1
|
Fuchs C, Stalnaker KJ, Dalgard CL, Sukumar G, Hupalo D, Dreyfuss JM, Pan H, Wang Y, Pham L, Wu X, Jozic I, Anderson RR, Cho S, Meyerle JH, Tam J. Plantar Skin Exhibits Altered Physiology, Constitutive Activation of Wound-Associated Phenotypes, and Inherently Delayed Healing. J Invest Dermatol 2024; 144:1633-1648.e14. [PMID: 38237729 DOI: 10.1016/j.jid.2023.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 06/24/2024]
Abstract
Wound research has typically been performed without regard for where the wounds are located on the body, despite well-known heterogeneities in physical and biological properties between different skin areas. The skin covering the palms and soles is highly specialized, and plantar ulcers are one of the most challenging and costly wound types to manage. Using primarily the porcine model, we show that plantar skin is molecularly and functionally more distinct from nonplantar skin than previously recognized, with unique gene and protein expression profiles, broad alterations in cellular functions, constitutive activation of many wound-associated phenotypes, and inherently delayed healing. This unusual physiology is likely to play a significant but underappreciated role in the pathogenesis of plantar ulcers as well as the last 25+ years of futility in therapy development efforts. By revealing this critical yet unrecognized pitfall, we hope to contribute to the development of more effective therapies for these devastating nonhealing wounds.
Collapse
Affiliation(s)
- Christiane Fuchs
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine J Stalnaker
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clifton L Dalgard
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Gauthaman Sukumar
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Daniel Hupalo
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ying Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Linh Pham
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xunwei Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillp Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunghun Cho
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Jon H Meyerle
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Joshua Tam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
2
|
Ma C, Gu C, Lian P, Wazir J, Lu R, Ruan B, Wei L, Li L, Pu W, Peng Z, Wang W, Zong Y, Huang Z, Wang H, Lu Y, Su Z. Sulforaphane alleviates psoriasis by enhancing antioxidant defense through KEAP1-NRF2 Pathway activation and attenuating inflammatory signaling. Cell Death Dis 2023; 14:768. [PMID: 38007430 PMCID: PMC10676357 DOI: 10.1038/s41419-023-06234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 11/27/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects millions of people worldwide. Sulforaphane (SFN) has been shown to have anti-inflammatory and antioxidant properties. In this study, we investigated the effects of SFN on a mouse model of psoriasis induced by imiquimod (IMQ) and its underlying molecular mechanism. Mice treated with SFN showed significant improvement in psoriatic symptoms, including reduced erythema, scales, and cutaneous thickness. Histopathological analysis and immunohistochemical staining revealed decreased expression of K16, K17, and Ki67 in SFN-treated mice, indicating reduced abnormal differentiation of keratinocytes and cutaneous inflammation. SFN treatment also reduced the activation of STAT3 and NF-κB pathways and downregulated pro-inflammatory cytokines IL-1β, IL-6, and CCL2. In vitro experiments using HaCaT cells demonstrated that SFN inhibited IL-22 and TNF-α-induced activation of inflammatory pathways and keratinocyte proliferation. Network pharmacology analysis suggested that the KEAP1-NRF2 pathway might be involved in the protective effects of SFN on psoriasis. We observed reduced NRF2 expression in human psoriatic lesions, and subsequent experiments showed that SFN activated KEAP1-NRF2 pathway in vivo and in vitro. Importantly, NRF2-deficient mice exhibited aggravated psoriasis-like symptoms and reduced response to SFN treatment. Our findings indicate that SFN ameliorates psoriasis symptoms and inflammation through the KEAP1-NRF2 pathway, suggesting a potential therapeutic role for SFN in the treatment of psoriasis.
Collapse
Affiliation(s)
- Chujun Ma
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
- Department of Dermatology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China
| | - Chaode Gu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Panpan Lian
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Renwei Lu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Binjia Ruan
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Ziqi Peng
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Wentong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Yangyongyi Zong
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, PR China.
| | - Yan Lu
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
3
|
Zhu JG, Xie P, Zheng MD, Meng Y, Wei ML, Liu Y, Liu TW, Gong DQ. Dynamic changes in protein concentrations of keratins in crop milk and related gene expression in pigeon crops during different incubation and chick rearing stages. Br Poult Sci 2023; 64:100-109. [PMID: 36069156 DOI: 10.1080/00071668.2022.2119836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
1. The objective of this study was to examine the keratin composition of crop milk, the variation of epithelial thickness and keratin (K) gene expression in samples from young pigeon during incubation and chick rearing.2. Crop milk was collected from 1-, 3- and 5-day-old squab crops for keratin content analysis. Results showed that K4 accounted for the highest proportion of all detected keratins.3. In total, 42 pairs of adult pigeons were allocated to seven groups according to different stages to collect crop samples. Gene expression studies showed that the K3 gene expression was maximised at rearing Day 15 (15) and R1 in males and females, respectively. K6a gene level was the greatest at R15 in females, whereas it peaked at incubation Day 4 (I4) in males. The K12, K13, K23 and K80 gene levels were inhibited at the peak period of crop milk formation in comparison with I4. In females, K cochleal expression peaked at I10, whereas it was the greatest at R25 in males. K4 and K14 gene expression was the highest at I10 in females, while K4 and K14 were minimised at I17 and R7 in males, respectively. Gene expressions of K5, K8, K19 and K20 in males and K19 in females were maximised at R1. The K5, K20 and K75 gene levels in females peaked at R7. K75 and K8 expressions in males and females reached a maximum value at R25 and I17, respectively.4. The epithelial thickness of male and female crops reached their greatest levels at R1 and had the highest correlation with K19.5. These results emphasised the importance of keratinisation in crop milk formation, and different keratins probably play various roles during this period. The K19 was probably a marker for pigeon crop epithelium development. The sex of the parent pigeon affected keratin gene expression profiles.
Collapse
Affiliation(s)
- J G Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, Huaiyin, China
| | - P Xie
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, Huaiyin, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, Huaiyin, China
| | - M D Zheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Y Meng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - M L Wei
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, Huaiyin, China
| | - Y Liu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, Huaiyin, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, Huaiyin, China
| | - T W Liu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, Huaiyin, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, Huaiyin, China
| | - D Q Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
4
|
Jia J, Han Y, Jin L, Lu C, Wang W, Tong P, Li N, Sun X, Gu W, Dai J. miR-204-3p downregulates KRT16 and promotes corneal repair in tree shrew fungal keratitis model. Am J Transl Res 2022; 14:7336-7349. [PMID: 36398250 PMCID: PMC9641470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Corneal repair is critical for the treatment and recovery of corneal injuries. However, the molecular mechanism underlying corneal repair remains unclear. METHODS A tree shrew model of corneal fungal infection was established by injecting Fusarium solani into the corneal stroma to study the role of miR-204-3p in repairing corneal injury induced by fungal keratitis and to explore the potential mechanisms underlying the repair process. RESULTS miR-204-3p expression was significantly downregulated, while KRT16 expression was significantly upregulated after F. solani infection in the cornea of tree shrews. Moreover, miR-204-3p injection promoted corneal injury repair post-infection, potentially by downregulating KRT16 expression. Results of a luciferase reporter gene assay showed that miR-204-3p had a targeted relationship with KRT16. KRT16 protein expression levels decreased after miR-204-3p injection into the cornea with fungal keratitis, reducing the degree of corneal injury. CONCLUSIONS In this study, we report for the first time that miR-204-3p and KRT16 influence the repair of corneal injury. In addition, their effects on the repair of corneal injury were studied in a tree shrew model, providing an experimental basis for the study of pathogenesis of human fungal keratitis.
Collapse
Affiliation(s)
- Jie Jia
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
- Scientific Research Laboratory Center, The First Affiliated Hospital of Kunming Medical UniversityKunming, China
| | - Yuanyuan Han
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Liangzi Jin
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Caixia Lu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Wenguang Wang
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Pinfen Tong
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Na Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Xiaomei Sun
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Wenpeng Gu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| | - Jiejie Dai
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical CollegeKunming, China
| |
Collapse
|
5
|
Lee HP, Kim DS, Park SH, Shin CY, Woo JJ, Kim JW, An RB, Lee C, Cho JY. Antioxidant Capacity of Potentilla paradoxa Nutt. and Its Beneficial Effects Related to Anti-Aging in HaCaT and B16F10 Cells. PLANTS (BASEL, SWITZERLAND) 2022; 11:873. [PMID: 35406853 PMCID: PMC9003520 DOI: 10.3390/plants11070873] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022]
Abstract
Skin aging is a natural process influenced by intrinsic and extrinsic factors, and many skin anti-aging strategies have been developed. Plants from the genus Potentilla has been used in Europe and Asia to treat various diseases. Potentilla paradoxa Nutt. has been used as a traditional medicinal herb in China and has recently been shown to have anti-inflammatory effects. Despite the biological and pharmacological potential of Potentilla paradoxa Nutt., its skin anti-aging effects remain unclear. Therefore, this study evaluated the free radical scavenging, moisturizing, anti-melanogenic, and wound-healing effects of an ethanol extract of Potentilla paradoxa Nutt. (Pp-EE). Pp-EE was found to contain phenolics and flavonoids and exhibits in vitro antioxidant activities. α-Linolenic acid was found to be a major component of Pp-EE on gas chromatography-mass spectrometry. Pp-EE promoted the expression of hyaluronic acid (HA) synthesis-related enzymes and suppressed the expression of HA degradation-related enzymes in keratinocytes, so it may increase skin hydration. Pp-EE also showed inhibitory effects on the production and secretion of melanin in melanocytes. In a scratch assay, Pp-EE improved skin wound healing. Taken together, Pp-EE has several effects that may delay skin aging, suggesting its potential benefits as a natural ingredient in cosmetic or pharmaceutical products.
Collapse
Affiliation(s)
- Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.P.L.); (D.S.K.); (J.W.K.)
| | - Dong Seon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.P.L.); (D.S.K.); (J.W.K.)
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (C.Y.S.); (J.J.W.)
| | - Chae Yun Shin
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (C.Y.S.); (J.J.W.)
| | - Jin Joo Woo
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (C.Y.S.); (J.J.W.)
| | - Ji Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.P.L.); (D.S.K.); (J.W.K.)
| | - Ren-Bo An
- College of Pharmacy, Yanbian University, Yanji 133002, China;
| | - Changyoung Lee
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea;
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.P.L.); (D.S.K.); (J.W.K.)
| |
Collapse
|
6
|
Elazezy M, Schwentesius S, Stegat L, Wikman H, Werner S, Mansour WY, Failla AV, Peine S, Müller V, Thiery JP, Ebrahimi Warkiani M, Pantel K, Joosse SA. Emerging Insights into Keratin 16 Expression during Metastatic Progression of Breast Cancer. Cancers (Basel) 2021; 13:cancers13153869. [PMID: 34359774 PMCID: PMC8345379 DOI: 10.3390/cancers13153869] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The mechanisms leading to tumor metastasis remain poorly understood, and therefore, phenotyping of circulating tumor cells from cancer patients may contribute to translating these mechanisms. In in silico analysis, high expression of keratin 16 was associated with higher tumor aggressiveness. According to our results, keratin 16 is a metastasis-associated protein that promotes EMT and acts as a positive regulator of cellular motility by reorganizing the actin cytoskeleton, which is the driving force behind disrupting intercellular adhesion and directional migration. In metastatic breast cancer patients, circulating tumor cells expressing keratin 16 were associated with shorter relapse-free survival. This is an important issue for future research to determine the exact function of keratin 16 in tumor dissemination and metastasis development by analyzing keratin 16 status in disseminating tumor cells. Furthermore, gaining a better knowledge of keratin 16’s biology would give crucial mechanistic insights that might lead to a unique treatment option. Abstract Keratins are the main identification markers of circulating tumor cells (CTCs); however, whether their deregulation is associated with the metastatic process is largely unknown. Previously we have shown by in silico analysis that keratin 16 (KRT16) mRNA upregulation might be associated with more aggressive cancer. Therefore, in this study, we investigated the biological role and the clinical relevance of K16 in metastatic breast cancer. By performing RT-qPCR, western blot, and immunocytochemistry, we investigated the expression patterns of K16 in metastatic breast cancer cell lines and evaluated the clinical relevance of K16 expression in CTCs of 20 metastatic breast cancer patients. High K16 protein expression was associated with an intermediate mesenchymal phenotype. Functional studies showed that K16 has a regulatory effect on EMT and overexpression of K16 significantly enhanced cell motility (p < 0.001). In metastatic breast cancer patients, 64.7% of the detected CTCs expressed K16, which was associated with shorter relapse-free survival (p = 0.0042). Our findings imply that K16 is a metastasis-associated protein that promotes EMT and acts as a positive regulator of cellular motility. Furthermore, determining K16 status in CTCs provides prognostic information that helps to identify patients whose tumors are more prone to metastasize.
