Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.502
Filter
1.
Nat Commun ; 15(1): 8637, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39366968

ABSTRACT

The cell transition from an inflammatory phase to a subsequent proliferative phase is crucial for wound healing, yet the driving mechanism remains unclear. By profiling lncRNA expression changes during human skin wound healing and screening lncRNA functions, we identify SNHG26 as a pivotal regulator in keratinocyte progenitors underpinning this phase transition. Snhg26-deficient mice exhibit impaired wound repair characterized by delayed re-epithelization accompanied by exacerbated inflammation. Single-cell transcriptome analysis combined with gain-of-function and loss-of-function of SNHG26 in vitro and ex vivo reveals its specific role in facilitating inflammatory-to-proliferative state transition of keratinocyte progenitors. A mechanistic study unravels that SNHG26 interacts with and relocates the transcription factor ILF2 from inflammatory genomic loci, such as JUN, IL6, IL8, and CCL20, to the genomic locus of LAMB3. Collectively, our findings suggest that lncRNAs play cardinal roles in expediting tissue repair and regeneration and may constitute an invaluable reservoir of therapeutic targets in reparative medicine.


Subject(s)
Cell Proliferation , Keratinocytes , RNA, Long Noncoding , Stem Cells , Wound Healing , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Keratinocytes/metabolism , Animals , Humans , Wound Healing/genetics , Cell Proliferation/genetics , Stem Cells/metabolism , Mice , Inflammation/genetics , Inflammation/pathology , Inflammation/metabolism , Skin/pathology , Skin/metabolism , Mice, Knockout , Mice, Inbred C57BL , Male
2.
Life Sci Alliance ; 7(12)2024 Dec.
Article in English | MEDLINE | ID: mdl-39366762

ABSTRACT

Mucosal healing is associated with better clinical outcomes in patients with inflammatory bowel disease. But the epithelial-specific contribution to mucosal healing in vivo is poorly understood. We evaluated mucosal healing in an acute dextran sulfate sodium mouse model that shows an alleviated colitis response after epithelial-specific loss of Smad4. We find that enhanced epithelial wound healing alleviates the fibrotic response. Dextran sulfate sodium caused increased mesenchymal collagen deposition-indicative of fibrosis-within a week in the WT but not in the Smad4 KO colon. The fibrotic response correlated with decreased epithelial proliferation in the WT, whereas uninterrupted proliferation and an expanded zone of proliferation were observed in the Smad4 KO colon epithelium. Furthermore, the Smad4 KO colon showed epithelial extracellular matrix alterations that promote epithelial regeneration. Our data suggest that epithelium is a key determinant of the mucosal healing response in vivo, implicating mucosal healing as a strategy against fibrosis in inflammatory bowel disease patients.


Subject(s)
Colitis , Dextran Sulfate , Disease Models, Animal , Fibrosis , Intestinal Mucosa , Mice, Knockout , Smad4 Protein , Wound Healing , Animals , Smad4 Protein/metabolism , Smad4 Protein/genetics , Mice , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Dextran Sulfate/adverse effects , Wound Healing/genetics , Colon/metabolism , Colon/pathology , Mice, Inbred C57BL , Cell Proliferation , Male , Extracellular Matrix/metabolism , Epithelial Cells/metabolism
3.
Clin Transl Med ; 14(10): e70026, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39350473

ABSTRACT

BACKGROUND: The immunoglobulin superfamily protein Trem2 (triggering receptor expressed on myeloid cells 2) is primarily expressed on myeloid cells where it functions to regulate macrophage-related immune response induction. While macrophages are essential mediators of diabetic wound healing, the specific regulatory role that Trem2 plays in this setting remains to be established. OBJECTIVE: This study was developed to explore the potential importance of Trem2 signalling in diabetic wound healing and to clarify the underlying mechanisms through which it functions. METHODS AND RESULTS: Following wound induction, diabetic model mice exhibited pronounced upregulation of Trem2 expression, which was primarily evident in macrophages. No cutaneous defects were evident in mice bearing a macrophage-specific knockout of Trem2 (T2-cKO), but they induced more pronounced inflammatory responses and failed to effectively repair cutaneous wounds, with lower levels of neovascularization, slower rates of wound closure, decreased collagen deposition following wounding. Mechanistically, we showed that interleukin (IL)-4 binds directly to Trem2, inactivating MAPK/AP-1 signalling to suppress the expression of inflammatory and chemoattractant factors. Co-culture of fibroblasts and macrophages showed that macrophages from T2-cKO mice suppressed the in vitro activation and proliferation of dermal fibroblasts through upregulation of leukaemia inhibitory factor (Lif). Injecting soluble Trem2 in vivo was also sufficient to significantly curtail inflammatory responses and to promote diabetic wound healing. CONCLUSIONS: These analyses offer novel insight into the role of IL-4/Trem2 signalling as a mediator of myeloid cell-fibroblast crosstalk that may represent a viable therapeutic target for efforts to enhance diabetic wound healing.


