Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Biomed Pharmacother ; 143: 112151, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34507115

ABSTRACT

Wound healing is a public health concern. Licorice gained a great attention for its antioxidant and anti-inflammatory properties which expand its valuable effects as a herbal medicine. In this study, we pointed out to the wound healing potential and the mechanism by which licorice alcoholic extract can modulate cutaneous wound healing through immune, antioxidant, histopathological, immunohistochemical (IHC) and molecular studies. 24 Wister rats were assigned into 3 groups (n = 8 each); control group, topical and oral supplied groups. Licorice extract administration significantly increased total and differential leucocyte counts, phagocytic activity of neutrophils, antioxidant biomarkers as superoxide dismutase (SOD), glutathione peroxidase activities (GPx) and reduced glutathione (GSH) content with a notable reduction in oxidative stress marker malondialdehyde (MDA). Moreover, histopathological findings detected complete re-epithelialization with increasing collagen synthesis while IHC results revealed a significant enhancement in the expression of α-SMA, PDGFR-α, FGFR1 and Cytokeratin 14 in licorice treated groups compared with the control group. Licorice extract supplementation accelerated wound healing by increasing angiogenesis and collagen deposition through up-regulation of bFGF, VEGF and TGF-ß gene expression levels compared with the control group. UPLC-PDA-MS/MS aided to authenticate the studied Glycyrrihza species and recognized 101 potential constituents that may be responsible for licorice-exhibited potentials. Based on our observations we concluded that licorice enhanced cutaneous wound healing via its free radical-scavenging potential, potent antioxidant activities, and anti-inflammatory actions. Therefore, licorice could be used as a potential alternative therapy for wound injury which could overcome the associated limitations of modern therapeutic products.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Glycyrrhiza , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Skin/drug effects , Wound Healing/drug effects , Wounds, Penetrating/drug therapy , Angiogenesis Inducing Agents/isolation & purification , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Disease Models, Animal , Gene Expression Regulation , Glycyrrhiza/chemistry , Inflammation Mediators/metabolism , Male , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Rats, Wistar , Reactive Oxygen Species/metabolism , Skin/injuries , Skin/metabolism , Skin/pathology , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism , Wounds, Penetrating/pathology
2.
STAR Protoc ; 2(3): 100650, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34278336

ABSTRACT

Platelet preparations are commonly used in the clinic in combination with mesenchymal stem cells (MSCs) to improve their wound healing capacity and optimize their therapeutic efficacy following their delivery into diseased tissues. To investigate the mechanisms by which platelets enhance the repair properties of MSCs, we detail a protocol using a humanized mouse model for excisional wounds to study by reverse transcription real-time PCR whether human platelets alter the therapeutic efficacy of grafted human MSCs. For complete details on the use and execution of this protocol, please refer to Levoux et al. (2021).


Subject(s)
Gene Expression Profiling/methods , Mesenchymal Stem Cell Transplantation/methods , Platelet Transfusion/methods , Wounds, Penetrating/therapy , Animals , Blood Platelets , Heterografts , Humans , Male , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Wounds, Penetrating/genetics
3.
Physiol Genomics ; 52(12): 575-587, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33017228

ABSTRACT

As a consequence of military operations, many veterans suffer from penetrating wounds and long-term retention of military-grade heavy metal fragments. Fragments vary in size and location, and complete surgical removal may not be feasible or beneficial in all cases. Increasing evidence suggests retention of heavy metal fragments may have serious biological implications, including increased risks for malignant transformation. Previous studies assessed the tumorigenic effects of metal alloys in rats, demonstrating combinations of metals are sufficient to induce tumor formation after prolonged retention in skeletal muscle tissue. In this study, we analyzed transcriptional changes in skeletal muscle tissue in response to eight different military-relevant pure metals over 12 mo. We found that most transcriptional changes occur at 1 and 3 mo after metal pellets are embedded in skeletal muscle and these effects resolve at 6 and 12 mo. We also report significant immunogenic effects of nickel and cobalt and suppressive effects of lead and depleted uranium on gene expression. Overall, skeletal muscle exhibits a remarkable capacity to adapt to and recover from internalized metal fragments; however, the cellular response to chronic exposure may be restricted to the metal-tissue interface. These data suggest that unless affected regions are specifically captured by biopsy, it would be difficult to reliably detect changes in muscle gene expression that would be indicative of long-term adverse health outcomes.


