Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.841
Filter
1.
Biomaterials ; 312: 122711, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088911

ABSTRACT

The unsuitable deformation stimulus, harsh urine environment, and lack of a regenerative microenvironment (RME) prevent scaffold-based urethral repair and ultimately lead to irreversible urethral scarring. The researchers clarify the optimal elastic modulus of the urethral scaffolds for urethral repair and design a multilayered PVA hydrogel scaffold for urethral scar-free healing. The inner layer of the scaffold has self-healing properties, which ensures that the wound effectively resists harsh urine erosion, even when subjected to sutures. In addition, the scaffold's outer layer has an extracellular matrix-like structure that synergizes with adipose-derived stem cells to create a favorable RME. In vivo experiments confirm successful urethral scar-free healing using the PVA multilayered hydrogel scaffold. Further mechanistic study shows that the PVA multilayer hydrogel effectively resists the urine-induced inflammatory response and accelerates the transition of urethral wound healing to the proliferative phase by regulating macrophage polarization, thus providing favorable conditions for urethral scar-free healing. This study provides mechanical criteria for the fabrication of urethral tissue-engineered scaffolds, as well as important insights into their design.


Subject(s)
Elastic Modulus , Hydrogels , Tissue Scaffolds , Urethra , Wound Healing , Tissue Scaffolds/chemistry , Animals , Hydrogels/chemistry , Tissue Engineering/methods , Mice , Regeneration , Cicatrix/pathology , Male , Cellular Microenvironment , Rats, Sprague-Dawley , Stem Cells/cytology
2.
Cell Commun Signal ; 22(1): 467, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350161

ABSTRACT

Traumatic brain injury (TBI) is an acquired insult to the brain caused by an external mechanical force, potentially resulting in temporary or permanent impairment. Microglia, the resident immune cells of the central nervous system, are activated in response to TBI, participating in tissue repair process. However, the underlying epigenetic mechanisms in microglia during TBI remain poorly understood. ARID1A (AT-Rich Interaction Domain 1 A), a pivotal subunit of the multi-protein SWI/SNF chromatin remodeling complex, has received little attention in microglia, especially in the context of brain injury. In this study, we generated a Arid1a cKO mouse line to investigate the potential roles of ARID1A in microglia in response to TBI. We found that glial scar formation was exacerbated due to increased microglial migration and a heightened inflammatory response in Arid1a cKO mice following TBI. Mechanistically, loss of ARID1A led to an up-regulation of the chemokine CCL5 in microglia upon the injury, while the CCL5-neutralizing antibody reduced migration and inflammatory response of LPS-stimulated Arid1a cKO microglia. Importantly, administration of auraptene (AUR), an inhibitor of CCL5, repressed the microglial migration and inflammatory response, as well as the glial scar formation after TBI. These findings suggest that ARID1A is critical for microglial response to injury and that AUR has a therapeutic potential for the treatment of TBI.


Subject(s)
Brain Injuries, Traumatic , Chemokine CCL5 , DNA-Binding Proteins , Mice, Knockout , Microglia , Transcription Factors , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/genetics , Microglia/metabolism , Microglia/pathology , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Movement , Cicatrix/pathology , Cicatrix/metabolism , Mice, Inbred C57BL , Male
3.
ACS Appl Mater Interfaces ; 16(39): 53042-53059, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39298643

ABSTRACT

Pathological scarring has been a challenge in skin injury repair since ancient times, and prophylactic treatment in the early stages of wound healing usually results in delayed wound healing. In this study, poly(ethylene oxide) (PEO) and chitosan (CTS) were used as carrier materials to construct multifunctional pirfenidone (PFD)/CTS/PEO (PCP) nanofiber membranes (NFMs) loaded with PFD by microfluidic blow-spinning (MBS). MBS is a good method for quickly, safely, and greenly constructing large-area manufacturing of inexpensive NFMs. PCP NFMs were uniform in external morphology, with diameters ranging from 200 to 500 nm. The encapsulation efficiency of the drug-loaded PCP NFMs was above 80%, which had a good slow release, visualization, water absorption, and biocompatibility. The inhibitory effect of PCP NFMs on normal human dermal fibroblasts was dose-dependent and inhibited the expression of the transforming growth factor-ß1/SMAD family member 3 (TGF-ß1/SMAD3) signaling pathway. PCP NFMs showed significant antibacterial effects against both Staphylococcus aureus and Escherichia coli. In the rabbit ear scar experiment, the wound healed about 70% on day 5 and almost completely on day 10 after PCP-3 NFMs treatment, with the thinnest scar tissue, skin color, tenderness close to normal tissue, and a Vancouver scar scale score of less than 5. PCP-3 NFMs had good effects on anti-inflammatory, wound healing, and collagen-I deposition reducing effects. In conclusion, PCP-3 NFMs can both promote wound healing and intervene to inhibit pathological scarring in advance, making them a potential multifunctional wound dressing for early prevention and treatment of pathological scarring.


