organs. Sequencing technology is currently moving beyond the standard transcriptome to the single-cell level, which is likely to bring new insights into the function of breast cells. In this study, we examine the primary cell types involved in breast development, as well as achievements in the study of scRNA-seq in the microenvironment, stressing the finding of novel cell subsets using single-cell approaches and analyzing the problems and solutions to scRNA-seq. Furthermore, we are excited about the field's promising future.
Collapse
Affiliation(s)
- Zhenyu Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoming Ma
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Yongfu La
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xian Guo
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Min Chu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Pengjia Bao
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Ping Yan
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoyun Wu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Chunnian Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| |
Collapse
|
50
|
Vu L, Xu F, Li T, Hua Q, Kuang X, Jiang Y, Liang Y, Niu X, Chen Y, Huang C, Mo W, Wang K, Tang K, Mo J, Lu KE, Mo Y, Mo S, Yang D, Zhao J. Analysis of immune cell activation in patients with diabetes foot ulcer from the perspective of single cell. Eur J Med Res 2024; 29:606. [PMID: 39702546 PMCID: PMC11657181 DOI: 10.1186/s40001-024-02179-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) can cause severe complications, including diabetic foot ulcers (DFU). There is a significant gap in understanding the single-cell ecological atlas of DM and DFU tissues. METHODS Single-cell RNA sequencing data were used to create a detailed single-cell ecological landscape of DM and DFU. Enrichment analysis identified pathways involved in cellular subpopulations, and pseudo-time analysis inferred cell development processes. A gene regulatory network explored the role of transcription factors in DFU progression, and a potential herbal drug-target gene interaction network was constructed. RESULTS In the DFU group, immune cells were activated, with notable changes in several subpopulations. ATP5E was significantly overexpressed in Naive T cells, fibroblasts, endothelial cells, and CD8+ T cells in DM patients. Specific immune cell subsets, such as Naive T_RGCC, CTL_TYROBP_CL4, Mac_SLC40A1, and M1_CCL3L1, likely contribute to DFU formation through overactivation and proliferation, leading to tissue damage and ulcer exacerbation. Key genes TPP1, TLR4, and RIPK2 were identified, and 88 active ingredients in the herbal drug-target network showed strong correlations with these targets. Herbs like Angelica dahurica, Angelica sinensis, Boswellia carterii, liquorice, myrrh, and Semen armeniacae amarae were included. CONCLUSIONS This study offers insights into DM and DFU cytology. T cells in DFU are activated, attacking normal tissues and worsening tissue damage. The ATP5E gene may be related to the ecological remodeling of DM, and TPP1, TLR4, and RIPK2 are potential targets for DFU treatment.
Collapse
Affiliation(s)
- Lehoanganh Vu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Fei Xu
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Ting Li
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Qikai Hua
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaocong Kuang
- Department of Physiology and Pathophysiology, Yulin Campus of Guangxi Medical University, Yulin, 537000, Guangxi, China
| | - Yongqiang Jiang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
| | - Yanfei Liang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Xing Niu
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Yixuan Chen
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Chengyu Huang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Weiliang Mo
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Kejian Wang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Kaihua Tang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Jianwen Mo
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Ke-Er Lu
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China
| | - Yan Mo
- Department of Pathology, Yulin Campus of Guangxi Medical University, Yulin, 537000, Guangxi, China
| | - Steven Mo
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China.
- Department of Basic Science, YuanDong International Academy Of Life Sciences, Hong Kong, 999077, China.
| | - Dengfeng Yang
- Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning , 530007, Guangxi, China.
| | - Jinmin Zhao
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|