Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Invest Dermatol ; 142(4): 1206-1216.e8, 2022 04.
Article En | MEDLINE | ID: mdl-34710388

Nonhealing wounds are a major area of unmet clinical need remaining problematic to treat. Improved understanding of prohealing mechanisms is invaluable. The enzyme arginase1 (ARG1) is involved in prohealing responses, with its role in macrophages best characterized. ARG1 is also expressed by keratinocytes; however, ARG1 function in these critical wound repair cells is not understood. We characterized ARG1 expression in keratinocytes during normal cutaneous repair and reveal de novo temporal and spatial expression at the epidermal wound edge. Interestingly, epidermal ARG1 expression was decreased in both human and murine delayed healing wounds. We therefore generated a keratinocyte-specific ARG1-null mouse model (K14-cre;Arg1fl/fl) to explore arginase function. Wound repair, linked to changes in keratinocyte proliferation, migration, and differentiation, was significantly delayed in K14-cre;Arg1fl/fl mice. Similarly, using the arginase inhibitor N(omega)-hydroxy-nor-L-arginine, human in vitro and ex vivo models further confirmed this finding, revealing the importance of the downstream polyamine pathway in repair. Indeed, restoring the balance in ARG1 activity through the addition of putrescine proved beneficial in wound closure. In summary, we show that epidermal ARG1 plays, to our knowledge, a previously unreported intrinsic role in cutaneous healing, highlighting epidermal ARG1 and the downstream mediators as potential targets for the therapeutic modulation of wound repair.


Arginase , Skin Abnormalities , Animals , Arginase/genetics , Arginase/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Skin/metabolism , Skin Abnormalities/metabolism
2.
J Invest Dermatol ; 141(7): 1633-1645.e13, 2021 07.
Article En | MEDLINE | ID: mdl-33493531

Hair follicles (HFs) are immersed within dermal white adipose tissue (dWAT), yet human adipocyte‒HF communication remains unexplored. Therefore, we investigated how perifollicular adipocytes affect the physiology of human anagen scalp HFs. Quantitative immunohistomorphometry, X-ray microcomputed tomography, and transmission electron microscopy showed that the number and size of perifollicular adipocytes declined during anagen‒catagen transition, whereas fluorescence-lifetime imaging revealed increased lipid oxidation in adipocytes surrounding the bulge and/or sub-bulge region. Ex vivo, dWAT tendentially promoted hair shaft production, and significantly stimulated hair matrix keratinocyte proliferation and HF pigmentation. Both dWAT pericytes and PREF1/DLK1+ adipocyte progenitors secreted HGF during human HF‒dWAT co-culture, for which the c-Met receptor was expressed in the hair matrix and dermal papilla. These effects were reproduced using recombinant HGF and abrogated by an HGF-neutralizing antibody. Laser-capture microdissection‒based microarray analysis of the hair matrix showed that dWAT-derived HGF upregulated keratin (K) genes (K27, K73, K75, K84, K86) and TCHH. Mechanistically, HGF stimulated Wnt/ß-catenin activity in the human hair matrix (increased AXIN2, LEF1) by upregulating WNT6 and WNT10B, and inhibiting SFRP1 in the dermal papilla. Our study demonstrates that dWAT regulates human hair growth and pigmentation through HGF secretion, and thus identifies dWAT and HGF as important novel molecular and cellular targets for therapeutic intervention in human hair growth and pigmentation disorders.


Hair Color , Hair Follicle/growth & development , Hepatocyte Growth Factor/metabolism , Pigmentation , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Cells, Cultured , Coculture Techniques , Hair Follicle/diagnostic imaging , Hair Follicle/metabolism , Humans , Keratinocytes/physiology , Laser Capture Microdissection , Primary Cell Culture , Wnt Signaling Pathway , X-Ray Microtomography
3.
Biol Rev Camb Philos Soc ; 95(3): 592-624, 2020 06.
Article En | MEDLINE | ID: mdl-31970855