Collapse
Affiliation(s)
- Maha Elazezy
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Sandra Schwentesius
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Luisa Stegat
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Wael Y. Mansour
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Antonio Virgilio Failla
- UKE Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Jean Paul Thiery
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510320, China;
| | | | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
| | - Simon A. Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (M.E.); (S.S.); (L.S.); (H.W.); (S.W.); (K.P.)
- Correspondence: ; Tel.: +49-(0)-40-7410-51970
| |
Collapse
|
7
|
Haensel D, Jin S, Sun P, Cinco R, Dragan M, Nguyen Q, Cang Z, Gong Y, Vu R, MacLean AL, Kessenbrock K, Gratton E, Nie Q, Dai X. Defining Epidermal Basal Cell States during Skin Homeostasis and Wound Healing Using Single-Cell Transcriptomics. Cell Rep 2021; 30:3932-3947.e6. [PMID: 32187560 PMCID: PMC7218802 DOI: 10.1016/j.celrep.2020.02.091] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 01/17/2023] Open
Abstract
Our knowledge of transcriptional heterogeneities in epithelial stem and progenitor cell compartments is limited. Epidermal basal cells sustain cutaneous tissue maintenance and drive wound healing. Previous studies have probed basal cell heterogeneity in stem and progenitor potential, but a comprehensive dissection of basal cell dynamics during differentiation is lacking. Using single-cell RNA sequencing coupled with RNAScope and fluorescence lifetime imaging, we identify three non-proliferative and one proliferative basal cell state in homeostatic skin that differ in metabolic preference and become spatially partitioned during wound re-epithelialization. Pseudotemporal trajectory and RNA velocity analyses predict a quasi-linear differentiation hierarchy where basal cells progress from Col17a1Hi/Trp63Hi state to early-response state, proliferate at the juncture of these two states, or become growth arrested before differentiating into spinous cells. Wound healing induces plasticity manifested by dynamic basal-spinous interconversions at multiple basal transcriptional states. Our study provides a systematic view of epidermal cellular dynamics, supporting a revised “hierarchical-lineage” model of homeostasis. Haensel et al. performed a comprehensive dissection of the cellular makeup of skin during homeostasis and wound healing and the molecular heterogeneity and cellular dynamics within its stem-cell-containing epidermal basal layer. Their work provides insights and stimulates further investigation into the mechanism of skin maintenance and repair.
Collapse
Affiliation(s)
- Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- These authors contributed equally
| | - Suoqin Jin
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- These authors contributed equally
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Rachel Cinco
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Morgan Dragan
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Zixuan Cang
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Yanwen Gong
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Remy Vu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Adam L. MacLean
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Qing Nie
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Correspondence: (Q.N.), (X.D.)
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Lead Contact
- Correspondence: (Q.N.), (X.D.)
| |
Collapse
|
8
|
Pora A, Yoon S, Dreissen G, Hoffmann B, Merkel R, Windoffer R, Leube RE. Regulation of keratin network dynamics by the mechanical properties of the environment in migrating cells. Sci Rep 2020; 10:4574. [PMID: 32165652 PMCID: PMC7067805 DOI: 10.1038/s41598-020-61242-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/24/2020] [Indexed: 01/19/2023] Open
Abstract
Keratin intermediate filaments provide mechanical resilience for epithelia. They are nevertheless highly dynamic and turn over continuously, even in sessile keratinocytes. The aim of this study was to characterize and understand how the dynamic behavior of the keratin cytoskeleton is integrated in migrating cells. By imaging human primary keratinocytes producing fluorescent reporters and by using standardized image analysis we detect inward-directed keratin flow with highest rates in the cell periphery. The keratin flow correlates with speed and trajectory of migration. Changes in fibronectin-coating density and substrate stiffness induces concordant changes in migration speed and keratin flow. When keratinocytes are pseudo-confined on stripes, migration speed and keratin flow are reduced affecting the latter disproportionately. The regulation of keratin flow is linked to the regulation of actin flow. Local speed and direction of keratin and actin flow are very similar in migrating keratinocytes with keratin flow lagging behind actin flow. Conversely, reduced actin flow in areas of high keratin density indicates an inhibitory function of keratins on actin dynamics. Together, we propose that keratins enhance persistence of migration by directing actin dynamics and that the interplay of keratin and actin dynamics is modulated by matrix adhesions.
Collapse
Affiliation(s)
- Anne Pora
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Sungjun Yoon
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Georg Dreissen
- Institute of Biological Information Processing 2, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Hoffmann
- Institute of Biological Information Processing 2, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Rudolf Merkel
- Institute of Biological Information Processing 2, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Reinhard Windoffer
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074, Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
9
|
Louis F, Fujii N, Katsuyama M, Okumoto S, Matsusaki M. Effects of radiofrequency and ultrasound on the turnover rate of skin aging components (skin extracellular matrix and epidermis) via HSP47-induced stimulation. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30286-2. [PMID: 32081434 DOI: 10.1016/j.bbrc.2020.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 10/25/2022]
Abstract
Skin aging cannot be escaped, being due to both intrinsic and extrinsic stimuli. They lead to a reduced extracellular collagen matrix in the dermis, along with a higher degradation by metalloproteases (MMPs) activity, as well as a lower differentiation and function of epidermis keratinocytes, characterized by wrinkling and loss of skin elasticity. One of the recent technology to overcome this skin aging process is the use of radiofrequency (RF) and ultrasound (US) technologies which use thermal stimulation to induce neocollagenesis in the skin. But no explanations exist on the involved pathways. Our hypothesis is that RF-US generated heat increases the collagen formation via the heat shock protein 47 (HSP47) induction, a heat sensitive protein related to the collagen expression. To confirm this hypothesis, normal human skin substitutes were subjected to RF-US treatment and results were monitored after 24 and 44 h. RNA sequencing showed a significant induction for the genes related to the epidermis differentiation processes. Almost all keratin genes were thus found upregulated from 2 to 15 times, while collagen type XVII and collagen type IV were increased 12 and 5 times respectively. In parallel, most of MMP genes were observed downregulated. RF-US treatment significantly increased levels of HSP47 proteins, while collagen XVII proteins showed a tendency to be increased and glycosaminoglycans were found 1.4 times significantly enhanced. Finally, histology assessment showed a higher expression of cytokeratins 10 and 14 which can testify a possible reactivation of the skin proliferative state as a rejuvenation strategy.
Collapse
Affiliation(s)
- Fiona Louis
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Narumi Fujii
- Materials Solution Department, Product Analysis Center, Panasonic Corporation, Kadoma, Osaka, 571-8686, Japan.
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Corporation, Kadoma, Osaka, 571-8686, Japan.
| | - Satoshi Okumoto
- Materials Solution Department, Product Analysis Center, Panasonic Corporation, Kadoma, Osaka, 571-8686, Japan.
| | - Michiya Matsusaki
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
10
|
Zeng Q, Jiang J, Wang J, Zhou Q, Zhang X. N-Terminal Acetylation and C-Terminal Amidation of Spirulina platensis-Derived Hexapeptide: Anti-Photoaging Activity and Proteomic Analysis. Mar Drugs 2019; 17:md17090520. [PMID: 31487895 PMCID: PMC6780235 DOI: 10.3390/md17090520] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/25/2019] [Accepted: 09/01/2019] [Indexed: 02/06/2023] Open
Abstract
Ultraviolet (UV) irradiation is a potent inducer for skin photoaging. This paper investigated the anti-photoaging effects of the acetylated and amidated hexapeptide (AAH), originally identified from Spirulina platensis, in (Ultraviolet B) UVB-irradiated Human immortalized keratinocytes (Hacats) and mice. The results demonstrated that AAH had much lower toxicity on Hacats than the positive matrixyl (81.52% vs. 5.32%). Moreover, AAH reduced MDA content by 49%; increased SOD, CAT, and GSH-Px activities by 103%, 49%, and 116%, respectively; decreased MMP-1 and MMP-3 expressions by 27% and 29%, respectively, compared to UVB-irradiated mice. Employing isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics, 60 differential proteins were identified, and major metabolic pathways were determined. Network analysis indicated that these differential proteins were mapped into an interaction network composed of two core sub-networks. Collectively, AAH is protective against UVB-induced skin photoaging and has potential application in skin care cosmetics.
Collapse
Affiliation(s)
- Qiaohui Zeng
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
- Department of Food Science, Foshan University, Foshan 528000, China.
| | - Jianguo Jiang
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Jingjing Wang
- Department of Food Science, Foshan University, Foshan 528000, China.
| | - Qiuchan Zhou
- Institute of Laboratory animal science, Jinan University, Guangzhou 510632, China.
| | - Xuewu Zhang
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
11
|
Zhang X, Yin M, Zhang LJ. Keratin 6, 16 and 17-Critical Barrier Alarmin Molecules in Skin Wounds and Psoriasis. Cells 2019; 8:E807. [PMID: 31374826 PMCID: PMC6721482 DOI: 10.3390/cells8080807] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/17/2022] Open
Abstract
Located at the skin surface, keratinocytes (KCs) are constantly exposed to external stimuli and are the first responders to invading pathogens and injury. Upon skin injury, activated KCs secrete an array of alarmin molecules, providing a rapid and specific innate immune response against danger signals. However, dysregulation of the innate immune response of KCs may lead to uncontrolled inflammation and psoriasis pathogenesis. Keratins (KRT) are the major structural intermediate filament proteins in KCs and are expressed in a highly specific pattern at different differentiation stages of KCs. While KRT14-KRT5 is restricted to basal proliferative KCs, and KRT10-KRT1 is restricted to suprabasal differentiated KCs in normal skin epidermis, the wound proximal KCs downregulate KRT10-K1 and upregulate KRT16/KRT17-KRT6 upon skin injury. Recent studies have recognized KRT6/16/17 as key early barrier alarmins and upregulation of these keratins alters proliferation, cell adhesion, migration and inflammatory features of KCs, contributing to hyperproliferation and innate immune activation of KCs in response to an epidermal barrier breach, followed by the autoimmune activation of T cells that drives psoriasis. Here, we have reviewed how keratins are dysregulated during skin injury, their roles in wound repairs and in initiating the innate immune system and the subsequent autoimmune amplification that arises in psoriasis.
