Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.686
Filter
1.
Science ; 385(6713): eadi1650, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39236183

ABSTRACT

Skin identity is controlled by intrinsic features of the epidermis and dermis and their interactions. Modifying skin identity has clinical potential, such as the conversion of residual limb and stump (nonvolar) skin of amputees to pressure-responsive palmoplantar (volar) skin to enhance prosthesis use and minimize skin breakdown. Greater keratin 9 (KRT9) expression, higher epidermal thickness, keratinocyte cytoplasmic size, collagen length, and elastin are markers of volar skin and likely contribute to volar skin resiliency. Given fibroblasts' capacity to modify keratinocyte differentiation, we hypothesized that volar fibroblasts influence these features. Bioprinted skin constructs confirmed the capacity of volar fibroblasts to induce volar keratinocyte features. A clinical trial of healthy volunteers demonstrated that injecting volar fibroblasts into nonvolar skin increased volar features that lasted up to 5 months, highlighting a potential cellular therapy.


Subject(s)
Biomedical Enhancement , Bioprinting , Dermis , Epidermis , Fibroblasts , Keratinocytes , Adult , Female , Humans , Male , Amputees , Cell Differentiation , Collagen/metabolism , Dermis/cytology , Dermis/metabolism , Elastin/metabolism , Epidermis/metabolism , Fibroblasts/cytology , Fibroblasts/transplantation , Hand , Keratin-9/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Biomedical Enhancement/methods
2.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125802

ABSTRACT

The hair follicle is the basis of hair regeneration, and the dermal papilla is one of the most important structures in hair regeneration. New intervention and reversal strategies for hair loss may arise due to the prevention of oxidative stress. GC/MS analysis was used to determine the compounds contained in NSO. Then, NSO was applied to DPC for cell proliferation and oxidative stress experiments. RNA-seq was performed in cells treated with NSO and minoxidil. The quantitative real-time polymerase chain reaction (qRT-PCR) was applied to verify the gene expression. The effects of NSO on hair length, weight, the number and depth of hair follicles, and the dermal thickness were also studied. GC/MS analysis showed that the main components of NSO were eicosapentaenoic acid, palmitic acid, and linoleic acid. NSO promotes DPC proliferation and reduces H2O2-mediated oxidative damage. NSO can also activate hair growth-related pathways and upregulate antioxidant-related genes analyzed by gene profiling. The topical application of NSO significantly promotes hair growth and increases hair length and weight in mice. NSO extract promotes hair growth and effectively inhibits oxidative stress, which is beneficial for the prevention and treatment of hair loss.


Subject(s)
Cell Proliferation , Hair Follicle , Hair , Oxidative Stress , Cell Proliferation/drug effects , Animals , Humans , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/growth & development , Hair Follicle/cytology , Mice , Oxidative Stress/drug effects , Hair/drug effects , Hair/growth & development , Antioxidants/pharmacology , Dermis/metabolism , Dermis/cytology , Dermis/drug effects
3.
Methods Mol Biol ; 2835: 17-27, 2024.
Article in English | MEDLINE | ID: mdl-39105902

ABSTRACT

Human mesenchymal stromal cells (MSCs) have gained significant interest as cell-based therapeutics for organ restoration in the field of regenerative medicine. More recently, substantial attention has been directed toward cell-free therapy, achieved through the utilization of soluble factors possessing trophic and immunomodulatory properties present in the MSC secretome. This collection of soluble factors can be found either freely in the secretome or packed within its vesicular fraction, known as extracellular vesicles (EVs). MSCs can be derived from various tissue sources, each involving different extraction methods and yielding varying cell amounts. In this study, we describe a nonenzymatic procedure for a straightforward isolation of MSCs from the fetal dermis and the adult dermis. The results demonstrate the isolation of a cell population with a uniform MSC immunophenotype from the earliest passages (approximately 90% positive for the classical MSC markers CD90, CD105, and CD73, while negative for the hematopoietic markers CD34 and CD45, as well as HLA-DR). Additionally, we describe the procedures for cell expansion, banking, and secretome collection.