The nervous system communicates with peripheral tissues through nerve fibres and the systemic release of hypothalamic and pituitary neurohormones. Communication between the nervous system and the largest human organ, skin, has traditionally received little attention. In particular, the neuro-regulation of sebaceous glands (SGs), a major skin appendage, is rarely considered. Yet, it is clear that the SG is under stringent pituitary control, and forms a fascinating, clinically relevant peripheral target organ in which to study the neuroendocrine and neural regulation of epithelia. Sebum, the major secretory product of the SG, is composed of a complex mixture of lipids resulting from the holocrine secretion of specialised epithelial cells (sebocytes). It is indicative of a role of the neuroendocrine system in SG function that excess circulating levels of growth hormone, thyroxine or prolactin result in increased sebum production (seborrhoea). Conversely, growth hormone deficiency, hypothyroidism, and adrenal insufficiency result in reduced sebum production and dry skin. Furthermore, the androgen sensitivity of SGs appears to be under neuroendocrine control, as hypophysectomy (removal of the pituitary) renders SGs largely insensitive to stimulation by testosterone, which is crucial for maintaining SG homeostasis. However, several neurohormones, such as adrenocorticotropic hormone and α-melanocyte-stimulating hormone, can stimulate sebum production independently of either the testes or the adrenal glands, further underscoring the importance of neuroendocrine control in SG biology. Moreover, sebocytes synthesise several neurohormones and express their receptors, suggestive of the presence of neuro-autocrine mechanisms of sebocyte modulation. Aside from the neuroendocrine system, it is conceivable that secretion of neuropeptides and neurotransmitters from cutaneous nerve endings may also act on sebocytes or their progenitors, given that the skin is richly innervated. However, to date, the neural controls of SG development and function remain poorly investigated and incompletely understood. Botulinum toxin-mediated or facial paresis-associated reduction of human sebum secretion suggests that cutaneous nerve-derived substances modulate lipid and inflammatory cytokine synthesis by sebocytes, possibly implicating the nervous system in acne pathogenesis. Additionally, evidence suggests that cutaneous denervation in mice alters the expression of key regulators of SG homeostasis. In this review, we examine the current evidence regarding neuroendocrine and neurobiological regulation of human SG function in physiology and pathology. We further call attention to this line of research as an instructive model for probing and therapeutically manipulating the mechanistic links between the nervous system and mammalian skin.


Brain/physiology , Sebaceous Glands/innervation , Sebaceous Glands/metabolism , Skin Physiological Phenomena , Skin/pathology , Animals , Dopamine/metabolism , Growth Hormone/metabolism , Humans , Hypothalamo-Hypophyseal System/physiology , Neurosecretory Systems/physiology , Peripheral Nervous System/physiology , Prolactin/metabolism , Sebaceous Glands/anatomy & histology , Sebaceous Glands/cytology , Sebum/chemistry , Sebum/metabolism , Somatomedins/metabolism , Stem Cells , Thyroid Gland/physiology
4.
Wound Repair Regen ; 27(1): 126-133, 2019 01.
Article En | MEDLINE | ID: mdl-30575205

Ex vivo wounded human skin organ culture is an invaluable tool for translationally relevant preclinical wound healing research. However, studies incorporating this system are still underutilized within the field because of the low throughput of histological analysis required for downstream assessment. In this study, we use intravital fluorescent dye to lineage trace epidermal cells, demonstrating that wound re-epithelialization of human ex vivo wounds occurs consistent with an extending shield mechanism of collective migration. Moreover, we also report a relatively simple method to investigate global epithelial closure of explants in culture using daily fluorescent dye treatment and en face imaging. This study is the first to quantify healing of ex vivo wounds in a longitudinal manner, providing global assessments for re-epithelialization and tissue contraction. We show that this approach can identify alterations to healing with a known healing promoter. This methodological study highlights the utility of human ex vivo wounds in enhancing our understanding of mechanisms of human skin repair and in evaluating novel therapies to improve healing outcome.


Cells, Cultured/pathology , Optical Imaging/methods , Re-Epithelialization/physiology , Skin/diagnostic imaging , Wound Healing/physiology , Fluorescent Dyes , Humans , Organ Culture Techniques , Skin/cytology , Skin/injuries
5.
Exp Dermatol ; 27(6): 589-602, 2018 06.
Article En | MEDLINE | ID: mdl-29603400

