Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 226
Filter
1.
Skin Res Technol ; 30(5): e13686, 2024 May.
Article in English | MEDLINE | ID: mdl-38682767

ABSTRACT

BACKGROUND: Our study aims to delineate the miRSNP-microRNA-gene-pathway interactions in the context of hypertrophic scars (HS) and keloids. MATERIALS AND METHODS: We performed a computational biology study involving differential expression analysis to identify genes and their mRNAs in HS and keloid tissues compared to normal skin, identifying key hub genes and enriching their functional roles, comprehensively analyzing microRNA-target genes and related signaling pathways through bioinformatics, identifying MiRSNPs, and constructing a pathway-based network to illustrate miRSNP-miRNA-gene-signaling pathway interactions. RESULTS: Our results revealed a total of 429 hub genes, with a strong enrichment in signaling pathways related to proteoglycans in cancer, focal adhesion, TGF-ß, PI3K/Akt, and EGFR tyrosine kinase inhibitor resistance. Particularly noteworthy was the substantial crosstalk between the focal adhesion and PI3K/Akt signaling pathways, making them more susceptible to regulation by microRNAs. We also identified specific miRNAs, including miRNA-1279, miRNA-429, and miRNA-302e, which harbored multiple SNP loci, with miRSNPs rs188493331 and rs78979933 exerting control over a significant number of miRNA target genes. Furthermore, we observed that miRSNP rs188493331 shared a location with microRNA302e, microRNA202a-3p, and microRNA20b-5p, and these three microRNAs collectively targeted the gene LAMA3, which is integral to the focal adhesion signaling pathway. CONCLUSIONS: The study successfully unveils the complex interactions between miRSNPs, miRNAs, genes, and signaling pathways, shedding light on the genetic factors contributing to HS and keloid formation.


Subject(s)
Cicatrix, Hypertrophic , Keloid , MicroRNAs , Humans , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Computational Biology , Keloid/genetics , Keloid/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Polymorphism, Single Nucleotide , Signal Transduction/genetics
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670440

ABSTRACT

BACKGROUND & AIMS: Hypertrophic scar (HS) is a skin fibroproliferative disorder occurring after burns, surgeries or traumatic injuries, and it has caused a tremendous economic and medical burden. Its molecular mechanism is associated with the abnormal proliferation and transition of fibroblasts and excessive deposition of extracellular matrix. Cartilage intermediate layer protein 2 (CILP2), highly homologous to cartilage intermediate layer protein 1 (CILP1), is mainly secreted predominantly from chondrocytes in the middle/deeper layers of articular cartilage. Recent reports indicate that CILP2 is involved in the development of fibrotic diseases. We investigated the role of CILP2 in the progression of HS. METHODS AND RESULTS: It was found in this study that CILP2 expression was significantly higher in HS than in normal skin, especially in myofibroblasts. In a clinical cohort, we discovered that CILP2 was more abundant in the serum of patients with HS, especially in the early stage of HS. In vitro studies indicated that knockdown of CILP2 suppressed proliferation, migration, myofibroblast activation and collagen synthesis of hypertrophic scar fibroblasts (HSFs). Further, we revealed that CILP2 interacts with ATP citrate lyase (ACLY), in which CILP2 stabilizes the expression of ACLY by reducing the ubiquitination of ACLY, therefore prompting Snail acetylation and avoiding reduced expression of Snail. In vivo studies indicated that knockdown of CILP2 or ACLY inhibitor, SB-204990, significantly alleviated HS formation. CONCLUSION: CILP2 exerts a vital role in hypertrophic scar formation and might be a detectable biomarker reflecting the progression of hypertrophic scar and a therapeutic target for hypertrophic scar.


Subject(s)
Cicatrix, Hypertrophic , Snail Family Transcription Factors , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/genetics , Humans , Acetylation , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Male , Animals , Cell Proliferation , Female , Myofibroblasts/metabolism , Myofibroblasts/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Adult , Mice , Cell Movement
3.
Burns ; 50(5): 1247-1258, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38503573