Subject(s)
Cell Separation , Dermis , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Dermis/cytology , Dermis/metabolism , Cell Separation/methods , Immunophenotyping , Cell Culture Techniques/methods , Biomarkers , Cells, Cultured , Extracellular Vesicles/metabolism , Secretome/metabolism
4.
Aging (Albany NY) ; 16(16): 11776-11795, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39197170

ABSTRACT

Crucial for skin homeostasis, synthesis and degradation of extracellular matrix components are orchestrated by dermal fibroblasts. During aging, alterations of component expression, such as collagens and enzymes, lead to reduction of the mechanical cutaneous tension and defects of skin wound healing. The aim of this study was to better understand the molecular alterations underwent by fibroblasts during aging by comparing secretomic and proteomic signatures of fibroblasts from young (<35years) and aged (>55years) skin donors, in quiescence or TGF-stimulated conditions, using HLPC/MS. The comparison of the secretome from young and aged fibroblasts revealed that 16 proteins in resting condition, and 11 proteins after a 24h-lasting TGF-ß1-treatment, were expressed in significant different ways between the two cell groups (fold change>2, p-value <0.05), with a 77% decrease in the number of secreted proteins in aged cells. Proteome comparison between young and aged fibroblasts identified a significant change of 63 proteins in resting condition, and 73 proteins in TGF-ß1-stimulated condition, with a 67% increase in the number of proteins in aged fibroblasts. The majority of the differentially-expressed molecules belongs to the cytoskeleton-associated proteins and aging was characterized by an increase in Coronin 1C (CORO1C), and Filamin B (FLNB) expression in fibroblasts together with a decrease in Cofilin (CFL1), and Actin alpha cardiac muscle 1 (ACTC1) detection in aged cells, these proteins being involved in actin-filament polymerization and sharing co-activity in cell motility. Our present data reinforce knowledge about an age-related alteration in the synthesis of major proteins linked to the migratory and contractile functions of dermal human fibroblasts.


Subject(s)
Aging , Cytoskeleton , Fibroblasts , Proteomics , Humans , Fibroblasts/metabolism , Cytoskeleton/metabolism , Adult , Middle Aged , Aging/metabolism , Transforming Growth Factor beta1/metabolism , Aged , Skin/metabolism , Skin/cytology , Proteome/metabolism , Cells, Cultured , Male , Secretome/metabolism , Female , Dermis/cytology , Dermis/metabolism
5.
Tissue Cell ; 90: 102495, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094367

ABSTRACT

There is a need in clinical practice for new wound healing techniques to address full thickness skin injuries, particularly in individuals with diabetes. Herein we investigated whether dermal derived matrix hydrogel (DMH) loaded with curcumin (Cur) could promote healing in diabetic rats. Sixty diabetic rats were randomly assigned into the non-treated group, DMH group, Cur group, and DMH+Cur group. According to the phases of wound healing, sampling was done on days 7, 14, and 21 for further assessments. Our results indicated that the wound contraction rate, new epidermal length and thickness, number of fibroblasts and vascular length, collagen deposition, and strength properties of the healed wounds were meaningfully increased in the treatment groups than in the non-treated group, and these changes were more obvious in the DMH+Cur ones. In addition, the expression of VEGF and IL-10 genes were meaningfully upregulated in all treatment groups compared to the non-treated group and were greater in the DMH+Cur group. This is while the number of neutrophils and expression levels of TNF-α and IL-1ß genes decreased more significantly in the DMH+Cur group compared to the other groups. In conclusion, it was found that using both DMH and curcumin has a greater impact on diabetic wound healing.


Subject(s)
Curcumin , Diabetes Mellitus, Experimental , Hydrogels , Wound Healing , Animals , Curcumin/pharmacology , Wound Healing/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Hydrogels/chemistry , Rats , Male , Rats, Sprague-Dawley , Dermis/metabolism , Dermis/pathology , Dermis/drug effects , Dermis/injuries
6.
J Nanobiotechnology ; 22(1): 425, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030543