Subject(s)
Anti-Bacterial Agents , Chitosan , Cicatrix , Nanofibers , Staphylococcus aureus , Wound Healing , Nanofibers/chemistry , Animals , Rabbits , Humans , Cicatrix/prevention & control , Cicatrix/pathology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Chitosan/chemistry , Chitosan/pharmacology , Wound Healing/drug effects , Pyridones/chemistry , Pyridones/pharmacology , Escherichia coli/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Polyethylene Glycols/chemistry , Transforming Growth Factor beta1/metabolism , Membranes, Artificial , Microfluidics/methods
4.
Cells ; 13(18)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39329731

ABSTRACT

Spinal cord injury (SCI) often results in permanent loss of motor and sensory function. After SCI, the blood-spinal cord barrier (BSCB) is disrupted, causing the infiltration of neutrophils and macrophages, which secrete several kinds of cytokines, as well as matrix metalloproteinases (MMPs). MMPs are proteases capable of degrading various extracellular matrix (ECM) proteins, as well as many non-matrix substrates. The tissue inhibitor of MMPs (TIMP)-1 is significantly upregulated post-SCI and operates via MMP-dependent and MMP-independent pathways. Through the MMP-dependent pathway, TIMP-1 directly reduces inflammation and destruction of the ECM by binding and blocking the catalytic domains of MMPs. Thus, TIMP-1 helps preserve the BSCB and reduces immune cell infiltration. The MMP-independent pathway involves TIMP-1's cytokine-like functions, in which it binds specific TIMP surface receptors. Through receptor binding, TIMP-1 can stimulate the proliferation of several types of cells, including keratinocytes, aortic smooth muscle cells, skin epithelial cells, corneal epithelial cells, and astrocytes. TIMP-1 induces astrocyte proliferation, modulates microglia activation, and increases myelination and neurite extension in the central nervous system (CNS). In addition, TIMP-1 also regulates apoptosis and promotes cell survival through direct signaling. This review provides a comprehensive assessment of TIMP-1, specifically regarding its contribution to inflammation, ECM remodeling, and scar formation after SCI.


Subject(s)
Cicatrix , Spinal Cord Injuries , Tissue Inhibitor of Metalloproteinases , Spinal Cord Injuries/pathology , Spinal Cord Injuries/metabolism , Humans , Animals , Cicatrix/pathology , Cicatrix/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism
5.
PLoS One ; 19(9): e0311147, 2024.
Article in English | MEDLINE | ID: mdl-39331598

ABSTRACT

Scar tissue formation following skin wound healing is a challenging public health problem. Skin regeneration and preventing the formation of scar tissue by currently available commercial products are largely ineffective. This study aimed to test the efficacy of a novel topical metformin lotion (ML) in inhibiting scar tissue formation during skin wound healing in rats and to determine the mechanisms of action involved. A 6% ML was prepared in our laboratory. A skin wound healing model in rats was used. The wounded rats were divided into two groups and treated daily for 10 days as follows: Group 1 received a daily application of 50 mg of control lotion, or 0% ML (totaling 100 mg of lotion per rat), and Group 2 received a daily application of 50 mg of 6% ML (totaling 100 mg of 6% ML per rat). Blood samples from the heart of each rat were analyzed for inflammatory markers, HMGB1 and IL-1ß, using ELISA, and immunological and histological analyses were performed on skin tissue sections. ML decreased levels of inflammatory markers HMGB1 and IL-1ß in the serum of rats and inhibited the release of HMGB1 from cell nuclei into the skin tissue matrix. Additionally, ML demonstrated anti-fibrotic properties by enhancing AMPK activity, decreasing the expression of TGF-ß1, reducing the number of myofibroblasts, decreasing the production of collagen III, and increasing the expression of collagen I. ML promotes the regeneration of high-quality skin during wound healing by reducing scar tissue formation. This effect is mediated through the activation of AMPK, inhibition of TGF-ß1, and a decrease in the number of myofibroblasts.