Subject(s)
Interleukin-4 , Membrane Glycoproteins , Receptors, Immunologic , Wound Healing , Animals , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Wound Healing/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice , Interleukin-4/metabolism , Interleukin-4/genetics , Mice, Knockout , Disease Models, Animal , Diabetes Mellitus, Experimental/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL
4.
Mol Genet Genomics ; 299(1): 89, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39317785

ABSTRACT

A burn is a type of injury to the skin or other tissues caused by heat, chemicals, electricity, sunlight, or radiation. Burn injuries have been proven to have the potential for long-term detrimental effects on the human body. The conventional therapeutic approaches are not able to effectively and easily heal these burn wounds completely. The main potential drawbacks of these treatments include hypertrophic scarring, contracture, infection, necrosis, allergic reactions, prolonged healing times, and unsatisfactory cosmetic results. The existence of these drawbacks and limitations in current treatment approaches necessitates the need to search for and develop better, more efficient therapies. The regenerative potential of microRNAs (miRNAs) and the exosomal miRNAs derived from various cell types, especially stem cells, offer advantages that outweigh traditional burn wound healing treatment procedures. The use of multiple types of stem cells is gaining interest due to their improved healing efficiency for various applications. Stem cells have several key distinguishing characteristics, including the ability to promote more effective and rapid healing of burn wounds, reduced inflammation levels at the wound site, and less scar tissue formation and fibrosis. In this review, we have discussed the stages of wound healing, the role of exosomes and miRNAs in improving thermal-induced wounds, and the impact of miRNAs in preventing the formation of hypertrophic scars. Research studies, pre-clinical and clinical, on the use of different cell-derived exosomal miRNAs and miRNAs for the treatment of thermal burns have been documented from the year 2000 up to the current time. Studies show that the use of different cell-derived exosomal miRNAs and miRNAs can improve the healing of burn wounds. The migration of exosomal miRNAs to the site of a wound leads to inhibition of apoptosis, induction of autophagy, re-epithelialization, granulation, regeneration of skin appendages, and angiogenesis. In conclusion, this study underscores the importance of integrating miRNA and exosome research into treatment strategies for burn injuries, paving the way for novel therapeutic approaches that could significantly improve patient outcomes and recovery times.


Subject(s)
Burns , Exosomes , MicroRNAs , Skin , Wound Healing , Exosomes/genetics , Exosomes/metabolism , MicroRNAs/genetics , Wound Healing/genetics , Humans , Burns/genetics , Burns/pathology , Burns/therapy , Animals , Skin/pathology , Skin/injuries , Skin/metabolism , Cicatrix/genetics , Cicatrix/pathology , Stem Cells/metabolism
5.
Int J Mol Med ; 54(5)2024 11.
Article in English | MEDLINE | ID: mdl-39301661

ABSTRACT

Diabetic foot ulcer (DFU) is a destructive complication of diabetes. Negative pressure wound therapy (NPWT) promotes DFU wound healing through an undetermined mechanism. In the present study, RNA sequencing was performed on wound granulation tissue from 3 patients with DFU before and after 1 week of NPWT. The fused in sarcoma (FUS) and interleukin enhancer binding factor 2 (ILF2) encoding RNA­binding proteins (RBPs) were screened from the sequencing data, and wound tissue samples from 24 patients with DFU were validated and analyzed before and after receiving NPWT by reverse transcription­quantitative PCR, western blotting and immunohistochemistry. In addition, in vitro and in vivo experiments were conducted to determine the effect of the expression of FUS and ILF2 on the function of human epidermal keratinocyte cells (HaCaT cells) and the healing of diabetic skin wounds. The results indicated that NPWT induced the upregulation of 101 genes and the downregulation of 98 genes in DFU wound granulation tissue. After NPWT, the expression of FUS and ILF2 was significantly upregulated (P<0.05). Pearson's correlation coefficient showed that the changes in FUS and ILF2 before and after NPWT were negatively correlated with changes in white blood cells, the neutrophil percentage, C­reactive protein, tumor necrosis factor­α, reactive oxygen species, lipid peroxides, matrix metalloproteinase (MMP) 2 and MMP9 (P<0.05), but positively correlated with the anti­inflammatory factor, IL­4 (P<0.01). There was also a positive correlation (P<0.05) with the 4­week ulcer healing rate. Additionally, the knockdown of FUS and ILF2 expression inhibited the proliferation and migration of HaCaT cells, while increasing cell apoptosis. In vivo, the knockdown of FUS and ILF2 significantly reduced the rate of skin wound healing in diabetic mice. The results of the present study therefore provide new insights into the mechanism by which NPWT promotes DFU wound healing. In conclusion, the RBPs, FUS and ILF2, promoted DFU wound healing by regulating the function of keratinocytes and reducing the inflammatory response and oxidative stress.