ABSTRACT

OBJECTIVE: Research indicates that long noncoding RNAs (lncRNAs) contribute significantly to fibrotic diseases. Although lncRNAs may play a role in hypertrophic scars after burns, its mechanisms remain poorly understood. METHODS: Using chip technology, we compared the lncRNA expression profiles of burn patients and healthy controls (HCs). Microarray results were examined by quantitative reverse-transcription polymerase chain reaction (RT-PCR) to verify their reliability. The biological functions of differentially expressed mRNAs and the relationships between genes and signaling pathways were investigated by Gene Ontology (GO) and pathway analyses, respectively. RESULTS: In contrast with HCs, it was found that 2738 lncRNAs (1628 upregulated) and 2166 mRNAs (1395 upregulated) were differentially expressed in hypertrophic scars after burn. Results from RT-PCR were consistent with those from microarray. GO and pathway analyses revealed that the differentially expressed mRNAs are mainly associated with processes related to cytokine secretion in the immune system, notch signaling, and MAPK signaling. CONCLUSION: The lncRNA expression profiles of hypertrophic scars after burn changed significantly compared with HCs. It was believed that the transcripts could be used as potential targets for inhibiting abnormal scar formation in burn patients.


Subject(s)
Burns , Cicatrix, Hypertrophic , RNA, Long Noncoding , RNA, Messenger , Humans , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/etiology , Burns/metabolism , Burns/complications , Burns/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Male , Female , Adult , RNA, Messenger/metabolism , RNA, Messenger/genetics , Case-Control Studies , Middle Aged , Young Adult , Up-Regulation , Gene Expression Profiling , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Adolescent , Oligonucleotide Array Sequence Analysis , Gene Ontology
4.
Genomics ; 116(2): 110797, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38262564

ABSTRACT

BACKGROUND: Hypertrophic scar (HTS) is a prevalent chronic inflammatory skin disorder characterized by abnormal proliferation and extracellular matrix deposition and the precise mechanisms underlying HTS remain elusive. This study aimed to identify and validate potential immune-related genes associated with hypertrophic scar formation. METHODS: Skin samples from normal (n = 12) and hypertrophic scar tissues (n = 12) were subjected to RNA-seq analysis. Differentially expressed genes (DEGs) and significant modular genes in Weighted gene Co-expression Network Analysis (WGCNA) were identified. Subsequently, functional enrichment analysis was performed on the intersecting genes. Additionally, eight immune-related genes were matched from the ImmPort database. Validation of NRG1 and CRLF1 was carried out using an external cohort (GSE136906). Furthermore, the association between these two genes and immune cells was assessed by Spearman correlation analysis. Finally, RNA was extracted from normal and hypertrophic scar samples, and RT-qPCR, Immunohistochemistry staining and Western Blot were employed to validate the expression of characteristic genes. RESULTS: A total of 940 DEGs were identified between HTS and normal samples, and 288 key module genes were uncovered via WGCNA. Enrichment analysis in key module revealed involvement in many immune-related pathways, such as Th17 cell differentiation, antigen processing and presentation and B cell receptor signaling pathway. The eight immune-related genes (IFI30, NR2F2, NRG1, ESM1, NFATC2, CRLF1, COLEC12 and IL6) were identified by matching from the ImmPort database. Notably, we observed that activated mast cell positively correlated with CRLF1 expression, while CD8 T cells exhibited a positive correlation with NRG1. The expression of NRG1 and CRLF1 was further validated in clinical samples. CONCLUSION: In this study, two key immune-related genes (CRLF1 and NRG1) were identified as characteristic genes associated with HTS. These findings provide valuable insights into the immune-related mechanisms underlying hypertrophic scar formation.


Subject(s)
Cicatrix, Hypertrophic , Neuregulin-1 , Receptors, Cytokine , Humans , Cell Differentiation , Cicatrix, Hypertrophic/genetics , Databases, Factual , Extracellular Matrix , Skin , Receptors, Cytokine/genetics
5.
Int Wound J ; 21(1): e14384, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37697692