Subject(s)
AMP-Activated Protein Kinases , Cicatrix , Metformin , Myofibroblasts , Skin , Transforming Growth Factor beta1 , Wound Healing , Animals , Metformin/pharmacology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , Rats , Wound Healing/drug effects , AMP-Activated Protein Kinases/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Cicatrix/prevention & control , Cicatrix/pathology , Cicatrix/drug therapy , Cicatrix/metabolism , Male , Interleukin-1beta/metabolism , HMGB1 Protein/metabolism , Rats, Sprague-Dawley
6.
Nat Commun ; 15(1): 8379, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333481

ABSTRACT

Scar tissue formation is a hallmark of wound repair in adults and can chronically affect tissue architecture and function. To understand the general phenomena, we sought to explore scar-driven imbalance in tissue homeostasis caused by a common, and standardized surgical procedure, the uterine scar due to cesarean surgery. Deep uterine scar is associated with a rapidly increasing condition in pregnant women, placenta accreta spectrum (PAS), characterized by aggressive trophoblast invasion into the uterus, frequently necessitating hysterectomy at parturition. We created a model of uterine scar, recapitulating PAS-like invasive phenotype, showing that scar matrix activates mechanosensitive ion channel, Piezo1, through glycolysis-fueled cellular contraction. Piezo1 activation increases intracellular calcium activity and Protein kinase C activation, leading to NF-κB nuclear translocation, and MafG stabilization. This inflammatory transformation of decidua leads to production of IL-8 and G-CSF, chemotactically recruiting invading trophoblasts towards scar, initiating PAS. Our study demonstrates aberrant mechanics of scar disturbs stroma-epithelia homeostasis in placentation, with implications in cancer dissemination.


Subject(s)
Cicatrix , Inflammation , Ion Channels , Placenta Accreta , Trophoblasts , Female , Pregnancy , Humans , Placenta Accreta/metabolism , Placenta Accreta/pathology , Cicatrix/metabolism , Cicatrix/pathology , Ion Channels/metabolism , Ion Channels/genetics , Animals , Inflammation/metabolism , Inflammation/pathology , Trophoblasts/metabolism , Trophoblasts/pathology , Decidua/pathology , Decidua/metabolism , Mice , NF-kappa B/metabolism , Cesarean Section/adverse effects , Protein Kinase C/metabolism , Protein Kinase C/genetics , Interleukin-8/metabolism , Uterus/pathology , Uterus/metabolism
7.
J Med Life ; 17(6): 582-587, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39296433

ABSTRACT

Follicular unit hair extraction (FUE) is effective for hair restoration but is less successful on scarred tissue due to reduced vascularity and altered tissue architecture. Stem cell therapy can enhance tissue regeneration, possibly improving FUE outcomes on scarred tissue. This study investigated the impact of stem cell therapy prior to FUE on scarred tissue. Sixty patients with scalp scars from trauma or previous surgeries were divided into two groups. Group A (n = 30) received autologous stem cell therapy followed by FUE, while Group B (n = 30) underwent FUE without prior stem cell treatment. Autologous stem cells were harvested from patients' adipose tissue and injected into the scarred area four weeks before FUE. Outcomes were assessed at 3-, 6-, and 12-months post-transplantation, focusing on hair density, graft survival rate, and patient satisfaction. Histological examinations evaluated tissue regeneration. Group A showed significantly higher hair density (mean increase of 45%) and graft survival rates (87%) compared to Group B (mean increase of 25%, graft survival rate of 60%) at all follow-up points (P < 0.05). Histological analysis revealed enhanced neovascularization and reduced fibrosis in the stem cell-treated group, with 70% more new blood vessels and 50% less fibrotic tissue compared to the control group. Patient satisfaction scores were higher in Group A (average score of 8.5 out of 10) versus Group B (6.0), indicating better aesthetic outcomes and reduced scar visibility. Pre-treatment with autologous stem cell therapy significantly improved FUE effectiveness on scarred tissue, enhancing graft survival, hair density, and patient satisfaction. Further research is recommended to optimize this therapeutic strategy.


Subject(s)
Cicatrix , Hair Follicle , Stem Cell Transplantation , Humans , Cicatrix/therapy , Cicatrix/pathology , Hair Follicle/transplantation , Female , Adult , Stem Cell Transplantation/methods , Male , Middle Aged , Transplantation, Autologous , Alopecia/therapy , Scalp , Hair/transplantation , Young Adult
8.
Arch Dermatol Res ; 316(8): 600, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225836

ABSTRACT

Wounds are most commonly caused by accidents, surgery and burns, and can be internal or external. Naturally, the wound healing process can take a long time and lead to scarring. In this study we present a technique to shorten wound healing time and prevent or mitigate scarring using D-mannose that is applied directly on the wound. The results showed that the healing time is almost halved compared to treatment with povidone-iodine solution which is an antiseptic widely used in surgery. D-Mannose is neither an antisepsis nor an antibiotic. We propose a posssible mechanism by which D-mannose binds to D-mannose binding lectin and immediately activates the innate immune system that ultimately phagocytizes pathogens and clears the wound of degraded cells and materials, which reduces inflammation and implicitly wound healing time. D-Mannose also intervenes in the coagulation process by binding to fibrinogen, generating a finer and denser fibrin, which visibly reduces collagen scars. Our findings show that applying D-mannose directly on the wound as a powder shortens wound healing time and visibly attenuates scarring. Apart from the unaesthetic appearance, these scars can also cause a certain tissue dysfunction, regardless of the affected organ.