Subject(s)
Diabetic Foot , Negative-Pressure Wound Therapy , RNA-Binding Protein FUS , Wound Healing , Humans , Wound Healing/genetics , Diabetic Foot/therapy , Diabetic Foot/metabolism , Diabetic Foot/genetics , Diabetic Foot/pathology , Negative-Pressure Wound Therapy/methods , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Animals , Male , Mice , Middle Aged , Nuclear Factor 45 Protein/metabolism , Nuclear Factor 45 Protein/genetics , Female , Keratinocytes/metabolism , Aged
6.
Commun Biol ; 7(1): 1155, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39300285

ABSTRACT

Oral mucosal wounds exhibit accelerated healing with reduced scarring compared to cutaneous wounds, representing an optimal wound healing paradigm. However, the specific cellular subtypes orchestrating the efficient healing of mucosal tissues remain elusive. Through a comprehensive analysis integrating bulk-mRNA and single-cell sequencing data during the wound healing process in oral mucosa and skin, we have delineated a distinct set of genes markedly upregulated during tissue repair. This collection of wound healing-associated genesets was highly enriched in a specific keratinocyte subpopulation identified as STAT3-activated SPRR1B+ keratinocytes. Notably, despite the inherent rapidity of oral mucosal healing, the induction of SPRR1B+ keratinocytes is evident in both skin and mucosal wound healing processes in murine model. Intriguingly, these wound healing-promoting SPRR1B+ keratinocytes, which are induced via STAT3 activation, inherently abundant in unwounded normal mucosa but absent in normal skin. SPRR1B knockdown significantly inhibits mucosal keratinocyte migration, a critical attribute for effective wound healing. In summary, through analysis of human oral and skin wound healing processes at single-cell resolution, coupled with validation in murine model, suggests STAT3-activated SPRR1B+ keratinocytes are associated with the rapid mucosal repair process. This discovery underscores the potential application of SPRR1B+ keratinocytes in the therapeutic management of chronic or non-healing wounds.


Subject(s)
Keratinocytes , Mouth Mucosa , STAT3 Transcription Factor , Wound Healing , Animals , Humans , Male , Mice , Cell Movement , Keratinocytes/metabolism , Mice, Inbred C57BL , Mouth Mucosa/metabolism , Skin/metabolism , Skin/injuries , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Wound Healing/genetics
7.
Int J Biol Macromol ; 279(Pt 4): 135621, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39276896

ABSTRACT

Skin wound healing is a multifaceted biological process that encompasses a variety of cell types and intricate signaling pathways. Recent research has uncovered that exosomes derived from adipose stem cells, commonly referred to as ADSC exosomes, play a crucial role in facilitating the healing process. Moreover, it has been demonstrated that an anoxic, or low-oxygen, environment significantly enhances the effectiveness of these exosomes in promoting skin repair. The primary objective of this study was to investigate the underlying mechanisms through which ADSC exosomes contribute to Skin wound healing, particularly by regulating the long non-coding RNA known as NORAD under hypoxic conditions. A significant focus of our research was to examine the interplay between the microRNA miR-524-5p and the Pumilio protein, as we aimed to understand how these molecular interactions might influence the overall healing process. In this study, ADSC exosomes were extracted by simulating hypoxia in vitro and their effects on the proliferation and migration of skin fibroblasts (FB) were evaluated. The expression levels of NORAD, miR-524-5p and Pumilio were analyzed by fluorescence quantitative PCR. Pumilio protein was silenced by siRNA technique to evaluate its role in ADSC exosome-mediated wound healing. The experimental results showed that under hypoxia conditions, NORAD levels in ADSC exosomes increased significantly and could effectively regulate the expression of miR-524-5p. After Pumilio protein silencing, the proliferation and migration ability of fibroblasts were significantly reduced, indicating that Pumilio protein played a role in the process of wound healing. By inhibiting miR-524-5p, the expression of Pumilio protein was restored, further confirming its regulatory mechanism.