ABSTRACT

Hair follicle (HF) regeneration during wound healing continues to present a significant clinical challenge. Dermal papilla cell-derived exosomes (DPC-Exos) hold immense potential for inducing HF neogenesis. However, the accurate role and underlying mechanisms of DPC-Exos in HF regeneration in wound healing remain to be fully explained. This study, represents the first analysis into the effects of DPC-Exos on fibroblasts during wound healing. Our findings demonstrated that DPC-Exos could stimulate the proliferation and migration of fibroblasts, more importantly, enhance the hair-inducing capacity of fibroblasts. Fibroblasts treated with DPC-Exos were capable of inducing HF neogenesis in nude mice when combined with neonatal mice epidermal cells. In addition, DPC-Exos accelerated wound re-epithelialization and promoted HF regeneration during the healing process. Treatment with DPC-Exos led to increased expression levels of the Wnt pathway transcription factors ß-catenin and Lef1 in both fibroblasts and the dermis of skin wounds. Specifically, the application of a Wnt pathway inhibitor reduced the effects of DPC-Exos on fibroblasts and wound healing. Accordingly, these results offer evidence that DPC-Exos promote HF regeneration during wound healing by enhancing the hair-inducing capacity of fibroblasts and activating the Wnt/ß-catenin signaling pathway. This suggests that DPC-Exos may represent a promising therapeutic strategy for achieving regenerative wound healing.


Subject(s)
Cell Proliferation , Exosomes , Fibroblasts , Hair Follicle , Mice, Nude , Regeneration , Vibrissae , Wnt Signaling Pathway , Wound Healing , beta Catenin , Animals , Mice , Fibroblasts/metabolism , Exosomes/metabolism , Vibrissae/physiology , beta Catenin/metabolism , Dermis/metabolism , Cell Movement , Lymphoid Enhancer-Binding Factor 1/metabolism
7.
Int J Mol Sci ; 25(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39063192

ABSTRACT

Wool is generated by hair follicles (HFs), which are crucial in defining the length, diameter, and morphology of wool fibers. However, the regulatory mechanism of HF growth and development remains largely unknown. Dermal papilla cells (DPCs) are a specialized cell type within HFs that play a crucial role in governing the growth and development of HFs. This study aims to investigate the proliferation and induction ability of ovine DPCs to enhance our understanding of the potential regulatory mechanisms underlying ovine HF growth and development. Previous research has demonstrated that microRNA-181a (miR-181a) was differentially expressed in skin tissues with different wool phenotypes, which indicated that miR-181a might play a crucial role in wool morphogenesis. In this study, we revealed that miR-181a inhibited the proliferation and induction ability of ovine DPCs by quantitative Real-time PCR (qRT-PCR), cell counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and alkaline phosphatase staining. Then, we also confirmed G protein subunit alpha i2 (GNAI2) is a target gene of miR-181a by dual luciferase reporter assay, qRT-PCR, and Western blot, and that it could promote the proliferation and induction ability of ovine DPCs. In addition, GNAI2 could also activate the Wnt/ß-Catenin signaling pathway in ovine DPCs. This study showed that miR-181a can inhibit the proliferation and induction ability of ovine DPCs by targeting GNAI2 through the Wnt/ß-Catenin signaling pathway.


Subject(s)
Cell Proliferation , Hair Follicle , MicroRNAs , Wnt Signaling Pathway , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Sheep , Hair Follicle/metabolism , Hair Follicle/cytology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Dermis/cytology , Dermis/metabolism , Cells, Cultured , Wool/metabolism , beta Catenin/metabolism , beta Catenin/genetics
8.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38990714

ABSTRACT

Dermal fibroblasts deposit type I collagen, the dominant extracellular matrix molecule found in skin, during early postnatal development. Coincident with this biosynthetic program, fibroblasts proteolytically remodel pericellular collagen fibrils by mobilizing the membrane-anchored matrix metalloproteinase, Mmp14. Unexpectedly, dermal fibroblasts in Mmp14-/- mice commit to a large-scale apoptotic program that leaves skin tissues replete with dying cells. A requirement for Mmp14 in dermal fibroblast survival is recapitulated in vitro when cells are embedded within, but not cultured atop, three-dimensional hydrogels of crosslinked type I collagen. In the absence of Mmp14-dependent pericellular proteolysis, dermal fibroblasts fail to trigger ß1 integrin activation and instead actuate a TGF-ß1/phospho-JNK stress response that leads to apoptotic cell death in vitro as well as in vivo. Taken together, these studies identify Mmp14 as a requisite cell survival factor that maintains dermal fibroblast viability in postnatal dermal tissues.