Subject(s)
Cicatrix , Mannose-Binding Lectin , Mannose , Wound Healing , Wound Healing/drug effects , Cicatrix/pathology , Cicatrix/prevention & control , Mannose-Binding Lectin/metabolism , Animals , Humans , Male , Skin/pathology , Skin/drug effects , Skin/metabolism , Female , Immunity, Innate/drug effects
9.
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
10.
ACS Appl Mater Interfaces ; 16(38): 50305-50320, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39255049

ABSTRACT

Excessive fibrotic scar formation during skin defect repair poses a formidable challenge, impeding the simultaneous acceleration of wound healing and prevention of scar formation and hindering the restoration of skin integrity and functionality. Drawing inspiration from the structural, compositional, and biological attributes of skin, we developed a hydrogel containing modified recombinant human collagen type III and thiolated hyaluronic acid to address the challenges of regenerating skin appendages and improving the recovery of skin functions after injury by reducing fibrotic scarring. The hydrogel displayed favorable biocompatibility, antioxidant properties, angiogenic potential, and fibroblast migration stimulation in vitro. In a rat full-layer defect model, it reduced inflammation, promoted microvascular formation, and significantly enhanced the wound healing speed and effectiveness. Additionally, by upregulating fibrosis-associated genes, such as TGFB1, it facilitated collagen accumulation and a beneficial balance between type I and type III collagen, potentially expediting skin regeneration and functional recovery. In conclusion, the utilization of rhCol III-HS demonstrated considerable potential as a wound dressing, offering a highly effective strategy for the restoration and rejuvenation of complete skin defects.


Subject(s)
Cicatrix , Collagen Type III , Hydrogels , Recombinant Proteins , Wound Healing , Wound Healing/drug effects , Collagen Type III/metabolism , Collagen Type III/genetics , Collagen Type III/chemistry , Animals , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Rats , Cicatrix/pathology , Cicatrix/drug therapy , Recombinant Proteins/pharmacology , Recombinant Proteins/chemistry , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/pathology , Male , Fibroblasts/drug effects , Fibroblasts/metabolism , Polysaccharides/chemistry , Polysaccharides/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology
11.
Ren Fail ; 46(2): 2387432, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39177245

ABSTRACT

BACKGROUND: Ureteral stricture (US) is a pathological stenosis in the urinary tract characterized by increased collagen synthesis and inflammation. Autophagy activation has been shown to ameliorate tissue fibrosis and protect against fibrotic diseases. Verapamil has beneficial therapeutic benefits on fibrotic disorders. The pharmacological effects of verapamil on fibroblast autophagy in US and the underlying mechanism need to be investigated further. METHODS: US patients were recruited to isolate scar tissues, hematoxylin-eosin (HE) and Masson trichrome staining were performed to analyze histopathological changes. The US animal model was established and administered with verapamil (0.05 mg/kg) in the drinking water. Transforming growth factor (TGF)-ß1 was adopted to facilitate collagen synthesis in fibroblasts. The mRNA and protein expressions were examined by qRT-PCR, western blot, immunofluorescence and immunohistochemistry. ELISA was adopted to measure interleukin (IL)-1ß and IL-6 levels. Molecular interaction experiments like dual luciferase reporter and chromatin immunoprecipitation (ChIP) assays were performed to analyze the interaction between signal transducers and activators of transcription 3 (STAT3) and RNA polymerase II associated factor 1 (PAF1). RESULTS: Herein, our results revealed that verapamil activated TGF-ß1-treated fibroblast autophagy and inhibited inflammation and fibrosis by repressing Ca2+/calmodulin-dependent protein kinase II (CaMK II) δ-mediated STAT3 activation. Our following tests revealed that STAT3 activated PAF1 transcription. PAF1 upregulation abrogated the regulatory effect of verapamil on fibroblast autophagy and fibrosis during US progression. Finally, verapamil mitigated US in vivo by activating fibroblast autophagy. CONCLUSION: Taken together, verapamil activated TGF-ß1-treated fibroblast autophagy and inhibited fibrosis by repressing the CaMK IIδ/STAT3/PAF1 axis.