Subject(s)
Gene Expression , Metals, Heavy , Muscle, Skeletal , Transcriptome , Wounds, Penetrating/genetics , Animals , Carcinogens , Male , Models, Animal , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Time Factors
4.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998232

ABSTRACT

Estrogenic steroids and adenosine A2A receptors promote the wound healing and angiogenesis processes. However, so far, it is unclear whether estrogen may regulate the expression and pro-angiogenic activity of A2A receptors. Using in vivo analyses, we showed that female wild type (WT) mice have a more rapid wound healing process than female or male A2A-deficient mice (A2AKO) mice. We also found that pulmonary endothelial cells (mPEC) isolated from female WT mice showed higher expression of A2A receptor than mPEC from male WT mice. mPEC from female WT mice were more sensitive to A2A-mediated pro-angiogenic response, suggesting an ER and A2A crosstalk, which was confirmed using cells isolated from A2AKO. In those female cells, 17ß-estradiol potentiated A2A-mediated cell proliferation, an effect that was inhibited by selective antagonists of estrogen receptors (ER), ERα, and ERß. Therefore, estrogen regulates the expression and/or pro-angiogenic activity of A2A adenosine receptors, likely involving activation of ERα and ERß receptors. Sexual dimorphism in wound healing observed in the A2AKO mice process reinforces the functional crosstalk between ER and A2A receptors.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Neovascularization, Physiologic/drug effects , Receptor, Adenosine A2A/genetics , Wounds, Penetrating/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Phenethylamines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Cross-Talk , Receptor, Adenosine A2A/metabolism , Sex Factors , Signal Transduction , Wound Healing/drug effects , Wound Healing/genetics , Wounds, Penetrating/drug therapy , Wounds, Penetrating/metabolism , Wounds, Penetrating/pathology
5.
Dis Model Mech ; 13(6)2020 06 15.
Article in English | MEDLINE | ID: mdl-32541065

ABSTRACT

Tissue fibrosis is the deposition of excessive extracellular matrix and can occur as part of the body's natural wound healing process upon injury, or as a consequence of diseases such as systemic sclerosis. Skin fibrosis contributes to significant morbidity due to the prevalence of injuries resulting from trauma and burn. Fibroblasts, the principal cells of the dermis, synthesize extracellular matrix to maintain the skin during homeostasis and also play a pivotal role in all stages of wound healing. Although it was previously believed that fibroblasts are homogeneous and mostly quiescent cells, it has become increasingly recognized that numerous fibroblast subtypes with unique functions and morphologies exist. This Review provides an overview of fibroblast heterogeneity in the mammalian dermis. We explain how fibroblast identity relates to their developmental origin, anatomical site and precise location within the skin tissue architecture in both human and mouse dermis. We discuss current evidence for the varied functionality of fibroblasts within the dermis and the relationships between fibroblast subtypes, and explain the current understanding of how fibroblast subpopulations may be controlled through transcriptional regulatory networks and paracrine communications. We consider how fibroblast heterogeneity can influence wound healing and fibrosis, and how insight into fibroblast heterogeneity could lead to novel therapeutic developments and targets for skin fibrosis. Finally, we contemplate how future studies should be shaped to implement knowledge of fibroblast heterogeneity into clinical practice in order to lessen the burden of skin fibrosis.


Subject(s)
Cicatrix/pathology , Extracellular Matrix/pathology , Fibroblasts/pathology , Skin/pathology , Wound Healing , Wounds, Penetrating/pathology , Animals , Cicatrix/genetics , Cicatrix/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Humans , Phenotype , Signal Transduction , Skin/injuries , Skin/metabolism , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
6.
J Pathol ; 249(4): 435-446, 2019 12.
Article in English | MEDLINE | ID: mdl-31342513

ABSTRACT

Diabetes induces dysregulation throughout the spectrum of myeloid lineage cells from progenitors to terminally differentiated cells. Another complication of diabetes is persistent inflammation, including prolonged accumulation of macrophages, which contributes to impaired wound healing. However, it remains unclear whether diabetes disrupts the response of bone marrow progenitors to peripheral injury and whether such dysregulation leads to sustained inflammation and impaired healing. Here, we demonstrated that diabetic mice (db/db, referred to here as DB) exhibit myeloid lineage bias during homeostasis and following injury. In addition, cells in the LSK (Lin- Sca-1+ cKit+ ) population of DB mice are preprogrammed towards myeloid commitment at the transcriptional level, and cultured myeloid progenitors from DB mice produce more monocytes ex vivo than their non-diabetic counterparts. We also show via bone marrow transfer between interleukin-1 receptor 1 KO (Il1r1-/- ) and DB mice that IL-1R1 signaling is likely not involved in myeloid skewing in DB mice. Furthermore, in vitro experiments indicated that macrophage colony-stimulating factor receptor signaling is not likely involved in enhanced myeloid transcription factor expression in LSK cells of DB mice. Our findings indicate that myeloid lineage commitment in bone marrow may contribute to increased macrophage numbers observed in diabetic skin wounds, and that strategies to regulate monopoiesis during homeostasis or post-wounding may improve diabetic wound healing. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Lineage , Diabetes Mellitus, Type 2/pathology , Macrophages/pathology , Myeloid Progenitor Cells/pathology , Skin/pathology , Wound Healing , Wounds, Penetrating/pathology , Animals , Bone Marrow Transplantation , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/metabolism , Myelopoiesis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , Skin/injuries , Skin/metabolism , Stem Cell Transplantation , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
7.
Arch Dermatol Res ; 310(9): 737-750, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30209580

ABSTRACT

CD271, a receptor of nerve growth factor (NGF), affects the biological properties of epidermal stem cells (eSCs) which are essential for skin wound closure. Tropomyosin-receptor kinase A (TrkA), another receptor of NGF, combined with CD271 has been involved with nervous system and skin keratinocytes. However, the exact role of TrkA combined with CD271 in eSCs during skin wound closure is still unclear. This study aimed to reveal the role of TrkA in the promoting wounding-healing effect of CD271 on eSCs. We obtained CD271-vo (over-expression of CD271) eSCs by lentiviral infection. K252a was used to inhibit TrkA expression. Full-thickness skin mouse wound closure model (5 mm in diameter) was used to detect the ability of CD271 over-expressed/TrkA-deficient during wound healing. The biological characteristics of eSCs and their proliferation and apoptosis were detected using immunohistochemistry and western blot. The expressions of protein kinase B (pAkt)/Akt, phosphorylated extracellular-signal-related kinase (pERK)/ERK1/2, and c-Jun N-terminal kinase (pJNK)/JNK were also detected by western blot. We found that over-expression of CD271 promoted the biological functions of eSCs. Interestingly, over-expression of CD271 in the absence of TrkA neither promoted eSCs' migration and proliferation nor promoted wound healing in a mouse model. In addition, we observed the reduced expression of pAkt/Akt and pERK/ERK1/2 following TrkA inhibition in vitro. Our studies demonstrated that the role of TrkA in the promoting wounding-healing effect of CD271 on eSCs.


Subject(s)
Epidermis/metabolism , Nerve Tissue Proteins/physiology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/physiology , Stem Cells/metabolism , Wound Healing , Wounds, Penetrating/metabolism , Animals , Carbazoles/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epidermis/drug effects , Epidermis/injuries , Epidermis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Indole Alkaloids/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/antagonists & inhibitors , Signal Transduction , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/pathology , Wound Healing/drug effects , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology
8.
Nat Cell Biol ; 20(4): 422-431, 2018 04.
Article in English | MEDLINE | ID: mdl-29593327

ABSTRACT

During fetal development, mammalian back-skin undergoes a natural transition in response to injury, from scarless regeneration to skin scarring. Here, we characterize dermal morphogenesis and follow two distinct embryonic fibroblast lineages, based on their history of expression of the engrailed 1 gene. We use single-cell fate-mapping, live three dimensional confocal imaging and in silico analysis coupled with immunolabelling to reveal unanticipated structural and regional complexity and dynamics within the dermis. We show that dermal development and regeneration are driven by engrailed 1-history-naive fibroblasts, whose numbers subsequently decline. Conversely, engrailed 1-history-positive fibroblasts possess scarring abilities at this early stage and their expansion later on drives scar emergence. The transition can be reversed, locally, by transplanting engrailed 1-naive cells. Thus, fibroblastic lineage replacement couples the decline of regeneration with the emergence of scarring and creates potential clinical avenues to reduce scarring.


Subject(s)
Cell Lineage , Cell Proliferation , Cicatrix/pathology , Fibroblasts/metabolism , Regeneration , Skin/metabolism , Wounds, Penetrating/pathology , Animals , Cell Movement , Cell Tracking , Cells, Cultured , Cicatrix/genetics , Cicatrix/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibroblasts/transplantation , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Morphogenesis , Phenotype , Signal Transduction , Single-Cell Analysis , Skin/injuries , Skin/pathology , Skin Transplantation , Time Factors , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
9.
Acta Derm Venereol ; 97(5): 593-600, 2017 May 08.
Article in English | MEDLINE | ID: mdl-28127619

ABSTRACT

Stem cells have recently been shown to play important roles in wound healing. The aim of this study was to investigate the role of dermal CD271+ cells in wound healing. Full-thickness wounds were produced on the backs of 5-year-old and 24-week-old mice, and time-course of wound closure, CD271+ cell counts, and gene expression levels were compared. Delayed wound healing was observed in 24-week-old mice. The peak of CD271+ cell increase was delayed in 24-week-old mice, and gene expression levels of growth factors in wounded tissue were significantly increased in 5-year-old mice. Dermal CD271+ cells purified by fluorescence-activated cell sorting (FACS) expressed higher growth factors than CD271- cells, suggesting that CD271+ cells play important roles by producing growth factors. This study also investigated dermal CD271+ cells in patients with chronic skin ulcers. Dermal CD271+ cells in patients were significantly reduced compared with in healthy controls. Thus, dermal CD271+ cells are closely associated with wound healing.


Subject(s)
Adapalene/immunology , Cell Proliferation , Nerve Tissue Proteins/immunology , Receptors, Nerve Growth Factor/immunology , Skin Ulcer/immunology , Skin/immunology , Stem Cells/immunology , Wound Healing , Wounds, Penetrating/immunology , Adapalene/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Aging/pathology , Animals , Case-Control Studies , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Male , Mice , Middle Aged , Phenotype , Skin/injuries , Skin/metabolism , Skin/pathology , Skin Ulcer/metabolism , Skin Ulcer/pathology , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism , Wounds, Penetrating/mortality
10.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 151-168, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27816443

ABSTRACT

Lucidone, which comprises a naturally occurring cyclopentenedione, has been investigated for its in vitro and in vivo wound healing properties, and the underlying molecular signaling cascades in the wound healing mechanism have been elucidated. We demonstrated the cell-/dose-specific responses of lucidone (0.5-8µM) on proliferation and migration/invasion of keratinocyte HaCaT and fibroblast Hs68 cells. In keratinocytes, lucidone-induced nuclear translocation of ß-catenin was accompanied by increased transcriptional target genes, including c-Myc and cyclin-D1, through GSK3ß-dependent pathway. Correspondingly, lucidone promoted the cell-cycle by increasing PCNA/CDK4 and decreasing p21/p27 expressions. Lucidone induced EMT through the downregulation of epithelial (E-cadherin/occludin) and upregulation of mesenchymal (vimentin/Twist/Snail) marker proteins. Activated MMP-9/-2 and uPA/uPAR as well as suppressed TIMP-1/-2 and PAI-1 expressions by lucidone may promote the migration/invasion of keratinocytes. Notably, lucidone activated NF-κB signaling via IKK-mediated-IκB degradation, and its inhibition abolished MMP-9 activation and keratinocyte migration. Inhibition of PI3K/AKT signaling impaired the lucidone-induced proliferation/migration with corresponding suppression of ß-catenin/c-Myc/cyclin-D1 and NF-κB/MMP-9 expressions. Results indicate that lucidone-induced PI3K/AKT signaling anchored the ß-catenin/NF-κB-mediated healing mechanism. ß-catenin knockdown substantially diminished lucidone-induced keratinocyte migration. Furthermore, lucidone increased endothelial cell proliferation/migration and triggered angiogenesis (MMP-9/uPA/ICAM-1). In macrophages, lucidone-activated NF-κB-mediated inflammation (COX-2/iNOS/NO) and VEGF, which may contribute to the growth of keratinocytes/fibroblasts and endothelial cells. Punched wounds on mice were rapidly healed with the topical application of lucidone (5mM) compared with control ointment-treated mice. Taken together, lucidone accelerates wound healing through the cooperation of keratinocyte/fibroblast/endothelial cell growth and migration and macrophage inflammation via PI3K/AKT, Wnt/ß-catenin and NF-κB signaling cascade activation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclopentanes/pharmacology , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Wound Healing/drug effects , Wounds, Penetrating/drug therapy , beta Catenin/genetics , Animals , Cell Line , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Wound Healing/physiology , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism , Wounds, Penetrating/pathology , beta Catenin/metabolism
11.
Adv Exp Med Biol ; 913: 177-191, 2016.
Article in English | MEDLINE | ID: mdl-27796888

ABSTRACT

In this chapter, we outline the role of human CD34+ stromal cells/telocytes (CD34+ SC/TCs) as progenitor cells during repair. The in vivo activation phenomena of CD34+ SC/TCs in this process include increased size; separation from the neighbouring structures (mainly of the vascular walls); association with inflammatory cells, predominantly macrophages; development of the organelles of synthesis (rough endoplasmic reticulum and Golgi apparatus); cell proliferation with presence of mitosis and high proliferative index (transit-amplifying cells); and fibroblastic and myofibroblastic differentiation. A procedure to study these tissue-resident cells, comparison of their behaviour in vivo and in vitro and different behaviour depending on location, time, type of injury (including tumour stroma) and greater or lesser proximity to the injury are also considered.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Macrophages/pathology , Neoplasms/pathology , Stem Cells/cytology , Telocytes/cytology , Wounds, Penetrating/pathology , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Biomarkers/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression , Humans , Inflammation , Macrophages/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Stem Cells/metabolism , Telocytes/metabolism , Wound Healing , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
12.
Nat Commun ; 7: 11394, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27099134

ABSTRACT

Notch has a well-defined role in controlling cell fate decisions in the embryo and the adult epidermis and immune systems, yet emerging evidence suggests Notch also directs non-cell-autonomous signalling in adult tissues. Here, we show that Notch1 works as a damage response signal. Epidermal Notch induces recruitment of immune cell subsets including RORγ(+) ILC3s into wounded dermis; RORγ(+) ILC3s are potent sources of IL17F in wounds and control immunological and epidermal cell responses. Mice deficient for RORγ(+) ILC3s heal wounds poorly resulting from delayed epidermal proliferation and macrophage recruitment in a CCL3-dependent process. Notch1 upregulates TNFα and the ILC3 recruitment chemokines CCL20 and CXCL13. TNFα, as a Notch1 effector, directs ILC3 localization and rates of wound healing. Altogether these findings suggest that Notch is a key stress/injury signal in skin epithelium driving innate immune cell recruitment and normal skin tissue repair.


Subject(s)
Epidermis/immunology , Immunity, Innate , Lymphocyte Subsets/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Receptor, Notch1/immunology , Wounds, Penetrating/immunology , Animals , Cell Movement/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Epidermis/injuries , Female , Gene Expression Regulation , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptor, Notch1/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Wound Healing/genetics , Wound Healing/immunology , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology
13.
J Steroid Biochem Mol Biol ; 164: 379-385, 2016 11.
Article in English | MEDLINE | ID: mdl-26282157

ABSTRACT

Wound healing is essential for survival. This is a multistep process involving a number of different cell types. In the skin wounding triggers an acute inflammatory response, with the innate immune system contributing both to protection against invasive organisms and to triggering the invasion of inflammatory cells into the wounded area. These cells release a variety of cytokines and growth factors that stimulate the proliferation and migration of dermal and epidermal cells to close the wound. In particular, wounding activates stem cells in the interfollicular epidermis (IFE) and hair follicles (HF) to proliferate and send their progeny to re-epithelialize the wound. ß-catenin and calcium signaling are important for this activation process. Mice lacking the VDR when placed on a low calcium diet have delayed wound healing. This is associated with reduced ß-catenin transcriptional activity and proliferation in the cells at the leading edge of wound closure. These data suggest that vitamin D and calcium signaling are necessary components of the epidermal response to wounding, likely by regulating stem cell activation through increased ß-catenin transcriptional activity.


Subject(s)
Calcium/metabolism , Epidermis/metabolism , Receptors, Calcitriol/genetics , Vitamin D/metabolism , Wound Healing/genetics , Wounds, Penetrating/metabolism , beta Catenin/genetics , Animals , Calcium Signaling , Cell Movement , Cell Proliferation , Epidermal Cells , Epidermis/injuries , Female , Gene Expression Profiling , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Calcitriol/deficiency , Stem Cells/cytology , Stem Cells/metabolism , Transcription, Genetic , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology , beta Catenin/metabolism
14.
PLoS One ; 10(11): e0142046, 2015.
Article in English | MEDLINE | ID: mdl-26535894

ABSTRACT

DNA methylation is a key epigenetic mechanism controlling DNA accessibility and gene expression. Blockade of DNA methylation can significantly affect pain behaviors implicated in neuropathic and inflammatory pain. However, the role of DNA methylation with regard to postoperative pain has not yet been explored. In this study we sought to investigate the role of DNA methylation in modulating incisional pain and identify possible targets under DNA methylation and contributing to incisional pain. DNA methyltranferase (DNMT) inhibitor 5-Aza-2'-deoxycytidine significantly reduced incision-induced mechanical allodynia and thermal sensitivity. Aza-2'-deoxycytidine also reduced hindpaw swelling after incision, suggesting an anti-inflammatory effect. Global DNA methylation and DNMT3b expression were increased in skin after incision, but none of DNMT1, DNMT3a or DNMT3b was altered in spinal cord or DRG. The expression of proopiomelanocortin Pomc encoding ß-endorphin and Oprm1 encoding the mu-opioid receptor were upregulated peripherally after incision; moreover, Oprm1 expression was further increased under DNMT inhibitor treatment. Finally, local peripheral injection of the opioid receptor antagonist naloxone significantly exacerbated incision-induced mechanical hypersensitivity. These results suggest that DNA methylation is functionally relevant to incisional nociceptive sensitization, and that mu-opioid receptor signaling might be one methylation regulated pathway controlling sensitization after incision.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation , Edema/drug therapy , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Nociception/drug effects , Wounds, Penetrating/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Decitabine , Edema/etiology , Edema/pathology , Hindlimb/drug effects , Hindlimb/injuries , Hindlimb/metabolism , Male , Mice , Mice, Inbred C57BL , Pain Measurement , Real-Time Polymerase Chain Reaction , Receptors, Opioid, mu/genetics , Signal Transduction/drug effects , Wounds, Penetrating/genetics
15.
Cell Cycle ; 14(21): 3408-17, 2015.
Article in English | MEDLINE | ID: mdl-26398918

ABSTRACT

Upon wounding, multiple stem cell populations in the hair follicle (HF) and interfollicular epidermis (IFE) converge at the site of injury. Although these cells can contribute permanently to the regenerating epithelium, it remains unclear whether these contributions vary among cells originating from diverse compartments in the skin. By comparing the fates of several keratinocyte lineages, we observed here an initial decrease in both HF- and IFE-derived cells within the transient acanthotic layers of the regenerating epithelium. At the same time, the relative abundance of early-arriving IFE-derived cells specifically in the wound basal layer declined as later-arriving HF-derived cells entered the site of injury. Although laggard bulge-derived cells were typically constrained at the regenerative periphery, these cells persisted in the wound basal layer. Finally, suppressing Notch enabled IFE-derived cells to out-compete HF-derived cells. Taken together, these findings indicate that IFE-, HF- and bulge-derived cells make distinct contributions to regeneration over time. Furthermore, we speculate that extrinsic, non-genetic factors such as spatial constraint, distance from the wound, and basal versus suprabasal position may largely determine whether a cell ultimately persists.


Subject(s)
Epidermis/pathology , Hair Follicle/pathology , Stem Cells/pathology , Wound Healing , Wounds, Penetrating/pathology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Disease Models, Animal , Epidermis/injuries , Epidermis/metabolism , Genes, Reporter , Genotype , Hair Follicle/injuries , Hair Follicle/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Transgenic , Phenotype , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism , Time Factors , Wounds, Penetrating/genetics , Wounds, Penetrating/metabolism
16.
Reprod Sci ; 22(11): 1377-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25868581

ABSTRACT

OBJECTIVE: To establish a mouse model for endometrial injury and determine the underlying mechanism regarding its favorable effect on embryo implantation. STUDY DESIGN: Female Kunming mice were randomly allocated into 4 groups: group I, normal control; group II, injury procedure control; and group III and group IV, the mice being scratched with a blunt syringe on the right uterine horn or both, respectively. All the mice were mated with the males during the next estrus phase. The number of implanted embryos on each side of uterus was calculated on day 8 of pregnancy. The endometrial samples were taken on day 4 of pregnancy, and the local morphological changes and cytokine expressions were examined. RESULTS: Compared to group II, our results showed that in group IV (1) there were significantly higher numbers of implanted embryos, (2) the endometrial glands and vasculatures in stroma were obviously increased and the pinopodes were abundant and well developed, and (3) the local levels of cytokines leukemia inhibitory factor (LIF) and oncostatin M (OSM) messenger RNA and protein expression were significantly increased. CONCLUSIONS: Local mechanical injury on mouse uteri enhanced endometrial receptivity and improved embryo implantation, which were correlated with the characteristic changes in endometrial morphology and the upregulation of LIF and OSM gene and protein expression.


Subject(s)
Cytokines/metabolism , Embryo Implantation , Endometrium/metabolism , Wounds, Penetrating/metabolism , Animals , Cytokines/genetics , Disease Models, Animal , Endometrium/injuries , Endometrium/physiopathology , Endometrium/ultrastructure , Female , Gene Expression Regulation, Developmental , Gestational Age , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Mice , Oncostatin M/genetics , Oncostatin M/metabolism , Pregnancy , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Up-Regulation , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology , Wounds, Penetrating/physiopathology
17.
Cell Mol Biol (Noisy-le-grand) ; 61(8): 89-96, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26718435

ABSTRACT

The larvae of Lucilia sericata have been used for centuries as medicinal maggots in the healing of wounds. The present study aimed to screen potential microRNAs related to ES-induced wound healing in rat skin wounds and to investigate the potential mechanisms contributing to accelerated wound healing. Healthy, male, 12 weeks old Wistar albino rats weighing 250-300 g were supplied by the Animal Experimental Center. All animal studies were performed in accordance with the NIH Guide for the Care and Use of Laboratory Animals. Wistar albino rats were treated by ES after post wounding and the differentially expressed miRNAs in wound biopsies were screened by microarray analysis at the end of treatments for 4,7 and 10 days. In addition, bioinformatics approaches were used to identify the potential target genes of differentially expressed miRNAs and the functions of their target genes. We found a significant up-regulation of rno-miR-99a* and rno-mir-877 in response to ES treatment. Further investigation of rno-miR-99a* and rno-mir-877 and their target genes (TGFa, TNF, TAGLN, MAPK1, MMP-9) implicated in present study could provide new insight for an understanding lead to the development of new treatment strategies. The identified miRNAs can be new biomarkers for ES- induced wound healing.


Subject(s)
Bodily Secretions/chemistry , Complementary Therapies/methods , MicroRNAs/genetics , Wound Healing/genetics , Wounds, Penetrating/therapy , Animals , Bodily Secretions/metabolism , Computational Biology/methods , Diptera/chemistry , Diptera/physiology , Gene Expression Profiling , Gene Expression Regulation , Larva/chemistry , Larva/physiology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Rats , Rats, Wistar , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology
19.
Clin Orthop Relat Res ; 472(2): 396-404, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136804

ABSTRACT

BACKGROUND: Heterotopic ossification (HO) is a frequent complication of modern wartime extremity injuries. The biological mechanisms responsible for the development of HO in traumatic wounds remain elusive. QUESTION/PURPOSES: The aims of our study were to (1) characterize the expression profile of osteogenesis-related gene transcripts in traumatic war wounds in which HO developed; and (2) determine whether expression at the mRNA level correlated with functional protein expression and HO formation. METHODS: Biopsy specimens from 54 high-energy penetrating extremity wounds obtained at the initial and final surgical débridements were evaluated. The levels of selected osteogenic-related gene transcripts from RNA extracts were assessed by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) analysis. As a result of its key role in osteogenesis, the concentration of BMP-2 in the effluent of 29 wounds also was determined. RESULTS: The transcripts of 13 genes (ALPL [p = 0.006], BMP-2 [p < 0.001], BMP-3 [p = 0.06], COL2A1 [p < 0.001], COLL10A1 [p < 0.001], COL11A1 [p = 0.006], COMP [p = 0.02], CSF2 [p = 0.003], CSF3 [p = 0.012], MMP8 [p < 0.001], MMP9 [p = 0.014], SMAD1 [p = 0.024], and VEGFA [p = 0.017]) were upregulated greater than twofold in wounds in which HO developed compared with wounds in which it did not develop. Gene transcript expression of BMP-2 also correlated directly with functional protein expression in the wounds that formed HO (p = 0.029). CONCLUSIONS: Important differences exist in the osteogenic gene expression profile of wounds in which HO developed compared with wounds in which it did not develop. The upregulation of multiple osteogenesis-related gene transcripts indicates the presence of a proosteogenic environment necessary for ectopic bone formation in traumatic wounds. CLINICAL RELEVANCE: Understanding the osteogenic environment associated with war wounds may allow for the development of novel therapeutic strategies for HO.


Subject(s)
Afghan Campaign 2001- , Iraq War, 2003-2011 , Military Medicine , Ossification, Heterotopic/genetics , Osteogenesis/genetics , Wounds, Penetrating/genetics , Adolescent , Adult , Biopsy , Bone Morphogenetic Protein 2/analysis , Bone Morphogenetic Protein 2/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Markers , Humans , Male , Military Personnel , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/prevention & control , Prognosis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Wounds, Penetrating/complications , Wounds, Penetrating/metabolism , Wounds, Penetrating/therapy , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...