Subject(s)
Apoptosis , Cell Survival , Fibroblasts , Matrix Metalloproteinase 14 , Animals , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/genetics , Fibroblasts/metabolism , Mice , Mice, Knockout , Collagen Type I/metabolism , Collagen Type I/genetics , Integrin beta1/metabolism , Integrin beta1/genetics , Transforming Growth Factor beta1/metabolism , Dermis/metabolism , Dermis/cytology , Cells, Cultured , Extracellular Matrix/metabolism , Mice, Inbred C57BL , Skin/metabolism
9.
ACS Appl Mater Interfaces ; 16(29): 37698-37706, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38980897

ABSTRACT

Despite the remarkable advances of dermal fillers that reduce wrinkles caused by dermis thickness reduction, they still lack effective hydrogel systems that stimulate collagen generation along with injection convenience. Here, we develop a stem cell-derived extracellular vesicle (EV)-bearing thermosensitive hydrogel (EVTS-Gel) for effective in vivo collagen generation. The TS-Gel undergoes sol-gel transition at 32.6 °C, as demonstrated by the storage and loss moduli crossover. Moreover, the TS-Gel and the EVTS-Gel have comparable rheological properties. Both hydrogels are injected in a sol state; hence, they require lower injection forces than conventional hydrogel-based dermal fillers. When locally administered to mouse skin, the TS-Gel extends the retention time of EVs by 2.23 times. Based on the nature of the controlled EV release, the EVTS-Gel significantly inhibits the dermis thickness reduction caused by aging compared to the bare EV treatment for 24 weeks. After a single treatment, the collagen layer thickness of the EVTS-Gel-treated dermis becomes 2.64-fold thicker than that of the bare EV-treated dermis. Notably, the collagen generation efficacy of the bare EV is poorer than that of the EVTS-Gel of a 10× lesser dose. Overall, the EVTS-Gel shows potential as an antiaging dermal filler for in vivo collagen generation.


Subject(s)
Collagen , Dermis , Extracellular Vesicles , Hydrogels , Animals , Mice , Dermis/metabolism , Dermis/drug effects , Collagen/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Humans , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/drug effects , Dermal Fillers/chemistry , Dermal Fillers/pharmacology
10.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000592

ABSTRACT

Prostaglandin E2 (PGE2) is known to be effective in regenerating tissues, and bimatoprost, an analog of PGF2α, has been approved by the FDA as an eyelash growth promoter and has been proven effective in human hair follicles. Thus, to enhance PGE2 levels while improving hair loss, we found dihydroisoquinolinone piperidinylcarboxy pyrazolopyridine (DPP), an inhibitor of 15-hydroxyprostaglandin dehydrogenase (15-PGDH), using DeepZema®, an AI-based drug development program. Here, we investigated whether DPP improved hair loss in human follicle dermal papilla cells (HFDPCs) damaged by dihydrotestosterone (DHT), which causes hair loss. We found that DPP enhanced wound healing and the expression level of alkaline phosphatase in DHT-damaged HFDPCs. We observed that DPP significantly down-regulated the generation of reactive oxygen species caused by DHT. DPP recovered the mitochondrial membrane potential in DHT-damaged HFDPCs. We demonstrated that DPP significantly increased the phosphorylation levels of the AKT/ERK and activated Wnt signaling pathways in DHT-damaged HFDPCs. We also revealed that DPP significantly enhanced the size of the three-dimensional spheroid in DHT-damaged HFDPCs and increased hair growth in ex vivo human hair follicle organ culture. These data suggest that DPP exhibits beneficial effects on DHT-damaged HFDPCs and can be utilized as a promising agent for improving hair loss.


Subject(s)
Hair Follicle , Hydroxyprostaglandin Dehydrogenases , Humans , Hair Follicle/drug effects , Hair Follicle/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Dihydrotestosterone/pharmacology , Dihydrotestosterone/metabolism , Reactive Oxygen Species/metabolism , Dermis/metabolism , Dermis/cytology , Dermis/drug effects , Cells, Cultured , Wnt Signaling Pathway/drug effects , Alopecia/drug therapy , Alopecia/metabolism , Wound Healing/drug effects , Hair/drug effects , Hair/growth & development , Membrane Potential, Mitochondrial/drug effects , Enzyme Inhibitors/pharmacology
11.
Cells ; 13(14)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39056788