Subject(s)
Autophagy , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Fibroblasts , Fibrosis , STAT3 Transcription Factor , Transforming Growth Factor beta1 , Ureteral Obstruction , Verapamil , Verapamil/pharmacology , Verapamil/therapeutic use , Autophagy/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism , STAT3 Transcription Factor/metabolism , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Transforming Growth Factor beta1/metabolism , Cicatrix/pathology , Cicatrix/metabolism , Cicatrix/drug therapy , Cicatrix/etiology , Cicatrix/prevention & control , Disease Models, Animal , Inflammation/metabolism , Signal Transduction/drug effects , Female , Middle Aged
12.
Circ Arrhythm Electrophysiol ; 17(9): e012922, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39193754

ABSTRACT

BACKGROUND: Cardiac magnetic resonance imaging (CMR)-defined ventricular scar and anatomic conduction channels (CMR-CCs) offer promise in delineating ventricular tachycardia substrate. No studies have validated channels with coregistered histology, nor have they ascertained the histological characteristics of deceleration zones (DZs) within these channels. We aimed to validate CMR scar and CMR-CCs with whole-heart histology and electroanatomic mapping in a postinfarction model. METHODS: Five sheep underwent anteroseptal infarction. CMR (116±20 days post infarct) was postprocessed using ADAS-3D, varying pixel intensity thresholds (5545, 6040, 6535, and 7030). DZs were identified by electroanatomic mapping (129±12 days post infarct). Explanted hearts were sectioned and stained with Picrosirius red, and whole-heart histopathologic shells were generated. Scar topography as well as percentage fibrosis, adiposity, and remaining viable myocardium within 3 mm histological biopsies and within CMR-CCs were determined. RESULTS: Using the standard 6040 thresholding, CMR had 83.8% accuracy for identifying histological scar in the endocardium (κ, 0.666) and 61.4% in the epicardium (κ, 0.276). Thirty-seven CMR-CCs were identified by varying thresholding; 23 (62%) were unique. DZs colocalized to 19 of 23 (83%) CMR-CCs. Twenty (87%) CMR-CCs were histologically confirmed. Within-channel histological fibrosis did not differ by the presence of DZs (P=0.242). Within-channel histological adiposity was significantly higher at sites with versus without DZs (24.1% versus 8.3%; P<0.001). CONCLUSIONS: Postprocessed CMR-derived scars and channels were validated by histology and electroanatomic mapping. Regions of CMR-CCs at sites of DZs had higher adiposity but similar fibrosis than regions without DZs, suggesting that lipomatous metaplasia may contribute to arrhythmogenicity of postinfarction scar.


Subject(s)
Cicatrix , Disease Models, Animal , Myocardium , Tachycardia, Ventricular , Animals , Cicatrix/pathology , Cicatrix/physiopathology , Cicatrix/diagnostic imaging , Cicatrix/etiology , Myocardium/pathology , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/pathology , Fibrosis , Heart Conduction System/physiopathology , Sheep , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/complications , Magnetic Resonance Imaging , Electrophysiologic Techniques, Cardiac , Predictive Value of Tests , Biopsy , Reproducibility of Results
13.
Semin Immunopathol ; 46(5): 13, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39186134

ABSTRACT

Skin scar formation following Bacille Calmette-Guérin (BCG) or smallpox (Vaccinia) vaccination is an established marker of successful vaccination and 'vaccine take'. Potent pathogen-specific (tuberculosis; smallpox) and pathogen-agnostic (protection from diseases unrelated to the intentionally targeted pathogen) effects of BCG and smallpox vaccines hold significant translational potential. Yet despite their use for centuries, how scar formation occurs and how local skin-based events relate to systemic effects that allow these two vaccines to deliver powerful health promoting effects has not yet been determined. We review here what is known about the events occurring in the skin and place this knowledge in the context of the overall impact of these two vaccines on human health with a particular focus on maternal-child health.


Subject(s)
BCG Vaccine , Cicatrix , Skin , Smallpox Vaccine , Vaccination , Animals , Humans , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , Cicatrix/etiology , Cicatrix/pathology , Cicatrix/immunology , Skin/pathology , Skin/immunology , Smallpox/prevention & control , Smallpox/immunology , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/immunology
14.
Sci Rep ; 14(1): 19035, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152163

ABSTRACT

Glial scar formation represents a fundamental response to central nervous system (CNS) injuries. It is mainly characterized by a well-defined spatial rearrangement of reactive astrocytes and microglia. The mechanisms underlying glial scar formation have been extensively studied, yet quantitative descriptors of the spatial arrangement of reactive glial cells remain limited. Here, we present a novel approach using point pattern analysis (PPA) and topological data analysis (TDA) to quantify spatial patterns of reactive glial cells after experimental ischemic stroke in mice. We provide open and reproducible tools using R and Julia to quantify spatial intensity, cell covariance and conditional distribution, cell-to-cell interactions, and short/long-scale arrangement, which collectively disentangle the arrangement patterns of the glial scar. This approach unravels a substantial divergence in the distribution of GFAP+ and IBA1+ cells after injury that conventional analysis methods cannot fully characterize. PPA and TDA are valuable tools for studying the complex spatial arrangement of reactive glia and other nervous cells following CNS injuries and have potential applications for evaluating glial-targeted restorative therapies.