Subject(s)
Adipose Tissue , Exosomes , MicroRNAs , RNA, Long Noncoding , RNA-Binding Proteins , Skin , Wound Healing , MicroRNAs/genetics , MicroRNAs/metabolism , Wound Healing/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Exosomes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Skin/metabolism , Cell Proliferation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Cell Movement/genetics , Cell Hypoxia/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Fibroblasts/metabolism , Signal Transduction
8.
BMC Biol ; 22(1): 193, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256768

ABSTRACT

BACKGROUND: Foxn1-/- deficient mice are a rare model of regenerative skin wound healing among mammals. In wounded skin, the transcription factor Foxn1 interacting with hypoxia-regulated factors affects re-epithelialization, epithelial-mesenchymal transition (EMT) and dermal white adipose tissue (dWAT) reestablishment and is thus a factor regulating scar-forming/reparative healing. Here, we hypothesized that transcriptional crosstalk between Foxn1 and Hif-1α controls the switch from scarless (regenerative) to scar-present (reparative) skin wound healing. To verify this hypothesis, we examined (i) the effect of hypoxia/normoxia and Foxn1 signalling on the proteomic signature of Foxn1-/- (regenerative) dermal fibroblasts (DFs) and then (ii) explored the effect of Hif-1α or Foxn1/Hif-1α introduced by a lentiviral (LV) delivery vector to injured skin of regenerative Foxn1-/- mice with particular attention to the remodelling phase of healing. RESULTS: We showed that hypoxic conditions and Foxn1 stimulation modified the proteome of Foxn1-/- DFs. Hypoxic conditions upregulated DF protein profiles, particularly those related to extracellular matrix (ECM) composition: plasminogen activator inhibitor-1 (Pai-1), Sdc4, Plod2, Plod1, Lox, Loxl2, Itga2, Vldlr, Ftl1, Vegfa, Hmox1, Fth1, and F3. We found that Pai-1 was stimulated by hypoxic conditions in regenerative Foxn1-/- DFs but was released by DFs to the culture media exclusively upon hypoxia and Foxn1 stimulation. We also found higher levels of Pai-1 protein in DFs isolated from Foxn1+/+ mice (reparative/scar-forming) than in DFs isolated from Foxn1-/- (regenerative/scarless) mice and triggered by injury increase in Foxn1 and Pai-1 protein in the skin of mice with active Foxn1 (Foxn1+/+ mice). Then, we demonstrated that the introduction of Foxn1 and Hif-1α via lentiviral injection into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing by increasing the wounded skin area and decreasing hyaluronic acid deposition and the collagen type III to I ratio. We also identified a stimulatory effect of LV-Foxn1 + LV-Hif-1α injection in the wounded skin of Foxn1-/- mice on Pai-1 protein levels. CONCLUSIONS: The present data highlight the effect of hypoxia and Foxn1 on the protein profile and functionality of regenerative Foxn1-/- DFs and demonstrate that the introduction of Foxn1 and Hif-1α into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing.


Subject(s)
Cicatrix , Fibroblasts , Forkhead Transcription Factors , Wound Healing , Animals , Wound Healing/physiology , Wound Healing/genetics , Fibroblasts/metabolism , Mice , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Cicatrix/metabolism , Skin/metabolism , Skin/injuries , Mice, Knockout , Proteome/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Proteomics/methods , Hypoxia/metabolism
9.
Invest Ophthalmol Vis Sci ; 65(11): 43, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39330987

ABSTRACT

Purpose: Corneal wounding healing is critical for maintaining clear vision, however, a complete understanding of its dynamic regulatory mechanisms remains elusive. Here, we used single-cell RNA sequencing (scRNA-seq) to analyze the cellular activities and transcriptional changes of corneal limbal epithelial cells at different stages after wound healing in cynomolgus monkeys, which exhibit a closer transcriptomic similarity to humans. Methods: Corneal limbal tissues were collected during uninjured, 1-day and 3-day healing stages, dissociated into single cells, and subjected to scRNA-seq using the 10× Genomics platform. Cell types were clustered by graph-based visualization methods and unbiased computational analysis. Additionally, cell migration assays and immunofluorescent staining were performed on cultured human corneal epithelial cells. Results: We characterized nine cell clusters by scRNA-seq analysis of the cynomolgus monkey corneal epithelium. By comparing heterogeneous transcriptional changes in major cell types during corneal healing, we highlighted the importance of limbal epithelial cells (LEPCs) and basal epithelial cells (BEPCs) in extracellular matrix (ECM) formation and wound healing, as well as suprabasal epithelial cells (SEPCs) in epithelial differentiation during the healing processes. We further identified five different sub-clusters in LEPC, including the transit amplifying cell (TAC) sub-cluster that promotes early healing through the activation of thrombospondin-1 (THBS1) expression. Conclusions: Our study represents the first comprehensive exploration of the detailed transcriptome profile of individual corneal cells during the wound healing process in nonhuman primates. We demonstrate the intricate mechanisms involved in corneal healing and provide a promising avenue for potential therapies in corneal wound healing.