Collapse
Affiliation(s)
- Xiaowei Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Meimei Yin
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Zhang J, Li L, Zhang Q, Wang W, Zhang D, Jia J, Lv Y, Yuan H, Song H, Xiang F, Hu J, Huang Y. Microtubule-associated protein 4 phosphorylation regulates epidermal keratinocyte migration and proliferation. Int J Biol Sci 2019; 15:1962-1976. [PMID: 31523197 PMCID: PMC6743305 DOI: 10.7150/ijbs.35440] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Both cell migration and proliferation are indispensable parts of reepithelialization during skin wound healing, which is a complex process for which the underlying molecular mechanisms are largely unknown. Here, we identify a novel role for microtubule-associated protein 4 (MAP4), a cytosolic microtubule-binding protein that regulates microtubule dynamics through phosphorylation modification, as a critical regulator of epidermal wound repair. We showed that MAP4 phosphorylation was induced in skin wounds. In an aberrant phosphorylated MAP4 mouse model, hyperphosphorylation of MAP4 (S737 and S760) accelerated keratinocyte migration and proliferation and skin wound healing. Data from both primary cultured keratinocytes and HaCaT cells in vitro revealed the same results. The promigration and proproliferation effects of MAP4 phosphorylation depended on microtubule rearrangement and could be abolished by MAP4 dephosphorylation. We also identified p38/MAPK as an upstream regulator of MAP4 phosphorylation in keratinocytes. Our findings provide new insights into the molecular mechanisms underlying wound-associated keratinocyte migration and proliferation and identify potential targets for the remediation of defective wound healing.
Collapse
Affiliation(s)
- Junhui Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lingfei Li
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wensheng Wang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiezhi Jia
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanling Lv
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongping Yuan
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huapei Song
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Xiang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiongyu Hu
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Endocrinology Department, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
13
|
LRG1 Promotes Keratinocyte Migration and Wound Repair through Regulation of HIF-1α Stability. J Invest Dermatol 2019; 140:455-464.e8. [PMID: 31344385 DOI: 10.1016/j.jid.2019.06.143] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/13/2019] [Accepted: 06/30/2019] [Indexed: 01/10/2023]
Abstract
Re-epithelialization is a complex process during skin wound healing, and cell migration is an integral part of this phenomenon. Here we identified a role for LRG1 as a key regulator of epidermal keratinocyte migration where LRG1 acts via enhancement of HIF-1α stability. We showed that LRG1 is upregulated at murine skin wound edges and that addition of recombinant human LRG1 accelerates keratinocyte migration and skin wound healing. Furthermore, we identified transcription factor ELK3 as a downstream effector of LRG1. We confirmed that elevated ELK3 levels manipulated by LRG1 can promote cell migration through upregulation of HIF-1α stability. Because hyperglycemia complicatedly affects HIF-1α stability and activation, our findings provide insights into the molecular controls of wound-associated cell migration and identify potential therapeutic targets for the treatment of chronic diabetic wounds. In conclusion, we demonstrated that LRG1 promotes wound repair through keratinocyte migration and is important for normalization of an abnormal process of diabetic wound healing where HIF-1α stability is insufficient.
Collapse
|
14
|
Coates M, Mariottoni P, Corcoran DL, Kirshner HF, Jaleel T, Brown DA, Brooks SR, Murray J, Morasso MI, MacLeod AS. The skin transcriptome in hidradenitis suppurativa uncovers an antimicrobial and sweat gland gene signature which has distinct overlap with wounded skin. PLoS One 2019; 14:e0216249. [PMID: 31059533 PMCID: PMC6502346 DOI: 10.1371/journal.pone.0216249] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/16/2019] [Indexed: 12/15/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a debilitating chronic inflammatory skin disease resulting in non-healing wounds affecting body areas of high hair follicle and sweat gland density. The pathogenesis of HS is not well understood but appears to involve dysbiosis-driven aberrant activation of the innate immune system leading to excessive inflammation. Marked dysregulation of antimicrobial peptides and proteins (AMPs) in HS is observed, which may contribute to this sustained inflammation. Here, we analyzed HS skin transcriptomes from previously published studies and integrated these findings through a comparative analysis with a published wound healing data set and with immunofluorescence and qPCR analysis from new HS patient samples. Among the top differently expressed genes between lesional and non-lesional HS skin were members of the S100 family as well as dermcidin, the latter known as a sweat gland-associated AMP and one of the most downregulated genes in HS lesions. Interestingly, many genes associated with sweat gland function, such as secretoglobins and aquaporin 5, were decreased in HS lesional skin and we discovered that these genes demonstrated opposite expression profiles in healing skin. Conversely, HS lesional and wounded skin shared a common gene signature including genes encoding for S100 proteins, defensins, and genes encoding antiviral proteins. Overall, our results suggest that the pathogenesis of HS may be driven by changes in AMP expression and altered sweat gland function, and may share a similar pathology with chronic wounds.
Collapse
Affiliation(s)
- Margaret Coates
- Department of Dermatology, Duke University, Durham, NC, United States of America
| | - Paula Mariottoni
- Department of Dermatology, Duke University, Durham, NC, United States of America
| | - David L. Corcoran
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, United States of America
| | - Hélène Fradin Kirshner
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, United States of America
| | - Tarannum Jaleel
- Department of Dermatology, Duke University, Durham, NC, United States of America
| | - David A. Brown
- Department of Surgery, Duke University, Durham, NC, United States of America
| | - Stephen R. Brooks
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, MD, United States of America
| | - John Murray
- Department of Dermatology, Duke University, Durham, NC, United States of America
| | - Maria I. Morasso
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, MD, United States of America
| | - Amanda S. MacLeod
- Department of Dermatology, Duke University, Durham, NC, United States of America
- Department of Immunology, Duke University, Durham, NC, United States of America
- Pinnell Center for Investigative Dermatology, Duke University, Durham, NC, United States of America
- * E-mail:
| |
Collapse
|
15
|
Ma J, Xu S, Wang X, Zhang J, Wang Y, Liu M, Jin L, Wu M, Qian D, Li X, Zhen Q, Guo H, Gao J, Yang S, Zhang X. Noninvasive analysis of skin proteins in healthy Chinese subjects using an Orbitrap Fusion Tribrid mass spectrometer. Skin Res Technol 2019; 25:424-433. [PMID: 30657212 DOI: 10.1111/srt.12668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/29/2018] [Accepted: 12/08/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Jie Ma
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Shuangjun Xu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xiaomeng Wang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Jing Zhang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Yaochi Wang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Mengting Liu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Ling Jin
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Mingshun Wu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Danfeng Qian
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xueying Li
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Qi Zhen
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Huimin Guo
- Center for Biological TechnologyAnhui Agricultural University Hefei China
| | - Jinping Gao
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Sen Yang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xuejun Zhang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| |
Collapse
|
16
|
Wang F, Chen S, Liu HB, Parent CA, Coulombe PA. Keratin 6 regulates collective keratinocyte migration by altering cell-cell and cell-matrix adhesion. J Cell Biol 2018; 217:4314-4330. [PMID: 30389720 PMCID: PMC6279382 DOI: 10.1083/jcb.201712130] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 09/18/2018] [Accepted: 10/16/2018] [Indexed: 01/21/2023] Open
Abstract
Keratin 6 (K6) isoforms are induced in wound-proximal keratinocytes after injury to skin. Paradoxically, absence of K6 isoforms leads to faster directional cell migration. Wang et al. report that K6 promotes collective keratinocyte migration by interacting with desmoplakin and myosin IIA and stabilizing cell adhesion. The a and b isoforms of keratin 6 (K6), a type II intermediate filament (IF) protein, are robustly induced upon injury to interfollicular epidermis. We previously showed that complete loss of K6a/K6b stimulates keratinocyte migration, correlating with enhanced Src activity. In this study, we demonstrate that this property is cell autonomous, depends on the ECM, and results from elevated speed, enhanced directionality, and an increased rate of focal adhesion disassembly. We show that myosin IIA interacts with K6a/K6b, that its levels are markedly reduced in Krt6a/Krt6b-null keratinocytes, and that inhibiting myosin ATPase activity normalizes the enhanced migration potential of Krt6a/Krt6b-null cells. Desmoplakin, which mediates attachment of IFs to desmosomes, is also expressed at reduced levels and is mislocalized to the nucleus in Krt6a/Krt6b-null cells, correlating with defects in cell adhesion. These findings reveal that K6a/K6b modulate keratinocyte migration by regulating cell–matrix and cell–cell adhesion and highlight a role for keratins in collective cell migration.
Collapse
Affiliation(s)
- Fengrong Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI.,Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Song Chen
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Hans B Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Carole A Parent
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI .,Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD.,Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
17
|
Lange SS, Bhetawal S, Reh S, Powell KL, Kusewitt DF, Wood RD. DNA polymerase ζ deficiency causes impaired wound healing and stress-induced skin pigmentation. Life Sci Alliance 2018; 1. [PMID: 30046772 PMCID: PMC6055517 DOI: 10.26508/lsa.201800048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mice harboring DNA polymerase ζ–defective keratinocytes are shown to have a defect in wound healing and a striking p53-dependent migration of melanocytes to the skin following UV radiation or wounding. DNA polymerase ζ (pol ζ) is well established as a specialized enzyme important for DNA damage tolerance, facilitating DNA synthesis past lesions caused by radiation or chemical damage. We report that disruption of Rev3l (encoding the catalytic subunit of pol ζ) in mouse epidermis leads to a defect in proliferation that impairs cutaneous wound healing. A striking increase in epidermal skin pigmentation accompanied both wound healing and UV irradiation in these mice. This was a consequence of stress-induced migration of Rev3l-proficient melanocytes to the Rev3l-defective epidermis. We found that this pigmentation corresponded with p53 activation in keratinocytes and was absent in p53-negative areas of the epidermis. Expression of the kit ligand (Kitl) gene, a p53-controlled mediator of keratinocyte to melanocyte signaling, was enhanced during wound healing or following UV irradiation. This study extends the function of pol ζ to the process of proliferation during wound healing. Rev3l-deficient epidermis may be a useful mouse model system for examining communication between damaged keratinocytes and melanocytes, including signaling relevant to human disease.
Collapse
Affiliation(s)
- Sabine S Lange
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| | - Sarita Bhetawal
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| | - Shelley Reh
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| | - Katherine Leslie Powell
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| | - Donna F Kusewitt
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| | - Richard D Wood
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, and the Graduate School of Biomedical Sciences at Houston, Smithville, Texas, P.O. Box 389, Smithville, TX, 78957, USA
| |
Collapse
|
18
|
Kauffmann F, Meert E, de Jonge K, Elkrim Y, Hanot Mambres D, Denis O, Muraille E, Magez S, De Trez C. STAT6 Mediates Footpad Immunopathology in the Absence of IL-12p40 Following Infection of Susceptible BALB/c Mice With Leishmania major. Front Immunol 2018; 9:503. [PMID: 29593739 PMCID: PMC5861353 DOI: 10.3389/fimmu.2018.00503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/26/2018] [Indexed: 01/19/2023] Open
Abstract
Leishmania major (L. major) parasites are intracellular parasites belong to the Trypanosomatidae family and are the causative agent of cutaneous leishmaniasis. This disease affects approximately 1.5 million per year worldwide and there is currently no prophylactic vaccine available. L. major is transmitted by the bite of an infected sandfly and has been considered for decades now as a mouse model of choice to identify the factors implicated in T helper (Th)1 and Th2 polarization due to the natural resistance and susceptibility to infection of C57BL/6 and BALB/c mice, respectively. In this study, we refine the role of IL-12p40 cytokine, which is implicated the development of a protective Th1 response, and STAT6, a transcription factor involved in the signaling via detrimental interleukin (IL)-4 and IL-13 associated Th2 cytokines during L. major infection in the BALB/c model. In the absence of STAT6 and IL-12p40 signaling, double knockout (DKO) susceptible BALB/c mice displayed reduced footpad swelling and ulcerative lesion compared to IL-12p40−/− mice upon L. major infection. Hence, they expressed slower upregulation of keratinocyte markers implicated in the inhibition of wound healing, such as keratin 6a (Krt6a) and Krt16. This coincides with the presence of neutrophils displaying an altered phenotype characterized by a lower expression of surface markers Ly6C, CD11b, and Ly6G. These neutrophils exhibited very lower levels of apoptosis similarly to neutrophils present in resistant STAT6−/− mice. Interestingly, the reduced footpad swelling in DKO mice is associated with a high footpad parasite level similar to susceptible IL-12p40−/− mice. In conclusion, this study demonstrate that in the absence of both STAT6 and IL-12p40 signaling, L. major-infected mice display smaller and less ulcerated lesions, which does, however, not correlate with reduced parasite load. In addition, the presence of neutrophils with an altered phenotype is associated with reduced apoptosis and delayed immunopathologies, demonstrating the detrimental role of STAT6 in infected susceptible BALB/c mice.