Subject(s)
Astrocytes , Cicatrix , Neuroglia , Animals , Mice , Cicatrix/pathology , Neuroglia/pathology , Astrocytes/pathology , Microglia/pathology , Ischemic Stroke/pathology , Data Analysis , Disease Models, Animal , Male , Glial Fibrillary Acidic Protein/metabolism , Mice, Inbred C57BL
15.
Exp Neurol ; 380: 114915, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122167

ABSTRACT

BACKGROUND: The inflammatory response and scar formation after spinal cord injury (SCI) limit nerve regeneration and functional recovery. Our research group has previously shown that the expression of astrocyte-derived lipocalin 2 (Lcn2) is upregulated after SCI, which correlates with neuronal apoptosis and functional recovery. Therefore, we speculate that astrocyte-specific knockdown of Lcn2 after SCI may lead to a better prognosis. METHODS: Tissue RNA sequencing, Western blotting, PCR, and immunofluorescence assays were conducted to assess the expression of Lcn2 following SCI in mice. Adeno-associated virus 9 (AAV9) transfection was employed to specifically reduce the expression of Lcn2 in astrocytes, and subsequent evaluations of scarring and inflammation were conducted. In vitro experiments involved treating primary astrocytes with TGF-ß or an A1-induced mixture (C1q, TNF-α and IL-1α) following Lcn2 knockdown. Finally, the intrathecal injection of recombinant Lcn2 (ReLcn2) protein was conducted post-injury to further confirm the role of Lcn2 and its underlying mechanism in SCI. RESULTS: Lcn2 expression was elevated in astrocytes after SCI at 7 dpi (days post injury). Lcn2 knockdown in astrocytes is beneficial for neuronal survival and functional recovery after SCI, and is accompanied by a reduced inflammatory response and inhibited scar formation. The inhibition of SMAD-associated signaling activation was identified as a possible mechanism, and in vitro experiments further confirmed this finding. ReLcn2 further activated SMAD-associated signaling and aggravated motor function after SCI. CONCLUSION: The upregulation of Lcn2 expression in astrocytes is involved in neuroinflammation and scar formation after SCI, and the activation of SMAD-associated signaling is one of the underlying mechanisms.


Subject(s)
Astrocytes , Cicatrix , Lipocalin-2 , Mice, Inbred C57BL , Smad Proteins , Spinal Cord Injuries , Animals , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/genetics , Lipocalin-2/genetics , Lipocalin-2/metabolism , Mice , Astrocytes/metabolism , Cicatrix/etiology , Cicatrix/pathology , Cicatrix/metabolism , Smad Proteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Inflammation/etiology , Male , Neuroinflammatory Diseases/etiology , Female , Recovery of Function/physiology , Cells, Cultured
16.
Cells ; 13(16)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39195271

ABSTRACT

Scars may represent more than a cosmetic concern for patients; they may impose functional limitations and are frequently associated with the sensation of itching or pain, thus impacting both psychological and physical well-being. From an aesthetic perspective, scars display variances in color, thickness, texture, contour, and their homogeneity, while the functional aspect encompasses considerations of functionality, pliability, and sensory perception. Scars located in critical anatomic areas have the potential to induce profound impairments, including contracture-related mobility restrictions, thereby significantly impacting daily functioning and the quality of life. Conventional approaches to scar management may suffice to a certain extent, yet there are cases where tailored interventions are warranted. Autologous fat grafting emerges as a promising therapeutic avenue in such instances. Fundamental mechanisms underlying scar formation include chronic inflammation, fibrogenesis and dysregulated wound healing, among other contributing factors. These mechanisms can potentially be alleviated through the application of adipose-derived stem cells, which represent the principal cellular component utilized in the process of lipofilling. Adipose-derived stem cells possess the capacity to secrete proangiogenic factors such as fibroblast growth factor, vascular endothelial growth factor and hepatocyte growth factor, as well as neurotrophic factors, such as brain-derived neurotrophic factors. Moreover, they exhibit multipotency, remodel the extracellular matrix, act in a paracrine manner, and exert immunomodulatory effects through cytokine secretion. These molecular processes contribute to neoangiogenesis, the alleviation of chronic inflammation, and the promotion of a conducive milieu for wound healing. Beyond the obvious benefit in restoring volume, the adipose-derived stem cells and their regenerative capacities facilitate a reduction in pain, pruritus, and fibrosis. This review elucidates the regenerative potential of autologous fat grafting and its beneficial and promising effects on both functional and aesthetic outcomes when applied to scar tissue.