ABSTRACT

Fibroblasts are among the most abundant cell types in the human body, playing crucial roles in numerous physiological processes, including the structural maintenance of the dermis, production of extracellular matrix components, and mediation of inflammatory responses. Despite their importance, fibroblasts remain one of the least characterized cell populations. The advent of single-cell analysis techniques, particularly single-cell RNA sequencing (scRNA-seq) and fluorescence-activated cell sorting (FACS), has enabled detailed investigations into fibroblast biology. In this study, we present an extensive analysis of fibroblast surface markers suitable for cell sorting and subsequent functional studies. We reviewed over three thousand research articles describing fibroblast populations and their markers, characterizing and comparing subtypes based on their surface markers, as well as their intra- and extracellular proteins. Our detailed analysis identified a variety of distinct fibroblast subpopulations, each with unique markers, characteristics dependent on their location, and the physiological or pathophysiological environment. These findings underscore the diversity of fibroblasts as a cellular population and could lead to the development of novel diagnostic and therapeutic tools.


Subject(s)
Biomarkers , Cell Separation , Fibroblasts , Flow Cytometry , Fibroblasts/metabolism , Fibroblasts/cytology , Humans , Cell Separation/methods , Biomarkers/metabolism , Flow Cytometry/methods , Dermis/cytology , Dermis/metabolism , Single-Cell Analysis/methods , Cell Survival , Animals
12.
Biomed Pharmacother ; 177: 117051, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959608

ABSTRACT

Due to the limitations of the current skin wound treatments, it is highly valuable to have a wound healing formulation that mimics the extracellular matrix (ECM) and mechanical properties of natural skin tissue. Here, a novel biomimetic hydrogel formulation has been developed based on a mixture of Agarose-Collagen Type I (AC) combined with skin ECM-related components: Dermatan sulfate (DS), Hyaluronic acid (HA), and Elastin (EL) for its application in skin tissue engineering (TE). Different formulations were designed by combining AC hydrogels with DS, HA, and EL. Cell viability, hemocompatibility, physicochemical, mechanical, and wound healing properties were investigated. Finally, a bilayered hydrogel loaded with fibroblasts and mesenchymal stromal cells was developed using the Ag-Col I-DS-HA-EL (ACDHE) formulation. The ACDHE hydrogel displayed the best in vitro results and acceptable physicochemical properties. Also, it behaved mechanically close to human native skin and exhibited good cytocompatibility. Environmental scanning electron microscopy (ESEM) analysis revealed a porous microstructure that allows the maintenance of cell growth and ECM-like structure production. These findings demonstrate the potential of the ACDHE hydrogel formulation for applications such as an injectable hydrogel or a bioink to create cell-laden structures for skin TE.


Subject(s)
Biomimetic Materials , Hydrogels , Tissue Engineering , Hydrogels/chemistry , Humans , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Tissue Engineering/methods , Cell Survival/drug effects , Mesenchymal Stem Cells/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Wound Healing/drug effects , Collagen Type I/metabolism , Skin/drug effects , Skin/metabolism , Dermatan Sulfate/chemistry , Dermatan Sulfate/pharmacology , Fibroblasts/drug effects , Elastin/chemistry , Extracellular Matrix/metabolism , Biomimetics/methods , Sepharose/chemistry , Dermis/drug effects , Dermis/metabolism , Dermis/cytology , Animals
13.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062918

ABSTRACT

The aging process is linked to numerous cellular changes, among which are modifications in the functionality of dermal fibroblasts. These fibroblasts play a crucial role in sustaining the healing of skin wounds. Reduced cell proliferation is a hallmark feature of aged dermal fibroblasts. Long intergenic non-coding RNA (lincRNAs), such as LincRNA-EPS (Erythroid ProSurvival), has been implicated in various cellular processes. However, its role in aged dermal fibroblasts and its impact on the cell cycle and its regulator, Cyclin D1 (CCND1), remains unclear. Primary dermal fibroblasts were isolated from the skin of 17-week-old (young) and 88-week-old (aged) mice. Overexpression of LincRNA-EPS was achieved through plasmid transfection. Cell proliferation was detected using the MTT assay. Real-time PCR was used to quantify relative gene expressions. Our findings indicate a noteworthy decline in the expression of LincRNA-EPS in aged dermal fibroblasts, accompanied by reduced levels of CCND1 and diminished cell proliferation in these aging cells. Significantly, the overexpression of LincRNA-EPS in aged dermal fibroblasts resulted in an upregulation of CCND1 expression and a substantial increase in cell proliferation. Mechanistically, LincRNA-EPS induces CCND1 expression by sequestering miR-34a, which was dysregulated in aged dermal fibroblasts, and directly targeting CCND1. These outcomes underscore the crucial role of LincRNA-EPS in regulating CCND1 and promoting cell proliferation in aged dermal fibroblasts. Our study provides novel insights into the molecular mechanisms underlying age-related changes in dermal fibroblasts and their implications for skin wound healing. The significant reduction in LincRNA-EPS expression in aged dermal fibroblasts and its ability to induce CCND1 expression and enhance cell proliferation highlight its potential as a therapeutic target for addressing age-related skin wound healing.