Collapse
Affiliation(s)
- Florence Kauffmann
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Elyn Meert
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Kaat de Jonge
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yvon Elkrim
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Laboratory of Myeloid Cell Immunology, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Delphine Hanot Mambres
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, Université de Namur, Namur, Belgium
| | - Olivier Denis
- Scientific Service Immunology, Scientific Institute of Public Health (WIV-ISP), Brussels, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, Université de Namur, Namur, Belgium.,Laboratoire de Parasitologie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Stefan Magez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Ghent University Global Campus, Incheon, South Korea
| | - Carl De Trez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
19
|
Elentner A, Schmuth M, Yannoutsos N, Eichmann TO, Gruber R, Radner FPW, Hermann M, Del Frari B, Dubrac S. Epidermal Overexpression of Xenobiotic Receptor PXR Impairs the Epidermal Barrier and Triggers Th2 Immune Response. J Invest Dermatol 2017; 138:109-120. [PMID: 28927887 PMCID: PMC6217923 DOI: 10.1016/j.jid.2017.07.846] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/29/2017] [Accepted: 07/03/2017] [Indexed: 12/11/2022]
Abstract
The skin is in daily contact with environmental pollutants, but the long-term effects of such exposure remain underinvestigated. Many of these toxins bind and activate the pregnane X receptor (PXR), a ligand-activated transcription factor that regulates genes central to xenobiotic metabolism. The objective of this work was to investigate the effect of constitutive activation of PXR in the basal layer of the skin to mimic repeated skin exposure to noxious molecules. We designed a transgenic mouse model that overexpresses the human PXR gene linked to the herpes simplex VP16 domain under the control of the keratin 14 promoter. We show that transgenic mice display increased transepidermal water loss and elevated skin pH, abnormal stratum corneum lipids, focal epidermal hyperplasia, activated keratinocytes expressing more thymic stromal lymphopoietin, a T helper type 2/T helper type 17 skin immune response, and increased serum IgE. Furthermore, the cutaneous barrier dysfunction precedes development of the T helper type 2/T helper type 17 inflammation in transgenic mice, thereby mirroring the time course of atopic dermatitis development in humans. Moreover, further experiments suggest increased PXR signaling in the skin of patients with atopic dermatitis when compared with healthy skin. Thus, PXR activation by environmental pollutants may compromise epidermal barrier function and favor an immune response resembling atopic dermatitis.
Collapse
Affiliation(s)
- Andreas Elentner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolaos Yannoutsos
- Gene Regulation and Immunology Laboratory, Department of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Franz P W Radner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Hermann
- KMT Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Center for Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Esthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
20
|
Cheng F, Eriksson JE. Intermediate Filaments and the Regulation of Cell Motility during Regeneration and Wound Healing. Cold Spring Harb Perspect Biol 2017; 9:9/9/a022046. [PMID: 28864602 DOI: 10.1101/cshperspect.a022046] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SUMMARYIntermediate filaments (IFs) comprise a diverse group of flexible cytoskeletal structures, the assembly, dynamics, and functions of which are regulated by posttranslational modifications. Characteristically, the expression of IF proteins is specific for tissues, differentiation stages, cell types, and functional contexts. Recent research has rapidly expanded the knowledge of IF protein functions. From being regarded as primarily structural proteins, it is now well established that IFs act as powerful modulators of cell motility and migration, playing crucial roles in wound healing and tissue regeneration, as well as inflammatory and immune responses. Although many of these IF-associated functions are essential for tissue repair, the involvement of IF proteins has been established in many additional facets of tissue healing and regeneration. Here, we review the recent progress in understanding the multiple functions of cytoplasmic IFs that relate to cell motility in the context of wound healing, taking examples from studies on keratin, vimentin, and nestin. Wound healing and regeneration include orchestration of a broad range of cellular processes, including regulation of cell attachment and migration, proliferation, differentiation, immune responses, angiogenesis, and remodeling of the extracellular matrix. In this respect, IF proteins now emerge as multifactorial and tissue-specific integrators of tissue regeneration, thereby acting as essential guardian biopolymers at the interface between health and disease, the failing of which contributes to a diverse range of pathologies.
Collapse
Affiliation(s)
- Fang Cheng
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland.,Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| | - John E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland.,Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| |
Collapse
|
21
|
Exploring a Role for Regulatory miRNAs In Wound Healing during Ageing:Involvement of miR-200c in wound repair. Sci Rep 2017; 7:3257. [PMID: 28607463 PMCID: PMC5468284 DOI: 10.1038/s41598-017-03331-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/26/2017] [Indexed: 01/08/2023] Open
Abstract
Multiple factors and conditions can lead to impaired wound healing. Chronic non-healing wounds are a common problem among the elderly. To identify microRNAs negatively impacting the wound repair, global miRNA profiling of wounds collected from young and old mice was performed. A subset of miRNAs that exhibited an age-dependent expression pattern during wound closure was identified, including miR-31 and miR-200c. The expression of miR-200 family members was markedly downregulated upon wounding in both young and aged mice, with an exception of acute upregulation of miR-200c at the early phase of wound healing in aged skin. In unwounded aged skin (versus unwounded younger skin), the level of miR-200c was also found elevated in both human and mice. Overexpression of miR-200c in human ex vivo wounds delayed re-epithelialisation and inhibited cell proliferation in the wound epithelium. Modulation of miR-200c expression in both human and mouse keratinocytes in vitro revealed inhibitory effects of miR-200c on migration, but not proliferation. Accelerated wound closure in vitro induced by anti-miR-200c was associated with upregulation of genes controlling cell migration. Thus, our study identified miR-200c as a critical determinant that inhibits cell migration during skin repair after injury and may contribute to age-associated alterations in wound repair.
Collapse
|
22
|
Boink MA, Roffel S, Breetveld M, Thon M, Haasjes MSP, Waaijman T, Scheper RJ, Blok CS, Gibbs S. Comparison of advanced therapy medicinal product gingiva and skin substitutes and their in vitro wound healing potentials. J Tissue Eng Regen Med 2017; 12:e1088-e1097. [PMID: 28388010 PMCID: PMC5836907 DOI: 10.1002/term.2438] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/25/2017] [Accepted: 04/03/2017] [Indexed: 01/23/2023]
Abstract
Skin and oral mucosa substitutes are a therapeutic option for closing hard‐to‐heal skin and oral wounds. Our aim was to develop bi‐layered skin and gingiva substitutes, from 3 mm diameter biopsies, cultured under identical conditions, which are compliant with current European regulations for advanced therapy medicinal products. We present in vitro mode of action methods to (i) determine viability: epithelial expansion, proliferation (Ki‐67), metabolic activity (MTT assay); (ii) characterize skin and gingiva substitutes: histology and immunohistochemistry; and (iii) determine potency: soluble wound healing mediator release (enzyme‐linked immunosorbent assay). Both skin and gingiva substitutes consist of metabolically active autologous reconstructed differentiated epithelium expanding from the original biopsy sheet on a fibroblast populated connective tissue matrix (donor dermis). Gingival epithelium expanded 1.7‐fold more than skin epithelium during the 3 week culture period. The percentage of proliferating Ki‐67‐positive cells located in the basal layer of the gingiva substitute was >1.5‐fold higher than in the skin substitute. Keratins 16 and 17, which are upregulated during normal wound healing, were expressed in both the skin and gingiva substitutes. Notably, the gingiva substitute secreted higher amounts of key cytokines involved in mitogenesis, motogenesis and chemotaxis (interleukin‐6 > 23‐fold, CXCL8 > 2.5‐fold) as well as higher amounts of the anti‐fibrotic growth factor, hepatocyte growth factor (>7‐fold), compared with the skin substitute. In conclusion, while addressing the viability, characterization and potency of the tissue substitutes, important intrinsic differences between skin and gingiva were discovered that may explain in part the superior quality of wound healing observed in the oral mucosa compared with skin.
Collapse
Affiliation(s)
- Mireille A Boink
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Sanne Roffel
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Melanie Breetveld
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Maria Thon
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands.,A-Skin BV, Amsterdam, the Netherlands
| | - Michiel S P Haasjes
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Taco Waaijman
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Chantal S Blok
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Gimblet C, Loesche MA, Carvalho L, Carvalho EM, Grice EA, Artis D, Scott P. IL-22 Protects against Tissue Damage during Cutaneous Leishmaniasis. PLoS One 2015; 10:e0134698. [PMID: 26285207 PMCID: PMC4540492 DOI: 10.1371/journal.pone.0134698] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/13/2015] [Indexed: 01/11/2023] Open
Abstract
Cutaneous leishmaniasis is a disease characterized by ulcerating skin lesions, the resolution of which requires an effective, but regulated, immune response that limits parasite growth without causing permanent tissue damage. While mechanisms that control the parasites have been well studied, the factors regulating immunopathologic responses are less well understood. IL-22, a member of the IL-10 family of cytokines, can contribute to wound healing, but in other instances promotes pathology. Here we investigated the role of IL-22 during leishmania infection, and found that IL-22 limits leishmania-induced pathology when a certain threshold of damage is induced by a high dose of parasites. Il22-/- mice developed more severe disease than wild-type mice, with significantly more pathology at the site of infection, and in some cases permanent loss of tissue. The increased inflammation was not due to an increased parasite burden, but rather was associated with the loss of a wound healing phenotype in keratinocytes. Taken together, these studies demonstrate that during cutaneous leishmaniasis, IL-22 can play a previously unappreciated role in controlling leishmania-induced immunopathology.
Collapse
Affiliation(s)
- Ciara Gimblet
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Michael A. Loesche
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Lucas Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia—Doenças Tropicais, Salvador, Bahia, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Edgar M. Carvalho
- Serviço de Imunologia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia—Doenças Tropicais, Salvador, Bahia, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - David Artis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| |
Collapse
|
24
|
Castro-Muñozledo F, Velez-DelValle C, Marsch-Moreno M, Hernández-Quintero M, Kuri-Harcuch W. Vimentin is necessary for colony growth of human diploid keratinocytes. Histochem Cell Biol 2014; 143:45-57. [PMID: 25142512 DOI: 10.1007/s00418-014-1262-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2014] [Indexed: 01/08/2023]
Abstract
The role of vimentin (Vim) in diploid epithelial cells is not well known. To understand its biological function, we cultured human epidermal keratinocytes under conditions that support migration, proliferation, stratification and terminal differentiation. We identified a keratinocyte subpopulation that shows a p63(+)/α5β1(bright) phenotype and displays Vim intermediate filaments (IFs) besides their keratin IF network. These cells were mainly located at the proliferative/migratory rim of the growing colonies; but also, they were scarce and scattered or formed small groups of basal cells in confluent stratified epithelia. Stimulation of cells with EGF and wounding experiments in confluent arrested epithelia increased the number of Vim(+) keratinocytes in an extent higher to the expected for a cell population doubling. BrdU labeling demonstrated that most of the proliferative cells located at the migratory border of the colony have Vim, in contrast with proliferative cells located at the basal layer at the center of big colonies which lacked of Vim IFs, suggesting that Vim expression was not solely linked to proliferation. Therefore, we silenced Vim mRNA in the cultured keratinocytes and observed an inhibition of colony growth. Such results, together with long-term cultivation assays which showed that Vim might be associated to pattern formation in cultured epithelia, suggest that Vim expression is essential for a highly motile phenotype, which is necessary for keratinocyte colony growth and possibly for development and wound healing. Vim(+)/p63(+)/α5β1(bright) epithelial cells may play a significant physiological role in embryonic morphogenetic movements; wound healing and other pathologies such as carcinomas and hyperproliferative diseases.
Collapse
Affiliation(s)
- Federico Castro-Muñozledo
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN Apdo, Postal 14-740, 07000, Mexico City, Mexico,
| | | | | | | | | |
Collapse
|
25
|
Tcf3 promotes cell migration and wound repair through regulation of lipocalin 2. Nat Commun 2014; 5:4088. [PMID: 24909826 PMCID: PMC4052366 DOI: 10.1038/ncomms5088] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/09/2014] [Indexed: 01/25/2023] Open
Abstract
Cell migration is an integral part of re-epithelialization during skin wound healing, a complex process involving molecular controls that are still largely unknown. Here we identify a novel role for Tcf3, an essential transcription factor regulating embryonic and adult skin stem cell functions, as a key effector of epidermal wound repair. We show that Tcf3 is upregulated in skin wounds and that Tcf3 overexpression accelerates keratinocyte migration and skin wound healing. We also identify Stat3 as an upstream regulator of Tcf3. We show that the pro-migration effects of Tcf3 are non-cell autonomous and occur independently of its ability to interact with β-catenin. Finally, we identify lipocalin-2 as the key secreted factor downstream of Tcf3 that promotes cell migration in vitro and wound healing in vivo. Our findings provide new insights into the molecular controls of wound-associated cell migration and identify potential therapeutic targets for the treatment of defective wound repair.
Collapse
|
26
|
Skazik C, Amann PM, Heise R, Marquardt Y, Czaja K, Kim A, Rühl R, Kurschat P, Merk HF, Bickers DR, Baron JM. Downregulation of STRA6 expression in epidermal keratinocytes leads to hyperproliferation-associated differentiation in both in vitro and in vivo skin models. J Invest Dermatol 2013; 134:1579-1588. [PMID: 24284421 DOI: 10.1038/jid.2013.507] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 10/11/2013] [Accepted: 10/22/2013] [Indexed: 11/09/2022]
Abstract
Retinoids are known to affect skin cell proliferation and differentiation and are key molecules that target retinoid and retinoic acid receptors (RXRs and RARs), leading to physiological and pharmacologic effects. Our aim was to elucidate the role of the retinol-binding protein receptor STRA6, mediating cellular uptake of retinol, on skin structure and function. Our results indicate that STRA6 is constitutively expressed in human epidermal keratinocytes and dermal fibroblasts and is regulated via RAR/RXR-mediated pathways. HaCaT (Human adult low Calcium high Temperature) cells with stable STRA6 knockdown (STRA6KD) showed increased proliferation. Consistently, human organotypic 3D skin models using stable STRA6KD HaCaT cells showed a significantly thicker epidermis and enhanced expression of activation, differentiation, and proliferation markers. The effects were reversible after treatment with free retinol. Human skin reconstitution employing STRA6KD HaCaT cells leads to massive epithelial thickening under in vivo conditions in SCID mice. We propose that STRA6KD could lead to cellular vitamin A deficiency in keratinocytes. Consequently, STRA6 has a role for regulating retinoid homeostasis and in helping to program signaling that drives proliferation and differentiation of human skin cells. By its influence on hyperproliferation-associated differentiation, STRA6 could also have a role in skin regeneration and could be a target for pharmacological approaches to improve wound healing.
Collapse
Affiliation(s)
- Claudia Skazik
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Philipp M Amann
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Ruth Heise
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Yvonne Marquardt
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Katharina Czaja
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Arianna Kim
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Ralph Rühl
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary and
| | - Peter Kurschat
- Department of Dermatology, University Hospital of Cologne, Cologne, Germany
| | - Hans F Merk
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - David R Bickers
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jens M Baron
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
27
|
Bone morphogenetic protein signaling suppresses wound-induced skin repair by inhibiting keratinocyte proliferation and migration. J Invest Dermatol 2013; 134:827-837. [PMID: 24126843 PMCID: PMC3945401 DOI: 10.1038/jid.2013.419] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 09/02/2013] [Accepted: 09/16/2013] [Indexed: 12/29/2022]
Abstract
Bone morphogenetic protein (BMP) signalling plays a key role in the control of skin development and postnatal remodelling by regulating keratinocyte proliferation, differentiation and apoptosis. To study the role of BMPs in wound-induced epidermal repair, we used transgenic mice overexpressing the BMP downstream component Smad1 under the control of a K14 promoter as an in vivo model, as well as ex vivo and in vitro assays. K14-caSmad1 mice exhibited retarded wound healing associated with significant inhibition of proliferation and increased apoptosis in healing wound epithelium. Furthermore, microarray and qRT-PCR analyses revealed decreased expression of a number of cytoskeletal/cell motility-associated genes including wound-associated keratins (Krt16, Krt17) and Myo5a, in the epidermis of K14-caSmad1 mice versus wild-type controls during wound healing. BMP treatment significantly inhibited keratinocyte migration ex vivo, and primary keratinocytes of K14-caSmad1 mice showed retarded migration compared to wild-type controls. Finally, siRNA-mediated silencing of Bmpr-1B in primary mouse keratinocytes accelerated cell migration and was associated with increased expression of Krt16, Krt17 and Myo5a compared to controls. Thus, this study demonstrates that BMPs inhibit keratinocyte proliferation, cytoskeletal organization and migration in regenerating skin epithelium during wound healing, and raises a possibility for using BMP antagonists for the management of chronic wounds.
Collapse
|
28
|
Somarelli JA, Schaeffer D, Bosma R, Bonano VI, Sohn JW, Kemeny G, Ettyreddy A, Garcia-Blanco MA. Fluorescence-based alternative splicing reporters for the study of epithelial plasticity in vivo. RNA (NEW YORK, N.Y.) 2013; 19:116-127. [PMID: 23185039 PMCID: PMC3527723 DOI: 10.1261/rna.035097.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 10/22/2012] [Indexed: 05/31/2023]
Abstract
Alternative splicing generates a vast diversity of protein isoforms from a limited number of protein-coding genes, with many of the isoforms possessing unique, and even contrasting, functions. Fluorescence-based splicing reporters have the potential to facilitate studies of alternative splicing at the single-cell level and can provide valuable information on phenotypic transitions in almost real time. Fibroblast growth factor receptor 2 (FGFR2) pre-mRNA is alternatively spliced to form the epithelial-specific and mesenchymal-specific IIIb and IIIc isoforms, respectively, which are useful markers of epithelial-mesenchymal transitions (EMT). We have used our knowledge of FGFR2 splicing regulation to develop a fluorescence-based reporter system to visualize exon IIIc regulation in vitro and in vivo. Here we show the application of this reporter system to the study of EMT in vitro in cell culture and in vivo in transgenic mice harboring these splicing constructs. In explant studies, the reporters revealed that FGFR2 isoform switching is not required for keratinocyte migration during cutaneous wound closure. Our results demonstrate the value of the splicing reporters as tools to study phenotypic transitions and cell fates at single cell resolution. Moreover, our data suggest that keratinocytes migrate efficiently in the absence of a complete EMT.
Collapse
Affiliation(s)
| | - Daneen Schaeffer
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Reggie Bosma
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Vivian I. Bonano
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
- University Program in Genetics and Genomics
| | - Jang Wook Sohn
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Gabor Kemeny
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Abhinav Ettyreddy
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Mariano A. Garcia-Blanco
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
29
|
Brouillard F, Fritsch J, Edelman A, Ollero M. Contribution of proteomics to the study of the role of cytokeratins in disease and physiopathology. Proteomics Clin Appl 2012; 2:264-85. [PMID: 21136830 DOI: 10.1002/prca.200780018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cytokeratins (CKs), the most abundant group of cytoskeletal intermediate filaments, and proteomics are strongly connected. On the one hand, proteomics has been extremely useful to uncover new features and functions of CKs, on the other, the highly abundant CKs serve as an exceptional tool to test new technological developments in proteomics. As a result, proteomics has contributed to finding valuable associations of CKs with diseases as diverse as cancer, cystic fibrosis, steatohepatitis, viral and bacterial infection, keratoconus, vitreoretinopathy, preeclampsia or the chronic fatigue syndrome, as well as to characterizing their participation in a number of physiopathological processes, including drug resistance, response to toxicants, inflammation, stem cell differentiation, embryo development, and tissue repair. In some cases, like in cystic fibrosis, CKs have been described as potential therapeutic targets. The development of a specific field of proteomics where CKs become the main subject of research aims and hypotheses is suggested.
Collapse
Affiliation(s)
- Franck Brouillard
- INSERM, Unité 845, Paris, France; Faculté de Médecine René Descartes, Université Paris-Descartes, Plateau Protéomes IFR94, Paris, France
| | | | | | | |
Collapse
|
30
|
Liang X, Bhattacharya S, Bajaj G, Guha G, Wang Z, Jang HS, Leid M, Indra AK, Ganguli-Indra G. Delayed cutaneous wound healing and aberrant expression of hair follicle stem cell markers in mice selectively lacking Ctip2 in epidermis. PLoS One 2012; 7:e29999. [PMID: 22383956 PMCID: PMC3283611 DOI: 10.1371/journal.pone.0029999] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 12/08/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND COUP-TF interacting protein 2 [(Ctip2), also known as Bcl11b] is an important regulator of skin homeostasis, and is overexpressed in head and neck cancer. Ctip2(ep-/-) mice, selectively ablated for Ctip2 in epidermal keratinocytes, exhibited impaired terminal differentiation and delayed epidermal permeability barrier (EPB) establishment during development, similar to what was observed in Ctip2 null (Ctip2(-/-)) mice. Considering that as an important role of Ctip2, and the fact that molecular networks which underlie cancer progression partially overlap with those responsible for tissue remodeling, we sought to determine the role of Ctip2 during cutaneous wound healing. METHODOLOGY/PRINCIPAL FINDINGS Full thickness excisional wound healing experiments were performed on Ctip2(L2/L2) and Ctip2(ep-/-) animals per time point and used for harvesting samples for histology, immunohistochemistry (IHC) and immunoblotting. Results demonstrated inherent defects in proliferation and migration of Ctip2 lacking keratinocytes during re-epithelialization. Mutant mice exhibited reduced epidermal proliferation, delayed keratinocyte activation, altered cell-cell adhesion and impaired ECM development. Post wounding, Ctip2(ep-/-) mice wounds displayed lack of E-Cadherin suppression in the migratory tongue, insufficient expression of alpha smooth muscle actin (alpha SMA) in the dermis, and robust induction of K8. Importantly, dysregulated expression of several hair follicle (HF) stem cell markers such as K15, NFATc1, CD133, CD34 and Lrig1 was observed in mutant skin during wound repair. CONCLUSIONS/SIGNIFICANCE Results confirm a cell autonomous role of keratinocytic Ctip2 to modulate cell migration, proliferation and/or differentiation, and to maintain HF stem cells during cutaneous wounding. Furthermore, Ctip2 in a non-cell autonomous manner regulated granulation tissue formation and tissue contraction during wound closure.
Collapse
Affiliation(s)
- Xiaobo Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
| | - Shreya Bhattacharya
- Molecular and Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Gaurav Bajaj
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
| | - Gunjan Guha
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
| | - Zhixing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
| | - Hyo-Sang Jang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
| | - Mark Leid
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
- Molecular and Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Science Centre, Oregon State University, Corvallis, Oregon, United States of America
| | - Arup Kumar Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
- Molecular and Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Science Centre, Oregon State University, Corvallis, Oregon, United States of America
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, United States of America
- Molecular and Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
31
|
Trost A, Desch P, Wally V, Haim M, Maier RH, Reitsamer HA, Hintner H, Bauer JW, Onder K. Aberrant heterodimerization of keratin 16 with keratin 6A in HaCaT keratinocytes results in diminished cellular migration. Mech Ageing Dev 2010; 131:346-53. [PMID: 20403371 DOI: 10.1016/j.mad.2010.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 03/15/2010] [Accepted: 04/09/2010] [Indexed: 11/28/2022]
Abstract
Keratin filaments form obligatory heterodimers consisting of one type I and one type II keratin that build the intermediate filaments. In keratinocytes, type II keratin 6 (K6) interacts with type I keratin 16 (K16). We previously showed that the intermediate filament protein K16 is up-regulated in aged human skin. Here, we report that there is an obvious imbalance of K16 to K6 mRNA in in vivo and in vitro aging, which possibly leads to cellular effects. To unveil a possible biological function of K16 overexpression we investigated the migration potential of keratinocytes having up-regulated K16 expression in vitro. Two cell lines were established by transfection of human keratinocytes (HaCaT cells) with K16 or control vectors and subsequent fluorescence-activated cell sorting. By performing migration assays we were able to show a 90% reduction in the migration ability of the K16-overexpressing keratinocytes. In addition, a delay in wound closure associated with K16-overexpressing cells was shown by scratch assays. Transient overexpression of K6A in K16-overexpressing keratinocytes partially corrected the cell-migration defect. By real-time PCR we excluded co-regulation of the annotated interaction partner, K6, in the K16 cell line. Finally, we observed a decreased level of tyrosine phosphorylation in K16-overexpressing cells. Taken together, these data highlight the possibility of a physiological role for K6/K16 heterodimers in keratinocyte cell migration, in addition to the heterodimer's known functions in cell differentiation and mechanical resilience.
Collapse
Affiliation(s)
- A Trost
- Division of Molecular Dermatology, Department of Dermatology, Paracelsus Medical University Salzburg, Müllner Hauptstrasse 48, A-5020 Salzburg, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Maher J, Yamamoto M. The rise of antioxidant signaling--the evolution and hormetic actions of Nrf2. Toxicol Appl Pharmacol 2010; 244:4-15. [PMID: 20122947 DOI: 10.1016/j.taap.2010.01.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 01/19/2010] [Accepted: 01/19/2010] [Indexed: 11/17/2022]
Abstract
Organisms have evolved sophisticated and redundant mechanisms to manage oxidative and electrophilic challenges that arise from internal metabolism or xenobiotic challenge for survival. NF-E2-related factor 2 (Nrf2) is a transcription factor that has evolved over millennia from primitive origins, with homologues traceable back to invertebrate Caenorhabditis and Drosophila species. The ancestry of Nrf2 clearly has deep-seated roots in hematopoiesis, yet has diversified into a transcription factor that can mediate a multitude of antioxidant signaling and detoxification genes. In higher organisms, a more sophisticated means of tightly regulating Nrf2 activity was introduced via the cysteine-rich kelch-like ECH-associated protein 1 (Keap1), thus suggesting a need to modulate Nrf2 activity. This is evidenced in Keap1(-/-) mice, which succumb to juvenile mortality due to hyperkeratosis of the gastrointestinal tract. Although Nrf2 activation protects against acute toxicity and prevents or attenuates several disease states, constitutive activation in some tumors leads to poor clinical outcomes, suggesting Nrf2 has evolved in response to a multitude of selective pressures. The purpose of this review is to examine the origins of Nrf2, while highlighting the versatility and protective abilities elicited upon activation. Various model systems in which Nrf2 is normally beneficial but in which exaggerated pharmacology exacerbates a physiological or pathological condition will be addressed. Although Darwinian principles have selected Nrf2 activity for maximal beneficial effect based on environmental and oxidative challenge, both sub- or super-physiological effects have been noted to be detrimental. The functions of Nrf2 thus suggest a hormetic factor that has evolved empirically over time.
Collapse
Affiliation(s)
- Jonathan Maher
- Department of Medicinal Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | | |
Collapse
|
33
|
Adra S, Sun T, MacNeil S, Holcombe M, Smallwood R. Development of a three dimensional multiscale computational model of the human epidermis. PLoS One 2010; 5:e8511. [PMID: 20076760 PMCID: PMC2799518 DOI: 10.1371/journal.pone.0008511] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 12/02/2009] [Indexed: 11/18/2022] Open
Abstract
Transforming Growth Factor (TGF-β1) is a member of the TGF-beta superfamily ligand-receptor network. and plays a crucial role in tissue regeneration. The extensive in vitro and in vivo experimental literature describing its actions nevertheless describe an apparent paradox in that during re-epithelialisation it acts as proliferation inhibitor for keratinocytes. The majority of biological models focus on certain aspects of TGF-β1 behaviour and no one model provides a comprehensive story of this regulatory factor's action. Accordingly our aim was to develop a computational model to act as a complementary approach to improve our understanding of TGF-β1. In our previous study, an agent-based model of keratinocyte colony formation in 2D culture was developed. In this study this model was extensively developed into a three dimensional multiscale model of the human epidermis which is comprised of three interacting and integrated layers: (1) an agent-based model which captures the biological rules governing the cells in the human epidermis at the cellular level and includes the rules for injury induced emergent behaviours, (2) a COmplex PAthway SImulator (COPASI) model which simulates the expression and signalling of TGF-β1 at the sub-cellular level and (3) a mechanical layer embodied by a numerical physical solver responsible for resolving the forces exerted between cells at the multi-cellular level. The integrated model was initially validated by using it to grow a piece of virtual epidermis in 3D and comparing the in virtuo simulations of keratinocyte behaviour and of TGF-β1 signalling with the extensive research literature describing this key regulatory protein. This research reinforces the idea that computational modelling can be an effective additional tool to aid our understanding of complex systems. In the accompanying paper the model is used to explore hypotheses of the functions of TGF-β1 at the cellular and subcellular level on different keratinocyte populations during epidermal wound healing.
Collapse
Affiliation(s)
- Salem Adra
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
- * E-mail: (SA); (RS)
| | - Tao Sun
- Centre for Cell Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Sheila MacNeil
- Department of Engineering Materials, University of Sheffield, Sheffield, United Kingdom
| | - Mike Holcombe
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Rod Smallwood
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
- * E-mail: (SA); (RS)
| |
Collapse
|
34
|
Exploring hypotheses of the actions of TGF-beta1 in epidermal wound healing using a 3D computational multiscale model of the human epidermis. PLoS One 2009; 4:e8515. [PMID: 20046881 PMCID: PMC2796169 DOI: 10.1371/journal.pone.0008515] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 12/02/2009] [Indexed: 11/19/2022] Open
Abstract
In vivo and in vitro studies give a paradoxical picture of the actions of the key regulatory factor TGF-beta1 in epidermal wound healing with it stimulating migration of keratinocytes but also inhibiting their proliferation. To try to reconcile these into an easily visualized 3D model of wound healing amenable for experimentation by cell biologists, a multiscale model of the formation of a 3D skin epithelium was established with TGF-beta1 literature-derived rule sets and equations embedded within it. At the cellular level, an agent-based bottom-up model that focuses on individual interacting units (keratinocytes) was used. This was based on literature-derived rules governing keratinocyte behavior and keratinocyte/ECM interactions. The selection of these rule sets is described in detail in this paper. The agent-based model was then linked with a subcellular model of TGF-beta1 production and its action on keratinocytes simulated with a complex pathway simulator. This multiscale model can be run at a cellular level only or at a combined cellular/subcellular level. It was then initially challenged (by wounding) to investigate the behavior of keratinocytes in wound healing at the cellular level. To investigate the possible actions of TGF-beta1, several hypotheses were then explored by deliberately manipulating some of these rule sets at subcellular levels. This exercise readily eliminated some hypotheses and identified a sequence of spatial-temporal actions of TGF-beta1 for normal successful wound healing in an easy-to-follow 3D model. We suggest this multiscale model offers a valuable, easy-to-visualize aid to our understanding of the actions of this key regulator in wound healing, and provides a model that can now be used to explore pathologies of wound healing.
Collapse
|
35
|
Gu LH, Coulombe PA. Hedgehog signaling, keratin 6 induction, and sebaceous gland morphogenesis: implications for pachyonychia congenita and related conditions. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:752-61. [PMID: 18688029 DOI: 10.2353/ajpath.2008.071089] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Keratins 6a and b (K6a, K6b) belong to a subset of keratin genes with constitutive expression in epithelial appendages, and inducible expression in additional epithelia, when subjected to environmental challenges or disease. Mutations in K6a or K6b cause a broad spectrum of epithelial lesions that differentially affect nail, hair, and glands in humans. Some lesions reflect a loss of the structural support function shared by K6, other keratins, and intermediate filament proteins. The formation of sebaceous gland-derived epithelial cysts does not fit this paradigm, raising the question of the unique functions of different K6 isoforms in this setting. Here, we exploit a mouse model of constitutively expressed Gli2, a Hedgehog (Hh) signal effector, to show that K6a expression correlates with duct fate in sebaceous glands (SGs). Whether in the setting of Gli2 transgenic mice skin, which develops a prominent SG duct and additional pairs of highly branched SGs, or in wild-type mouse skin, K6a expression consistently coincides with Hh signaling in ductal tissue. Gli2 expression modestly transactivates a K6a promoter-driven reporter in heterologous systems. Our findings thus identify K6 as a marker of duct fate in SGs, partly in response to Hh signaling, with implications for the pathological expansion of SGs that arises in the context of certain keratin-based diseases and related disorders.
Collapse
Affiliation(s)
- Li-Hong Gu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | | |
Collapse
|
36
|
Abstract
The keratins are the typical intermediate filament proteins of epithelia, showing an outstanding degree of molecular diversity. Heteropolymeric filaments are formed by pairing of type I and type II molecules. In humans 54 functional keratin genes exist. They are expressed in highly specific patterns related to the epithelial type and stage of cellular differentiation. About half of all keratins—including numerous keratins characterized only recently—are restricted to the various compartments of hair follicles. As part of the epithelial cytoskeleton, keratins are important for the mechanical stability and integrity of epithelial cells and tissues. Moreover, some keratins also have regulatory functions and are involved in intracellular signaling pathways, e.g. protection from stress, wound healing, and apoptosis. Applying the new consensus nomenclature, this article summarizes, for all human keratins, their cell type and tissue distribution and their functional significance in relation to transgenic mouse models and human hereditary keratin diseases. Furthermore, since keratins also exhibit characteristic expression patterns in human tumors, several of them (notably K5, K7, K8/K18, K19, and K20) have great importance in immunohistochemical tumor diagnosis of carcinomas, in particular of unclear metastases and in precise classification and subtyping. Future research might open further fields of clinical application for this remarkable protein family.
Collapse
|
37
|
SERPINE1 (PAI-1) is deposited into keratinocyte migration "trails" and required for optimal monolayer wound repair. Arch Dermatol Res 2008; 300:303-10. [PMID: 18386027 DOI: 10.1007/s00403-008-0845-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 12/27/2007] [Accepted: 03/05/2008] [Indexed: 01/12/2023]
Abstract
Cutaneous tissue injury, both in vivo and in vitro, initiates activation of a "wound repair" transcriptional program. One such highly induced gene encodes plasminogen activator inhibitor type-1 (PAI-1, SERPINE1). PAI-1-GFP, expressed as a fusion protein under inducible control of +800 bp of the wound-activated PAI-1 promoter, prominently "marked" keratinocyte migration trails during the real-time of monolayer scrape-injury repair. Addition of active recombinant PAI-1 to wounded wild-type keratinocyte monolayers as well as to PAI-1(-/-) MEFs and PAI-1(-/-) keratinocytes significantly stimulated directional motility above basal levels in all cell types. PAI-1 expression knockdown or antibody-mediated functional inhibition, in contrast, effectively attenuated injury repair. The defect in wound-associated migratory activity as a consequence of antisense-mediated PAI-1 down-regulation was effectively reversed by addition of recombinant PAI-1 immediately after scrape injury. One possible mechanism underlying the PAI-1-dependent motile response may involve fine control of the keratinocyte substrate detachment/re-attachment process. Exogenous PAI-1 significantly enhanced keratinocyte spread cell "footprint" area while PAI-1 neutralizing antibodies, but not control non-immune IgG, effectively inhibited spreading with apoptotic hallmarks evident within 24 h. Importantly, PAI-1 not only stimulated keratinocyte adhesion and wound-initiated planar migration but also rescued keratinocytes from plasminogen-induced substrate detachment/anoikis. The early transcriptional response of the PAI-1 gene to monolayer trauma and its prominence in the injury repair genetic signature are consistent with its function as both a survival factor and regulator of the time course of epithelial migration as part of the cutaneous injury response program.
Collapse
|
38
|
Rodgers KE, Ellefson DD, Espinoza T, Hsu YH, diZerega GS, Mehrian-Shai R. Expression of intracellular filament, collagen, and collagenase genes in diabetic and normal skin after injury. Wound Repair Regen 2006; 14:298-305. [PMID: 16808808 DOI: 10.1111/j.1743-6109.2006.00124.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Reports have shown differences in gene expression in the skin of diabetic and normal mice both at baseline and after injury. Cluster analysis identified distinct expression patterns within intermediate filaments and extracellular proteins. This report addresses the effect of diabetes and injury on the expression of keratin-associated proteins, keratin complexes, procollagen, and collagenase (matrix metalloproteinase; MMP) genes. At baseline keratin-associated proteins and keratin complexes gene expression was increased in diabetic mice. After surgery, the level of expression for keratin-associated proteins and keratin complexes genes decreased in diabetic mice, but did not change in normal mice. If the expression of a procollagen gene differed between diabetic and normal mice, the expression was lower in diabetic mice. Procollagen gene expression was elevated after skin excision compared with noninjured skin. At baseline, the level of MMP and tissue inhibitor of metalloproteinase gene expression was comparable between mouse strains. With injury, the expression of several MMP genes was increased in both mouse strains, but to higher levels in diabetic mice. At day 7, the level of MMP-9 activity in granulation tissue was elevated. This alteration may contribute to delayed healing in diabetic mice. Therefore, differences in gene expression exist between mouse strains and can assist in understanding of physiological manifestations, including delayed healing, in diabetic mice.
Collapse
Affiliation(s)
- Kathleen E Rodgers
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California 90033, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Ekmektzoglou KA, Zografos GC. A concomitant review of the effects of diabetes mellitus and hypothyroidism in wound healing. World J Gastroenterol 2006; 12:2721-9. [PMID: 16718759 PMCID: PMC4130981 DOI: 10.3748/wjg.v12.i17.2721] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This paper reviews the negative impact of diabetes mellitus or hypothyroidism on wound healing, both in experimental and clinical settings. Since both are metabolic disorders of great clinical importance, special attention is given, not only to their pathophysiology, but also to their biochemical and histological effects on tissue integrity and regeneration. Also, special focus is awarded on wound healing of the gastrointestinal tract, i.e. in intestinal anastomosis, and how these disorders can lead to wound dehiscence. Since diabetes mellitus and hypothyroidism can coexist in clinical settings, more research must be directed on their influence on wound healing, considering them as one clinical entity.
Collapse
Affiliation(s)
- Konstantinos A Ekmektzoglou
- Laboratory of Experimental Surgery and Surgical Research, N.S. Christeas, Athens School of Medicine, Athens, Greece.
| | | |
Collapse
|
40
|
Hernández-Quintero M, Kuri-Harcuch W, González Robles A, Castro-Muñozledo F. Interleukin-6 promotes human epidermal keratinocyte proliferation and keratin cytoskeleton reorganization in culture. Cell Tissue Res 2006; 325:77-90. [PMID: 16550359 DOI: 10.1007/s00441-006-0173-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 01/19/2006] [Indexed: 11/30/2022]
Abstract
We have studied the effects of interleukin-6 (IL-6) on human epidermal keratinocytes by using serum-free culture conditions that allow the serial transfer, differentiation, and formation of well-organized multilayered epithelia. IL-6 at 2.5 ng/ml or higher concentrations promoted keratinocyte proliferation, with an ED(50) of about 15 ng/ml and a maximum effect at 50 ng/ml. IL-6 was 10-fold less potent than epidermal growth factor (EGF) or transforming growth factor-alpha (TGF-alpha) and supported keratinocyte growth for up to eight cumulative cell generations. IL-6-treated keratinocytes formed highly stratified colonies with a narrower proliferative/migratory rim than those keratinocytes stimulated with EGF or TGF-alpha; confluent epithelial sheets treated with IL-6 also underwent an increase in the number of cell layers. We also examined the effect of IL-6 on the keratin cytoskeleton. Immunostaining with anti-K16 monoclonal antibodies showed that the keratin network was aggregated and reorganized around cell nucleus and that this was not attributable to changes in keratin levels. This is the first report concerning the induction of the reorganization of keratin intermediate filaments by IL-6 in human epidermal keratinocytes.
Collapse
Affiliation(s)
- Miriam Hernández-Quintero
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN, Apdo. Postal 14-740, México 07000, Mexico
| | | | | | | |
Collapse
|
41
|
Goto M, Sumiyoshi H, Sakai T, Fässler R, Ohashi S, Adachi E, Yoshioka H, Fujiwara S. Elimination of epiplakin by gene targeting results in acceleration of keratinocyte migration in mice. Mol Cell Biol 2006; 26:548-58. [PMID: 16382146 PMCID: PMC1346887 DOI: 10.1128/mcb.26.2.548-558.2006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epiplakin (EPPK) was originally identified as a human epidermal autoantigen. To identify the function of epiplakin, we generated epiplakin "knockout" mice. These mice developed normally, with apparently normal epidermis and hair. Electron microscopy after immunostaining revealed the presence of EPPK adjacent to keratin filaments in wild-type mice, suggesting that epiplakin might associate with keratin. The appearance and localization of keratin bundles in intact epidermal keratinocytes of EPPK-/- mice were similar to those in wild-type mice. Wounds on the backs of EPPK-/- mice closed more rapidly than those on the backs of wild-type and heterozygous mice. The outgrowth of keratinocytes from skin explants from knockout mice was enhanced compared to outgrowth from explants from wild-type mice, even in the presence of mitomycin C, suggesting that the difference in keratinocyte outgrowth might be due to a difference in the speed of migration of keratinocytes. At wound edges in wild-type mice, EPPK was expressed in proliferating keratinocytes in conjunction with keratin 6. In EPPK-/- mice, no similar proliferating keratinocytes were observed, but migrating keratinocytes weakly expressed keratin 6. EPPK was coexpressed with keratin 6 in some keratinocytes in explant cultures from wild mice. We propose that EPPK might be linked functionally with keratin 6.
Collapse
Affiliation(s)
- Mizuki Goto
- Department of Anatomy, Biology and Medicine (Dermatology and Biochemistry), Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama-machi, Yufu 879-5593, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Hosokawa R, Urata MM, Ito Y, Bringas P, Chai Y. Functional significance of Smad2 in regulating basal keratinocyte migration during wound healing. J Invest Dermatol 2006; 125:1302-9. [PMID: 16354202 DOI: 10.1111/j.0022-202x.2005.23963.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the transforming growth factor-beta (TGF-beta) superfamily are critical regulators for wound healing. Transduction of TGF-beta signaling depends on activation of Smad2 and Smad3 by heteromeric complexes of ligand-specific receptors. Mice lacking Smad3 show accelerated wound healing, whereas the biological significance of Smad2-mediated TGF-beta signaling in wound healing remains unknown. To understand the function of Smad2 in regulating wound healing, we investigated the effect of Smad2 overexpression on epithelialization of incision wounds. Cutaneous wounds made in K14-Smad2 mice showed delayed healing. This delay in wound healing resulted from a defect in basal keratinocyte migration in K14-Smad2 mice. Instead of basal keratinocytes, the suprabasal layer of keratinocytes migrated into the wound region. Furthermore, overexpression of Smad2 activated the Smad2/Smad4 complex in keratinocytes and inhibited keratin 16 (K16) expression. As K16 functions as a critical mediator for reorganization of keratin filaments following skin injury, we propose that altered K16 expression affects the migration of basal keratinocytes in the K14-Smad2 mice. Taken together, these findings demonstrate a crucial role of TGF-beta signaling mediator Smad2 in regulating keratinocyte migration and re-epithelialization during wound healing. The K14-Smad2 transgenic mice can serve as an animal model for the investigation of TGF-beta signaling mechanism in regulating wound healing.
Collapse
Affiliation(s)
- Ryoichi Hosokawa
- Center for craniofacial molecular biology, School of Dentistry, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|
43
|
Bhawan J, Whren K, Panova I, Yaar M. Keratin 16 Expression in Epidermal Melanocytes of Normal Human Skin. Am J Dermatopathol 2005; 27:476-81. [PMID: 16314702 DOI: 10.1097/01.dad.0000179627.81172.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although the prevailing dogma states that keratin filaments are the hallmark of keratinocytes and other epithelial cells, recent publications suggest that they may be expressed by a variety of normal and malignant cells of different embryonic origin. Keratin expression has been reported in fibroblasts and endothelial cells as well as in various sarcomas. Also, some human melanomas express keratins in addition to the traditional diagnostic markers of differentiation, such as S-100 and melanocyte-specific antigens. Many studies have shown that cultured cells obtained from various melanomas express keratin. Most recently, keratin expression has also been shown in cultured melanocytes of normal skin. We now report that normal human melanocytes in vivo express keratin 16 (K16) but not keratins 1, 5, 8, 10, 14, or even keratin 6, the type II partner that is normally expressed with K16 in keratinocytes. Similarly, melanocytes in vitro express K16 but not K6. Keratin 16 expression in vivo was present in basal melanocytes in specimens derived from donors (0-77 years) and from different anatomic locations, suggesting that keratin 16 is constitutively expressed by all melanocytes. It appears that keratin expression may be more prevalent than previously assumed, and that these cytoskeletal filaments may play important roles in tissues and cells other than epithelia.
Collapse
Affiliation(s)
- Jag Bhawan
- Dermatopathology Section, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | |
Collapse
|
44
|
Coulombe PA, Tong X, Mazzalupo S, Wang Z, Wong P. Great promises yet to be fulfilled: defining keratin intermediate filament function in vivo. Eur J Cell Biol 2005; 83:735-46. [PMID: 15679118 DOI: 10.1078/0171-9335-00443] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Keratins are abundant proteins in epithelial cells, in which they occur as a cytoplasmic network of 10 - 12 nm wide intermediate filaments (IFs). They are encoded by a large family of conserved genes in mammals, with more than 50 individual members partitioned into two sequence types. A strict requirement for the heteropolymerization of type I and type II keratin proteins during filament formation underlies the pairwise transcriptional regulation of keratin genes. In addition, individual pairs are regulated in a tissue-type and differentiation-specific manner. Elucidating the rationale behind the diversity and differential distribution of keratin proteins offers the promise of novel insight into epithelial biology. At present, we know that keratin IFs act as resilient yet pliable scaffolds that endow epithelial cells with the ability to sustain mechanical and non-mechanical stresses. Accordingly, inherited mutations altering the coding sequence of keratins underlie several epithelial fragility disorders. In addition, keratin IFs influence the cellular response to pro-apoptotic signals in specific settings, and the routing of membrane proteins in polarized epithelia. Here we review studies focused on a subset of keratin genes, K6, K16 and K17, showing a complex regulation in vivo, including a widely known upregulation during wound repair and in diseased skin. Progress in defining the function of these and other keratins through gene manipulation in mice has been hampered by functional redundancy within the family. Still, detailed studies of the phenotype exhibited by K6 and K17 null mice yielded novel insight into the properties and function of keratin IFs in vivo.
Collapse
Affiliation(s)
- Pierre A Coulombe
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
45
|
Bhawan J, Bansal C, Whren K, Schwertschlag U. K16 expression in uninvolved psoriatic skin: a possible marker of pre-clinical psoriasis. J Cutan Pathol 2005; 31:471-6. [PMID: 15239676 DOI: 10.1111/j.0303-6987.2004.0220.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND K16, a type I keratin, is upregulated in hyperproliferative states including psoriasis. It has been used as a marker of psoriasis and its expression is upregulated in relapsing psoriasis and downregulating in resolving. We evaluated non-lesional psoriatic skin for K16 expression. METHODS Sixty-seven non-lesional and lesional skin samples from patients with psoriasis and normal skin from 19 non-psoriatic patients were studied by immunohistochemistry on frozen sections with K16. RESULTS Seventeen of 19 normal skin samples showed staining of basal cells in the deeper part of the rete ridges. Sixty-two non-lesional psoriatic skin samples showed intense basal staining of K16. Of the remaining five non-lesional samples, diffuse intense suprabasal staining in one, pan-epidermal staining in two, and no staining was seen in two samples. Suprabasal (37), diffuse (14), sandwich (12), and basal (3) pattern staining were seen in psoriatic skin. One psoriatic skin sample did not show any expression. CONCLUSION Our results demonstrate that K16 expression is also observed in non-lesional psoriatic skin and may serve as a marker of preclinical psoriasis.
Collapse
Affiliation(s)
- Jag Bhawan
- Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | |
Collapse
|
46
|
Providence KM, Higgins PJ. PAI-1 expression is required for epithelial cell migration in two distinct phases of in vitro wound repair. J Cell Physiol 2004; 200:297-308. [PMID: 15174100 DOI: 10.1002/jcp.20016] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several proteases and their specific inhibitors modulate the interdependent processes of cell migration and matrix proteolysis as part of the global program of trauma repair. Expression of plasminogen activator inhibitor type-1 (PAI-1), a serine protease inhibitor (SERPIN) important in the control of barrier proteolysis and cell-to-matrix adhesion, for example, is spatially-temporally regulated following epithelial denudation injury in vitro as well as in vivo. PAI-1 mRNA/protein synthesis was induced early after epidermal monolayer scraping and restricted to keratinocytes comprising the motile cohort closely recapitulating, thereby, similar events during cutaneous healing. The time course of PAI-1 promoter-driven PAI-1-GFP fusion "reporter" expression in wound-juxtaposed cells approximated that of the endogenous PAI-1 gene confirming the location-specificity of gene regulation in this model. ERK activation was evident within 5 min after injury and particularly prominent in cells residing at the scrape-edge (suggesting a possible role in PAI-1 induction and/or the motile response) as was myosin light chain (MLC) phosphorylation. Indeed, MEK blockade with PD98059 or U0126 attenuated keratinocyte migration (by > or =60%), as did transient transfection of a dominant-negative ERK1 construct (40% decrease in monolayer repair), and completely inhibited PAI-1 transcript expression. Anti-sense down-regulation of PAI-1 synthesis (by 80-85%), or addition of PAI-1 neutralizing antibodies also inhibited injury site closure over a 24 h period establishing that PAI-1 was required for efficient long-term planar motility in this system. PAI-1 anti-sense transfection or actinomycin D transcriptional blockade, in contrast, did not affect the initial migratory response suggesting that residual PAI-1 protein levels (at least in transfectant cells and actinomycin D-treated cultures) may be sufficient to support early cell movement. Pharmacologic inhibition of keratinocyte MEK signaling effectively ablated scrape-induced PAI-1 mRNA expression but failed to attenuate wound-associated increases in cellular PAI-1 protein levels soon after monolayer injury. Collectively, these data suggest that basal PAI-1 transcripts may be mobilized for initial PAI-1 synthesis and, perhaps, the early motile response while maintenance of the normal rate of migration requires the prolonged PAI-1 expression that typically accompanies the repair response. To assess this possibility, scrape site closure studies were designed using keratinocytes isolated from PAI-1-/- mice. PAI-1-/- keratinocytes, in fact, had a significant wound healing defect evident even within the first 6 h following monolayer denudation injury. Addition of active PAI-1 protein to PAI-/- keratinocytes rescued the migratory phenotype that that approximating wild-type cells. These findings validate use of the present keratinocyte model to investigate injury-related controls on PAI-1 gene regulation and, collectively, implicate participation of PAI-1 in two distinct phases of epidermal wound repair.
Collapse
Affiliation(s)
- Kirwin M Providence
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, New York 12208, USA
| | | |
Collapse
|
47
|
Safer JD, Crawford TM, Holick MF. A role for thyroid hormone in wound healing through keratin gene expression. Endocrinology 2004; 145:2357-61. [PMID: 14736740 DOI: 10.1210/en.2003-1696] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The importance of thyroid hormone (TH) in wound healing is not well understood. To gain insight, we evaluated the impact of TH deficiency on wound-healing genes in cultured keratinocytes. By RT-PCR, keratin 6a (K6a) and 16 (K16) gene expression in TH replete cells was 3.8- (P < 0.005) and 1.9-fold (P < 0.05) greater, respectively, than expression in TH-deficient cells. By real-time PCR, TH replete cell expression of K6a, K16, and K17 was greater than in deficient cells: 18- (P < 0.001), 10- (P < 0.001), and 4-fold (P < 0.005), respectively. To examine TH requirement for optimal wound healing, we contrasted TH-deficient vs. ip T(3)-treated mice. Four days after wounding, ip T(3)-treated mice had twice the degree of wound closure as hypothyroid mice (P < 0.001). By RT-PCR, K6a and K17 gene expression from control mouse skin was greater than from hypothyroid mouse skin: 5- (P < 0.001) and 1.7-fold (P < 0.05), respectively. T(3) is necessary for the keratinocyte proliferation required for optimal wound healing. T(3) exerts influence by stimulating expression of the wound-healing keratin genes. Thus, for hypothyroid patients undergoing surgery that cannot be delayed until euthyroidism is achieved, our data support T(3) treatment for the perioperative period.
Collapse
Affiliation(s)
- Joshua D Safer
- Section of Endocrinology, Diabetes, and Nutrition, Boston University School of Medicine, 715 Albany Street, Room M-1022, Boston, Massachusetts 02118, USA.
| | | | | |
Collapse
|
48
|
Wong P, Coulombe PA. Loss of keratin 6 (K6) proteins reveals a function for intermediate filaments during wound repair. ACTA ACUST UNITED AC 2003; 163:327-37. [PMID: 14568992 PMCID: PMC2173512 DOI: 10.1083/jcb.200305032] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability to heal wounds is vital to all organisms. In mammalian tissues, alterations in intermediate filament (IF) gene expression represent an early reaction of cells surviving injury. We investigated the role of keratin IFs during the epithelialization of skin wounds using a keratin 6α and 6β (K6α/K6β)-null mouse model. In skin explant culture, null keratinocytes exhibit an enhanced epithelialization potential due to increased migration. The extent of the phenotype is strain dependent, and is accompanied by alterations in keratin IF and F-actin organization. However, in wounded skin in vivo, null keratinocytes rupture as they attempt to migrate under the blood clot. Fragility of the K6α/K6β-null epidermis is confirmed when applying trauma to chemically treated skin. We propose that the alterations in IF gene expression after tissue injury foster a compromise between the need to display the cellular pliability necessary for timely migration and the requirement for resilience sufficient to withstand the rigors of a wound site.
Collapse
Affiliation(s)
- Pauline Wong
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
49
|
Herrmann H, Hesse M, Reichenzeller M, Aebi U, Magin TM. Functional complexity of intermediate filament cytoskeletons: from structure to assembly to gene ablation. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 223:83-175. [PMID: 12641211 DOI: 10.1016/s0074-7696(05)23003-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The cell biology of intermediate filament (IF) proteins and their filaments is complicated by the fact that the members of the gene family, which in humans amount to at least 65, are differentially expressed in very complex patterns during embryonic development. Thus, different tissues and cells express entirely different sets and amounts of IF proteins, the only exception being the nuclear B-type lamins, which are found in every cell. Moreover, in the course of evolution the individual members of this family have, within one species, diverged so much from each other with regard to sequence and thus molecular properties that it is hard to envision a unifying kind of function for them. The known epidermolytic diseases, caused by single point mutations in keratins, have been used as an argument for a role of IFs in mechanical "stress resistance," something one would not have easily ascribed to the beaded chain filaments, a special type of IF in the eye lens, or to nuclear lamins. Therefore, the power of plastic dish cell biology may be limited in revealing functional clues for these structural elements, and it may therefore be of interest to go to the extreme ends of the life sciences, i.e., from the molecular properties of individual molecules including their structure at the atomic level to targeted inactivation of their genes in living animals, mouse, and worm to define their role more precisely in metazoan cell physiology.
Collapse
Affiliation(s)
- Harald Herrmann
- Division of Cell Biology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
50
|
Morley SM, D'Alessandro M, Sexton C, Rugg EL, Navsaria H, Shemanko CS, Huber M, Hohl D, Heagerty AI, Leigh IM, Lane EB. Generation and characterization of epidermolysis bullosa simplex cell lines: scratch assays show faster migration with disruptive keratin mutations. Br J Dermatol 2003; 149:46-58. [PMID: 12890194 DOI: 10.1046/j.1365-2133.2003.05493.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Epidermolysis bullosa simplex (EBS) is an inherited skin fragility disorder caused by mutations in keratin intermediate filament proteins. While discoveries of these mutations have increased understanding of the role of keratins and other intermediate filaments in epithelial tissues, progress towards the development of therapy for these disorders is much slower. OBJECTIVES Cell culture model systems that display these structural defects are needed for analysis of the cellular consequences of the mutations and to enable possible therapeutic strategies to be developed. Our aim was to generate immortalized cell lines as such model systems for the study of EBS. METHODS We generated a series of stable cell lines expressing EBS-associated keratin mutations, by immortalizing keratinocytes from EBS-affected skin biopsies with either simian virus 40 (SV40) T antigen or human papillomavirus 16 (HPV16) E6/E7, and assessed their keratin expression (by immunofluorescence), proliferation rates and migratory behaviour (in outgrowth and scratch wound assays). RESULTS Clonal immortalized keratinocyte cell lines KEB-1, KEB-2, KEB-3 (using SV40 T antigen) and KEB-4, KEB-7 and NEB-1 (using HPV16 E6/E7) were established. These include two lines from a single individual with Weber-Cockayne EBS (i.e. KEB-3 and KEB-4, mutation K14 V270M), and three cell lines from a second family, two from siblings carrying the same mutation (KEB-1, KEB-2 lines from Dowling-Meara EBS, mutation K5 E475G) and one from an unaffected relative (NEB-1). The sixth cell line (KEB-7), with a previously unreported severe mutation (K14 R125P), was the only one to show keratin aggregates in resting conditions. Despite variations in the immortalization procedure, there was no significant difference between cell lines in keratin expression, outgrowth capabilities or response to transient heat shock. However, cell migration, as measured by speed of scratch wound closure, was significantly faster in cells with severe EBS mutations. CONCLUSIONS These cell lines provide useful culture systems in which to assess aspects of EBS-induced cell changes. The faster migration after scratch wounding of the EBS keratinocytes may be a consequence of the known upregulation of stress-activated kinase pathways in these cells.
Collapse
Affiliation(s)
- S M Morley
- Cancer Research UK Cell Structure Research Group, Dundee University School of Life Sciences, MSI/WTB Complex, Dow Street, Dundee DD1 5EH, U.K
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|