Subject(s)
Epithelium, Corneal , Macaca fascicularis , Single-Cell Analysis , Transcriptome , Wound Healing , Animals , Wound Healing/physiology , Wound Healing/genetics , Epithelium, Corneal/metabolism , Corneal Injuries/metabolism , Corneal Injuries/genetics , Cell Movement/physiology , Gene Expression Profiling , Cells, Cultured , Disease Models, Animal , Humans , Limbus Corneae/cytology , Limbus Corneae/metabolism , Male
10.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337611

ABSTRACT

Treating diabetic wounds effectively remains a significant clinical challenge. Emerging studies suggest that microRNAs (miRNAs) play crucial roles in various physiological and pathological processes and hold promise as therapeutic tools. This study investigates the miRNA expression profile in keratinocytes using a cell model of diabetic wounds. Microarray analysis revealed that 43 miRNAs from wounded keratinocytes incubated under diabetic conditions (high glucose/hypoxia) exhibited a two-fold change in expression compared to those incubated under normal conditions (low glucose/normoxia). Quantitative RT-PCR confirmed significant differences in the expression of eight miRNAs, with miR-3138 and miR-3679-5p being further analyzed for their roles in keratinocyte migration. Transfection with a miR-3138 mimic and a miR-3679-5p inhibitor indicated that upregulation of miR-3138 and downregulation of miR-3679-5p enhance keratinocyte migration in both normal and diabetic wounds. Pathway and gene ontology (GO) analyses identified potential pathways and functional annotations associated with miR-3138 and miR-3679-5p in diabetic wound healing. Potential human gene targets of miR-3138 and miR-3679-5p were predicted using a three-way comparison of the TargetScan, miRDB, and DIANA databases. This study elucidates the miRNA expression signature of human keratinocytes in a diabetes-like environment, providing deeper insights into the pathogenesis of diabetic wounds.


Subject(s)
Cell Movement , Keratinocytes , MicroRNAs , Wound Healing , Keratinocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Wound Healing/genetics , Cell Movement/genetics , Gene Expression Profiling , Gene Expression Regulation , Diabetes Mellitus/metabolism , Diabetes Mellitus/genetics , Gene Ontology
11.
Int J Biol Sci ; 20(11): 4532-4550, 2024.
Article in English | MEDLINE | ID: mdl-39247808

ABSTRACT

Adenosine receptor-mediated signaling, especially adenosine A2A receptor (A2AR) signaling, has been implicated in wound healing. However, the role of endothelial cells (ECs) in A2AR-mediated wound healing and the mechanism underlying this effect are still unclear. Here, we showed that the expression of A2AR substantially increased after wounding and was especially prominent in granulation tissue. The delaying effects of A2AR knockout (KO) on wound healing are due mainly to the effect of A2AR on endothelial cells, as shown with A2AR-KO and EC-A2AR-KO mice. Moreover, the expression of c-Ski, which is especially prominent in CD31-positive cells in granulation tissue, increased after wounding and was decreased by both EC-A2AR KO and A2AR KO. In human microvascular ECs (HMECs), A2AR activation induced EC proliferation, migration, tubule formation and c-Ski expression, whereas c-Ski depletion by RNAi abolished these effects. Mechanistically, A2AR activation promotes the expression of c-Ski through an ERK/CREB-dependent pathway. Thus, A2AR-mediated angiogenesis plays a critical role in wound healing, and c-Ski is involved mainly in the regulation of angiogenesis by A2AR via the ERK/CREB pathway. These findings identify A2AR as a therapeutic target in wound repair and other angiogenesis-dependent tissue repair processes.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Mice, Knockout , Receptor, Adenosine A2A , Wound Healing , Wound Healing/physiology , Wound Healing/genetics , Animals , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/genetics , Mice , Humans , Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Signal Transduction , MAP Kinase Signaling System/physiology , Cell Proliferation/genetics , Cell Movement/genetics , Angiogenesis
12.
PLoS Genet ; 20(9): e1011387, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226333

ABSTRACT

A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.