Subject(s)
Adipose Tissue , Cicatrix , Transplantation, Autologous , Humans , Cicatrix/pathology , Adipose Tissue/transplantation , Wound Healing , Animals
17.
Matrix Biol ; 133: 14-32, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39098433

ABSTRACT

BACKGROUND: Members of the cellular communication network family (CCN) of matricellular proteins, like CCN1, have long been implicated in the regulation of cellular processes underlying wound healing, tissue fibrogenesis, and collagen dynamics. While many studies suggest antifibrotic actions for CCN1 in the adult heart through the promotion of myofibroblast senescence, they largely relied on exogenous supplementation strategies in in vivo models of cardiac injury where its expression is already induced-which may confound interpretation of its function in this process. The objective of this study was to interrogate the role of the endogenous protein on fibroblast function, collagen structural dynamics, and its associated impact on cardiac fibrosis after myocardial infarction (MI). METHODS/RESULTS: Here, we employed CCN1 loss-of-function methodologies, including both in vitro siRNA-mediated depletion and in vivo fibroblast-specific knockout mice to assess the role of the endogenous protein on cardiac fibroblast fibrotic signaling, and its involvement in acute scar formation after MI. In vitro depletion of CCN1 reduced cardiac fibroblast senescence and proliferation. Although depletion of CCN1 decreased the expression of collagen processing and stabilization enzymes (i.e., P4HA1, PLOD1, and PLOD2), it did not inhibit myofibroblast induction or type I collagen synthesis. Alone, fibroblast-specific removal of CCN1 did not negatively impact ventricular performance or myocardial collagen content but did contribute to disorganization of collagen fibrils and increased matrix compliance. Similarly, Ccn1 ablated animals subjected to MI showed no discernible alterations in cardiac structure or function one week after permanent coronary artery ligation, but exhibited marked increases in incidence of mortality and cardiac rupture. Consistent with our findings that CCN1 depletion does not assuage myofibroblast conversion or type I collagen synthesis in vitro, Ccn1 knockout animals revealed no measurable differences in collagen scar width or mass compared to controls; however, detailed structural analyses via SHG and TEM of scar regions revealed marked alterations in their scar collagen topography-exhibiting changes in numerous macro- and micro-level collagen architectural attributes. Specifically, Ccn1 knockout mice displayed heightened ECM structural complexity in post-MI scar regions, including diminished local alignment and heightened tortuosity of collagen fibers, as well as reduced organizational coherency, packing, and size of collagen fibrils. Associated with these changes in ECM topography with the loss of CCN1 were reductions in fibroblast-matrix interactions, as evidenced by reduced fibroblast nuclear and cellular deformation in vivo and reduced focal-adhesion formation in vitro; findings that ultimately suggest CCN1's ability to influence fibroblast-led collagen alignment may in part be credited to its capacity to augment fibroblast-matrix interactions. CONCLUSIONS: These findings underscore the pivotal role of endogenous CCN1 in the scar formation process occurring after MI, directing the appropriate arrangement of the extracellular matrix's collagenous components in the maturing scar-shaping the mechanical properties that support its structural stability. While this suggests an adaptive role for CCN1 in regulating collagen structural attributes crucial for supporting scar integrity post MI, the long-term protracted expression of CCN1 holds maladaptive implications, potentially diminishing collagen structural complexity and compliance in non-infarct regions.


Subject(s)
Cicatrix , Collagen , Cysteine-Rich Protein 61 , Fibrosis , Myocardial Infarction , Myofibroblasts , Animals , Humans , Male , Mice , Cicatrix/metabolism , Cicatrix/pathology , Cicatrix/genetics , Collagen/metabolism , Collagen/genetics , Cysteine-Rich Protein 61/metabolism , Cysteine-Rich Protein 61/genetics , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/genetics , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction
18.
Cell Tissue Res ; 398(1): 15-25, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39120736

ABSTRACT

In goldfish, spinal cord injury triggers the formation of a fibrous scar at the injury site. Regenerating axons are able to penetrate the scar tissue, resulting in the recovery of motor function. Previous findings suggested that regenerating axons enter the scar through tubular structures surrounded by glial elements with laminin-positive basement membranes and that glial processes expressing glial fibrillary acidic protein (GFAP) are associated with axonal regeneration. How glia contribute to promoting axonal regeneration, however, is unknown. Here, we revealed that glial processes expressing vimentin or brain lipid-binding protein (BLBP) also enter the fibrous scar after spinal cord injury in goldfish. Vimentin-positive glial processes were more numerous than GFAP- or BLBP-positive glial processes in the scar tissue. Regenerating axons in the scar tissue were more closely associated with vimentin-positive glial processes than GFAP-positive glial processes. Vimentin-positive glial processes co-expressed matrix metalloproteinase (MMP)-14. Our findings suggest that vimentin-positive glial processes closely associate with regenerating axons through tubular structures entering the scar after spinal cord injury in goldfish. In intact spinal cord, ependymo-radial glial cell bodies express BLBP and their radial processes express vimentin, suggesting that vimentin-positive glial processes derive from migrating ependymo-radial glial cells. MMP-14 expressed in vimentin-positive glial cells and their processes might provide a beneficial environment for axonal regeneration.


Subject(s)
Axons , Goldfish , Nerve Regeneration , Neuroglia , Spinal Cord Injuries , Vimentin , Animals , Goldfish/metabolism , Vimentin/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Neuroglia/metabolism , Axons/metabolism , Nerve Regeneration/physiology , Matrix Metalloproteinase 14/metabolism , Cicatrix/metabolism , Cicatrix/pathology , Fatty Acid-Binding Proteins/metabolism
19.
Skin Res Technol ; 30(8): e13859, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39096179

ABSTRACT

INTRODUCTION: Lupus erythematosus (LE) is an inflammatory autoimmune disease, that can affect the skin to varying degree. In particular, discoid LE (DLE) and the rare form of lupus panniculitis/profundus are associated with scarring alopecia. The heterogeneity of the clinical, dermatoscopic, and histologic presentation poses a major challenge to the clinician in the diagnosis and differential diagnosis of other forms of scarring alopecia. OBJECTIVE: While noninvasive imaging techniques using optical coherence tomography (OCT) and reflectance confocal microscopy (RCM) have proven to be helpful in the diagnosis of scarring alopecia in the context of LE, this study aimed to investigate line-field confocal OCT (LC-OCT) to identify characteristic features of cicatricial alopecia in LE. METHODS: Fifteen patients with cicatricial alopecia in LE were included and the most affected/inflamed areas of the scalp were prospectively examined. In analogy to histopathology and previously reported criteria in RCM, all images were evaluated according to seven established criteria and underwent descriptive analyses. RESULTS: LC-OCT revealed characteristic features of cicatricial alopecia, such as lymphocytic interface dermatitis (14/15; 93.3%) and basal cell vacuolization (13/15; 86.7%). The most impressive feature was the occurrence of prominent hyperreflective fibers in 14/15 patients (93.3%). CONCLUSION: LC-OCT imaging can noninvasively detect morphologic criteria such as lymphocytic and vacuolar interface dermatitis of cicatricial alopecia due to LE. In particular, the presence of hyperreflective collagen fibers appears to be a characteristic easily recognizable feature that may facilitate differential diagnosis with other forms of cicatricial alopecia. Further studies are mandatory to differentiate other forms of scarring alopecia.


Subject(s)
Alopecia , Cicatrix , Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Alopecia/pathology , Alopecia/diagnostic imaging , Female , Cicatrix/diagnostic imaging , Cicatrix/pathology , Adult , Middle Aged , Male , Diagnosis, Differential , Microscopy, Confocal/methods , Young Adult , Lupus Erythematosus, Discoid/pathology , Lupus Erythematosus, Discoid/diagnostic imaging , Lupus Erythematosus, Discoid/complications , Prospective Studies , Lupus Erythematosus, Cutaneous/pathology , Lupus Erythematosus, Cutaneous/diagnostic imaging , Aged
20.
J Neuroinflammation ; 21(1): 193, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095832

ABSTRACT

Lactate-derived histone lactylation is involved in multiple pathological processes through transcriptional regulation. The role of lactate-derived histone lactylation in the repair of spinal cord injury (SCI) remains unclear. Here we report that overall lactate levels and lactylation are upregulated in the spinal cord after SCI. Notably, H4K12la was significantly elevated in the microglia of the injured spinal cord, whereas exogenous lactate treatment further elevated H4K12la in microglia after SCI. Functionally, lactate treatment promoted microglial proliferation, scar formation, axon regeneration, and locomotor function recovery after SCI. Mechanically, lactate-mediated H4K12la elevation promoted PD-1 transcription in microglia, thereby facilitating SCI repair. Furthermore, a series of rescue experiments confirmed that a PD-1 inhibitor or microglia-specific AAV-sh-PD-1 significantly reversed the therapeutic effects of lactate following SCI. This study illustrates the function and mechanism of lactate/H4K12la/PD-1 signaling in microglia-mediated tissue repair and provides a novel target for SCI therapy.


Subject(s)
Histones , Lactic Acid , Microglia , Recovery of Function , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Microglia/metabolism , Microglia/drug effects , Histones/metabolism , Recovery of Function/drug effects , Recovery of Function/physiology , Lactic Acid/metabolism , Rats , Lysine/metabolism , Lysine/analogs & derivatives , Lysine/pharmacology , Mice , Cicatrix/metabolism , Cicatrix/pathology , Female , Rats, Sprague-Dawley , Mice, Inbred C57BL , Male , Locomotion/drug effects , Locomotion/physiology
SELECTION OF CITATIONS
SEARCH DETAIL