Subject(s)
Cell Proliferation , Cyclin D1 , Fibroblasts , RNA, Long Noncoding , Cyclin D1/metabolism , Cyclin D1/genetics , Fibroblasts/metabolism , Fibroblasts/cytology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Mice , Skin/metabolism , Skin/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Cells, Cultured , Skin Aging/genetics , Dermis/cytology , Dermis/metabolism , Cellular Senescence/genetics , Gene Expression Regulation , Wound Healing/genetics , Aging/genetics
14.
Sci Rep ; 14(1): 13899, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886354

ABSTRACT

The current investigation aims to study the embryonic dermis formed in the early stages of development and identify the initial interstitial components of the dermis that serve as biological and structural scaffolds for the development of the dermal tissue. To investigate the dermal structure, the current study used morphological and immunological techniques. TCs identified by TEM. They had a cell body and unique podomeres and podoms. They formed a 3D network spread throughout the dermis. Homocellular contact established between them, as well as heterocellular contacts with other cells. Immunohistochemical techniques using specific markers for TCss CD34, CD117, and VEGF confirmed TC identification. TCs represent the major interstitial component in the dermal tissue. They established a 3D network, enclosing other cells and structures. Expression of VEGF by TC promotes angiogenesis. TCs establish cellular contact with sprouting endothelial cells. At the site of cell junction with TCs, cytoskeletal filaments identified and observed to form the pseudopodium core that projects from endothelial cells. TCs had proteolytic properties that expressed MMP-9, CD68, and CD21. Proteolytic activity aids in the removal of components of the extracellular matrix and the phagocytosis of degraded remnants to create spaces to facilitate the development of new dermal structures. In conclusion, TCs organized the scaffold for the development of future dermal structures, including fibrous components and skin appendages. Studying dermal TCs would be interested in the possibility of developing therapeutic strategies for treating different skin disorders and diseases.


Subject(s)
Dermis , Immunohistochemistry , Telocytes , Telocytes/metabolism , Telocytes/cytology , Dermis/metabolism , Dermis/cytology , Humans , Antigens, CD34/metabolism , Animals , Vascular Endothelial Growth Factor A/metabolism , Antigens, CD/metabolism , Matrix Metalloproteinase 9/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Antigens, Differentiation, Myelomonocytic/metabolism , CD68 Molecule
15.
Skin Res Technol ; 30(6): e13810, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887125

ABSTRACT

BACKGROUND: Human dermal fibroblasts secrete diverse proteins that regulate wound repair and tissue regeneration. METHODS: In this study, dermal fibroblast-conditioned medium (DFCM) proteins potentially regulating nerve restoration were bioinformatically selected among the 337 protein lists identified by quantitative liquid chromatography-tandem mass spectrometry. Using these proteins, protein-protein interaction network analysis was conducted. In addition, the roles of DFCM proteins were reviewed according to their protein classifications. RESULTS: Gene Ontology protein classification categorized these 57 DFCM proteins into various classes, including protein-binding activity modulator (N = 11), cytoskeletal protein (N = 8), extracellular matrix protein (N = 6), metabolite interconversion enzyme (N = 5), chaperone (N = 4), scaffold/adapter protein (N = 4), calcium-binding protein (N = 3), cell adhesion molecule (N = 2), intercellular signal molecule (N = 2), protein modifying enzyme (N = 2), transfer/carrier protein (N = 2), membrane traffic protein (N = 1), translational protein (N = 1), and unclassified proteins (N = 6). Further protein-protein interaction network analysis of 57 proteins revealed significant interactions among the proteins that varied according to the settings of confidence score. CONCLUSIONS: Our bioinformatic analysis demonstrated that DFCM contains many secretory proteins that form significant protein-protein interaction networks crucial for regulating nerve restoration. These findings underscore DFCM proteins' critical roles in various nerve restoration stages during the wound repair process.