Subject(s)
DNA Damage , Drosophila melanogaster , Wings, Animal , Animals , Wings, Animal/growth & development , Wings, Animal/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Cell Proliferation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Apoptosis , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Wound Healing/genetics , Cellular Senescence , MAP Kinase Signaling System , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Cell Cycle
13.
Invest Ophthalmol Vis Sci ; 65(11): 13, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39240550

ABSTRACT

Purpose: Transdifferentiation of corneal fibroblasts to myofibroblasts in the stroma is a central mechanistic event in corneal wound healing. This study sought to characterize genes and pathways influencing transdifferentiation of human corneal fibroblasts (hCSFs) to human corneal myofibroblasts (hCMFs) using RNA sequencing (RNA-seq) to develop comprehensive mechanistic information and identify newer targets for corneal fibrosis management. Methods: Primary hCSFs were derived from donor human corneas. hCMFs were generated by treating primary hCSFs with transforming growth factor ß1 (TGFß1; 5 ng/mL) for 72 hours under serum-free conditions. RNA was extracted using the RNeasy Plus Mini Kit and subjected to RNA-seq analysis after quality control testing. Differential gene expression, pathway enrichment, and protein-protein network analyses were performed using DESeq2, GSEA/PANTHER/Reactome, and Cytoscape/cytoHubba, respectively. Results: RNA-seq analysis of hCMFs and hCSFs identified 3843 differentially expressed genes and transcripts (adjusted P < 0.05). The log(fold change) ≥ ±1.5 filter showed 816 upregulated and 739 downregulated genes between two cell types. Pathway enrichment analysis showed the highest normalized enrichment score for epithelial-to-mesenchymal transition (5.569), followed by mTORC1 signaling (2.949), angiogenesis (2.176), and TGFß signaling (2.008). Protein-protein interaction network analysis identified the top 20 nodes influencing corneal myofibroblast development. The expression of a novel MXRA5 in corneal stroma and its association with corneal fibrosis was verified by real-time quantitative reverse transcription PCR and immunofluorescence. RNA-seq and gene count files were submitted to the NCBI Gene Expression Omnibus (GSE260476). Conclusions: This study identified several novel genes involved in myofibroblast development, offering potential targets for developing newer therapeutic strategies for corneal fibrosis.


Subject(s)
Myofibroblasts , RNA-Seq , Wound Healing , Humans , Wound Healing/genetics , Myofibroblasts/metabolism , Cells, Cultured , Fibroblasts/metabolism , Cell Transdifferentiation/genetics , Gene Expression Regulation , Cornea/metabolism , Cornea/pathology , Corneal Injuries/genetics , Corneal Injuries/metabolism , Corneal Injuries/pathology , Transforming Growth Factor beta1/genetics , Signal Transduction , Sequence Analysis, RNA , Male , Female
14.
Mol Biol Rep ; 51(1): 954, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39230578

ABSTRACT

BACKGROUND: Leukocyte- and platelet-rich fibrin (L-PRF) is an autologous platelet concentrate, prepared by centrifugation of blood and consisting of a dense fibrin network with incorporated leukocytes and platelets. This study aims to perform an in-depth analysis of the cells, growth factors, and transcriptome of L-PRF. METHODS AND RESULTS: Fresh, 1 week and 2 weeks cultured human L-PRF membranes and liquid L-PRF glue were characterized on cellular and transcriptional level using flow cytometry (n = 4), single-cell RNA sequencing (n = 5) and RT-qPCR. Growth factor kinetics were investigated using ELISA (EGF, VEGF, PDGF-AB, TGF-ß1, bFGF). L-PRF contained a large number of viable cells (fresh 97.14 ± 1.09%, 1 week cultured 93.57 ± 1.68%), mainly granulocytes in fresh samples (53.9 ± 19.86%) and T cells in cultured samples (84.7 ± 6.1%), confirmed with scRNA-seq. Monocytes differentiate to macrophages during 1 week incubation. Specifically arterial L-PRF membranes were found to release significant amounts of VEGF, EGF, PDGF-AB and TGF-ß1. CONCLUSION: We characterized L-PRF using in vitro experiments, to obtain an insight in the composition of the material including a possible mechanistic role for tissue healing. This was the first study characterizing L-PRF at a combined cellular, proteomic, and transcriptional level.