Dermal white adipose tissue (DWAT) is a main component of human skin, composed of individual lipid-laden mesenchymal cells known as dermal adipocytes (DAs). Besides their well-known role in lipid storage and release, DAs also promote skin immunity, wound healing and hair follicle cycling and are important players in cutaneous neuroendocrinology. The ever-growing insights into DWAT functions, albeit mostly in mice, have invited speculation that it may be involved in multiple skin diseases ranging from fibrosis to alopecia and psoriasis, thus designating human DWAT a clinically relevant, but as yet insufficiently investigated skin compartment. Therefore, this practical, user-friendly guide aims to introduce the techniques available to study human DWAT in situ and ex vivo, including immunohistochemistry, immunofluorescence microscopy and analysis via quantitative immunohistomorphometry. Here, we provide information on a collection of stains comprising pre-adipocyte (Pref1) and mature adipocyte markers (Perilipin1, Caveolin1), as well as various lipid (OilRedO, BODIPY) and histochemical stains (H&E, trichrome) available for use on human DWAT. We offer the reader guidelines on fixing, processing and staining human DAs and highlight caveats and solutions to common problems that one may encounter when studying this fascinating skin compartment. We also suggest standard methods for conducting quantitative immunohistomorphometry on human DWAT and its individual adipocytes to quantify cell size, number, lipid content and fluorescence intensity of adipose-specific markers. Finally, we briefly introduce in situ hybridization, transmission electron microscopy and essentials of magnetic resonance imaging imaging as additional tools for instructively interrogating this largest, but still least-known compartment of human skin.


Adipocytes, White/cytology , Adipocytes, White/physiology , Immunohistochemistry , Skin/cytology , Apoptosis , Biomarkers/metabolism , Cell Culture Techniques , Cell Proliferation , Coloring Agents , Humans , Microscopy/methods , Organ Culture Techniques , Staining and Labeling
6.
J Invest Dermatol ; 138(3): 511-519, 2018 03.
Article En | MEDLINE | ID: mdl-29106928

Epithelial-to-mesenchymal transition (EMT) is critical for embryonic development and wound healing, and occurs in fibrotic disease and carcinoma. Here, we show that EMT also occurs within the bulge, the epithelial stem cell (eSC) niche of human scalp hair follicles, during the inflammatory permanent alopecia, lichen planopilaris. We show that a molecular EMT signature can be experimentally induced in healthy human eSCs in situ by antagonizing E-cadherin, combined with transforming growth factor-ß1, epidermal growth factor, and IFN-γ administration, which to our knowledge has not been reported previously. Moreover, induction of EMT within primary human eSCs can be prevented and even partially reversed ex vivo by peroxisome proliferator-activated receptor-γ agonists, likely through suppression of the transforming growth factor-ß signaling pathway. Furthermore, we show that peroxisome proliferator-activated receptor-γ agonists also attenuates the EMT signature even in lesional lichen planopilaris hair follicles ex vivo. We introduce lichen planopilaris as a model disease for pathological EMT in human adult eSCs, report a preclinical assay for therapeutically manipulating eSC EMT within a healthy human (mini-)organ, and show that peroxisome proliferator-activated receptor-γ agonists are promising agents for suppressing and partially reversing EMT in human hair follicles eSCs ex vivo, including in lichen planopilaris.


Epithelial-Mesenchymal Transition , Lichen Planus/pathology , Mesenchymal Stem Cells/pathology , Adult , Aged , Cells, Cultured , Female , Humans , Keratin-15/analysis , PPAR gamma/physiology , Peroxisomes/drug effects , Pioglitazone/pharmacology , Stem Cell Niche
8.
Heliyon ; 3(5): e00309, 2017 May.
Article En | MEDLINE | ID: mdl-28607955

Wound healing is a complex process regulated by various cell types and a plethora of mediators. While interactions between wounded skin and the hair follicles (HFs) could induce HF neogenesis or promote wound healing, it remains unknown whether the wound healing-associated signaling milieu can be manipulated to protect against alopecia, such as chemotherapy-induced alopecia (CIA). Utilizing a well-established neonatal rat model of CIA, we show here that skin wounding protects from alopecia caused by several clinically relevant chemotherapeutic regimens, and that protection is dependent on the time of wounding and hair cycle stage. Gene expression profiling unveiled a significant increase in interleukin-1 beta (IL-1ß) mediated signaling by skin wounding. Subsequently, we showed that IL-1ß is sufficient and indispensable for mediating the CIA-protective effect. Administration of IL-1ß alone to unwounded rats exhibited local CIA protection while IL-1ß neutralization abrogated CIA protection by wounding. Mechanistically, IL-1ß retarded postnatal HF morphogenesis, making HFs at the wound sites or IL-1ß treated areas damage-resistant while the rats developed total alopecia elsewhere. We conclude that wound healing switches the cutaneous cytokine milieu to an IL-1ß-dominated state thus retarding HF growth progression and rendering the HFs resistant to chemotherapy agents. In the future, manipulation of HF progression through interfering with the IL-1ß signaling milieu may provide therapeutic benefits to a variety of conditions, from prevention of CIA to inhibition of hair growth and treatment of hirsutism.