ABSTRACT

Pathological scarring resulting from traumas and wounds, such as hypertrophic scars and keloids, pose significant aesthetic, functional and psychological challenges. This study provides a comprehensive transcriptomic analysis of these conditions, aiming to illuminate underlying molecular mechanisms and potential therapeutic targets. We employed a co-expression and module analysis tool to identify significant gene clusters associated with distinct pathophysiological processes and mechanisms, notably lipid metabolism, sebum production, cellular energy metabolism and skin barrier function. This examination yielded critical insights into several skin conditions including folliculitis, skin fibrosis, fibrosarcoma and congenital ichthyosis. Particular attention was paid to Module Cluster (MCluster) 3, encompassing genes like BLK, TRPV1 and GABRD, all displaying high expression and potential implications in immune modulation. Preliminary immunohistochemistry validation supported these findings, showing elevated expression of these genes in non-fibrotic samples rich in immune activity. The complex interplay of different cell types in scar formation, such as fibroblasts, myofibroblasts, keratinocytes and mast cells, was also explored, revealing promising therapeutic strategies. This study underscores the promise of targeted gene therapy for pathological scars, paving the way for more personalised therapeutic approaches. The results necessitate further research to fully ascertain the roles of these identified genes and pathways in skin disease pathogenesis and potential therapeutics. Nonetheless, our work forms a strong foundation for a new era of personalised medicine for patients suffering from pathological scarring.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Humans , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/therapy , Cicatrix, Hypertrophic/metabolism , Keloid/genetics , Keloid/therapy , Keratinocytes/metabolism , Fibroblasts/metabolism , Myofibroblasts/metabolism
6.
Small ; 20(8): e2305374, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37724002

ABSTRACT

Hypertrophic scar (HS) is a common fibroproliferative disease caused by abnormal wound healing after deep skin injury. However, the existing approaches have unsatisfactory therapeutic effects, which promote the exploration of newer and more effective strategies. MiRNA-modified functional exosomes delivered by dissolvable microneedle arrays (DMNAs) are expected to provide new hope for HS treatment. In this study, a miRNA, miR-141-3p, which is downregulated in skin scar tissues and in hypertrophic scar fibroblasts (HSFs), is identified. MiR-141-3p mimics inhibit the proliferation, migration, and myofibroblast transdifferentiation of HSFs in vitro by targeting TGF-ß2 to suppress the TGF-ß2/Smad pathway. Subsequently, the engineered exosomes encapsulating miR-141-3p (miR-141-3pOE -Exos) are isolated from adipose-derived mesenchymal stem cells transfected with Lv-miR-141-3p. MiR-141-3pOE -Exos show the same inhibitive effects as miR-141-3p mimics on the pathological behaviors of HSFs in vitro. The DMNAs for sustained release of miR-141-3pOE -Exos are further fabricated in vivo. MiR-141OE -Exos@DMNAs effectively decrease the thickness of HS and improve fibroblast distribution and collagen fiber arrangement, and downregulate the expression of α-SMA, COL-1, FN, TGF-ß2, and p-Smad2/3 in the HS tissue. Overall, a promising, effective, and convenient exosome@DMNA-based miRNA delivery strategy for HS treatment is provided.


Subject(s)
Cicatrix, Hypertrophic , Exosomes , MicroRNAs , Humans , Cicatrix, Hypertrophic/therapy , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Transforming Growth Factor beta2/metabolism , Exosomes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Fibroblasts/metabolism , Cell Proliferation/genetics
7.
Int J Biol Macromol ; 256(Pt 1): 128334, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38007032

ABSTRACT

Hypertrophic scars (HS) and keloids (KD) are lesions that develop as a result of excessive fibroblast proliferation and collagen deposition in response to dermal injury, leading to dysregulation of the inflammatory, proliferative, and remodeling phases during wound healing. HS and KD affect up to 90 % of the population and are associated with lower quality of life, physical health, and mental status in patients. Efficient targeted treatment represents a significant challenge, primarily due to our limited understanding of their underlying pathogenesis. Non-coding RNAs (ncRNAs), which constitute a significant portion of the human transcriptome with minimal or no protein-coding capacity, have been implicated in various cellular physiologies and pathologies and may serve as diagnostic indicators or therapeutic targets. NcRNAs have been found to be aberrantly expressed and regulated in HS and KD. This review provides a summary of the expression profiles and molecular mechanisms of three common ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in HS and KD. It also discusses their potential as biomarkers for the diagnosis and treatment of these diseases and provides novel insights into epigenetic-based diagnosis and treatment strategies for HS and KD.


Subject(s)
Cicatrix, Hypertrophic , Keloid , MicroRNAs , RNA, Long Noncoding , Humans , Cicatrix, Hypertrophic/genetics , Keloid/genetics , Quality of Life , Wound Healing , MicroRNAs/genetics
8.
Cell Mol Biol (Noisy-le-grand) ; 69(7): 158-163, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37715390