Subject(s)
Computational Biology , Fibroblasts , Nerve Regeneration , Protein Interaction Maps , Humans , Fibroblasts/metabolism , Nerve Regeneration/physiology , Protein Interaction Maps/physiology , Culture Media, Conditioned , Wound Healing/physiology , Cells, Cultured , Tandem Mass Spectrometry , Dermis/cytology , Dermis/metabolism
16.
In Vivo ; 38(4): 1767-1774, 2024.
Article in English | MEDLINE | ID: mdl-38936924

ABSTRACT

BACKGROUND/AIM: Dermal papilla (DP) stem cells are known for their remarkable regenerative capacity, making them a valuable model for assessing the effects of natural products on cellular processes, including stemness, and autophagy. MATERIALS AND METHODS: Autophagy and stemness characteristics were assessed using real-time RT-PCR to analyze mRNA levels, along with immunofluorescence and western blot techniques for protein level evaluation. RESULTS: Butterfly Pea, Emblica Fruits, Kaffir Lime, and Thunbergia Laurifolia extracts induced autophagy in DP cells. Kaffir Lime-treated cells exhibited increase in the OCT4, NANOG, and SOX2 mRNA (6-, 5, and 5.5-fold, respectively), and protein levels (4-, 3-, and 1.5-fold, respectively). All extracts activated the survival protein kinase B (Akt) in DP cells. CONCLUSION: Natural products are a promising source for promoting hair growth by rejuvenating hair stem cells.


Subject(s)
Autophagy , Biological Products , Hair Follicle , Plant Extracts , Stem Cells , Autophagy/drug effects , Humans , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Biological Products/pharmacology , Plant Extracts/pharmacology , Hair Follicle/drug effects , Hair Follicle/cytology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Proto-Oncogene Proteins c-akt/metabolism , Dermis/cytology , Dermis/drug effects , Dermis/metabolism , Cell Differentiation/drug effects
17.
Tissue Cell ; 89: 102431, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870572

ABSTRACT

Tunneling nanotubes (TNTs) represent an innovative way for cells to communicate with one another, as they act as long conduits between cells. However, their roles in human dermal microvascular pericytes (HDMPCs) interaction remain elusive in vitro. In this work, we identified and characterized the TNT-like structures that connected two or more pericytes in two-dimensional cultures and formed a functional network in the human dermis. Immunofluorescence assay indicated that the F-actin was an essential element to form inter-pericyte TNT-like structures, as it decreased in actin polymer inhibitor-cytochalasin B treated groups, and microtubules were present in almost half of the TNT-like structures. Most importantly, we only found the presence of mitochondrial in TNT-like structures containing α-tubulin, and the application of microtubule assembly inhibitor-Nocodazole significantly reduced the percentage of TNT-like structures that contain α-tubulin, resulting in a sudden decrease in the positive rate of cytochrome c oxidase subunit 4 isoform 1 (COX IV, a marker of mitochondria) in TNT-like structures. In summary, we described a novel intercellular communication-TNT-like structures-between HDMPCs in vitro, and this work allows us to properly understand the cellular mechanisms of spreading materials between HDMPCs, shedding light on the role of HDMPCs.


Subject(s)
Pericytes , Humans , Pericytes/cytology , Pericytes/metabolism , Tubulin/metabolism , Microtubules/metabolism , Dermis/cytology , Dermis/metabolism , Cell Communication , Mitochondria/metabolism , Actins/metabolism , Nanotubes/chemistry , Microvessels/cytology , Microvessels/metabolism , Cells, Cultured , Cell Membrane Structures
18.
Bull Exp Biol Med ; 176(5): 636-639, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38727953

ABSTRACT

Immunohistochemical detection of the LYVE-1 marker in healthy human full-thickness skin (the epidermis and the dermis) was carried out. LYVE-1 expression was found in the endothelium of lymphatic capillaries located in the papillary dermis, in the endothelium of larger lymphatic vessels of the reticular dermis, and in fibroblasts, which indicates their joint participation in hyaluronan metabolism. LYVE-1+ staining detected for the first time in cells of the stratum basale, the stratum spinosum, and the stratum granulosum of healthy human epidermis indicates their participation in hyaluronan metabolism and allows us to consider the spaces between epidermis cells as prelimphatics.