Subject(s)
Intercellular Signaling Peptides and Proteins , Leukocytes , Platelet-Rich Fibrin , Transcriptome , Humans , Platelet-Rich Fibrin/metabolism , Leukocytes/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Transcriptome/genetics , Blood Platelets/metabolism , Wound Healing/genetics , Kinetics , Cells, Cultured
15.
Arch Biochem Biophys ; 760: 110134, 2024 10.
Article in English | MEDLINE | ID: mdl-39181381

ABSTRACT

Skin wound is an emerging health challenge on account of the high-frequency trauma, surgery and chronic refractory ulcer. Further study on the disease biology will help to develop new effective approaches for wound healing. Here, we identified a wound-stress responsive gene, activating transcription factor 3 (ATF3), and then investigated its biological action and mechanism in wound healing. In the full-thickness skin wound model, ATF3 was found to promote fibroblast activation and collagen production, resulted in accelerated wound healing. Mechanically, ATF3 transcriptionally activated TGF-ß receptor Ⅱ via directly binding to its specific promoter motif, followed by the enhanced TGF-ß/Smad pathway in fibroblasts. Moreover, the increased ATF3 upon skin injury was partly resulted from hypoxia stimulation with Hif-1α dependent manner. Altogether, this work gives novel insights into the biology and mechanism of stress-responsive gene ATF3 in wound healing, and provides a potential therapeutic target for treatment.


Subject(s)
Activating Transcription Factor 3 , Collagen , Fibroblasts , Skin , Wound Healing , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Wound Healing/genetics , Fibroblasts/metabolism , Animals , Collagen/metabolism , Skin/metabolism , Skin/injuries , Skin/pathology , Mice , Humans , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Promoter Regions, Genetic , Male , Transcriptional Activation , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Transcription, Genetic
16.
Development ; 151(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39177163

ABSTRACT

One of the key tissue movements driving closure of a wound is re-epithelialisation. Earlier wound healing studies describe the dynamic cell behaviours that contribute to wound re-epithelialisation, including cell division, cell shape changes and cell migration, as well as the signals that might regulate these cell behaviours. Here, we have used a series of deep learning tools to quantify the contributions of each of these cell behaviours from movies of repairing wounds in the Drosophila pupal wing epithelium. We test how each is altered after knockdown of the conserved wound repair signals Ca2+ and JNK, as well as after ablation of macrophages that supply growth factor signals believed to orchestrate aspects of the repair process. Our genetic perturbation experiments provide quantifiable insights regarding how these wound signals impact cell behaviours. We find that Ca2+ signalling is a master regulator required for all contributing cell behaviours; JNK signalling primarily drives cell shape changes and divisions, whereas signals from macrophages largely regulate cell migration and proliferation. Our studies show deep learning to be a valuable tool for unravelling complex signalling hierarchies underlying tissue repair.


Subject(s)
Cell Movement , Deep Learning , Signal Transduction , Wings, Animal , Wound Healing , Animals , Cell Movement/genetics , Wound Healing/physiology , Wound Healing/genetics , Wings, Animal/metabolism , Re-Epithelialization , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Pupa/metabolism , Macrophages/metabolism , Cell Proliferation , Calcium Signaling , Cell Shape , Epithelium/metabolism
18.
Nat Cardiovasc Res ; 3(6): 685-700, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39196227

ABSTRACT

Sterile inflammation after injury is important for tissue restoration. In injured human and mouse tissues, macrophages were recently found to accumulate perivascularly. This study investigates if macrophages adopt a mural cell phenotype important for restoration after ischemic injury. Single-cell RNA sequencing of fate-mapped macrophages from ischemic mouse muscles demonstrates a macrophage-toward-mural cell switch of a subpopulation of macrophages with downregulated myeloid cell genes and upregulated mural cell genes, including PDGFRß. This observation was further strengthened when including unspliced transcripts in the analysis. The macrophage switch was proven functionally relevant, as induction of macrophage-specific PDGFRß deficiency prevented their perivascular macrophage phenotype, impaired vessel maturation and increased vessel leakiness, which ultimately reduced limb function. In conclusion, macrophages in adult ischemic tissue were demonstrated to undergo a cellular program to morphologically, transcriptomically and functionally resemble mural cells while weakening their macrophage identity. The macrophage-to-mural cell-like phenotypic switch is crucial for restoring tissue function and warrants further exploration as a potential target for immunotherapies to enhance healing.