ABSTRACT

The formation of hypertrophic scar and keloid is considered to be a very complex pathological process. Our previous studies have shown that miR-15a-5p is an important miRNA in HTS tissues, and its expression level is significantly increased. Therefore, the potential mechanism of action of miR-15a-5p in scarring arouses our interest. This study preliminarily investigated the expression level of miR-15a-5p in HTS tissue and normal skin tissue and further explored the molecular mechanism. The results of this study once again confirmed that the expression level of miR-15a-5p was increased in HTS tissues and cells, and the closely related mRNA and protein levels of MyD88 and TGF-ß were also highly expressed. The relative expression levels of fibrosis-related indicators in HTsFb cells were up-regulated, such as collagen-Ⅰ, collagen-III and α-SMA. We constructed the HTS cell model and BALB/c nude animal model, and down-regulating miR-15a-5p, the HTsFb cells proliferation was inhibited, and qRT-PCR results showed that the fibrosis index mRNA was also reduced, and significantly reduce the pathological state of scar tissue. In conclusion, miR-15a-5p may participate in the formation and development of HTS through TLR/MyD88 signaling pathway and TGF-ß1 signaling pathway.


Subject(s)
Cicatrix, Hypertrophic , Keloid , MicroRNAs , Animals , Mice , Cicatrix, Hypertrophic/genetics , Keloid/genetics , Toll-Like Receptor 4/genetics , Myeloid Differentiation Factor 88/genetics , Adaptor Proteins, Signal Transducing , Mice, Nude , MicroRNAs/genetics , RNA, Messenger/genetics
9.
Front Immunol ; 14: 1207522, 2023.
Article in English | MEDLINE | ID: mdl-37409114

ABSTRACT

Hypertrophic scar (HS) is a chronic inflammatory skin disease characterized by excessive deposition of extracellular matrix, but the exact mechanisms related to its formation remain unclear, making it difficult to treat. This study aimed to investigate the potential role of cuproptosis in the information of HS. To this end, we used single-cell sequencing and bulk transcriptome data, and screened for cuproptosis-related genes (CRGs) using differential gene analysis and machine learning algorithms (random forest and support vector machine). Through this process, we identified a group of genes, including ATP7A, ULK1, and MTF1, as novel therapeutic targets for HS. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to confirm the mRNA expression of ATP7A, ULK1, and MTF1 in both HS and normal skin (NS) tissues. We also constructed a diagnostic model for HS and analyzed the immune infiltration characteristics. Additionally, we used the expression profiles of CRGs to perform subgroup analysis of HS. We focused mainly on fibroblasts in the transcriptional profile at single-cell resolution. By calculating the cuproptosis activity of each fibroblast, we found that cuproptosis activity of normal skin fibroblasts increased, providing further insights into the pathogenesis of HS. We also analyzed the cell communication network and transcription factor regulatory network activity, and found the existence of a fibroblast-centered communication regulation network in HS, where cuproptosis activity in fibroblasts affects intercellular communication. Using transcription factor regulatory activity network analysis, we obtained highly active transcription factors, and correlation analysis with CRGs suggested that CRGs may serve as potential target genes for transcription factors. Overall, our study provides new insights into the pathophysiological mechanisms of HS, which may inspire new ideas for the diagnosis and treatment.


Subject(s)
Apoptosis , Cicatrix, Hypertrophic , Humans , Algorithms , Cicatrix, Hypertrophic/genetics , Machine Learning , Single-Cell Analysis , Skin , Copper
10.
J Dermatol ; 50(9): 1170-1179, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37365986

ABSTRACT

Hypertrophic scars and keloids are fibroproliferative disorders caused by abnormal wound healing. Their exact cause has not been found, but abnormalities during the wound healing process including inflammatory, immune, genetic, and other factors are thought to predispose an individual to excessive scarring. In the present study, we performed transcriptome analysis of established keloid cell lines (KEL FIB), focusing on gene expression analysis and fusion gene detection for the first time. For gene expression analysis, fragments per kilobase per million map read values were calculated, which were validated by real-time PCR and immunohistochemistry. Fusion genes were predicted by transcriptome sequence, and validated by Sanger sequence and G-banding. As a result, GPM6A was shown in the expression analysis to be upregulated in KEL FIB compared with normal fibroblasts. The GPM6A upregulation in KEL FIB was confirmed by real-time PCR, and GPM6A messenger ribonucleic acid expression was consistently significantly elevated in the tissues of hypertrophic scar and keloid compared to normal skin. Immunohistochemistry also revealed that the number of fibroblast-like spindle-shaped cells positive for GPM6A was significantly increased in keloidal tissues. GPM6A inhibition by small interfering ribonucleic acid significantly reduced the number of KEL FIB. On the other hand, although we hypothesized that fusion genes are involved in the pathogenesis of keloids, the transcriptome analysis could not prove the presence of fusion genes in KEL FIB. Taken together, GPM6A upregulation may have an inducible effect on cell proliferation in keloidal fibroblasts. GPM6A can be a novel therapeutic target in hypertrophic scars and keloids. The inflammatory nature may be more prominent in the pathogenesis of keloids, rather than being skin tumors, as proposed by Ogawa et al. Future studies using several cell lines will be required.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Humans , Keloid/genetics , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Up-Regulation , Transcriptome , Fibroblasts/pathology , Gene Expression Profiling , Cell Proliferation/genetics , RNA , Glycoproteins/genetics
11.
Lasers Surg Med ; 55(5): 490-502, 2023 07.
Article in English | MEDLINE | ID: mdl-37051852

ABSTRACT

OBJECTIVES: One symptom of hypertrophic scar (HTS) that can develop after burn injury is dyschromia with hyper- and hypopigmentation. There are limited treatments for these conditions. Previously, we showed there is no expression of alpha melanocyte stimulating hormone (α-MSH) in hypopigmented scars, and if these melanocytes are treated with synthetic α-MSH in vitro, they respond by repigmenting. The current study tested the same hypothesis in the in vivo environment using laser-assisted drug delivery (LADD). METHODS: HTSs were created in red Duroc pigs. At Day 77 (pre), they were treated with CO2 fractional ablative laser (FLSR). Synthetic α-MSH was delivered as a topical solution dissolved in  l-tyrosine (n = 6, treated). Control scars received LADD of  l-tyrosine only (n = 2, control). Scars were treated and examined weekly through Week 4. Digital images and punch biopsies of hyper, hypo-, and normally pigmented scar and skin were collected. Digital pictures were analyzed with ImageJ by tracing the area of hyperpigmentation. Epidermal sheets were obtained from punch biopsies through dispase separation and RNA was isolated. qRT-PCR was run for melanogenesis-related genes: tyrosinase (TYR), tyrosinase-related protein-1 (TYRP1), and dopachrome tautomerase (DCT). Two-way ANOVA with multiple comparisons and Dunnett's correction compared the groups. RESULTS: The areas of hyperpigmentation were variable before treatment. Therefore, data is represented as fold-change where each scar was normalized to its own pre value. Within the LADD of NDP α-MSH + l-tyrosine group, hyperpigmented areas gradually increased each week, reaching 1.3-fold over pre by Week 4. At each timepoint, area of hyperpigmentation was greater in the treated versus the control (1.04 ± 0.05 vs. 0.89 ± 0.08, 1.21 ± 0.07 vs. 0.98 ± 0.24, 1.21 ± 0.08 vs. 1.04 ± 0.11, 1.28 ± 0.11 vs. 0.94 ± 0.25; fold-change from pre-). Within the treatment group, pretreatment, levels of TYR were decreased -17.76 ± 4.52 below the level of normal skin in hypopigmented scars. After 1 treatment, potentially due to laser fractionation, the levels decreased to -43.49 ± 5.52. After 2, 3, and 4 treatments, there was ever increasing levels of TYR to almost the level of normally pigmented skin (-35.74 ± 15.72, -23.25 ± 6.80, -5.52 ± 2.22 [p < 0.01, Week 4]). This pattern was also observed for TYRP1 (pre = -12.94 ± 1.82, Week 1 = -48.85 ± 13.25 [p < 0.01], Weeks 2, 3, and 4 = -34.45 ± 14.64, -28.19 ± 4.98, -6.93 ± 3.05 [p < 0.01, Week 4]) and DCT (pre = -214.95 ± 89.42, Week 1 = -487.93 ± 126.32 [p < 0.05], Weeks 2, 3, and 4 = -219.06 ± 79.33, -72.91 ± 20.45 [p < 0.001], -76.00 ± 24.26 [p < 0.001]). Similar patterns were observed for scars treated with LADD of  l-tyrosine alone without NDP α-MSH. For each gene, in hyperpigmented scar, levels increased at Week 4 of treatment compared to Week 1 (p < 0.01). CONCLUSIONS: A clinically-relevant FLSR treatment method can be combined with topical delivery of synthetic α-MSH and l-tyrosine to increase the area of pigmentation and expression of melanogenesis genes in hypopigmented HTS. LADD of  l-tyrosine alone leads to increased expression of melanogenesis genes. Future studies will aim to optimize drug delivery, timing, and dosing.


Subject(s)
Cicatrix, Hypertrophic , Hyperpigmentation , Hypopigmentation , Lasers, Gas , Animals , Swine , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Tyrosine , alpha-MSH/therapeutic use , alpha-MSH/metabolism , Pharmaceutical Preparations , Pigmentation , Hypopigmentation/drug therapy , Hypopigmentation/genetics , Hyperpigmentation/drug therapy , Hyperpigmentation/genetics , Lasers, Gas/therapeutic use , Melanins/metabolism
12.
Article in Chinese | MEDLINE | ID: mdl-36878530

ABSTRACT

Hypertrophic scar (HS) affects the function and beauty of patients, and brings a heavy psychological burden to patients. However, the specific pathogenesis mechanism of HS in molecular biology level is not yet clear, and this disease is still one of the clinical diseases difficult to prevent and cure. MicroRNA (miR) is a family of single-stranded endogenous noncoding RNAs that can regulate gene expression. The abnormal transcription of miR in hypertrophic scar fibroblasts can affect the transduction and expression of downstream signal pathway or protein, and the exploration of miR and its downstream signal pathway and protein helps deeply understand the occurrence and development mechanism of scar hyperplasia. This article summarized and analyzed how miR and multiple signal pathways involve in the formation and development of HS in recent years, and further outlined the interaction between miR and target genes in HS.


Subject(s)
Cicatrix, Hypertrophic , MicroRNAs , Humans , MicroRNAs/genetics , Cicatrix, Hypertrophic/genetics , Fibroblasts , Hyperplasia
14.
J Plast Surg Hand Surg ; 57(1-6): 163-171, 2023.
Article in English | MEDLINE | ID: mdl-35001812

ABSTRACT

Keloid is a disease that seriously affects the aesthetic appearance of the body. In contrast to normal skin or hypertrophic scars, keloid tissue extends beyond the initial site of injury. Patients may complain of pain, itching, or burning. Although multiple treatments exist, none is uniformly successful. Genetic advances have made it possible to explore differences in gene expression between keloids and normal skin. Identifying the biomarker for keloid is beneficial to the mechanism exploration and treatment development of keloid. In this study, we identified seven genes with significant differences in keloids through weighted gene co-expression network analysis(WGCNA) and differential expression analysis. Then, by the Lasso regression, we constructed a keloid diagnostic model using five of these genes. Further studies found that keloids could be divided into high-risk and low-risk groups by this model, with differences in immunity, m6A methylation, and pyroptosis. Finally, we verified the accuracy of the diagnostic model in clinical RNA-sequencing data.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Humans , Keloid/genetics , Keloid/pathology , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Gene Expression Profiling , Biomarkers , Pruritus
15.
Chinese Journal of Burns ; (6): 196-200, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-971170

ABSTRACT

Hypertrophic scar (HS) affects the function and beauty of patients, and brings a heavy psychological burden to patients. However, the specific pathogenesis mechanism of HS in molecular biology level is not yet clear, and this disease is still one of the clinical diseases difficult to prevent and cure. MicroRNA (miR) is a family of single-stranded endogenous noncoding RNAs that can regulate gene expression. The abnormal transcription of miR in hypertrophic scar fibroblasts can affect the transduction and expression of downstream signal pathway or protein, and the exploration of miR and its downstream signal pathway and protein helps deeply understand the occurrence and development mechanism of scar hyperplasia. This article summarized and analyzed how miR and multiple signal pathways involve in the formation and development of HS in recent years, and further outlined the interaction between miR and target genes in HS.


Subject(s)
Humans , MicroRNAs/genetics , Cicatrix, Hypertrophic/genetics , Fibroblasts , Hyperplasia
16.
Eur J Dermatol ; 33(6): 604-611, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38465540

ABSTRACT

Wound healing and skin regeneration after injury are complex biological processes, and deep injuries with a high degree of tissue destruction may result in severe scar formation. Clinically, scars can be classified into normal, hypertrophic and keloid scars. However, the molecular signature of each scar type is currently not known. The aim of this study was to reveal the transcriptional landscape of normal, hypertrophic and keloid skin scars following hand and plastic surgery based on total RNA sequencing. Eighteen skin scar samples from hand and plastic surgeries of human donors were minced directly after removal and stored in TRIzol (Thermo Fisher, USA). Samples were then subjected to RNA isolation, cDNA library preparation, bulk RNA sequencing and bioinformatics analysis. We show that keloid scars transcriptionally differed from normal and hypertrophic scars. Normal and hypertrophic scars presented overlapping clustering, and eight genes were shown to be commonly expressed between hypertrophic and normal scars. No genes were specifically expressed at a higher level in keloid and normal scars. Based on gene ontology pathway analysis, genes with a higher level of expression in keloid scars lead to increased (extra-) cellular matrix proliferation and cell interaction. Moreover, tumour-like genes were more highly expressed in keloid scars, supporting the clinical impression of strong and diffuse growth. This study furthers our understanding of the classification of differential scar types based on molecular signature, which may shed light on new diagnostic and therapeutic strategies for keloid scars in the future.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Humans , Keloid/genetics , Keloid/pathology , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Skin/pathology , Wound Healing/genetics , Hypertrophy/pathology
17.
Cells ; 11(24)2022 12 12.
Article in English | MEDLINE | ID: mdl-36552789

ABSTRACT

Background: As a fibrotic disease with a high incidence, the pathogenesis of hypertrophic scarring is still not fully understood, and the treatment of this disease is also challenging. In recent years, human adipose-derived mesenchymal stem cells (AD-MSCs) have been considered an effective treatment for hypertrophic scars. This study mainly explored whether the therapeutic effect of AD-MSCs on hypertrophic scars is associated with oxidative-stress-related proteins. Methods: AD-MSCs were isolated from adipose tissues and characterized through flow cytometry and a differentiation test. Afterwards, coculture, cell proliferation, apoptosis, and migration were detected. Western blotting and a quantitative real-time polymerase chain reaction (qRT-PCR) were used to detect oxidative stress-related genes and protein expression in hypertrophic scar fibroblasts (HSFs). Flow cytometry was used to detect reactive oxygen species (ROS). A nude mouse animal model was established; the effect of AD-MSCs on hypertrophic scars was observed; and hematoxylin and eosin staining, Masson's staining, and immunofluorescence staining were performed. Furthermore, the content of oxidative-stress-related proteins, including nuclear factor erythroid-2-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), B-cell lymphoma 2(Bcl2), Bcl2-associated X(BAX) and caspase 3, was detected. Results: Our results showed that AD-MSCs inhibited HSFs' proliferation and migration and promoted apoptosis. Moreover, after coculture, the expression of antioxidant enzymes, including HO-1, in HSFs decreased; the content of reactive oxygen species increased; and the expression of Nrf2 decreased significantly. In animal experiments, we found that, at 14 days after injection of AD-MSCs into human hypertrophic scar tissue blocks that were transplanted onto the dorsum of nude mice, the weight of the tissue blocks decreased significantly. Hematoxylin and eosin staining and Masson's staining demonstrated a rearrangement of collagen fibers. We also found that Nrf2 and antioxidant enzymes decreased significantly, while apoptotic cells increased after AD-MSC treatment. Conclusions: Our results demonstrated that AD-MSCs efficiently cured hypertrophic scars by promoting the apoptosis of HSFs and by inhibiting their proliferation and migration, which may be related to the inhibition of Nrf2 expression in HSFs, suggesting that AD-MSCs may provide an alternative therapeutic approach for the treatment of hypertrophic scars.


Subject(s)
Cicatrix, Hypertrophic , Fibroblasts , Mesenchymal Stem Cells , Animals , Humans , Mice , Antioxidants/metabolism , Apoptosis , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/therapy , Eosine Yellowish-(YS) , Fibroblasts/metabolism , Hematoxylin , Mesenchymal Stem Cells/metabolism , Mice, Nude , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism
18.
Arkh Patol ; 84(6): 23-31, 2022.
Article in Russian | MEDLINE | ID: mdl-36469714

ABSTRACT

OBJECTIVE: Evaluate the morphogenetic and pathogenetic features of hypertrophic and keloid scars of the head and neck. MATERIAL AND METHODS: The study included 286 patients, among them 176 (61.5%) patients with hypertrophic and 110 (38.5%) with keloid scars aged 18 to 65 years with a disease duration from 1 month to 2 years. Material for histological and immunohistochemical (IHC) studies of scar tissue was fixed in 10% buffered formalin. Serial paraffin sections were stained with H&E, according to Van Gieson and Weigert. IHC was performed using monoclonal mouse antibodies to collagen type I (clone 3G3, Santa Cruz, dilution 1:100), collagen type III (clone B-4, Santa Cruz, dilution 1:50), collagen type IV (clone COL-94, Santa Cruz, dilution 1:50), MMP-1 (clone 3B6, Santa Cruz, dilution 1:100), α-SMA1 (clone 1A4, Dako Agilent, dilution 1:100) and rabbit polyclonal anti-TGFß antibodies (clone 3C11, Santa Cruz, 1:100 dilution). RESULTS: Pathogenetic, morphological and immunohistochemical differences in hypertrophic and keloid scars were established depending on their degree of maturity. In the formation of hypertrophic scars, the key factor in sclerotic processes is TGF-b on the background of low MMP1 activity. Keloid scars were distinguished not only by the accumulation of hard-to-degrade collagens, but also by the development of an osteoclast-like reaction with a high content of MMP1. Immature scar tissue was characterized by the presence of myofibroblastic α-SMA1 positive focus and center of inflammatory changes. CONCLUSIONS: The data obtained allow substantiating new approaches to the treatment of patients with hypertrophic and keloid scars.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Collagen , Keloid/genetics , Keloid/pathology , Matrix Metalloproteinase 1 , Humans , Adolescent , Young Adult , Adult , Middle Aged , Aged
19.
Int J Biol Sci ; 18(15): 5724-5739, 2022.
Article in English | MEDLINE | ID: mdl-36263180

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling plays a key role in excessive fibrosis. As a class IIa family histone deacetylase (HDAC), HDAC5 shows a close relationship with TGF-ß signaling and fibrosis. However, the effect and regulatory mechanism of HDAC5 in hypertrophic scar (HS) formation remain elusive. We show that HDAC5 was overexpressed in HS tissues and depletion of HDAC5 attenuated HS formation in vivo and inhibited fibroblast activation in vitro. HDAC5 knockdown (KD) significantly downregulated TGF-ß1 induced Smad2/3 phosphorylation and increased Smad7 expression. Meanwhile, Smad7 KD rescued the Smad2/3 phosphorylation downregulation and scar hyperplasia inhibition mediated by HDAC5 deficiency. Luciferase reporter assays and ChIP-qPCR assays revealed that HDAC5 interacts with myocyte enhancer factor 2A (MEF2A) suppressing MEF2A binding to the Smad7 promoter region, which results in Smad7 promoter activity repression. HDAC4/5 inhibitor, LMK235, significantly alleviated hypertrophic scar formation. Our study provides clues for the development of HDAC5 targeting strategies for the therapy or prophylaxis of fibrotic diseases.


Subject(s)
Cicatrix, Hypertrophic , Histone Deacetylases , MEF2 Transcription Factors , Smad7 Protein , Humans , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/metabolism , Fibroblasts/metabolism , Fibrosis , Histone Deacetylases/metabolism , Luciferases/metabolism , MEF2 Transcription Factors/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/metabolism
20.
Int J Mol Sci ; 23(11)2022 May 30.
Article in English | MEDLINE | ID: mdl-35682793

ABSTRACT

The clinical aspects of hypertrophic scarring vary according to personal constitution and body part. However, the mechanism of hypertrophic scar (HS) formation remains unclear. MicroRNAs (miRNAs) are known to contribute to HS formation, however, their detailed role remains unknown. In this study, candidate miRNAs were identified and analyzed as biomarkers of hypertrophic scarring for future clinical applications. HSfibroblasts and normal skin fibroblasts from patients were used for profiling and validation of miRNAs. An HS mouse model with xenografted human skin on nude mice was established. The miRNA expression between normal human, normal mouse, and mouse HS skin tissues was compared. Circulating miRNA expression levels in the serum of normal mice and mice with HSs were also analyzed. Ten upregulated and twenty-one downregulated miRNAs were detected. Among these, miR-365a/b-3p and miR-16-5p were identified as candidate miRNAs with statistically significant differences; miR-365a/b-3p was significantly upregulated (p = 0.0244). In mouse studies, miR-365a/b-3p expression levels in skin tissue and serum were higher in mice with HSs than in the control group. These results indicate that miRNAs contribute to hypertrophic scarring and that miR-365a/b-3p may be considered a potential biomarker for HS formation.


Subject(s)
Cicatrix, Hypertrophic , Circulating MicroRNA , MicroRNAs , Animals , Biomarkers/metabolism , Cicatrix, Hypertrophic/genetics , Gene Expression Profiling , Humans , Mice , Mice, Nude , MicroRNAs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...