Subject(s)
Epidermis , Hyaluronic Acid , Lymphatic Vessels , Skin , Vesicular Transport Proteins , Humans , Hyaluronic Acid/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Skin/metabolism , Lymphatic Vessels/metabolism , Epidermis/metabolism , Ligands , Fibroblasts/metabolism , Dermis/metabolism , Lymphatic System/metabolism , Adult , Female , Male , Immunohistochemistry
19.
Commun Biol ; 7(1): 577, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755434

ABSTRACT

Pseudoxanthoma elasticum (PXE) is a rare disease characterized by ectopic calcification, however, despite the widely spread effect of pro/anti-calcifying systemic factors associated with this genetic metabolic condition, it is not known why elastic fibers in the same patient are mainly fragmented or highly mineralized in clinically unaffected (CUS) and affected (CAS) skin, respectively. Cellular morphology and secretome are investigated in vitro in CUS and CAS fibroblasts. Here we show that, compared to CUS, CAS fibroblasts exhibit: a) differently distributed and organized focal adhesions and stress fibers; b) modified cell-matrix interactions (i.e., collagen gel retraction); c) imbalance between matrix metalloproteinases and tissue inhibitor of metalloproteinases; d) differentially expressed pro- and anti-calcifying proteoglycans and elastic-fibers associated glycoproteins. These data emphasize that in the development of pathologic mineral deposition fibroblasts play an active role altering the stability of elastic fibers and of the extracellular matrix milieu creating a local microenvironment guiding the level of matrix remodeling at an extent that may lead to degradation (in CUS) or to degradation and calcification (in CAS) of the elastic component. In conclusion, this study contributes to a better understanding of the mechanisms of the mineral deposition that can be also associated with several inherited or age-related diseases (e.g., diabetes, atherosclerosis, chronic kidney diseases).


Subject(s)
Calcinosis , Elastin , Fibroblasts , Pseudoxanthoma Elasticum , Female , Humans , Male , Calcinosis/metabolism , Calcinosis/pathology , Cells, Cultured , Dermis/metabolism , Dermis/pathology , Elastic Tissue/metabolism , Elastic Tissue/pathology , Elastin/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Pseudoxanthoma Elasticum/metabolism , Pseudoxanthoma Elasticum/pathology , Pseudoxanthoma Elasticum/genetics
20.
Cells ; 13(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38786058

ABSTRACT

Fibrosis is a pathological condition consisting of a delayed deposition and remodeling of the extracellular matrix (ECM) by fibroblasts. This deregulation is mostly triggered by a chronic stimulus mediated by pro-inflammatory cytokines, such as TNF-α and IL-1, which activate fibroblasts. Due to their anti-inflammatory and immunosuppressive potential, dental pulp stem cells (DPSCs) could affect fibrotic processes. This study aims to clarify if DPSCs can affect fibroblast activation and modulate collagen deposition. We set up a transwell co-culture system, where DPSCs were seeded above the monolayer of fibroblasts and stimulated with LPS or a combination of TNF-α and IL-1ß and quantified a set of genes involved in inflammasome activation or ECM deposition. Cytokines-stimulated co-cultured fibroblasts, compared to unstimulated ones, showed a significant increase in the expression of IL-1ß, IL-6, NAIP, AIM2, CASP1, FN1, and TGF-ß genes. At the protein level, IL-1ß and IL-6 release as well as FN1 were increased in stimulated, co-cultured fibroblasts. Moreover, we found a significant increase of MMP-9 production, suggesting a role of DPSCs in ECM remodeling. Our data seem to suggest a crosstalk between cultured fibroblasts and DPSCs, which seems to modulate genes involved in inflammasome activation, ECM deposition, wound healing, and fibrosis.


Subject(s)
Collagen , Dental Pulp , Fibroblasts , Inflammasomes , Stem Cells , Dental Pulp/cytology , Dental Pulp/metabolism , Fibroblasts/metabolism , Humans , Inflammasomes/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Collagen/metabolism , Coculture Techniques , Extracellular Matrix/metabolism , Cells, Cultured , Cytokines/metabolism , Dermis/cytology , Dermis/metabolism , Interleukin-1beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL