1
|
Giannasi C, Cadelano F, Della Morte E, Baserga C, Mazzucato C, Niada S, Baj A. Unlocking the Therapeutic Potential of Adipose-Derived Stem Cell Secretome in Oral and Maxillofacial Medicine: A Composition-Based Perspective. BIOLOGY 2024; 13:1016. [PMCID: PMC11673083 DOI: 10.3390/biology13121016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025]
Abstract
The secretome of adipose-derived stem cells (ADSCs) holds significant promise for oral and maxillofacial medicine due to its rich composition of growth factors, cytokines, and other soluble or vesicle-embedded bioactive mediators that promote tissue regeneration and immunomodulation. Potential applications include enhancing wound healing, reducing inflammation, and stimulating the regeneration of hard and soft tissues. This could lead to improved outcomes in procedures such as bone grafting, soft tissue reconstruction, and the treatment of oral and facial defects. By harnessing the regenerative properties of ADSC secretome, clinicians may be able to achieve more effective tissue repair, ultimately benefiting patient recovery and quality of life. The adipose-derived stem cell (ADSC) secretome is widely studied for its immunomodulatory and regenerative properties, yet its potential in maxillofacial medicine remains largely underexplored. This review takes a composition-driven approach, beginning with a list of chemokines, cytokines, receptors, and inflammatory and growth factors quantified in the ADSC secretome to infer its potential applications in this medical field. First, a review of the literature confirmed the presence of 107 bioactive factors in the secretome of ADSCs or other types of mesenchymal stem cells. This list was then analyzed using the Search Tool for Retrieval of Interacting Genes/Proteins (STRING) software, revealing 844 enriched biological processes. From these, key processes were categorized into three major clinical application areas: immunoregulation (73 factors), bone regeneration (13 factors), and wound healing and soft tissue regeneration (27 factors), with several factors relevant to more than one area. The most relevant molecules were discussed in the context of existing literature to explore their therapeutic potential based on available evidence. Among these, TGFB1, IL10, and CSF2 have been shown to modulate immune and inflammatory responses, while OPG, IL6, HGF, and TIMP1 contribute to bone regeneration and tissue repair. Although the ADSC secretome holds great promise in oral and maxillofacial medicine, further research is needed to optimize its application and validate its clinical efficacy.
Collapse
Affiliation(s)
- Chiara Giannasi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Francesca Cadelano
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Elena Della Morte
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Baserga
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Mazzucato
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Stefania Niada
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Alessandro Baj
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| |
Collapse
|
2
|
Jiang Y, Cai R, Huang Y, Zhu L, Xiao L, Wang C, Wang L. Macrophages in organ fibrosis: from pathogenesis to therapeutic targets. Cell Death Discov 2024; 10:487. [PMID: 39632841 PMCID: PMC11618518 DOI: 10.1038/s41420-024-02247-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Fibrosis, an excessive self-repair response, is an age-related pathological process that universally affects various major organs such as the heart, liver, kidney, and lungs. Continuous accumulation of pathological tissue fibrosis destroys structural integrity and causes loss of function, with consequent organ failure and increased mortality. Although some differences exist in the triggering mechanisms and pathophysiologic manifestations of organ-specific fibrosis, they usually share similar cascading responses and features, including chronic inflammatory stimulation, parenchymal cell injury, and macrophage recruitment. Macrophages, due to their high plasticity, can polarize into different phenotypes in response to varied microenvironments and play a crucial role in the development of organ fibrosis. This review examined the relationship between macrophages and the pathogenesis of organ fibrosis. Moreover, it analyzed how fibrosis can be modulated by targeting macrophages, which may become a novel and promising therapeutic strategy for fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China
| | - Rong Cai
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China
| | - Yu Huang
- Department of Obstetrics and Gynecology, Zhangjiagang Hospital Affiliated to Soochow University, Zhangjiagang, 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China
| | - Caihong Wang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China.
| | - Lihong Wang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, Jiangsu, China.
| |
Collapse
|
3
|
Qiu P, Jiang Q, Song H. Unveiling the hidden world of transfer RNA-derived small RNAs in inflammation. J Inflamm (Lond) 2024; 21:46. [PMID: 39533297 PMCID: PMC11556027 DOI: 10.1186/s12950-024-00418-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Transfer RNA-derived small RNAs (tsRNAs) are a newly discovered class of small noncoding RNAs (sncRNAs) that include tRNA-derived stress-induced RNAs (tiRNAs) and tRNA-derived fragments (tRFs). Following the development of high-throughput sequencing technology, an increasing number of tsRNAs have been discovered with vital functions in different physiological and pathophysiological processes. Extensive research has revealed that tsRNAs are involved in various diseases, such as cancers, autoimmune illnesses and other diseases. This review focuses on the role and significance of tsRNAs in inflammation, such as the regulation of substances including inflammatory inducers, inflammatory cells and inflammatory factors, which contribute to the pathogenesis of inflammation-related diseases. Moreover, we discuss in-depth the molecular pathogenic mechanisms of tsRNAs in inflammation-related diseases through different signaling pathways and assess their clinical value, providing new perspectives for the exploration of tsRNA functions and inflammation-related diseases.
Collapse
Affiliation(s)
- Peiru Qiu
- Health Science Center, Ningbo University, Ningbo, China
| | - Qi Jiang
- Gastroenterology Department, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Haojun Song
- Gastroenterology Department, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- Department of Gastroenterology, Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Biobank, Ningbo, China.
| |
Collapse
|
4
|
Luo X, Wen W. MicroRNA in prostate cancer: from biogenesis to applicative potential. BMC Urol 2024; 24:244. [PMID: 39506720 PMCID: PMC11539483 DOI: 10.1186/s12894-024-01634-1] [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: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Prostate cancer is the most common solid malignant tumor in men, characterized by high morbidity and mortality. While current screening tools, such as prostate-specific antigen (PSA) testing and digital rectal examination, are available for early detection of prostate cancer, their sensitivity and specificity are limited. Tissue puncture biopsy, although capable of offering a definitive diagnosis, has poor positive predictive rates and burdens the patient more. Therefore, more reliable molecular diagnostic tools for prostate cancer urgently need to be developed. In recent years, microRNAs (miRNAs) have attracted much attention in prostate cancer research. miRNAs are extensively engaged in biological processes such as cell proliferation, differentiation, apoptosis, migration, and invasion by modulating gene expression post-transcriptionally. Dysregulation of miRNA expression in cancer is considered a critical factor in tumorigenesis and progression. This review first briefly introduces the biogenesis of miRNAs and their functions in cancer, then focuses on tumor-promoting miRNAs and tumor-suppressor miRNAs in prostate cancer. Finally, the potential application of miRNAs as multifunctional tools for cancer diagnosis, prognostic assessment, and therapy is discussed in detail. The concluding section summarizes the major points of the review and the challenges ahead.
Collapse
Affiliation(s)
- Xu Luo
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Wei Wen
- Department of Urology, West China Tianfu Hospital, Sichuan University, Chengdu, 610213, P.R. China.
| |
Collapse
|
5
|
Rai V. Transcriptomics Revealed Differentially Expressed Transcription Factors and MicroRNAs in Human Diabetic Foot Ulcers. Proteomes 2024; 12:32. [PMID: 39585119 PMCID: PMC11587442 DOI: 10.3390/proteomes12040032] [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: 07/23/2024] [Revised: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
Non-healing diabetic foot ulcers (DFUs) not only significantly increase morbidity and mortality but also cost a lot and drain healthcare resources. Persistent inflammation, decreased angiogenesis, and altered extracellular matrix remodeling contribute to delayed healing or non-healing. Recent studies suggest an increasing trend of DFUs in diabetes patients, and non-healing DFYs increase the incidence of amputation. Despite the current treatment with offloading, dressing, antibiotics use, and oxygen therapy, the risk of amputation persists. Thus, there is a need to understand the molecular and cellular factors regulating healing in DFUs. The ongoing research based on proteomics and transcriptomics has predicted multiple potential targets, but there is no definitive therapy to enhance healing in chronic DFUs. Increased or decreased expression of various proteins encoded by genes, whose expression transcriptionally and post-transcriptionally is regulated by transcription factors (TFs) and microRNAs (miRs), regulates DFU healing. For this study, RNA sequencing was conducted on 20 DFU samples of ulcer tissue and non-ulcerated nearby healthy tissues. The IPA analysis revealed various activated and inhibited transcription factors and microRNAs. Further network analysis revealed interactions between the TFs and miRs and the molecular targets of these TFs and miRs. The analysis revealed 30 differentially expressed transcription factors (21 activated and 9 inhibited), two translational regulators (RPSA and EIF4G2), and seven miRs, including mir-486, mir-324, mir-23, mir-186, mir-210, mir-199, and mir-338 in upstream regulators (p < 0.05), while causal network analysis (p < 0.05) revealed 28 differentially expressed TFs (19 activated and 9 inhibited), two translational regulators (RPSA and EIF4G2), and five miRs including mir-155, mir-486, mir-324, mir-210, and mir-1225. The protein-protein interaction analysis revealed the interaction of various novel proteins with the proteins involved in regulating DFU pathogenesis and healing. The results of this study highlight many activated and inhibited novel TFs and miRs not reported in the literature so far, as well as the targeted molecules. Since proteins are the functional units during biological processes, alteration of gene expression may result in different proteoforms and protein species, making the wound microenvironment a complex protein interaction (proteome complexity). Thus, investigating the effects of these TFs and miRs on protein expression using proteomics and combining these results with transcriptomics will help advance research on DFU healing and delineate potential therapeutic strategies.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766-1854, USA
| |
Collapse
|
6
|
Liu X, Wang Y, Wang C, Wang X, Tang G, Xiong Z, Zhou W. Role of non-coding RNA in exosomes for the diagnosis and treatment of osteosarcoma. Front Oncol 2024; 14:1469833. [PMID: 39512768 PMCID: PMC11540661 DOI: 10.3389/fonc.2024.1469833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Osteosarcoma (OS) is a malignancy characterized by the proliferation of osteoblasts that predominantly affects pediatric and adolescent populations. At present, early detection of OS is significantly lacking, coupled with treatment challenges such as high recurrence rates, increased side effects, and the development of drug resistance. Therefore, developing new diagnostic and therapeutic modalities is clinically significant. Exosomes are naturally occurring nanoparticles found in the body that contain various materials, including DNA, RNA, and proteins. Owing to their numerous beneficial properties, including histocompatibility and in vivo stability, they can be useful as drug carriers. With the development of competitive endogenous non-coding RNA (ncRNA) networks, the role of ncRNA in OS cell control has been increasingly studied. This review provides a thorough summary of multiple potential biogenetic pathways of different ncRNAs in exosomes, including microRNAs, long ncRNAs, and circular RNAs. Moreover, the review highlights their effects on OS cells and their potential applications in the diagnosis, treatment, and control of OS drug resistance. The interplay between different types of ncRNAs, which collectively affect OS through the networks of competing endogenous ncRNAs, is the primary focus of this research.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaling Wang
- Department of Nephropathy, Huanggang Hospital of Traditional Chinese Medicine, Hubei University of Traditional Chinese Medicine, Huanggang, China
| | - Chenwen Wang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyuan Wang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gangqiang Tang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Xiong
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhou
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
de Carvalho MR, Yang H, Stechmiller J, Lyon DE. MicroRNA Expression in Chronic Venous Leg Ulcers and Implications for Wound Healing: A Scoping Review. Biol Res Nurs 2024:10998004241291062. [PMID: 39412897 DOI: 10.1177/10998004241291062] [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: 10/18/2024]
Abstract
Purpose: Chronic venous leg ulcers (CVLUs) comprise the majority of lower-extremity wounds, yet their pathophysiology is not fully understood. While research has shown that microRNAs are an important component of wound inflammation, few have explored the role of microRNAs (miRNAs) in the healing of CVLUs. This scoping review examines miRNAs in CVLUs and the association with wound healing. Methods: In December 2023, we searched MEDLINE/PubMed, Embase, Scopus, and CINAHL for studies published in 2013-2023 examining miRNAs in CVLU healing. Results: Six studies met inclusion criteria. MicroRNAs were extracted from various specimens including serum, skin biopsy samples, and adipose tissue-derived mesenchymal cells from individuals with CVLUs. Overexpression of miR-221, miR-222, miR-92a, and miR-301a-3p hindered angiogenesis, while overexpression of miR-296, miR-126, miR-378, and miR-210 facilitated angiogenesis. Overexpression of miR-34a/c, miR-301a-3p, miR-450-5p, miR-424-5p, miR-516-5p, and miR-7704 increased local inflammatory responses and inhibited keratinocytes proliferation, impairing healing, while overexpression of miR-19a/b and miR-20 downregulated keratinocytes' inflammatory response, promoting healing. Downregulation of miR-205, miR-96-5p, and miR-218-5p enhanced cellular proliferation and promoted wound healing. Downregulation of miR-17-92 was linked with impaired healing. Discussion: MicroRNAs play a role in regulating angiogenesis, inflammatory responses, and cell migration in chronic-wound healing. However, studies of miRNAs in CVLUs are limited and lack a standardized approach to measurement and quantification. Further research is warranted to elucidate the mechanisms underlying microRNA involvement in CVLU healing to better understand the pathophysiology and for the future development of targeted therapies.
Collapse
Affiliation(s)
- Magali Rezende de Carvalho
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Hyehwan Yang
- Department of Family, Community and Health Systems Science,University of Florida College of Nursing, Gainesville, FL, USA
| | - Joyce Stechmiller
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Debra E Lyon
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| |
Collapse
|
8
|
Lee J, Lee MS, Kim Y. Effects of Green Tea and Java Pepper Mixture on Gut Microbiome and Colonic MicroRNA-221/222 in Mice with Dextran Sulfate Sodium-Induced Colitis. Prev Nutr Food Sci 2024; 29:279-287. [PMID: 39371512 PMCID: PMC11450278 DOI: 10.3746/pnf.2024.29.3.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
In this study, we aimed to investigate the regulatory effects of a green tea and java pepper mixture (GTP) on the gut microbiome and microRNA (miR)-221/222 expression in mice with dextran sulfate sodium (DSS)-induced colitis. Male C57BL/6J mice were divided into four groups: DSS-, DSS+, GTP50, and GTP100. In the GTP50 and GTP100 groups, GTP was orally administered to mice at doses of 50 and 100 mg/kg body weight, respectively, every day for 2 weeks, and colitis was induced in the DSS+, GTP50, and GTP100 groups by adding 3% DSS to their drinking water for 1 week. GTP was found to mitigate the severity of inflammation and the damage to goblet cells caused by DSS-induced colitis. The results showed that compared with the DSS- group, the relative abundance of Bacteroidetes was increased and that of Proteobacteria and Candidatus Melainabacteria was decreased in the GTP100 group. The ratio of Firmicutes to Bacteroidetes was also reduced in the GTP100 group. However, GTP administration did not modulate the microbial diversity. GTP administration upregulated the mRNA and protein levels of occludin and zonula occludens 1. In addition, GTP effectively downregulated the expression of miR-221 and miR-222. Overall, GTP altered the gut microbiota composition and downregulated colonic miR-221/222 expression in mice with DSS-induced colitis.
Collapse
Affiliation(s)
- Jumi Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Mak-Soon Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yangha Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
9
|
Alissa AI, McDonnell JM, Ross TD, Wu N, Sowa A, Wall J, Darwish S, Butler JS. Outcomes following spinal instrumented fusions in patients with parkinson's disease: a systematic review and meta-analysis. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:3420-3442. [PMID: 38937352 DOI: 10.1007/s00586-024-08307-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/09/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Parkinson's Disease (PD) patients represent challenging spinal surgery candidates due to associated frailty and deformity. This study consolidates the literature concerning spinal surgery outcomes in PD versus non-PD patients, to evaluate if PD predisposes patients to worse post-operative outcomes, so that treatment protocols can be optimised. METHODS A systematic review and meta-analysis was conducted of PubMed/Medline, Embase, and Google Scholar databases per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Studies of interest included comparative (PD versus non-PD) cohorts undergoing spinal instrumented fusions. Post-operative clinical outcomes were collated and compared for significance between cohorts. Further analysis was made on outcomes based on the different surgical procedures performed (Anterior Cervical Discectomy and Fusion (ACDF), Thoracolumbar or Lumbar fusions, Thoracolumbar or Lumbar fusions without Osteoporotic Vertebral Compression fracture (OVCF) patients). All statistical analysis was performed using The R Project for Statistical Computing (version 4.1.2), with a p-value of < 0.05 deemed statistically significant. RESULTS In total, 2,323,650 patients were included across 16 studies. Of those, 2,308,949 (99.37%) were patients without PD (non-PD), while 14,701 (0.63%) patients had PD at time of surgery. The collective mean age was 68.23 years (PD: 70.14 years vs non-PD: 64.86 years). Comparatively, there were 844,641 males (PD: 4,574; non-PD: 840,067) and 959,908 females (PD: 3,213; non-PD: 956,695). Overall, there were more post-operative complications in the PD cohort. Specifically, PD patients experienced significantly more surgical site infections (p = 0.01), increased rates of revision surgeries (p = 0.04) and increased venous thromboembolic events (p = 0.02) versus the non-PD cohort. In thoracolumbar/lumbar spinal fusions without OVCF patients, the PD cohort had increased rates of revision surgeries (p < 0.01) in comparison to the non-PD cohort. However, when including OVCF patients in thoracolumbar/lumbar spinal fusions, the PD cohort had significantly higher amounts of postoperative complications (p = 0.01), pneumonia (p = 0.02), and revision surgeries (p < 0.01) when compared to the non-PD cohort. CONCLUSION Although more robust prospective studies are needed, the results of this study highlight the need for advanced wound care management in the postoperative period, both in-hospital and in the community, in addition to comprehensive multidisciplinary care from allied health professionals, with potential for the use of Enhanced Recovery After Surgery (ERAS) protocols in PD patients undergoing spinal instrumented fusions.
Collapse
Affiliation(s)
- Ahmad Issa Alissa
- School of Medicine, University College Dublin, Dublin, Ireland
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
| | - Jake M McDonnell
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
- Centre of Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Tayler D Ross
- Department of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada
| | - Neil Wu
- School of Medicine, University College Dublin, Dublin, Ireland
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
| | - Aubrie Sowa
- School of Medicine, University College Dublin, Dublin, Ireland.
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland.
| | - Julia Wall
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
| | - Stacey Darwish
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
- Department of Orthopaedics, St. Vincent's University Hospital, Dublin, Ireland
| | - Joseph S Butler
- School of Medicine, University College Dublin, Dublin, Ireland
- National Spinal Injuries Unit, Mater Misericordiae University Hospital, Dublin, D07 R2WY, Ireland
| |
Collapse
|
10
|
Zhang G, Zhang Y, Wang B, Xu H, Xie D, Guo Z. miR-605-3p may affect caerulein-induced ductal cell injury and pyroptosis in acute pancreatitis by targeting the DUOX2/NLRP3/NF-κB pathway. PeerJ 2024; 12:e17874. [PMID: 39224819 PMCID: PMC11368084 DOI: 10.7717/peerj.17874] [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: 10/30/2023] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Acute pancreatitis (AP) is a sudden-onset disease of the digestive system caused by abnormal activation of pancreatic enzymes. Dual oxidase 2 (DUOX2) has been found to be elevated in the progression of a variety of inflammatory diseases. Therefore, we analyzed the specific roles of DUOX2 in AP development. Blood samples were collected from of AP patients and healthy people, and the caerulein- stimulated human pancreatic duct cells (H6C7) were utilized to establish an AP cell model. Cell growth and apoptosis were measured using an MTT assay and TUNEL staining. Additionally, RT-qPCR and western blot assays were conducted to assess the RNA and protein expressions of the cells. ELISA kits were used to determine TNF-α, IL-6, IL-8, and IL-1β levels. The interaction between DUOX2 and miR-605-3p was predicted using the Targetscan database and confirmed by dual-luciferase report assay. We found that DUOX2 increased while miR-605-3p decreased in the blood of AP patients and caerulein-stimulated H6C7 cells. DUOX2 was targeted by miR-605-3p. Furthermore, DUOX2 knockdown or miR-605-3p overexpression promoted cell viability, decreased the TNF-α, IL-6, IL-8, and IL-1β levels, and inhibited apoptosis rate in caerulein-stimulated H6C7 cells. DUOX2 knockdown or miR-605-3p overexpression also increased the Bcl-2 protein levels and down-regulated Bax, cleaved-caspase-1, NLRP3 and p-p65. Interestingly, DUOX2 overexpression reversed the miR-605-3p mimic function in the caerulein-treated H6C7 cells. In conclusion, our research demonstrated that DUOX2 knockdown relieved the injury and inflammation in caerulein-stimulated H6C7 cells.
Collapse
Affiliation(s)
- Gai Zhang
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Wannan Medical College Yijishan Hospital, Wuhu, Anhui, China
| | - Yuanyuan Zhang
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Bing Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Wannan Medical College Yijishan Hospital, Wuhu, Anhui, China
| | - Hao Xu
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Wannan Medical College Yijishan Hospital, Wuhu, Anhui, China
| | - Donghui Xie
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Wannan Medical College Yijishan Hospital, Wuhu, Anhui, China
| | - Zhenli Guo
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
11
|
Peng H, Du F, Wang J, Wu Y, Wei Q, Chen A, Duan Y, Shi S, Zhang J, Yu S. Adipose-Derived Stem-Cell-Membrane-Coated PLGA-PEI Nanoparticles Promote Wound Healing via Efficient Delivery of miR-21. Pharmaceutics 2024; 16:1113. [PMID: 39339150 PMCID: PMC11434648 DOI: 10.3390/pharmaceutics16091113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
miRNAs have been shown to be involved in the regulation of a variety of physiological and pathological processes, but their use in the treatment of diseases is still limited due to their instability. Biomimetic nanomaterials combine nanomaterials with cellular components that are readily modifiable and biocompatible, making them an emerging miRNA delivery vehicle. In this study, adipose-derived MSC membranes were wrapped around PLGA-PEI loaded with miR-21 through co-extrusion and later transplanted into C57BL/6 mice wounds. The wound-healing rate, epithelialization, angiogenesis, and collagen deposition were assessed after treatment and corroborated in vitro. Our study demonstrated that m/NP/miR-21 can promote wound healing in terms of epithelialization, dermal reconstruction, and neovascularization, and it can regulate the corresponding functions of keratinocytes, fibroblasts, and vascular endothelial cells. m/NP/miR-21 can inhibit the expression of PTEN, a gene downstream of miR-21, and increase the phosphorylation activation of AKT, which can then regulate the functions of fibroblasts. In conclusion, this provides a new approach to therapy for skin wounds using microRNA transporters and biomimetic nanoparticles.
Collapse
Affiliation(s)
- Huiyu Peng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Fangzhou Du
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Qian Wei
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Aoying Chen
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yuhan Duan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Shuaiguang Shi
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Shuang Yu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
12
|
Fan W, Lu H, Shi H, Yuan W, Liu G. Trends and challenges on inflammatory microenvironment in diabetic wound from 2014 to 2023: A bibliometric analysis. Int Wound J 2024; 21:e14913. [PMID: 39175213 PMCID: PMC11341833 DOI: 10.1111/iwj.14913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 08/24/2024] Open
Abstract
The disturbance of the inflammatory microenvironment is a frequent pathological trait of diabetic wounds, contributing to the emergence of numerous chronic illnesses. This is crucial in both the development and recovery of wounds caused by diabetes. This study aims to perform a bibliometric analysis of research on the inflammatory microenvironment within the domain of diabetic wounds (DW) over the past 10 years. The objective is to map out the current global research landscape, pinpoint the most significant areas of study and offer guidance for future research avenues. Our research involved querying the Web of Science Core Collection (WoSCC) database for all pertinent studies on the inflammatory microenvironment in diabetic wounds (DW). We utilized bibliometric tools such as CiteSpace, VOSviewer and R (version 4.3.1) to identify and highlight the most impactful studies in the field. The study encompassed a review of 1454 articles published from 2014 to 2023, highlighting China and the United States as pivotal nations in the research of the inflammatory microenvironment in diabetic wounds (DW). Within this sphere, the University of Michigan and Harvard University in the United States, along with Shanghai Jiaotong University in China, emerged as the most prolific institutions. WANG Y from China was identified as the most productive author, while KUNKEL SL from the United States received the most citations. The research primarily focuses on topics such as wound healing, repair processes, angiogenesis, oxidative stress and macrophage activity. Additionally, "macrophage" and "delivery" were pinpointed as the leading subjects with promising research potential in this area. Research on the inflammatory microenvironment of diabetic wounds is rapidly advancing through active international collaboration. The study of new mechanisms related to the inflammatory microenvironment and the development of novel materials for repair based on this microenvironment represent emerging fields of future research, particularly in terms of translational applications. This may offer guidance and novel perspectives for further research in the area of the diabetic wound inflammatory microenvironment.
Collapse
Affiliation(s)
- Weijing Fan
- Department of Vascular SurgeryShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huimin Lu
- Department of Vascular SurgeryShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hongshuo Shi
- Department of Vascular SurgeryShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weian Yuan
- Clinical Research CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Guobin Liu
- Department of Vascular SurgeryShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
13
|
Artimovič P, Špaková I, Macejková E, Pribulová T, Rabajdová M, Mareková M, Zavacká M. The ability of microRNAs to regulate the immune response in ischemia/reperfusion inflammatory pathways. Genes Immun 2024; 25:277-296. [PMID: 38909168 PMCID: PMC11327111 DOI: 10.1038/s41435-024-00283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
MicroRNAs play a crucial role in regulating the immune responses induced by ischemia/reperfusion injury. Through their ability to modulate gene expression, microRNAs adjust immune responses by targeting specific genes and signaling pathways. This review focuses on the impact of microRNAs on the inflammatory pathways triggered during ischemia/reperfusion injury and highlights their ability to modulate inflammation, playing a critical role in the pathophysiology of ischemia/reperfusion injury. Dysregulated expression of microRNAs contributes to the pathogenesis of ischemia/reperfusion injury, therefore targeting specific microRNAs offers an opportunity to restore immune homeostasis and improve patient outcomes. Understanding the complex network of immunoregulatory microRNAs could provide novel therapeutic interventions aimed at attenuating excessive inflammation and preserving tissue integrity.
Collapse
Affiliation(s)
- Peter Artimovič
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Ivana Špaková
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Ema Macejková
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Timea Pribulová
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Martina Zavacká
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia.
| |
Collapse
|
14
|
Wu L, Fu W, Cao Y, Zhao S, Zhang Y, Li X, Dong N, Qi W, Malik R, Wang J, Zhao RC. Inhibiting miR-618 Promotes Keratinocytes Proliferation and Migration to Enhance Wound Healing in Mice. Int J Mol Sci 2024; 25:7617. [PMID: 39062857 PMCID: PMC11277496 DOI: 10.3390/ijms25147617] [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: 05/21/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The delay in wound healing caused by chronic wounds or pathological scars is a pressing issue in clinical practice, imposing significant economic and psychological burdens on patients. In particular, with the aging of the population and the increasing incidence of diseases such as diabetes, impaired wound healing is one of the growing health problems. MicroRNA (miRNA) plays a crucial role in wound healing and regulates various biological processes. Our results show that miR-618 was significantly upregulated during the inflammatory phase of wound healing.Subsequently, miR-618 promotes the secretion of pro-inflammatory cytokines and regulates the proliferation and migration of keratinocytes. Mechanistically, miR-618 binds to the target gene-Atp11b and inhibits the PI3K-Akt signaling pathway, inhibiting the epithelial-mesenchymal transition (EMT) of keratinocytes. In addition, the PI3K-Akt signaling pathway induces the enrichment of nuclear miR-618, and miR-618 binds to the promoter of Lin7a to regulate gene transcription. Intradermal injection of miR-618 antagomir around full-thickness wounds in peridermal mice effectively accelerates wound closure compared to control. In conclusion, miR-618 antagomir can be a potential therapeutic agent for wound healing.
Collapse
Affiliation(s)
- Lingling Wu
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Wenjun Fu
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Yiyang Cao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Shuo Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Yuchen Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiaonan Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Naijun Dong
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Wenxin Qi
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Rabia Malik
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (L.W.); (W.F.); (Y.C.); (S.Z.); (Y.Z.); (X.L.); (N.D.); (W.Q.); (R.M.)
- School of Medicine, Shanghai University, Shanghai 200444, China
- Institute of Basic Medical, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- Centre of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing 100005, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| |
Collapse
|
15
|
Nadeem Butt E, Ali S, Summer M, Siddiqua Khan A, Noor S. Exploring the mechanistic role of silk sericin biological and chemical conjugates for effective acute and chronic wound repair and related complications. Drug Dev Ind Pharm 2024; 50:577-592. [PMID: 39087808 DOI: 10.1080/03639045.2024.2387814] [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: 03/07/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The current review is designed to elaborate and reveal the underlying mechanism of sericin and its conjugates of drug delivery during wounds and wound-related issues. SIGNIFICANCE Wound healing is a combination of different humoral, molecular, and cellular mechanisms. Various natural products exhibit potential in wound healing but among them, sericin, catches much attention of researchers due to its bio-functional properties such as being biodegradable, biocompatible, anti-oxidant, anti-bacterial, photo-protector, anti-inflammatory and moisturizing agent. METHODS AND RESULTS Sericin triggers the activity of anti-inflammatory cytokines which decrease cell adhesion and promote epithelial cell formation. Moreover, sericin enhances the anti-oxidant enzymes in the wounded area which scavenge the toxic consequences of reactive species (ROS). CONCLUSIONS This article highlights the mechanisms of how topical administration of sericin formulations along with 4-hexylresorcinol,\Chitosan\Ag@MOF-GO, polyvinyl alcohol (PVA), platelet lysate and UV photo cross-linked hydrogel sericin methacrylate which recruits a large number of cytokines on wounded area that stimulate fibroblasts and keratinocyte production as well as collagen deposition that led to early wound contraction. It also reviews the different sericin-based nanoparticles that play a significant role in rapid wound healing.
Collapse
Affiliation(s)
- Esham Nadeem Butt
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Ayesha Siddiqua Khan
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| |
Collapse
|
16
|
Hu Y, Yu L, Dai Q, Hu X, Shen Y. Multifunctional antibacterial hydrogels for chronic wound management. Biomater Sci 2024; 12:2460-2479. [PMID: 38578143 DOI: 10.1039/d4bm00155a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Chronic wounds have gradually evolved into a global health challenge, comprising long-term non-healing wounds, local tissue necrosis, and even amputation in severe cases. Accordingly, chronic wounds place a considerable psychological and economic burden on patients and society. Chronic wounds have multifaceted pathogenesis involving excessive inflammation, insufficient angiogenesis, and elevated reactive oxygen species levels, with bacterial infection playing a crucial role. Hydrogels, renowned for their excellent biocompatibility, moisture retention, swelling properties, and oxygen permeability, have emerged as promising wound repair dressings. However, hydrogels with singular functions fall short of addressing the complex requirements associated with chronic wound healing. Hence, current research emphasises the development of multifunctional antibacterial hydrogels. This article reviews chronic wound characteristics and the properties and classification of antibacterial hydrogels, as well as their potential application in chronic wound management.
Collapse
Affiliation(s)
- Yungang Hu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Lu Yu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Qiang Dai
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Xiaohua Hu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Yuming Shen
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
17
|
Zheng J, Yang B, Liu S, Xu Z, Ding Z, Mo M. Applications of Exosomal miRNAs from Mesenchymal Stem Cells as Skin Boosters. Biomolecules 2024; 14:459. [PMID: 38672475 PMCID: PMC11048182 DOI: 10.3390/biom14040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The skin is the outer layer of the human body, and it is crucial in defending against injuries and damage. The regenerative capacity of aging and damaged skin caused by exposure to external stimuli is significantly impaired. Currently, the rise in average life expectancy and the modern population's aesthetic standards have sparked a desire for stem-cell-based therapies that can address skin health conditions. In recent years, mesenchymal stem cells (MSCs) as therapeutic agents have provided a promising and effective alternative for managing skin regeneration and rejuvenation, attributing to their healing capacities that can be applied to damaged and aged skin. However, it has been established that the therapeutic effects of MSC may be primarily mediated by paracrine mechanisms, particularly the release of exosomes (Exos). Exosomes are nanoscale extracellular vesicles (EVs) that have lipid bilayer and membrane structures and can be naturally released by different types of cells. They influence the physiological and pathological processes of recipient cells by transferring a variety of bioactive molecules, including lipids, proteins, and nucleic acids such as messenger RNAs (mRNAs) and microRNAs (miRNAs) between cells, thus playing an important role in intercellular communication and activating signaling pathways in target cells. Among them, miRNAs, a type of endogenous regulatory non-coding RNA, are often incorporated into exosomes as important signaling molecules regulating protein biosynthesis. Emerging evidence suggests that exosomal miRNAs from MSC play a key role in skin regeneration and rejuvenation by targeting multiple genes and regulating various biological processes, such as participating in inflammatory responses, cell migration, proliferation, and apoptosis. In this review, we summarize the recent studies and observations on how MSC-derived exosomal miRNAs contribute to the regeneration and rejuvenation of skin tissue, with particular attention to the applications of bioengineering methods for manipulating the miRNA content of exosome cargo to improve their therapeutic potential. This review can provide new clues for the diagnosis and treatment of skin damage and aging, as well as assist investigators in exploring innovative therapeutic strategies for treating a multitude of skin problems with the aim of delaying skin aging, promoting skin regeneration, and maintaining healthy skin.
Collapse
Affiliation(s)
- Jinmei Zheng
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Beibei Yang
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Siqi Liu
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Zhenfeng Xu
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Zhimeng Ding
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Miaohua Mo
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
18
|
Hu K, Liu L, Tang S, Zhang X, Chang H, Chen W, Fan T, Zhang L, Shen B, Zhang Q. MicroRNA-221-3p inhibits the inflammatory response of keratinocytes by regulating the DYRK1A/STAT3 signaling pathway to promote wound healing in diabetes. Commun Biol 2024; 7:300. [PMID: 38461326 PMCID: PMC10924844 DOI: 10.1038/s42003-024-05986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
Diabetic foot ulcer (DFU), a serious complication of diabetes, remains a clinical challenge. MicroRNAs affect inflammation and may have therapeutic value in DFU. Here, we find that an miR-221-3p mimic reduces the inflammatory response and increases skin wound healing rates in a mouse model of diabetes, whereas miR-221-3p knockout produced the opposite result. In human keratinocytes cells, miR-221-3p suppresses the inflammatory response induced by high glucose. The gene encoding DYRK1A is a target of miR-221-3p. High glucose increases the expression of DYRK1A, but silencing DYRK1A expression decreases high glucose-induced inflammatory cytokine release via dephosphorylation of STAT3, a substrate of DYRK1A. Application of miR-221-3p mimic to human keratinocytes cells not only decreases DYRK1A expression but also inhibits high glucose-induced production of inflammatory cytokines to promote wound healing. This molecular mechanism whereby miR-221-3p regulates inflammation through the DYRK1A/STAT3 signaling pathway suggests targets and therapeutic approaches for treating DFU.
Collapse
Affiliation(s)
- Keyan Hu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Endocrinology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Lei Liu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Songtao Tang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongfeng Chang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenyang Chen
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Taotao Fan
- Center of Experimental Practice, Anhui Medical University, Hefei, China
| | - Lesha Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
19
|
Chen F, Wu P, Zhang H, Sun G. Signaling Pathways Triggering Therapeutic Hydrogels in Promoting Chronic Wound Healing. Macromol Biosci 2024; 24:e2300217. [PMID: 37831962 DOI: 10.1002/mabi.202300217] [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: 05/16/2023] [Revised: 10/08/2023] [Indexed: 10/15/2023]
Abstract
In recent years, there has been a significant increase in the prevalence of chronic wounds, such as pressure ulcers, diabetic foot ulcers, and venous ulcers of the lower extremities. The main contributors to chronic wound formation are bacterial infection, prolonged inflammation, and peripheral vascular disease. However, effectively treating these chronic wounds remains a global challenge. Hydrogels have extensively explored as wound healing dressing because of their excellent biocompatibility and structural similarity to extracellular matrix (ECM). Nonetheless, much is still unknown how the hydrogels promote wound repair and regeneration. Signaling pathways play critical roles in wound healing process by controlling and coordinating cells and biomolecules. Hydrogels, along with their therapeutic ingredients that impact signaling pathways, have the potential to significantly enhance the wound healing process and its ultimate outcomes. Understanding this interaction will undoubtedly provide new insights into developing advanced hydrogels for wound repair and regeneration. This paper reviews the latest studies on classical signaling pathways and potential targets influenced by hydrogel scaffolds in chronic wound healing. This work hopes that it will offer a different perspective in developing more efficient hydrogels for treating chronic wounds.
Collapse
Affiliation(s)
- Fang Chen
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China
- First Department of Bone Injury, Luzhou Municipal Hospital of Traditional Chinese Medicine, Luzhou, Sichuan, 646000, China
| | - Pingli Wu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Haisong Zhang
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Guoming Sun
- Sunogel Biotechnologies Inc., Lutherville Timonium, 9 W Ridgely Road Ste 270, Maryland, 21093, USA
| |
Collapse
|
20
|
Wicaksono S, Nugraha AP, Rahmahani J, Rantam FA, Kuncorojakti S, Susilowati H, Riawan W, Arundina I, Lestari P, Masya RN, Surboyo MDC, Ernawati DS. Adipose Mesenchymal Stem Cell Metabolites Oral Gel Enhance Pro-Angiogenic Factors Expression, Angiogenesis, and Clinical Outcome of Oral Ulcer Rat Model. Eur J Dent 2024; 18:117-123. [PMID: 36963426 PMCID: PMC10959621 DOI: 10.1055/s-0043-1761192] [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: 03/26/2023] Open
Abstract
OBJECTIVE Enhancing wound healing capacity is one of the main principles in oral ulcer management. Efficient oral ulcer management will accelerate clinical symptom amelioration and prevent complications. Adipose mesenchymal stem cell metabolites (AdMSCM), a novel biological product, contains a plethora of bioactive mediators that can induce a series of processes in wound healing. This study will analyze the clinical outcome, angiogenesis, and expression of FGF-2 and VEGFA in the oral ulcer rat model after AdMSCM oral gel application. MATERIALS AND METHODS Twenty healthy male Wistar rats (Rattus novergicus) were used to create oral ulcer animal models. AdMSCM oral gel treatment was performed three times daily for 3 and 7 days. Clinical outcome was assessed by measuring the major diameter of the ulcer; the angiogenesis was evaluated through histological assessment; the expression of VEGFA and FGF-2 was assessed using the immunohistochemistry method. STATISTICAL ANALYSIS This study uses parametric comparative analysis using one-way analysis of variance (ANOVA) and post-hoc Tukey's HSD test RESULTS: The application of AdMSCM oral gel in an oral ulcer rat model significantly enhanced the clinical outcome (p < 0.05). In addition, similar results were shown in the histologic assessment of angiogenesis and supported by the significant increase of VEGFA and FGF-2 expression. CONCLUSIONS AdMSCM oral gel accelerates oral ulcer healing processes, proven by the enhancement of angiogenesis, pro-angiogenic factors expression, and clinical outcomes.
Collapse
Affiliation(s)
- Satutya Wicaksono
- Master Program in Immunology, Postgraduate School, Universitas Airlangga, Surabaya, Indonesia
| | - Alexander Patera Nugraha
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Jola Rahmahani
- Division of Veterinary Microbiology, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Fedik Abdul Rantam
- Division of Veterinary Microbiology, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Suryo Kuncorojakti
- Division of Veterinary Anatomy, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Helen Susilowati
- Research Center for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Wibi Riawan
- Department of Biomolecular Biochemistry, Universitas Brawijaya, Malang, Indonesia
| | - Ira Arundina
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Pudji Lestari
- Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Resgita Nadila Masya
- Graduate Program in Dental Health Science, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Diah Savitri Ernawati
- Department of Oral Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
21
|
Mahmoud NN, Hamad K, Al Shibitini A, Juma S, Sharifi S, Gould L, Mahmoudi M. Investigating Inflammatory Markers in Wound Healing: Understanding Implications and Identifying Artifacts. ACS Pharmacol Transl Sci 2024; 7:18-27. [PMID: 38230290 PMCID: PMC10789122 DOI: 10.1021/acsptsci.3c00336] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024]
Abstract
Understanding the complex interplay of pro-inflammatory and anti-inflammatory cytokines is crucial in the field of wound healing, as it holds the key to developing effective therapeutics. In the initial stages of wound healing, pro-inflammatory cytokines like IL-1β, IL-6, TNF-α, and various chemokines play vital roles in recruiting cells for debris clearance and the recruitment of growth factors. Careful regulation and timely resolution of this early inflammation are essential for optimal wound repair. As the healing process progresses, anti-inflammatory proteins such as IL-10 and IL-4 become instrumental in facilitating the transition to later stages where pro-inflammatory cytokines promote angiogenesis and wound remodeling. This Perspective underscores the complexity of inflammatory cytokines in wound healing research and emphasizes the need for comprehensive and unbiased methodologies in their evaluation. For robust and reliable results in wound-healing research, a more holistic approach is necessary-one that considers the roles, interactions, and timing of biological molecules, alongside careful sampling and evaluation strategies.
Collapse
Affiliation(s)
- Nouf N. Mahmoud
- Faculty
of Pharmacy, Al-Zaytoonah University of
Jordan, Amman 11733, Jordan
- Department
of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Khawla Hamad
- School
of Medicine, Royal College of Surgeons in
Ireland-Bahrain, Busaiteen 228, Bahrain
| | - Aya Al Shibitini
- School
of Medicine, Royal College of Surgeons in
Ireland-Bahrain, Busaiteen 228, Bahrain
| | - Sarah Juma
- School
of Medicine, Royal College of Surgeons in
Ireland-Bahrain, Busaiteen 228, Bahrain
| | - Shahriar Sharifi
- Department
of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lisa Gould
- Warren
Alpert Medical School of Brown University, Providence, Rhode Island 02912, United
States
- South Shore
Health Center for Wound Healing, Weymouth, Massachusetts 02189, United States
| | - Morteza Mahmoudi
- Department
of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
22
|
Saifullah Q, Sharma A. Current Trends on Innovative Technologies in Topical Wound Care for Advanced Healing and Management. Curr Drug Res Rev 2024; 16:319-332. [PMID: 37807417 DOI: 10.2174/0125899775262048230925054922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/19/2023] [Accepted: 08/25/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVES To investigate critically traditional and modern techniques for cutaneous wound healing and to provide comprehensive information on these novel techniques to encounter the challenges with the existing wound healing methods. SIGNIFICANCE The financial burden and mortality associated with wounds is increasing, so managing wounds is essential. Traditional wound treatments include surgical and non-surgical methods, while modern techniques are advancing rapidly. This review examines the various traditional and modern techniques used for cutaneous wound healing. KEY FINDINGS Traditional wound treatments include surgical techniques such as debridement, skin flaps, and grafts. Non-surgical treatments include skin replacements, topical formulations, scaffold-based skin grafts, and hydrogel-based skin dressings. More modern techniques include using nanoparticles, growth factors, and bioactive substances in wound dressings. Bioengineered skin substitutes using biomaterials, cells, and growth factors are also being developed. Other techniques include stem cell therapy, growth factor/cytokine therapy, vacuum-assisted wound closure, and 3D-printed/bio-printed wound dressings. CONCLUSION Traditional wound treatments have been replaced by modern techniques such as stem cell therapy, growth factor/cytokine therapy, vacuum-assisted wound closure, and bioengineered skin substitutes. However, most of these strategies lack effectiveness and thorough evaluation. Therefore, further research is required to develop new techniques for cutaneous wound healing that are effective, cost-efficient, and appealing to patients.
Collapse
Affiliation(s)
- Qazi Saifullah
- Department of Pharmacy, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Punjab, 140413, India
| | - Abhishek Sharma
- Department of Pharmacy, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Punjab, 140413, India
| |
Collapse
|
23
|
Hu S, Liang Y, Chen J, Gao X, Zheng Y, Wang L, Jiang J, Zeng M, Luo M. Mechanisms of hydrogel-based microRNA delivery systems and its application strategies in targeting inflammatory diseases. J Tissue Eng 2024; 15:20417314241265897. [PMID: 39092451 PMCID: PMC11292707 DOI: 10.1177/20417314241265897] [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: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
Hydrogels, composed of three-dimensional polymer networks, are excellent delivery carriers and have been extensively employed in the biomedical field. Inflammation acts as a protective mechanism to prevent harmful substances from entering living organisms, but chronic, long-lasting inflammation can cause oxidative stress, which damages tissue and organs and adversely affects patients' quality of life. The aberrant expression of microRNAs (miRNAs) has been found to play a significant part in the etiology and progression of inflammatory diseases, as suggested by growing evidence. Numerous hydrogels that can act as gene carriers for the intracellular delivery of miRNA have been described during ongoing research into innovative hydrogel materials. MiRNA hydrogel delivery systems, which are loaded with exogenous miRNA inhibitors or mimics, enable targeted miRNA intervention in inflammatory diseases and effectively prevent environmental stressors from degrading or inactivating miRNA. In this review, we summarize the classification of miRNA hydrogel delivery systems, the basic strategies and mechanisms for loading miRNAs into hydrogels, highlight the biomedical applications of miRNA hydrogel delivery systems in inflammatory diseases, and share our viewpoints on potential opportunities and challenges in the promising region of miRNA delivery systems. These findings may provide a new theoretical basis for the prevention and treatment of inflammation-related diseases and lay the foundation for clinical translation.
Collapse
Affiliation(s)
- Shaorun Hu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinxiang Chen
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| |
Collapse
|
24
|
Wang Y, Li Y, Ni D, Wei Z, Fu Z, Li C, Sun H, Wu Y, Li Y, Zhang Y, Liu N, Liu Y, Wang Z, Li J, Sun D, He L, Yang Y, Wang Y, Yang X. miR-186-5p targets TGFβR2 to inhibit RAW264.7 cell migration and proliferation during mouse skin wound healing. ENVIRONMENTAL TOXICOLOGY 2023; 38:2826-2835. [PMID: 37565786 DOI: 10.1002/tox.23914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/11/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Active peptides play a vital role in the development of new drugs and the identification and discovery of drug targets. As the first reported native peptide homodimer with pro-regenerative potency, OA-GP11d could potentially be used as a novel molecular probe to help elucidate the molecular mechanism of skin wound repair and provide new drug targets. METHODS Bioinformatics analysis and luciferase assay were adopted to determine microRNAs (miRNAs) and its target. The prohealing potency of the miRNA was determined by MTS and a Transwell experiment against mouse macrophages. Enzyme-linked immunosorbent assay, realtime polymerase chain reaction, and western blotting were performed to explore the molecular mechanisms. RESULTS In this study, OA-GP11d was shown to induce Mus musculus microRNA-186-5p (mmu-miR-186-5p) down-regulation. Results showed that miR-186-5p had a negative effect on macrophage migration and proliferation as well as a targeted and negative effect on TGF-β type II receptor (TGFβR2) expression and an inhibitory effect on activation of the downstream SMAD family member 2 (Smad2) and protein-p38 kinase signaling pathways. Importantly, delivery of a miR-186-5p mimic delayed skin wound healing in mice. CONCLUSION miR-186-5p regulated macrophage migration and proliferation to delay wound healing through the TGFβR2/Smad2/p38 molecular axes, thus providing a promising new pro-repair drug target.
Collapse
Affiliation(s)
- Yinglei Wang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yuansheng Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Dan Ni
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Ziqi Wei
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Zhe Fu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Chao Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Huiling Sun
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yutong Wu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yilin Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yingxuan Zhang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Naixin Liu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yixiang Liu
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, China
| | - Zhuo Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, China
| | - Jiayi Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Dandan Sun
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Yang
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| |
Collapse
|
25
|
Kiani M, Moraffah F, Khonsari F, Kharazian B, Dinarvand R, Shokrgozar MA, Atyabi F. Co-delivery of simvastatin and microRNA-21 through liposome could accelerates the wound healing process. BIOMATERIALS ADVANCES 2023; 154:213658. [PMID: 37866233 DOI: 10.1016/j.bioadv.2023.213658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/10/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
The gene delivery approach, mainly microRNAs (miRNA) as key wound healing mediators, has recently received extensive attention. MicroRNA-21 (miR-21) has strongly impacted wound healing by affecting the inflammation and proliferation phases. Previous studies have also demonstrated the beneficial effect of simvastatin on wound healing. Therefore, we designed a dual-drug/gene delivery system using PEGylated liposomes that could simultaneously attain the co-encapsulation and co-delivery of miRNA and simvastatin (SIM) to explore the combined effect of this dual-drug delivery system on wound healing. The PEG-liposomes for simvastatin and miR-21 plasmid (miR-21-P/SIM/Liposomes) were prepared using the thin-film hydration method. The liposomes showed 85 % entrapment efficiency for SIM in the lipid bilayer and high physical entrapment of miR-21-P in the inner cavity. In vitro studies demonstrated no cytotoxicity for the carrier on normal human dermal fibroblast cells (NHDF) and 97 % cellular uptake over 2 h incubation. The scratch test revealed excellent cell proliferation and migration after treatment with miR-21-P/SIM/Liposomes. For the in vivo experiments, a full-thickness cutaneous wound model was used. The wound closure on day 8 was higher for Liposomal formulation containing miR-21-P promoting faster re-epithelialization. On day 12, all treated groups showed complete wound closure. However, following histological analysis, the miR-21-P/SIM/Liposomes revealed the best tissue regeneration, similar to normal functional skin, by reduced inflammation and increased re-epithelialization, collagen deposition and angiogenesis. In conclusion, the designed miR-21-P/SIM/Liposomes could significantly accelerate the process of wound healing, which provides a new strategy for the management of chronic wounds.
Collapse
Affiliation(s)
- Melika Kiani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moraffah
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khonsari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Kharazian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; School of Pharmacy, De Mont Fort University, Leicester, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Zhao M, Xie Y, Gao W, Li C, Ye Q, Li Y. Diabetes mellitus promotes susceptibility to periodontitis-novel insight into the molecular mechanisms. Front Endocrinol (Lausanne) 2023; 14:1192625. [PMID: 37664859 PMCID: PMC10469003 DOI: 10.3389/fendo.2023.1192625] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Diabetes mellitus is a main risk factor for periodontitis, but until now, the underlying molecular mechanisms remain unclear. Diabetes can increase the pathogenicity of the periodontal microbiota and the inflammatory/host immune response of the periodontium. Hyperglycemia induces reactive oxygen species (ROS) production and enhances oxidative stress (OS), exacerbating periodontal tissue destruction. Furthermore, the alveolar bone resorption damage and the epigenetic changes in periodontal tissue induced by diabetes may also contribute to periodontitis. We will review the latest clinical data on the evidence of diabetes promoting the susceptibility of periodontitis from epidemiological, molecular mechanistic, and potential therapeutic targets and discuss the possible molecular mechanistic targets, focusing in particular on novel data on inflammatory/host immune response and OS. Understanding the intertwined pathogenesis of diabetes mellitus and periodontitis can explain the cross-interference between endocrine metabolic and inflammatory diseases better, provide a theoretical basis for new systemic holistic treatment, and promote interprofessional collaboration between endocrine physicians and dentists.
Collapse
Affiliation(s)
- Mingcan Zhao
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Yuandong Xie
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Wenjia Gao
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Chunwang Li
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Qiang Ye
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Yi Li
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
27
|
Siu MC, Voisey J, Zang T, Cuttle L. MicroRNAs involved in human skin burns, wound healing and scarring. Wound Repair Regen 2023; 31:439-453. [PMID: 37268303 DOI: 10.1111/wrr.13100] [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: 10/17/2022] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 06/04/2023]
Abstract
MicroRNAs are small, non-coding RNAs that regulate gene expression, and consequently protein synthesis. Downregulation and upregulation of miRNAs and their corresponding genes can alter cell apoptosis, proliferation, migration and fibroproliferative responses following a thermal injury. This review summarises the evidence for altered human miRNA expression post-burn, and during wound healing and scarring. In addition, the most relevant miRNA targets and their roles in potential pathways are described. Previous studies using molecular techniques have identified 197 miRNAs associated with human wound healing, burn wound healing and scarring. Five miRNAs alter the expression of fibroproliferative markers, proliferation and migration of fibroblasts and keratinocytes post-burn: hsa-miR-21 and hsa-miR-31 are increased after wounding, and hsa-miR-23b, hsa-miR-200b and hsa-let-7c are decreased. Four of these five miRNAs are associated with the TGF-β pathway. In the future, large scale, in vivo, longitudinal human studies utilising a range of cell types, ethnicity and clinical healing outcomes are fundamental to identify burn wound healing and scarring specific markers. A comprehensive understanding of the underlying pathways will facilitate the development of clinical diagnostic or prognostic tools for better scar management and the identification of novel treatment targets for improved healing outcomes in burn patients.
Collapse
Affiliation(s)
- Man Ching Siu
- Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- Centre for Genomics and Personalised Health Research, QUT, Brisbane, Queensland, Australia
| | - Joanne Voisey
- Centre for Genomics and Personalised Health Research, QUT, Brisbane, Queensland, Australia
| | - Tuo Zang
- Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Leila Cuttle
- Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Bachtiar BM, Bachtiar EW, Kusumaningrum A, Sunarto H, Soeroso Y, Sulijaya B, Apriyanti E, Fragrantia Theodorea C, Putra Pratomo I, Yudhistira, Efendi D. Porphyromonas gingivalis association with inflammatory markers and exosomal miRNA-155 in saliva of periodontitis patients with and without diabetes diagnosed with COVID-19. Saudi Dent J 2023; 35:61-69. [PMID: 36540394 PMCID: PMC9756571 DOI: 10.1016/j.sdentj.2022.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/05/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Background /Purposes: Studies have indicated that salivary molecules from patients with periodontitis and diabetes are confounded with pathological conditions associated with SARS-CoV-2 infection. The study aimed to address whether the abundance of Porphyromonas gingivalis which causes periodontitis, differed compared with that of Aggregatibacter actinomycetemcomitans (used as control) and to analyze the correlation of periodontitis with the expression levels of severe acute respiratory syndrome coronavirus 2 receptor (ACE2) and periodontitis inflammatory markers (TLR-2/TLR-4, TNFα, and miR-155). Materials and Methods A saliva sample (5 mL) was obtained from 23 hospitalized patients with COVID-19, categorized into two groups: diabetic (G1, n = 10) and non-diabetic (G2, n = 13). Saliva from patients with periodontitis without diabetes and coronavirus disease 2019 (COVID-19; n = 6) were included as control. The quantitative real-time polymerase chain reaction measured the levels of P. gingivalis and A. actinomycetemcomitans, as well as periodontitis markers in saliva. The obtained data were analyzed using one-way ANOVA and the Spearman correlation test. Results The abundance of P. gingivalis was observed to be higher (p < 0.05) in saliva of patients with diabetes (G1) than in those without diabetes (G2). A contradictory trend was observed for A. actinomycetemcomitans. The transcription level of ACE2 was comparable in all groups tested, while the expression of periodontitis markers varied. The relationships and sensitivity/specificity among P. gingivalis infection ACE2 expression, and inflammatory markers were also evaluated. Conclusions This study showed that the association between P. gingivalis infection and ACE2 expression might reflect the characteristics of saliva in COVID-19 patients with and without diabetes. However, the relationships between TLR-4 and miR-155 are more specific in discriminating against COVID-19 patients with and without diabetes.
Collapse
Affiliation(s)
- Boy M. Bachtiar
- Department of Oral Biology, Faculty of Dentistry Universitas Indonesia, Indonesia,Oral Science Research Center, Faculty of Dentistry Universitas Indonesia, Indonesia
| | - Endang W. Bachtiar
- Department of Oral Biology, Faculty of Dentistry Universitas Indonesia, Indonesia,Oral Science Research Center, Faculty of Dentistry Universitas Indonesia, Indonesia,Corresponding author at: Department of oral Biology and Oral Science Research center, Faculty of Dentistry, Universitas Indonesia, Indonesia
| | - Ardiana Kusumaningrum
- Department of Microbiology, Faculty of Medicine, Universitas Indonesia, Indonesia,Clinical Microbiology Medicine Staff Group, Universitas Indonesia Hospital, Indonesia
| | - Hari Sunarto
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Indonesia,Dental Center Universitas Indonesia Hospital, Depok, Indonesia
| | - Yuniarti Soeroso
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Indonesia
| | - Benso Sulijaya
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Indonesia
| | - Efa Apriyanti
- Department of Pediatric Nursing, Faculty of Nursing Universitas Indonesia, and Paediatric Intensive Care Unit, Universitas Indonesia Hospital, Indonesia
| | - Citra Fragrantia Theodorea
- Department of Oral Biology, Faculty of Dentistry Universitas Indonesia, Indonesia,Oral Science Research Center, Faculty of Dentistry Universitas Indonesia, Indonesia
| | - Irandi Putra Pratomo
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Indonesia, Indonesia,Pulmonology and Respiratory Medicine Staff Group - COVID-19 Task Force, Universitas Indonesia Hospital, Universitas Indonesia, Depok, Indonesia,Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Indonesia
| | - Yudhistira
- Clinical Pathology Medicine Staff Group, Universitas Indonesia Hospital, Indonesia
| | - Defi Efendi
- Department of Pediatric Nursing, Faculty of Nursing Universitas Indonesia, and Neonatal Intensive Care Unit, Universitas Indonesia Hospital, Depok, Indonesia
| |
Collapse
|
29
|
Ma J, Yong L, Lei P, Li H, Fang Y, Wang L, Chen H, Zhou Q, Wu W, Jin L, Sun D, Zhang X. Advances in microRNA from adipose-derived mesenchymal stem cell-derived exosome: focusing on wound healing. J Mater Chem B 2022; 10:9565-9577. [PMID: 36398750 DOI: 10.1039/d2tb01987f] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Skin wounds are a common condition causing economic burden and they represent an urgent clinical need, especially chronic wounds. Numerous studies have been conducted on the applications of stem cell therapy in wound healing, with adipose-derived mesenchymal stem cells (ADMSCs) playing a major role since they can be isolated easily, yielding a high number of cells, the less invasive harvesting required, the longer life span and no ethical issues. However, the lack of standardized doses and protocols, the heterogeneity of clinical trials, as well as the incompatibility of the immune system limit its application. Recent studies have demonstrated that specific stem cell functions depend on paracrine factors, including extracellular vesicles, in which microRNAs in exosomes (Exo-miRNAs) are essential in controlling their functions. This paper describes the application and mechanism whereby ADMSC-Exo-miRNA regulates wound healing. ADMSC-Exo-miRNA is involved in various stages in wounds, including modulating the immune response and inflammation, accelerating skin cell proliferation and epithelialization, promoting vascular repair, and regulating collagen remodeling thereby reducing scar formation. In summary, this acellular therapy based on ADMSC-Exo-miRNA has considerable clinical potential, and provides reference values for developing new treatment strategies for wound healing.
Collapse
Affiliation(s)
- Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Ling Yong
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Hua Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Haojie Chen
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Qi Zhou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China. .,Wenzhou City and Kunlong Technology Co., Ltd Joint Doctoral Innovation Station, Wenzhou Association for Science and Technology, Wenzhou 325000, China
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, China.
| |
Collapse
|
30
|
Pucker AD, Ngo W, Postnikoff CK, Fortinberry H, Nichols JJ. Tear Film miRNAs and Their Association With Human Dry Eye Disease. Curr Eye Res 2022; 47:1479-1487. [PMID: 35930289 PMCID: PMC9762462 DOI: 10.1080/02713683.2022.2110597] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE miRNAs can regulate inflammatory pathways. The purpose of this work was to determine if inflammatory-related tear film miRNAs are associated with extracellular vesicles (EVs) in human non-Sjögren's Syndrome dry eye disease (DED) participants. METHODS Five DED and 5 non-DED human participants were recruited. Tears samples were collected by washing the ocular surface of both eyes with phosphate buffered saline, pooling samples from the right and left eyes, and purifying EVs from the samples with a polyethylene glycol (PEG) 8000 precipitation procedure. Samples were directly analyzed via ELISA or transmission electron microscopy (TEM), or RNA was isolated first from the EVs and evaluated with RNA-Seq. RESULTS EVs were identified in the tear film of both groups using TEM and ELISA. Following EV purification and RNA isolation, RNA-Seq determined that there were 126 EV miRNAs differentially expressed between the two groups when comparing their RNA cargoes. Ingenuity Pathways Analysis found 9 upregulated miRNAs that were associated with inflammation (miR-127-5p, miR-1273h-3p, miR-1288-5p, miR-130b-5p, miR-139-3p, miR-1910-5p, miR-203b-5p, miR-22-5p, and miR-4632-3p; all p < 0.049; fold regulation range = 1.43-1.67). CONCLUSION This study determined that EVs are present in the tear film and that tear EVs contain miRNAs that may be associated with DED inflammatory pathways.
Collapse
Affiliation(s)
- Andrew D Pucker
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham
| | - William Ngo
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham
| | - Cameron K Postnikoff
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham
| | - Henry Fortinberry
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham
| | - Jason J Nichols
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham
| |
Collapse
|
31
|
Xu Q, Dai X, Yang L, Liu X, Li Y, Gao F. ε-Polylysine-Based Macromolecules with Catalase-Like Activity to Accelerate Wound Healing by Clearing Bacteria and Attenuating Inflammatory Response. ACS Biomater Sci Eng 2022; 8:5018-5026. [PMID: 36256969 DOI: 10.1021/acsbiomaterials.2c00986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Wound healing has remained a critical challenge due to its susceptibility to bacterial infection and the unique biological inflammatory response. Safe and effective therapeutics are still lacking. Biodegradable macromolecules (ε-polylysine-g-ferrocene, EPL-g-Fc) were developed to accelerate wound healing by combating bacterial infection and attenuating inflammatory responses. The biodegradable macromolecules were prepared via a Schiff-based reaction between ferrocene carboxaldehyde (Fc) and ε-polylysine (EPL). Through the synergistic combination of positive-charged EPL and π-π stacked Fc, the macromolecules possess excellent antibacterial activities. EPL-g-Fc with catalase-like activity could modulate the oxidative microenvironment in mammalian cells and zebrafish by catalyzing H2O2 into H2O and O2. EPL-g-Fc could alleviate inflammatory response in vitro. Furthermore, the macromolecules could accelerate bacteria-infected wound healing in vivo. This work provides a versatile strategy for repairing bacteria-infected wounds by eliminating bacteria, modulating oxidative microenvironment, and alleviating inflammatory response.
Collapse
Affiliation(s)
- Qingqing Xu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| | - Xiaomei Dai
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| | - Lele Yang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| | - Xiaojun Liu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| | - Yu Li
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| | - Feng Gao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu241002, P. R. China
| |
Collapse
|
32
|
MicroRNA-34c-5p exhibits anticancer properties in gastric cancer by targeting MAP2K1 to inhibit cell proliferation, migration, and invasion. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7375661. [PMID: 36203485 PMCID: PMC9532111 DOI: 10.1155/2022/7375661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/27/2022] [Indexed: 12/02/2022]
Abstract
Purpose Gastric cancer(GC)is one of the deadliest digestive tract tumors worldwide,existing studies suggest that dysregulated expression of microRNAs (miRNAs) plays an important role in the pathogenesis and progression of GC. This study aimed to investigate the expression, biological function, and downstream mechanism of miR-34c-5p in GC, provide new targets for gastric cancer diagnosis and treatment. Methods The expression of miR-34c-5p in GC tissues and cell lines was examined by RT-qPCR. Cell wound healing, transwell and cell cloning assays were used to detect the effect of miR-34c-5p on the migration and invasion abilities, respectively, of GC cells. Western blot was performed to detect the expression of related proteins. Bioinformatics analysis was used to predict the binding of MAP2K1 to miR-34c-5p and the targeting relationship was confirmed by dual luciferase reporter assay. Results The expression level of miR-34c-5p was significantly decreased in GC tissues and cell lines. miR-34c-5p overexpression inhibited migration, invasion, and colony formation of gastric cancer cells, the related protein E-cadherin expression was significantly increased and N-cadherin, vimentin, and PCNA expression were significantly decreased, while miR-34c-5p knockdown exerted the opposite effects. In addition, the targeting relationship between miR-34c-5p and MAP2K1 was predicted and confirmed, and further confirmed by rescue experiments that MAP2K1 alleviated the inhibitory effect of miR-34c-5p in GC. Conclusion MiR-34c-5p is lowly expressed in GC, and it can target MAP2K1 to exert inhibitory effects on GC proliferation, invasion, and migration. These findings provide a promising biomarker and a potential therapeutic target for gastric cancer.
Collapse
|
33
|
Innovative Treatment Strategies to Accelerate Wound Healing: Trajectory and Recent Advancements. Cells 2022; 11:cells11152439. [PMID: 35954282 PMCID: PMC9367945 DOI: 10.3390/cells11152439] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/26/2022] Open
Abstract
Wound healing is highly specialized dynamic multiple phase process for the repair of damaged/injured tissues through an intricate mechanism. Any failure in the normal wound healing process results in abnormal scar formation, and chronic state which is more susceptible to infections. Chronic wounds affect patients’ quality of life along with increased morbidity and mortality and are huge financial burden to healthcare systems worldwide, and thus requires specialized biomedical intensive treatment for its management. The clinical assessment and management of chronic wounds remains challenging despite the development of various therapeutic regimens owing to its painstakingly long-term treatment requirement and complex wound healing mechanism. Various conventional approaches such as cell therapy, gene therapy, growth factor delivery, wound dressings, and skin grafts etc., are being utilized for promoting wound healing in different types of wounds. However, all these abovementioned therapies are not satisfactory for all wound types, therefore, there is an urgent demand for the development of competitive therapies. Therefore, there is a pertinent requirement to develop newer and innovative treatment modalities for multipart therapeutic regimens for chronic wounds. Recent developments in advanced wound care technology includes nanotherapeutics, stem cells therapy, bioengineered skin grafts, and 3D bioprinting-based strategies for improving therapeutic outcomes with a focus on skin regeneration with minimal side effects. The main objective of this review is to provide an updated overview of progress in therapeutic options in chronic wounds healing and management over the years using next generation innovative approaches. Herein, we have discussed the skin function and anatomy, wounds and wound healing processes, followed by conventional treatment modalities for wound healing and skin regeneration. Furthermore, various emerging and innovative strategies for promoting quality wound healing such as nanotherapeutics, stem cells therapy, 3D bioprinted skin, extracellular matrix-based approaches, platelet-rich plasma-based approaches, and cold plasma treatment therapy have been discussed with their benefits and shortcomings. Finally, challenges of these innovative strategies are reviewed with a note on future prospects.
Collapse
|
34
|
Anti-Inflammatory microRNAs for Treating Inflammatory Skin Diseases. Biomolecules 2022; 12:biom12081072. [PMID: 36008966 PMCID: PMC9405611 DOI: 10.3390/biom12081072] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 02/07/2023] Open
Abstract
Skin inflammation occurs due to immune dysregulation because of internal disorders, infections, and allergic reactions. The inflammation of the skin is a major sign of chronic autoimmune inflammatory diseases, such as psoriasis, atopic dermatitis (AD), and lupus erythematosus. Although there are many therapies for treating these cutaneous inflammation diseases, their recurrence rates are high due to incomplete resolution. MicroRNA (miRNA) plays a critical role in skin inflammation by regulating the expression of protein-coding genes at the posttranscriptional level during pathogenesis and homeostasis maintenance. Some miRNAs possess anti-inflammatory features, which are beneficial for mitigating the inflammatory response. miRNAs that are reduced in inflammatory skin diseases can be supplied transiently using miRNA mimics and agomir. miRNA-based therapies that can target multiple genes in a given pathway are potential candidates for the treatment of skin inflammation. This review article offers an overview of the function of miRNA in skin inflammation regulation, with a focus on psoriasis, AD, and cutaneous wounds. Some bioactive molecules can target and modulate miRNAs to achieve the objective of inflammation suppression. This review also reports the anti-inflammatory efficacy of these molecules through modulating miRNA expression. The main limitations of miRNA-based therapies are rapid biodegradation and poor skin and cell penetration. Consideration was given to improving these drawbacks using the approaches of cell-penetrating peptides (CPPs), nanocarriers, exosomes, and low-frequency ultrasound. A formulation design for successful miRNA delivery into skin and target cells is also described in this review. The possible use of miRNAs as biomarkers and therapeutic modalities could open a novel opportunity for the diagnosis and treatment of inflammation-associated skin diseases.
Collapse
|
35
|
Morawska-Kochman M, Śmieszek A, Marcinkowska K, Marycz KM, Nelke K, Zub K, Zatoński T, Bochnia M. Expression of Apoptosis-Related Biomarkers in Inflamed Nasal Sinus Epithelium of Patients with Chronic Rhinosinusitis with Nasal Polyps (CRSwNP)—Evaluation at mRNA and miRNA Levels. Biomedicines 2022; 10:biomedicines10061400. [PMID: 35740420 PMCID: PMC9220377 DOI: 10.3390/biomedicines10061400] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/19/2022] Open
Abstract
In chronic upper respiratory tract diseases, increased cell proliferative activity is observed, which is coordinated by BCL-2 proteins and small non-coding RNAs. This study aimed to determine the expression of critical apoptosis markers at the mRNA and miRNA levels in patients with chronic rhinosinusitis with nasal polyps (CSRwNP). The study group consisted of ten patients with CSRwNP and ten healthy subjects. To detect in situ apoptosis in the maxillary sinus mucosa, TUNEL staining was performed. The expression of transcripts was determined by RT-qPCR and included the detection of markers associated with cell survival and apoptosis, i.e., BAX, p53, p21, CASP3, CASP9, c-MYC, CCND1, BRIC5, and APAF1. Levels of miR-17-5p, miR-145-5p, miR-146a-5p, and miR-203a-3p were also measured by RT-qPCR. The obtained results indicated increased apoptosis determined by a TUNEL assay in CSRwNP patients and accompanied by an increased expression of BAX, P21, P53, CASP3, CASP9, c-MYC, and APAF-1 transcripts and decreased mRNA levels of BCL-2 and BIRC5. Furthermore, the nasal sinus epithelium of patients with CSRwNP showed increased levels of miR-203a-3p while also showing a decreased expression of miR-17-5p and miR-145-5p. Our results showed that pro-apoptotic transcripts detected at mRNA and miRNA levels might be involved in the pathogenesis of chronic sinusitis with polyps. The identification of those key molecular mediators may be applicable for the specific diagnostic and/or development of targeted therapies for chronic sinusitis with polyps.
Collapse
Affiliation(s)
- Monika Morawska-Kochman
- Department of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (K.Z.); (T.Z.); (M.B.)
- Correspondence:
| | - Agnieszka Śmieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland; (A.Ś.); (K.M.); (K.M.M.)
| | - Klaudia Marcinkowska
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland; (A.Ś.); (K.M.); (K.M.M.)
| | - Krzysztof Mariusz Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland; (A.Ś.); (K.M.); (K.M.M.)
| | - Kamil Nelke
- Department of Maxillofacial Surgery, 4th Military Clinical Hospital, Weigla 5, 50-981 Wroclaw, Poland;
| | - Krzysztof Zub
- Department of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (K.Z.); (T.Z.); (M.B.)
| | - Tomasz Zatoński
- Department of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (K.Z.); (T.Z.); (M.B.)
| | - Marek Bochnia
- Department of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (K.Z.); (T.Z.); (M.B.)
| |
Collapse
|