9.
Exp Dermatol ; 26(2): 101-104, 2017 02.
Article En | MEDLINE | ID: mdl-27574799

The hair follicle has an established role in wound re-epithelialisation, a phenomenon that has been appreciated since at least the first half of the last century. The bulge niche, one location of hair follicle epithelial stem cells has been of particular interest to researchers over recent years, with numerous studies showing its ability to directly contribute to epidermal repair. However, recent work has highlighted other progenitor regions of the hair follicle that appear to act as stem cells during epidermal repair. In addition, several studies within the last 12 months have questioned the importance of the bulge during re-epithelialisation, producing conflicting literature. Here we provide a new model to demonstrate how several important differences in experimental design between studies could account for these seemingly opposing findings, which may have implications for how future studies are conducted.


Epidermis/physiology , Hair Follicle/cytology , Re-Epithelialization , Stem Cells/physiology , Animals , Humans , Models, Biological
10.
Stem Cells ; 34(5): 1377-85, 2016 05.
Article En | MEDLINE | ID: mdl-26756547

The cutaneous healing response has evolved to occur rapidly, in order to minimize infection and to re-establish epithelial homeostasis. Rapid healing is achieved through complex coordination of multiple cell types, which importantly includes specific cell populations within the hair follicle (HF). Under physiological conditions, the epithelial compartments of HF and interfollicular epidermis remain discrete, with K15(+ve) bulge stem cells contributing progeny for HF reconstruction during the hair cycle and as a basis for hair shaft production during anagen. Only upon wounding do HF cells migrate from the follicle to contribute to the neo-epidermis. However, the identity of the first-responding cells, and in particular whether this process involves a direct contribution of K15(+ve) bulge cells to the early stage of epidermal wound repair remains unclear. Here we demonstrate that epidermal injury in murine skin does not induce bulge activation during early epidermal wound repair. Specifically, bulge cells of uninjured HFs neither proliferate nor appear to migrate out of the bulge niche upon epidermal wounding. In support of these observations, Diphtheria toxin-mediated partial ablation of K15(+ve) bulge cells fails to delay wound healing. Our data suggest that bulge cells only respond to epidermal wounding during later stages of repair. We discuss that this response may have evolved as a protective safeguarding mechanism against bulge stem cell exhaust and tumorigenesis. Stem Cells 2016;34:1377-1385.


Hair Follicle/cytology , Re-Epithelialization , Stem Cells/cytology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Integrases/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Stem Cell Niche
14.
Wound Repair Regen ; 22(2): 281-7, 2014.
Article En | MEDLINE | ID: mdl-24635179

Mice represent the most commonly used species for preclinical in vivo research. While incisional and excisional acute murine wound models are both frequently employed, there is little agreement on which model is optimum. Moreover, current lack of standardization of wounding procedure, analysis time point(s), method of assessment, and the use of individual wounds vs. individual animals as replicates makes it difficult to compare across studies. Here we have profiled secondary intention healing of incisional and excisional wounds within the same animal, assessing multiple parameters to determine the optimal methodology for future studies. We report that histology provides the least variable assessment of healing. Furthermore, histology alone (not planimetry) is able to detect accelerated healing in a castrated mouse model. Perhaps most importantly, we find virtually no correlation between wounds within the same animal, suggesting that use of wound (not animal) biological replicates is perfectly acceptable. Overall, these findings should guide and refine future studies, increasing the likelihood of detecting novel phenotypes while reducing the numbers of animals required for experimentation.


Skin/pathology , Wound Healing , Wounds, Penetrating/pathology , Animals , Disease Models, Animal , Mice , Reproducibility of Results , Skin/injuries , Translational Research, Biomedical
15.
Exp Dermatol ; 21(8): 581-5, 2012 Aug.
Article En | MEDLINE | ID: mdl-22775993

Current understanding of the complex process of wound repair is based on decades of study. Integral to this understanding has been the use of in vitro and in vivo models to uncover the key molecular players. Now that major wound processes are more fully understood, therapeutic strategies can be developed to manipulate wound repair. Particularly important areas for future research include developing therapies to aid treatment of healing pathologies such as chronic wounds, and manipulating the normal healing processes to drive a more regenerative phenotype in adults. Here, we discuss the benefits and limitations of current animal-based models and highlight the urgent need for improved predictive preclinical models for wound healing research. We conclude by suggesting directions where more robust models of chronic wound pathologies may arise, expediting the development of novel therapies.


Models, Animal , Models, Biological , Wound Healing/physiology , Ambystoma mexicanum , Animals , Mice , Regeneration/physiology , Skin/injuries , Skin Physiological Phenomena , Swine
16.
J Pathol ; 227(3): 346-56, 2012 Jul.
Article En | MEDLINE | ID: mdl-22407785

Non-healing wounds cause considerable patient morbidity and represent a significant economic burden. Central to wound repair is re-epithelialization, a crucial process involving the modulation of cell adhesion to allow keratinocyte migration to cover the exposed underlying tissues. The cellular mechanisms regulating the earliest stages of re-epithelialization are unclear. We present the first direct evidence that protein kinase Cα (PKCα) plays an important role in regulating wound re-epithelialization. In PKCα(-/-) mice re-epithelialization is delayed, while in novel bitransgenic mice over-expressing constitutively active PKCα it is accelerated. These effects are not due to changes in keratinocyte proliferation, apoptosis or intrinsic cell motility. Instead, they correlate with changes in desmosomal adhesiveness, delay being preceded by retained desmosomal hyper-adhesiveness and acceleration with a rapid switch to desmosomal Ca(2+) -dependence. We demonstrate mechanistic conservation in acute human wounds where PKCα localizes to wound edge desmosomes, which become Ca(2+) -dependent. However, in chronic wounds PKCα remains cytoplasmic and desmosomes fail to switch from the hyper-adhesive state. These results throw new mechanistic light on the earliest stages of wound re-epithelialization and suggest activation of PKCα as a new therapeutic strategy for non-healing wounds.


Cell Adhesion , Desmosomes/enzymology , Keratinocytes/enzymology , Protein Kinase C-alpha/metabolism , Wound Healing , Animals , Apoptosis , Calcium/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Movement , Cell Proliferation , Desmosomes/drug effects , Desmosomes/pathology , Dose-Response Relationship, Drug , Enzyme Activation , Genotype , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Point Mutation , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/deficiency , Protein Kinase C-alpha/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Wound Healing/drug effects
17.
J Invest Dermatol ; 131(2): 518-28, 2011 Feb.
Article En | MEDLINE | ID: mdl-20927125

When the skin is damaged, a variety of cell types must migrate, proliferate, and differentiate to reform a functional barrier to the external environment. Recent studies have shown that progenitor cells residing in hair follicles (HFs) are able to contribute to this re-epithelialization of wounds in vivo. However, the influence of the hair cycle on wound healing has not previously been addressed. Here, we have exploited spontaneous postnatal hair-cycle synchronicity in mice to systematically examine the influence of the different hair-cycle stages on murine skin wound healing. We report significant acceleration of healing during the anagen phase of HF cycling in vivo, associated with alterations in epithelial, endothelial, and inflammatory cell types. Intriguingly, gene profiling data reveal a clear correlation between the transcription of genes beneficial for wound healing and those upregulated during the anagen phase of the hair cycle in unwounded skin. These findings, which demonstrate a previously unappreciated association between HF cycling and wound healing, reveal numerous molecular correlates for further investigation.


Cell Cycle/physiology , Hair Follicle/cytology , Hair Follicle/physiology , Skin Physiological Phenomena , Wound Healing/physiology , Animals , Cell Movement/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Hair Follicle/growth & development , Mice , Mice, Inbred C57BL , Models, Animal , Neovascularization, Physiologic/physiology , Regeneration/physiology , Skin/blood supply , Skin/cytology
...