Subject(s)
Disease Models, Animal , Ischemia , Macrophages , Animals , Macrophages/metabolism , Macrophages/immunology , Ischemia/metabolism , Ischemia/pathology , Ischemia/genetics , Phenotype , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/injuries , Wound Healing/genetics , Wound Healing/physiology , Mice, Inbred C57BL , Mice , Male , Hindlimb/blood supply , Neovascularization, Physiologic/genetics , Up-Regulation , Transcriptome , Single-Cell Analysis , Biomarkers/metabolism , Recovery of Function , Mice, Knockout
19.
Fish Shellfish Immunol ; 152: 109794, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089638

ABSTRACT

To ensure welfare-friendly and effective internal tagging, the tagging process should not cause a long-term burden on individuals given that tagged fish serve as representatives for the entire population in telemetry applications. To some extent, stress is inevitable within regular aquaculture practices, and thus, the consequences of long-term stress should be described in terms of their effects on internal tagging. In fish, stressors activate the Hypothalamus-Pituitary-Interrenal (HPI) and Brain-Sympathetic-Chromaffin Cell (BSC) axes, leading to neuroimmunoendocrine communication and paracrine interactions among stress hormones. The interrelation between wound healing and stress is complex, owing to their shared components, pathways, and energy demands. This study assessed 14 genes (mmp9, mmp13, il-2, il-4, il-8a, il-10, il-12, il-17d, il-1b, tnfa, ifng, leg-3, igm, and crh) in the skin (1.5 cm from the wound) and head kidney over eight weeks. These genes, associated with cell signaling in immunity, wound healing, and stress, have previously been identified as influenced and regulated by these processes. Half of a group of Atlantic salmon (n = 90) with surgically implanted dummy smart-tags were exposed to daily crowding stress. The goal was to investigate how this gene panel responds to a wound alone and then to the combined effects of wounding and daily crowding stress. Our observations indicate that chronic stress impacts inflammation and impedes wound healing, as seen through the expression of matrix metalloproteinases genes in the skin but not in the head kidney. This difference is likely due to the ongoing internal wound repair, in contrast to the externally healed wound incision. Cytokine expression, when significant in the skin, was mainly downregulated in both treatments compared to control values, particularly in the study's first half. Conversely, the head kidney showed initial cytokine downregulation followed by upregulation. Across all weeks observed and combining both tissues, the significantly expressed gene differences were 12 % between the Wound and Stress+ groups, 28 % between Wound and Control, and 25 % between Stress+ and Control. Despite significant fluctuations in cytokines, sustained variations across multiple weeks are only evident in a few select genes. Furthermore, Stress+ individuals demonstrated the most cytokine correlations within the head kidney, which may suggest that chronic stress affects cytokine expression. This investigation unveils that the presence of stress and prolonged activation of the HPI axis in an eight weeklong study has limited yet detectable effects on the selected gene expression within immunity, wound healing, and stress, with notable tissue-specific differences.


Subject(s)
Head Kidney , Salmo salar , Skin , Stress, Physiological , Animals , Head Kidney/immunology , Head Kidney/metabolism , Salmo salar/genetics , Salmo salar/immunology , Skin/immunology , Crowding , Fish Proteins/genetics , Gene Expression Regulation/immunology , Gene Expression , Wound Healing/genetics
20.
PLoS One ; 19(7): e0306248, 2024.
Article in English | MEDLINE | ID: mdl-38950058

ABSTRACT

Diabetic foot ulcers (DFUs) pose a significant challenge in diabetes care. Yet, a comprehensive understanding of the underlying biological disparities between healing and non-healing DFUs remains elusive. We conducted bioinformatics analysis of publicly available transcriptome sequencing data in an attempt to elucidate these differences. Our analysis encompassed differential analysis to unveil shifts in cell composition and gene expression profiles between non-healing and healing DFUs. Cell communication alterations were explored employing the Cellchat R package. Pseudotime analysis and cytoTRACE allowed us to dissect the heterogeneity within fibroblast subpopulations. Our findings unveiled disruptions in various cell types, localized low-grade inflammation, compromised systemic antigen processing and presentation, and extensive extracellular matrix signaling disarray in non-healing DFU patients. Some of these anomalies partially reverted in healing DFUs, particularly within the abnormal ECM-receptor signaling pathway. Furthermore, we distinguished distinct fibroblast subpopulations in non-healing and healing DFUs, each with unique biological functions. Healing-associated fibroblasts exhibited heightened extracellular matrix (ECM) remodeling and a robust wound healing response, while non-healing-associated fibroblasts showed signs of cellular senescence and complement activation, among other characteristics. This analysis offers profound insights into the wound healing microenvironment, identifies pivotal cell types for DFU healing promotion, and reveals potential therapeutic targets for DFU management.


Subject(s)
Diabetic Foot , Fibroblasts , Single-Cell Analysis , Transcriptome , Wound Healing , Diabetic Foot/genetics , Diabetic Foot/pathology , Diabetic Foot/metabolism , Humans , Wound Healing/genetics , Single-Cell Analysis/methods , Fibroblasts/metabolism , Fibroblasts/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/genetics , Gene Expression Profiling , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL