1
|
Malik B, Vokic I, Mohr T, Poppelaars M, Holcmann M, Novoszel P, Timelthaler G, Lendl T, Krauss D, Elling U, Mildner M, Penninger JM, Petzelbauer P, Sibilia M, Csiszar A. FAM3C/ILEI protein is elevated in psoriatic lesions and triggers psoriasiform hyperproliferation in mice. EMBO Mol Med 2023; 15:e16758. [PMID: 37226685 PMCID: PMC10331587 DOI: 10.15252/emmm.202216758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
FAM3C/ILEI is an important cytokine for tumor progression and metastasis. However, its involvement in inflammation remains elusive. Here, we show that ILEI protein is highly expressed in psoriatic lesions. Inducible keratinocyte-specific ILEI overexpression in mice (K5-ILEIind ) recapitulates many aspects of psoriasis following TPA challenge, primarily manifested by impaired epidermal differentiation and increased neutrophil recruitment. Mechanistically, ILEI triggers Erk and Akt signaling, which then activates STAT3 via Ser727 phosphorylation. Keratinocyte-specific ILEI deletion ameliorates TPA-induced skin inflammation. A transcriptomic ILEI signature obtained from the K5-ILEIind model shows enrichment in several signaling pathways also found in psoriasis and identifies urokinase as a targetable enzyme to counteract ILEI activity. Pharmacological inhibition of urokinase in TPA-induced K5-ILEIind mice results in significant improvement of psoriasiform symptoms by reducing ILEI secretion. The ILEI signature distinguishes psoriasis from healthy skin with uPA ranking among the top "separator" genes. Our study identifies ILEI as a key driver in psoriasis, indicates the relevance of ILEI-regulated genes for disease manifestation, and shows the clinical impact of ILEI and urokinase as novel potential therapeutic targets in psoriasis.
Collapse
Affiliation(s)
- Barizah Malik
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
- Present address:
School of Biochemistry and Biotechnology, Quaid‐e‐Azam CampusUniversity of the PunjabLahorePakistan
| | - Iva Vokic
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Thomas Mohr
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
- Department of Analytical Chemistry, Faculty of ChemistryUniversity of ViennaViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University ViennaViennaAustria
| | - Marle Poppelaars
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Martin Holcmann
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Philipp Novoszel
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Gerald Timelthaler
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Thomas Lendl
- Research Institute of Molecular PathologyViennaAustria
| | - Dana Krauss
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Ulrich Elling
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)ViennaAustria
| | - Michael Mildner
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)ViennaAustria
- Department of Medical Genetics, Life Science InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | - Maria Sibilia
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Agnes Csiszar
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| |
Collapse
|
2
|
Parsa S, Soheili ZS, Alizadeh Otaghvar H, Behrangi E, Mansouri P, Lubberts E, Mowla SJ. BCL11B Is Involved in Stress-Induced Differentiation of Keratinocytes and Has A Potential Role in Psoriasis Pathogenesis. CELL JOURNAL 2023; 25:300-306. [PMID: 37300291 PMCID: PMC10257055 DOI: 10.22074/cellj.2023.558003.1090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/17/2022] [Accepted: 02/08/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVE Psoriasis is a common, auto-immune skin disease characterized by abnormal proliferation and differentiation of keratinocytes. Studies revealed the role of stress stimulators in the pathogenesis of psoriasis. Oxidative stress and heat shock are two important stress factors tuning differentiation and proliferation of keratinocytes, regarding to psoriasis disease. BCL11B is a transcription factor with critical role in embryonic keratinocyte differentiation and proliferation. Given this, in keratinocytes we have investigated potential role of BCL11B in stress-induced differentiation. Furthermore, we searched for a potential intercommunication between BCL11B expression and psoriasis-related keratinocyte stress factors. MATERIALS AND METHODS In this experimental study, data sets of psoriatic and healthy skin samples were downloaded in silico and BCL11B was chosen as a potential transcription factor to analyze. Next, a synchronized in vitro model was designed for keratinocyte proliferation and differentiation. Oxidative stress and heat shock treatments were employed on HaCaT keratinocytes in culture, and BCL11B expression level was measured. Cell proliferation rate and differentiation were analyzed by synchronized procedure test. Flow cytometry was done to analyze cell cycle alterations due to the oxidative stress. RESULTS Quantitative reverse transcription polymerase chain reaction (qRT-PCR) data revealed a significant upregulation of BCL11B expression in keratinocytes, by 24 hours after initiating differentiation. However, it was followed by a significant down-regulation in almost all the experiments, including the synchronized model. Flow cytometer data demonstrated a G1 cell cycle arrest in the treated cells. CONCLUSION Results indicated a remarkable role of BCL11B in differentiation and proliferation of HaCaT keratinocytes. This data along with the results of flow cytometer suggested a probable role for BCL11B in stress-induced differentiation, which is similar to what is happening during initiation and progression of normal differentiation.
Collapse
Affiliation(s)
- Sara Parsa
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | | | - Elham Behrangi
- Department of Dermatology, Rasoul-E Akram Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvin Mansouri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Erik Lubberts
- Department of Immunology, Erasmus Mc, University Medical Center, Rotterdam, The Netherlands
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Dou J, Zhang Z, Xu X, Zhang X. Exploring the effects of Chinese herbal ingredients on the signaling pathway of alopecia and the screening of effective Chinese herbal compounds. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115320. [PMID: 35483562 DOI: 10.1016/j.jep.2022.115320] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE alopecia is a hair disorder that can add a significant medical and psychological burden to patients. Currently, the FDA-approved drugs for the treatment of androgenetic alopecia (AGA) are minoxidil and finasteride and immunosuppressives are therapeutic options for alopecia areata (AA), but the objective adverse effects and high cost of these treatments reduce patient compliance and thus the effectiveness of the drugs. Traditional Chinese medicine (TCM) has good efficacy, a high safety profile and low treatment costs, but its mechanism of action is still not fully understood. The use of signaling pathways to modulate hair loss is a major direction in the study of the pathogenesis and pharmacology of alopecia. AIM OF THE STUDY This review aims to collect the results of experimental studies related to alopecia, to screen previously documented combinations of herbs claimed to be effective based on the herbs and their constituent compounds used in the identified studies, and to uncover other useful information that we hope will better guide the clinical application and scientific research of drug combinations or individual herbs for the treatment of alopecia. MATERIALS AND METHODS We have reviewed experimental studies to determine the methods used and the mechanisms of action of the herbs and constituent compounds. The following keywords were searched in databases, including PubMed, EMBASE, CNKI and CSTJ." Medicinal plants" "Chinese herbal medicine", "hair loss", " alopecia", "androgenetic alopecia" and " alopecia areata ". We also collected combinations of drugs from books approved by various schools for screening. RESULTS Using known combinations of compounds within herbal medicine to match the documented combinations, 34 topical combinations and 74 oral combinations were identified, and among the 108 herbal combinations screened Angelica, Rehmannia glutinosaLigusticum chuanxiong hort, Radix Rehmanniae, etc. The number of occurrences was very high, and the association with vascular drugs was also found to be very close. CONCLUSIONS This review further elucidates the therapeutic mechanisms of the compounds within the herbal components associated with alopecia and screens for other combinations that may be dominated by this component for the treatment of alopecia, uncovering compounds from other drugs that may be key factors in the treatment of alopecia. This improvement will provide a better quality of evidence for the effectiveness of herbs and compounds used to treat alopecia.
Collapse
Affiliation(s)
- Jinjin Dou
- The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Zhiming Zhang
- The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Xianrong Xu
- The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Xiwu Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China.
| |
Collapse
|
4
|
Dermal extracellular matrix molecules in skin development, homeostasis, wound regeneration and diseases. Semin Cell Dev Biol 2022; 128:137-144. [PMID: 35339360 DOI: 10.1016/j.semcdb.2022.02.027] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/22/2022] [Indexed: 12/14/2022]
Abstract
The extracellular matrix (ECM) is a dynamic structure that surrounds and anchors cellular components in tissues. In addition to functioning as a structural scaffold for cellular components, ECMs also regulate diverse biological functions, including cell adhesion, proliferation, differentiation, migration, cell-cell interactions, and intracellular signaling events. Dermal fibroblasts (dFBs), the major cellular source of skin ECM, develop from a common embryonic precursor to the highly heterogeneous subpopulations during development and adulthood. Upon injury, dFBs migrate into wound granulation tissue and transdifferentiate into myofibroblasts, which play a critical role in wound contraction and dermal ECM regeneration and deposition. In this review, we describe the plasticity of dFBs during development and wound healing and how various dFB-derived ECM molecules, including collagen, proteoglycans, glycosaminoglycans, fibrillins and matricellular proteins are expressed and regulated, and in turn how these ECM molecules play a role in regulating the function of dFBs and immune cells. Finally, we describe how dysregulation of ECM matrix is associated the pathogenesis of wound healing related skin diseases, including chronic wounds and keloid.
Collapse
|
5
|
Bhattacharya N, Indra AK, Ganguli-Indra G. Selective Ablation of BCL11A in Epidermal Keratinocytes Alters Skin Homeostasis and Accelerates Excisional Wound Healing In Vivo. Cells 2022; 11:cells11132106. [PMID: 35805190 PMCID: PMC9265695 DOI: 10.3390/cells11132106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/27/2023] Open
Abstract
Transcriptional regulator BCL11A plays a crucial role in coordinating a suite of developmental processes including skin morphogenesis, barrier functions and lipid metabolism. There is little or no reports so far documenting the role of BCL11A in postnatal adult skin homeostasis and in the physiological process of tissue repair and regeneration. The current study establishes for the first time the In Vivo role of epidermal BCL11A in maintaining adult epidermal homeostasis and as a negative regulator of cutaneous wound healing. Conditional ablation of Bcl11a in skin epidermal keratinocytes (Bcl11aep−/−mice) enhances the keratinocyte proliferation and differentiation program, suggesting its critical role in epidermal homeostasis of adult murine skin. Further, loss of keratinocytic BCL11A promotes rapid closure of excisional wounds both in a cell autonomous manner likely via accelerating wound re-epithelialization and in a non-cell autonomous manner by enhancing angiogenesis. The epidermis specific Bcl11a knockout mouse serves as a prototype to gain mechanistic understanding of various downstream pathways converging towards the manifestation of an accelerated healing phenotype upon its deletion.
Collapse
Affiliation(s)
- Nilika Bhattacharya
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Science Center, Oregon State University, Corvallis, OR 97331, USA
- OHSU Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Department of Dermatology, OHSU, Portland, OR 97239, USA
- Correspondence: (A.K.I.); (G.G.-I.)
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
- OHSU Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Correspondence: (A.K.I.); (G.G.-I.)
| |
Collapse
|
6
|
Permatasari HK, Nakahata S, Ichikawa T, Fauzi YR, Kiyonari H, Shide K, Kameda T, Shimoda K, Ono M, Taki T, Taniwaki M, Futakuchi M, Morishita K. Oncogenic isoform switch of tumor suppressor BCL11B in adult T-cell leukemia/lymphoma. Exp Hematol 2022; 111:41-49. [PMID: 35421541 DOI: 10.1016/j.exphem.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022]
Abstract
B-cell leukemia/lymphoma 11B (BCL11B) is a transcription factor important for T-cell development and acts as a tumor suppressor gene in T-cell acute lymphoblastic leukemia. Here, we identified BCL11B as a candidate leukemia-associated gene in human T-cell leukemia virus type 1 (HTLV-1)-induced adult T-cell leukemia/lymphoma (ATLL). Interestingly, the short-form lacking exon 3 (BCL11B/S) protein was more highly expressed than the full-length BCL11B (BCL11B/L) in leukemic cells from most of the ATLL patients, although expression ratios of BCL11B/L to BCL11B/S were almost equally expressed in control CD4+ T cells. BCL11B/S and BCL11B/L exhibited distinct subcellular localization and differential effects on cellular growth; BCL11B/L expression showed nuclear localization and inhibited cell growth in ATLL cells, whereas BCL11B/S showed nucleo-cytoplasmic distribution and accelerated cell growth. Furthermore, BCL11B/S expression accelerated the development of T-cell leukemia/lymphomas in transgenic mice carrying HTLV-1/HBZ, a critical viral factor in leukemogenesis, whereas these phenotypes did not occur in the double transgenic mice carrying BCL11B/L and HTLV-1/HBZ. In HTLV-1-infected T-cell lines, BCL11B expression is downregulated by HTLV-1/Tax, a viral factor necessary at the early stage of leukemogenesis. These results suggest that downregulation of BCL11B/L expression and upregulation of BCL11B/S may contribute to the development and progression of ATLL.
Collapse
Affiliation(s)
- Happy Kurnia Permatasari
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Biochemistry and Biomolecular, Faculty of Medicine, Universitas Brawijaya, 65145 Malang, East Java, Indonesia
| | - Shingo Nakahata
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Tomonaga Ichikawa
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Yanuar Rahmat Fauzi
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuou-ku, Kobe 650-0047, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Takuro Kameda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Masaya Ono
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Tomohiko Taki
- Department of Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; Center for Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masafumi Taniwaki
- Center for Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Mitsuru Futakuchi
- Department of Pathology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Project for Advanced Medical Research and Development, Project Research Division, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| |
Collapse
|
7
|
Bhattacharya N, Ganguli-Indra G, Indra AK. CTIP2 and lipid metabolism: regulation in skin development and associated diseases. Expert Rev Proteomics 2021; 18:1009-1017. [PMID: 34739354 PMCID: PMC9119322 DOI: 10.1080/14789450.2021.2003707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/02/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION COUP-TF INTERACTING PROTEIN 2 (CTIP2) is a crucial transcription factor exhibiting its control through coupled modulation of epigenetic modification and transcriptional regulation of key genes related to skin, immune, and nervous system development. Previous studies have validated the essential role of CTIP2 in skin development and maintenance, propagating its effects in epidermal permeability barrier (EPB) homeostasis, wound healing, inflammatory diseases, and epithelial cancers. Lipid metabolism dysregulation, on the other hand, has also established its independent emerging role over the years in normal skin development and various skin-associated ailments. This review focuses on the relatively unexplored connections between CTIP2-mediated control of lipid metabolism and alteration of EPB homeostasis, delayed wound healing, inflammatory diseases exacerbation, and cancer promotion and progression. AREAS COVERED Here we have discussed the intricate interplay of various endogenous lipids and lipoproteins accompanying skin development and associated disease processes and the possible link to CTIP2-mediated regulation of lipid metabolism. EXPERT OPINION Establishing the link between CTIP2 and lipid metabolism alterations in the context of skin morphogenesis and diverse types of skin diseases including cancer can help us identify novel targets for effective therapeutic intervention.
Collapse
Affiliation(s)
- Nilika Bhattacharya
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
- Department of Biochemistry and Biophysics, OSU, Corvallis, OR, USA
- Linus Pauling Science Center, OSU, Corvallis, OR, USA
- Department of Dermatology, OHSU, Portland, OR, USA
| |
Collapse
|
8
|
Imbert F, Langford D. Viruses, SUMO, and immunity: the interplay between viruses and the host SUMOylation system. J Neurovirol 2021; 27:531-541. [PMID: 34342851 PMCID: PMC8330205 DOI: 10.1007/s13365-021-00995-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022]
Abstract
The conjugation of small ubiquitin-like modifier (SUMO) proteins to substrates is a well-described post-translational modification that regulates protein activity, subcellular localization, and protein-protein interactions for a variety of downstream cellular activities. Several studies describe SUMOylation as an essential post-translational modification for successful viral infection across a broad range of viruses, including RNA and DNA viruses, both enveloped and un-enveloped. These viruses include but are not limited to herpes viruses, human immunodeficiency virus-1, and coronaviruses. In addition to the SUMOylation of viral proteins during infection, evidence shows that viruses manipulate the SUMO pathway for host protein SUMOylation. SUMOylation of host and viral proteins greatly impacts host innate immunity through viral manipulation of the host SUMOylation machinery to promote viral replication and pathogenesis. Other post-translational modifications like phosphorylation can also modulate SUMO function. For example, phosphorylation of COUP-TF interacting protein 2 (CTIP2) leads to its SUMOylation and subsequent proteasomal degradation. The SUMOylation of CTIP2 and subsequent degradation prevents CTIP2-mediated recruitment of a multi-enzymatic complex to the HIV-1 promoter that usually prevents the transcription of integrated viral DNA. Thus, the "SUMO switch" could have implications for CTIP2-mediated transcriptional repression of HIV-1 in latency and viral persistence. In this review, we describe the consequences of SUMO in innate immunity and then focus on the various ways that viral pathogens have evolved to hijack the conserved SUMO machinery. Increased understanding of the many roles of SUMOylation in viral infections can lead to novel insight into the regulation of viral pathogenesis with the potential to uncover new targets for antiviral therapies.
Collapse
Affiliation(s)
- Fergan Imbert
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, PA, 19140, Philadelphia, USA
| | - Dianne Langford
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, PA, 19140, Philadelphia, USA.
| |
Collapse
|
9
|
Daher MT, Bausero P, Agbulut O, Li Z, Parlakian A. Bcl11b/Ctip2 in Skin, Tooth, and Craniofacial System. Front Cell Dev Biol 2020; 8:581674. [PMID: 33363142 PMCID: PMC7758212 DOI: 10.3389/fcell.2020.581674] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022] Open
Abstract
Ctip2/Bcl11b is a zinc finger transcription factor with dual action (repression/activation) that couples epigenetic regulation to gene transcription during the development of various tissues. It is involved in a variety of physiological responses under healthy and pathological conditions. Its role and mechanisms of action are best characterized in the immune and nervous systems. Furthermore, its implication in the development and homeostasis of other various tissues has also been reported. In the present review, we describe its role in skin development, adipogenesis, tooth formation and cranial suture ossification. Experimental data from several studies demonstrate the involvement of Bcl11b in the control of the balance between cell proliferation and differentiation during organ formation and repair, and more specifically in the context of stem cell self-renewal and fate determination. The impact of mutations in the coding sequences of Bcl11b on the development of diseases such as craniosynostosis is also presented. Finally, we discuss genome-wide association studies that suggest a potential influence of single nucleotide polymorphisms found in the 3’ regulatory region of Bcl11b on the homeostasis of the cardiovascular system.
Collapse
Affiliation(s)
- Marie-Thérèse Daher
- Biological Adaptation and Ageing, Inserm ERL U1164, UMR CNRS 8256, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Pedro Bausero
- Biological Adaptation and Ageing, Inserm ERL U1164, UMR CNRS 8256, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Onnik Agbulut
- Biological Adaptation and Ageing, Inserm ERL U1164, UMR CNRS 8256, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Zhenlin Li
- Biological Adaptation and Ageing, Inserm ERL U1164, UMR CNRS 8256, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Ara Parlakian
- Biological Adaptation and Ageing, Inserm ERL U1164, UMR CNRS 8256, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| |
Collapse
|
10
|
Simon R, Wiegreffe C, Britsch S. Bcl11 Transcription Factors Regulate Cortical Development and Function. Front Mol Neurosci 2020; 13:51. [PMID: 32322190 PMCID: PMC7158892 DOI: 10.3389/fnmol.2020.00051] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
Transcription factors regulate multiple processes during brain development and in the adult brain, from brain patterning to differentiation and maturation of highly specialized neurons as well as establishing and maintaining the functional neuronal connectivity. The members of the zinc-finger transcription factor family Bcl11 are mainly expressed in the hematopoietic and central nervous systems regulating the expression of numerous genes involved in a wide range of pathways. In the brain Bcl11 proteins are required to regulate progenitor cell proliferation as well as differentiation, migration, and functional integration of neural cells. Mutations of the human Bcl11 genes lead to anomalies in multiple systems including neurodevelopmental impairments like intellectual disabilities and autism spectrum disorders.
Collapse
Affiliation(s)
- Ruth Simon
- Institute of Molecular and Cellular Anatomy, Ulm University, Germany
| | | | - Stefan Britsch
- Institute of Molecular and Cellular Anatomy, Ulm University, Germany
| |
Collapse
|
11
|
Panagiotakos G, Haveles C, Arjun A, Petrova R, Rana A, Portmann T, Paşca SP, Palmer TD, Dolmetsch RE. Aberrant calcium channel splicing drives defects in cortical differentiation in Timothy syndrome. eLife 2019; 8:51037. [PMID: 31868578 PMCID: PMC6964969 DOI: 10.7554/elife.51037] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/21/2019] [Indexed: 01/02/2023] Open
Abstract
The syndromic autism spectrum disorder (ASD) Timothy syndrome (TS) is caused by a point mutation in the alternatively spliced exon 8A of the calcium channel Cav1.2. Using mouse brain and human induced pluripotent stem cells (iPSCs), we provide evidence that the TS mutation prevents a normal developmental switch in Cav1.2 exon utilization, resulting in persistent expression of gain-of-function mutant channels during neuronal differentiation. In iPSC models, the TS mutation reduces the abundance of SATB2-expressing cortical projection neurons, leading to excess CTIP2+ neurons. We show that expression of TS-Cav1.2 channels in the embryonic mouse cortex recapitulates these differentiation defects in a calcium-dependent manner and that in utero Cav1.2 gain-and-loss of function reciprocally regulates the abundance of these neuronal populations. Our findings support the idea that disruption of developmentally regulated calcium channel splicing patterns instructively alters differentiation in the developing cortex, providing important in vivo insights into the pathophysiology of a syndromic ASD.
Collapse
Affiliation(s)
- Georgia Panagiotakos
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States.,Eli & Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Christos Haveles
- Eli & Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Arpana Arjun
- Eli & Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States.,Graduate Program in Developmental and Stem Cell Biology, University of California, San Francisco, San Francisco, United States
| | - Ralitsa Petrova
- Eli & Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Anshul Rana
- Graduate Program in Biochemistry, Stanford University School of Medicine, Stanford, United States
| | - Thomas Portmann
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Sergiu P Paşca
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Theo D Palmer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, United States
| | - Ricardo E Dolmetsch
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
12
|
Giffen KP, Liu H, Kramer KL, He DZ. Expression of Protein-Coding Gene Orthologs in Zebrafish and Mouse Inner Ear Non-sensory Supporting Cells. Front Neurosci 2019; 13:1117. [PMID: 31680844 PMCID: PMC6813431 DOI: 10.3389/fnins.2019.01117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/03/2019] [Indexed: 11/13/2022] Open
Abstract
Non-mammalian vertebrates, including zebrafish, retain the ability to regenerate hair cells (HCs) due to unknown molecular mechanisms that regulate proliferation and conversion of non-sensory supporting cells (nsSCs) to HCs. This regenerative capacity is not conserved in mammals. Identification of uniquely expressed orthologous genes in zebrafish nsSCs may reveal gene candidates involved in the proliferation and transdifferentiation of zebrafish nsSCs to HCs in the inner ear. A list of orthologous protein-coding genes was generated based on an Ensembl Biomart comparison of the zebrafish and mouse genomes. Our previously published RNA-seq-based transcriptome datasets of isolated inner ear zebrafish nsSCs and HCs, and mouse non-sensory supporting pillar and Deiters’ cells, and HCs, were merged to analyze gene expression patterns between the two species. Out of 17,498 total orthologs, 11,752 were expressed in zebrafish nsSCs and over 10,000 orthologs were expressed in mouse pillar and Deiters’ cells. Differentially expressed genes common among the zebrafish nsSCs and mouse pillar and Deiters’ cells, compared to species-specific HCs, included 306 downregulated and 314 upregulated genes; however, over 1,500 genes were uniquely upregulated in zebrafish nsSCs. Functional analysis of genes uniquely expressed in nsSCs identified several transcription factors associated with cell fate determination, cell differentiation and nervous system development, indicating inherent molecular properties of nsSCs that promote self-renewal and transdifferentiation into new HCs. Our study provides a means of characterizing these orthologous genes, involved in proliferation and transdifferentiation of nsSCs to HCs in zebrafish, which may lead to identification of potential targets for HC regeneration in mammals.
Collapse
Affiliation(s)
- Kimberlee P Giffen
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Kenneth L Kramer
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - David Z He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
13
|
Zhang LJ. Type1 Interferons Potential Initiating Factors Linking Skin Wounds With Psoriasis Pathogenesis. Front Immunol 2019; 10:1440. [PMID: 31293591 PMCID: PMC6603083 DOI: 10.3389/fimmu.2019.01440] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/07/2019] [Indexed: 12/18/2022] Open
Abstract
Psoriasis is a chronic autoimmune skin disease that can often be triggered upon skin injury, known as Koebner phenomenon. Type 1 interferons (IFNα and IFNβ), key cytokines that activate autoimmunity during viral infection, have been suggested to play an indispensable role in initiating psoriasis during skin injury. Type 1 IFN-inducible gene signature has been identified as one of the major upregulated gene signatures in psoriatic skin. Type 1 IFNs treatments often directly induce or exacerbate psoriasis, whereas blocking type 1 IFNs signaling pathway in animal models effectively inhibits the development of T cell-mediated skin inflammation and psoriasis-like inflammatory diseases. Epidermal keratinocytes (KCs) occupy the outermost position in the skin and are the first responder to skin injury. Skin injury rapidly induces IFNβ from KCs and IFNα from dermal plasmacytoid dendritic cells (pDCs) through distinct mechanisms. Host antimicrobial peptide LL37 potentiates double-stranded RNA (dsRNA) immune pathways in keratinocytes and single-stranded RNA or DNA pathways in pDCs, leading to production of distinct type 1 IFN genes. IFNβ from KC promotes dendritic cell maturation and the subsequent T cell proliferation, contributing to autoimmune activation during skin injury and psoriasis pathogenesis. Accumulating evidences have indicated an important role of this dsRNA immune pathway in psoriasis pathogenesis. Together, this review describes how skin injury induces type 1 IFNs from skin cells and how this may initiate autoimmune cascades that trigger psoriasis. Targeting keratinocytes or type 1 IFNs in combination with T cell therapy may result in more sustainable effect to treat auto-inflammatory skin diseases such as psoriasis.
Collapse
Affiliation(s)
- Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Department of Dermatology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
14
|
Kuntz S, Rudloff S, Kunz C. Milk Oligosaccharides From Different Cattle Breeds Influence Growth-Related Characteristics of Intestinal Cells. Front Nutr 2019; 6:31. [PMID: 30984764 PMCID: PMC6447651 DOI: 10.3389/fnut.2019.00031] [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: 11/24/2018] [Accepted: 03/04/2019] [Indexed: 01/02/2023] Open
Abstract
Oligosaccharides are present in human milk (HMO) in large amounts and in a high variety: Among other functions they are considered to influence the gut microbiota and gut maturation in infants. Due to the large volume of milk available bovine milk oligosaccharides (BMO) may be an alternative source of functional ingredients to potentially mimic HMO functions. Thus, we investigated direct effects of bovine milk oligosaccharides (BMO) from different cattle breeds on proliferation, differentiation and apoptosis in transformed (HT-29 and Caco-2) and non-transformed human intestinal cells (HIE cells). We observed a profound growth-inhibition effect induced by all BMO isolates in HT-29, Caco-2, and HIE cells in a dose-dependent manner. The effects varied not only between cell lines, i.e., HT-29 and Caco-2 cells were more sensitive than HIE cells, but also between the cattle breeds. Regarding the induction of differentiation, BMO induced differentiation only in HIE cells without affecting apoptosis. Cell cycle analysis via flow cytometry showed that growth inhibition was associated with a G2/M arrest in all cell lines. Expression levels detected by quantitative real-time RT-PCR revealed that this G2/M arrest was associated with changes in mRNA expression levels of cyclin A and B. Cyclin-dependent kinase inhibitors p21cip1 and p27kip1 and the tumor suppressor p53 were only enhanced in HIE cells necessary for arresting cells in the G2/M phase and induction of differentiation. In HT-29 and Caco-2 cells, a loss of p53 expression failed to induce G2/M associated induction of differentiation. The HIE cell specific differentiation induced by BMO was a result of influencing the phosphorylation states of EGFR (epidermal growth factor receptor) and MAP kinase, i.e., ERK1/2 (extracellular signal-regulated kinase 1/2), p38-α, and Akt2 phosphorylation. These results suggest that BMO inhibited intestinal cell proliferation and altered cell cycle dynamics by affecting corresponding regulator genes and mitogen-activated protein kinase signaling. As the development and maturation of digestive and absorptive processes depends on gut differentiation processes, our in vitro experiments show that breed-specific BMO are natural substances influencing various parameter which may be important in vivo in gastrointestinal development. This, however, needs to be proven in future studies.
Collapse
Affiliation(s)
- Sabine Kuntz
- Institute of Nutritional Sciences, Justus-Liebig-University Giessen, Giessen, Germany
| | - Silvia Rudloff
- Institute of Nutritional Sciences, Justus-Liebig-University Giessen, Giessen, Germany.,Department of Pediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Clemens Kunz
- Institute of Nutritional Sciences, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
15
|
Abstract
Epidermis, the outermost layer of the skin, plays a critical role as both a physical and immunological barrier protecting the internal tissues from external environmental insults, such as pathogenic bacteria, fungi, viruses, UV irradiation, and water loss. Epidermal keratinocytes (KC), the predominant cell type in the skin epidermis, are in the front line of skin defense. Here we describe methods to isolate and culture primary epidermal KC from neonatal and adult mouse skin and describe in vitro assays to study and characterize KC proliferation and differentiation and pro-inflammatory responses to viral products and UVB irradiation. These methods will be useful for researchers in the field of epidermal biology to set up in vitro assays to study the barrier and pro-inflammatory function of epidermal keratinocytes.
Collapse
Affiliation(s)
- Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
16
|
Wang P, Zhou Y, Yang JQ, Landeck L, Min M, Chen XB, Chen JQ, Li W, Cai SQ, Zheng M, Man XY. The role of Sprouty1 in the proliferation, differentiation and apoptosis of epidermal keratinocytes. Cell Prolif 2018; 51:e12477. [PMID: 30039569 DOI: 10.1111/cpr.12477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/03/2018] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Sprouty (SPRY) 1 is one of the SPRY proteins that inhibits signalling from various growth factors pathways and has also been known as a tumour suppressor in various malignancies. However, no study elucidates the role of SPRY1 in the skin. Our study was conducted to determine the function of SPRY1 in human keratinocytes and the epidermis. MATERIALS AND METHODS In vitro primary cultured epidermal keratinocytes were used to investigate the proliferation, differentiation and apoptosis of these cells. We also established overexpression of SPRY1 in vitro and K14-SPRY1 transgenic mice. RESULTS SPRY1 was mainly located in the cytoplasm of the epidermal keratinocytes from the granular epidermal layer of the skin and cultured cells. Overexpressed SPRY1 in keratinocytes resulted in up-regulation of P21, P27 and down-regulation of cyclin B1; decrease in MMP3 and integrin α6. SPRY1-overexpressed primary keratinocytes exhibited a lower proliferation and migration capability and higher rates of apoptosis. Epidermis of SPRY1-TG mice represented delayed wound healing. Proteomics analysis and GO enrichment showed DEPs of SPRY1 TG mice epidermis is significantly enriched in immune- and inflammatory-associated biological process. CONCLUSIONS In summary, SPRY1 expression was inversely correlated with cell proliferation, migration and promote cell apoptosis of keratinocytes. SPRY1 maybe a negative feedback regulator in normal human epidermal keratinocytes and cutaneous inflammatory responses. Our study raised the possibility that enhancing expression of SPRY1 may have the potential to promote anti-inflammatory effects.
Collapse
Affiliation(s)
- Ping Wang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Zhou
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Qiang Yang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lilla Landeck
- Ernst von Bergmann General Hospital, Teaching Hospital of the Charité-University Medicine Berlin, Humboldt University, Potsdam, Germany
| | - Min Min
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi-Bei Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Qi Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Wang CY, Sun YT, Fang KM, Ho CH, Yang CS, Tzeng SF. Function of B-Cell CLL/Lymphoma 11B in Glial Progenitor Proliferation and Oligodendrocyte Maturation. Front Mol Neurosci 2018; 11:4. [PMID: 29416501 PMCID: PMC5787563 DOI: 10.3389/fnmol.2018.00004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/03/2018] [Indexed: 01/04/2023] Open
Abstract
B-cell CLL/lymphoma 11B (Bcl11b) – a C2H2 zinc finger transcriptional factor – is known to regulate neuronal differentiation and function in the development of the central nervous system (CNS). Although its expression is reduced during oligodendrocyte (OLG) differentiation, its biological role in OLGs remains unknown. In this study, we found that the downregulation of Bcl11b gene expression in glial progenitor cells (GPCs) by lentivirus-mediated gene knockdown (KD) causes a reduction in cell proliferation with inhibited expression of stemness-related genes, while increasing the expression of cell cyclin regulator p21. In contrast, OLG specific transcription factors (Olig1) and OLG cell markers, including myelin proteolipid protein (PLP) and myelin oligodendrocyte glycoprotein (MOG), were upregulated in Bcl11b-KD GPCs. Chromatin immunoprecipitation (ChIP) analysis indicated that Bcl11b bound to the promoters of Olig1 and PLP, suggesting that Bcl11b could act as a repressor for Olig1 and PLP, similar to its action on p21. An increase in the number of GC+- or PLP+- OLGs derived from Bcl11b-KD GPCs or OLG precursor cells was also observed. Moreover, myelin basic protein (MBP) expression in OLGs derived from Bcl11b-KD GPCs was enhanced in hippocampal neuron co-cultures and in cerebellar brain-slice cultures. The in vivo study using a lysolecithin-induced demyelinating animal model also indicated that larger amounts of MBP+-OLGs and PLP+-OLGs derived from implanted Bcl11b-KD GPCs were present at the lesioned site of the white matter than in the scramble group. Taken together, our results provide insight into the functional role of Bcl11b in the negative regulation of GPC differentiation through the repression of OLG differentiation-associated genes.
Collapse
Affiliation(s)
- Chih-Yen Wang
- Institute of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Ting Sun
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Min Fang
- Institute of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Ho
- Institute of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Shi Yang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Shun-Fen Tzeng
- Institute of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
18
|
Transcription Factor CTIP1/ BCL11A Regulates Epidermal Differentiation and Lipid Metabolism During Skin Development. Sci Rep 2017; 7:13427. [PMID: 29044125 PMCID: PMC5647389 DOI: 10.1038/s41598-017-13347-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022] Open
Abstract
The epidermal permeability barrier (EPB) prevents organisms from dehydration and infection. The transcriptional regulation of EPB development is poorly understood. We demonstrate here that transcription factor COUP-TF-interacting protein 1 (CTIP1/BCL11A; hereafter CTIP1) is highly expressed in the developing murine epidermis. Germline deletion of Ctip1 (Ctip1−/−) results in EPB defects accompanied by compromised epidermal differentiation, drastic reduction in profilaggrin processing, reduced lamellar bodies in granular layers and significantly altered lipid composition. Transcriptional profiling of Ctip1−/− embryonic skin identified altered expression of genes encoding lipid-metabolism enzymes, skin barrier-associated transcription factors and junctional proteins. CTIP1 was observed to interact with genomic elements within the regulatory region of the gene encoding the differentiation-associated gene, Fos-related antigen2 (Fosl2) and lipid-metabolism-related gene, Fatty acid elongase 4 (Elvol4), and the expression of both was altered in Ctip1−/− mice. CTIP1 appears to play a role in EPB establishment of via direct or indirect regulation of a subset of genes encoding proteins involved in epidermal differentiation and lipid metabolism. These results identify potential, CTIP1-regulated avenues for treatment of skin disorders involving EBP defects.
Collapse
|
19
|
Zhang LJ, Sen GL, Ward NL, Johnston A, Chun K, Chen Y, Adase C, Sanford JA, Gao N, Chensee M, Sato E, Fritz Y, Baliwag J, Williams MR, Hata T, Gallo RL. Antimicrobial Peptide LL37 and MAVS Signaling Drive Interferon-β Production by Epidermal Keratinocytes during Skin Injury. Immunity 2017; 45:119-30. [PMID: 27438769 DOI: 10.1016/j.immuni.2016.06.021] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/30/2016] [Accepted: 05/02/2016] [Indexed: 02/02/2023]
Abstract
Type 1 interferons (IFNs) promote inflammation in the skin but the mechanisms responsible for inducing these cytokines are not well understood. We found that IFN-β was abundantly produced by epidermal keratinocytes (KCs) in psoriasis and during wound repair. KC IFN-β production depended on stimulation of mitochondrial antiviral-signaling protein (MAVS) by the antimicrobial peptide LL37 and double stranded-RNA released from necrotic cells. MAVS activated downstream TBK1 (TANK-Binding Kinase 1)-AKT (AKT serine/threonine kinase 1)-IRF3 (interferon regulatory factor 3) signaling cascade leading to IFN-β production and then promoted maturation of dendritic cells. In mice, the production of epidermal IFN-β by LL37 required MAVS, and human wounded and/or psoriatic skin showed activation of MAVS-associated IRF3 and induction of MAVS and IFN-β gene signatures. These findings show that KCs are an important source of IFN-β and MAVS is critical to this function, and demonstrates how the epidermis triggers unwanted skin inflammation under disease conditions.
Collapse
Affiliation(s)
- Ling-Juan Zhang
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, San Diego, La Jolla, CA 92093, USA
| | - Nicole L Ward
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andrew Johnston
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kimberly Chun
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yifang Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, San Diego, La Jolla, CA 92093, USA
| | - Christopher Adase
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - James A Sanford
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nina Gao
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Melanie Chensee
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emi Sato
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yi Fritz
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jaymie Baliwag
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael R Williams
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
20
|
Kulkarni NN, Adase CA, Zhang LJ, Borkowski AW, Li F, Sanford JA, Coleman DJ, Aguilera C, Indra AK, Gallo RL. IL-1 Receptor-Knockout Mice Develop Epidermal Cysts and Show an Altered Innate Immune Response after Exposure to UVB Radiation. J Invest Dermatol 2017; 137:2417-2426. [PMID: 28754339 DOI: 10.1016/j.jid.2017.07.814] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022]
Abstract
In this study, we observed that mice lacking the IL-1 receptor (IL-1R) (IL1r-/-) or deficient in IL1-β developed multiple epidermal cysts after chronic UVB exposure. Cysts that developed in IL1r-/- mice were characterized by the presence of the hair follicle marker Sox 9, keratins 10 and 14, and normal melanocyte distribution and retinoid X receptor-α expression. The increased incidence of cysts in IL1r-/- mice was associated with less skin inflammation as characterized by decreased recruitment of macrophages, and their skin also maintained epidermal barrier function compared with wild-type mice. Transcriptional analysis of the skin of IL1r-/- mice after UVB exposure showed decreased gene expression of proinflammatory cytokines such as tumor necrosis factor-α and IL-6. In vitro, primary keratinocytes derived from IL1r-/- mice were more resistant to UVB-triggered cell death compared with wild-type cells, and tumor necrosis factor-α release was completely blocked in the absence of IL-1R. These observations illustrate an unexpected yet prominent phenotype associated with the lack of IL-1R signaling in mice and support further investigation into the role of IL-1 ligands in epidermal repair and innate immune response after damaging UVB exposure.
Collapse
Affiliation(s)
- Nikhil N Kulkarni
- Department of Dermatology, University of California, San Diego, California, USA
| | - Christopher A Adase
- Department of Dermatology, University of California, San Diego, California, USA
| | - Ling-Juan Zhang
- Department of Dermatology, University of California, San Diego, California, USA
| | - Andrew W Borkowski
- Department of Dermatology, University of California, San Diego, California, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, California, USA
| | - James A Sanford
- Department of Dermatology, University of California, San Diego, California, USA
| | - Daniel J Coleman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA; Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Carlos Aguilera
- Department of Dermatology, University of California, San Diego, California, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA; Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA; Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA; Linus Pauling Science Center, Oregon State University, Corvallis, Oregon, USA; Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Portland, Oregon, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, California, USA.
| |
Collapse
|
21
|
Li F, Adase CA, Zhang LJ. Isolation and Culture of Primary Mouse Keratinocytes from Neonatal and Adult Mouse Skin. J Vis Exp 2017. [PMID: 28745643 DOI: 10.3791/56027] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The keratinocyte (KC) is the predominant cell type in the epidermis, the outermost layer of the skin. Epidermal KCs play a critical role in providing skin defense by forming an intact skin barrier against environmental insults, such as UVB irradiation or pathogens, and also by initiating an inflammatory response upon those insults. Here we describe methods to isolate KCs from neonatal mouse skin and from adult mouse tail skin. We also describe culturing conditions using defined growth supplements (dGS) in comparison to chelexed fetal bovine serum (cFBS). Functionally, we show that both neonatal and adult KCs are highly responsive to high calcium-induced terminal differentiation, tight junction formation and stratification. Additionally, cultured adult KCs are susceptible to UVB-triggered cell death and can release large amounts of TNF upon UVB irradiation. Together, the methods described here will be useful to researchers for the setup of in vitro models to study epidermal biology in the neonatal mouse and/or the adult mouse.
Collapse
Affiliation(s)
- Fengwu Li
- Department of Dermatology, School of Medicine, UC San Diego
| | | | - Ling-Juan Zhang
- Department of Dermatology, School of Medicine, UC San Diego;
| |
Collapse
|
22
|
Richardson R, Mitchell K, Hammond NL, Mollo MR, Kouwenhoven EN, Wyatt ND, Donaldson IJ, Zeef L, Burgis T, Blance R, van Heeringen SJ, Stunnenberg HG, Zhou H, Missero C, Romano RA, Sinha S, Dixon MJ, Dixon J. p63 exerts spatio-temporal control of palatal epithelial cell fate to prevent cleft palate. PLoS Genet 2017; 13:e1006828. [PMID: 28604778 PMCID: PMC5484519 DOI: 10.1371/journal.pgen.1006828] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/26/2017] [Accepted: 05/17/2017] [Indexed: 12/01/2022] Open
Abstract
Cleft palate is a common congenital disorder that affects up to 1 in 2500 live births and results in considerable morbidity to affected individuals and their families. The aetiology of cleft palate is complex with both genetic and environmental factors implicated. Mutations in the transcription factor p63 are one of the major individual causes of cleft palate; however, the gene regulatory networks in which p63 functions remain only partially characterized. Our findings demonstrate that p63 functions as an essential regulatory molecule in the spatio-temporal control of palatal epithelial cell fate to ensure appropriate fusion of the palatal shelves. Initially, p63 induces periderm formation and controls its subsequent maintenance to prevent premature adhesion between adhesion-competent, intra-oral epithelia. Subsequently, TGFβ3-induced down-regulation of p63 in the medial edge epithelia of the palatal shelves is a pre-requisite for palatal fusion by facilitating periderm migration from, and reducing the proliferative potential of, the midline epithelial seam thereby preventing cleft palate. Cleft palate is a serious congenital condition which affects approximately 1 in every 2500 births. Cleft palate occurs when the palatal shelves fail to grow, adhere or fuse during development. Mutations in the gene encoding the transcription factor p63 result in cleft palate in humans and mice. However, the role of p63 and how it controls the network of genes to regulate palate development is not well understood.In this study, we demonstrate that p63 controls the spatio-temporal regulation of palatal epithelial cell fate to ensure appropriate palatal adhesion: p63 induces the formation of a flattened layer of epithelial (periderm) cells and controls its subsequent maintenance. We also demonstrate that TGFβ3-induced, down-regulation of p63 in the medial edge epithelial cells, through which the palatal shelves adhere and fuse, controls Jag2-induced periderm migration to the oral and nasal epithelial triangles. In addition, p63 plays a central role in maintaining the proliferative potential of the basal layer of the medial edge epithelia. Our study provides significant new insights into the mechanisms that regulate development of the palate by establishing the role of p63 in governing the fate of the midline epithelial cells.
Collapse
Affiliation(s)
- Rose Richardson
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Karen Mitchell
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Nigel L. Hammond
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Maria Rosaria Mollo
- CEINGE Biotecnologie Avanzate Scarl (Center for Genetic Engineering), Napoli, Italy
| | - Evelyn N. Kouwenhoven
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | - Niki D. Wyatt
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Ian J. Donaldson
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Leo Zeef
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Tim Burgis
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Rognvald Blance
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Simon J. van Heeringen
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | - Hendrik G. Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate Scarl (Center for Genetic Engineering), Napoli, Italy
- Department of Biology, University of Naples, Federico II, Napoli, Italy
| | - Rose Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michael J. Dixon
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
- * E-mail: (JD); (MD)
| | - Jill Dixon
- Faculty of Biology, Medicine & Health, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
- * E-mail: (JD); (MD)
| |
Collapse
|
23
|
Li Z, Chen G, Yang Y, Guo W, Tian W. Bcl11b regulates enamel matrix protein expression and dental epithelial cell differentiation during rat tooth development. Mol Med Rep 2016; 15:297-304. [PMID: 27959403 DOI: 10.3892/mmr.2016.6030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 09/27/2016] [Indexed: 11/05/2022] Open
Abstract
Amelogenesis, beginning with thickened epithelial aggregation and ending with highly mineralized enamel formation, is a process mediated by a complex signaling network that involves several molecules, including growth and transcription factors. During early tooth development, the transcription factor B‑cell CLL/lymphoma 11B (Bcl11b) participates in dental epithelial cell proliferation and differentiation. However, whether it affects the postnatal regulation of enamel matrix protein expression and ameloblast differentiation remains unclear. To clarify the role of Bcl11b in enamel development, the present study initially detected the protein expression levels of Bcl11b during tooth development using immunohistochemistry, from the embryonic lamina stage to the postnatal period, and demonstrated that Bcl11b is predominantly restricted to cervical loop epithelial cells at the cap and bell stages, whereas expression is reduced in ameloblasts. Notably, the expression pattern of Bcl11b during tooth development differed between rats and mice. Knockdown of Bcl11b by specific small interfering RNA attenuated the expression of enamel‑associated genes, including amelogenin, X‑linked (Amelx), ameloblastin (Ambn), enamelin (Enam), kallikrein related peptidase 4 (Klk4), matrix metallopeptidase 20 and Msh homeobox 2 (Msx2). Chromatin immunoprecipitation assay verified that Msx2 was a transcriptional target of Bcl11b. However, overexpression of Msx2 resulted in downregulation of enamel‑associated genes, including Ambn, Amelx, Enam and Klk4. The present study suggested that Bcl11b serves a potentially important role in the regulation of ameloblast differentiation and enamel matrix protein expression. In addition, a complex feedback regulatory network may exist between Bcl11b and Msx2.
Collapse
Affiliation(s)
- Ziyue Li
- National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Guoqing Chen
- National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yaling Yang
- National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weihua Guo
- National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weidong Tian
- National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
24
|
Nishiguchi Y, Ohmoto M, Koki J, Enomoto T, Kominami R, Matsumoto I, Hirota J. Bcl11b/Ctip2 is required for development of lingual papillae in mice. Dev Biol 2016; 416:98-110. [PMID: 27287879 DOI: 10.1016/j.ydbio.2016.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 11/26/2022]
Abstract
Molecular mechanisms underlying the development and morphogenesis of oral epithelia, comprising the gustatory and nongustatory epithelium, remain unclear. Here, we show that Bcl11b, a zinc finger transcription factor, plays an important role in the development of lingual papillae, especially filiform papillae. In both gustatory and nongustatory epithelium, Bcl11b was expressed in keratin 14-positive epithelial basal cells, which differentiate into keratinocytes and/or taste cells. Loss of Bcl11b function resulted in abnormal morphology of the gustatory papillae: flattened fungiform papillae, shorter trench wall in the foliate and circumvallate papillae, and ectopic invagination in more than half of circumvallate papillae. However, Bcl11b loss caused no effect on differentiation of taste receptor cells. In nongustatory epithelium, the impact of Bcl11b deficiency was much more striking, resulting in a smooth surface on the tongue tip and hypoplastic filiform papillae in the dorsal lingual epithelium. Immunohistochemical analyses revealed that a keratinocyte differentiation marker, Tchh expression was severely decreased in the Bcl11b(-/-) filiform papillae. In addition, expression of Pax9, required for morphogenesis of filiform papillae and its downstream target genes, hard keratins, almost disappeared in the tongue tip and was decreased in the dorsal tongue of Bcl11b(-/-) mice. Gene expression analyses demonstrated a delayed onset of expression of epithelial differentiation complex genes, which disturbed barrier formation in the mutant tongue. These results indicate that Bcl11b regulates the differentiation of keratinocytes in the tongue and identify Bcl11b as an essential factor for the lingual papilla morphogenesis.
Collapse
Affiliation(s)
- Yugo Nishiguchi
- Department of Bioengineering, Graduate School of Bioscience and Bioengineering, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Makoto Ohmoto
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| | - Jun Koki
- Center for Advanced Materials Analysis, Tokyo Institute of Technology, Meguro-ku, Tokyo 152-8550, Japan
| | - Takayuki Enomoto
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Ryo Kominami
- Department of Molecular Genetics, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8122, Japan
| | | | - Junji Hirota
- Department of Bioengineering, Graduate School of Bioscience and Bioengineering, Tokyo Institute of Technology, Yokohama 226-8501, Japan; Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama 226-8501, Japan.
| |
Collapse
|
25
|
Labott AT, Lopez-Pajares V. Epidermal differentiation gene regulatory networks controlled by MAF and MAFB. Cell Cycle 2016; 15:1405-9. [PMID: 27097296 DOI: 10.1080/15384101.2016.1172148] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Numerous regulatory factors in epidermal differentiation and their role in regulating different cell states have been identified in recent years. However, the genetic interactions between these regulators over the dynamic course of differentiation have not been studied. In this Extra-View article, we review recent work by Lopez-Pajares et al. that explores a new regulatory network in epidermal differentiation. They analyze the changing transcriptome throughout epidermal regeneration to identify 3 separate gene sets enriched in the progenitor, early and late differentiation states. Using expression module mapping, MAF along with MAFB, are identified as transcription factors essential for epidermal differentiation. Through double knock-down of MAF:MAFB using siRNA and CRISPR/Cas9-mediated knockout, epidermal differentiation was shown to be impaired both in-vitro and in-vivo, confirming MAF:MAFB's role to activate genes that drive differentiation. Lopez-Pajares and collaborators integrated 42 published regulator gene sets and the MAF:MAFB gene set into the dynamic differentiation gene expression landscape and found that lncRNAs TINCR and ANCR act as upstream regulators of MAF:MAFB. Furthermore, ChIP-seq analysis of MAF:MAFB identified key transcription factor genes linked to epidermal differentiation as downstream effectors. Combined, these findings illustrate a dynamically regulated network with MAF:MAFB as a crucial link for progenitor gene repression and differentiation gene activation.
Collapse
Affiliation(s)
- Andrew T Labott
- a Program in Epithelial Biology, Stanford University , Stanford , CA , USA
| | | |
Collapse
|
26
|
Adase CA, Borkowski AW, Zhang LJ, Williams MR, Sato E, Sanford JA, Gallo RL. Non-coding Double-stranded RNA and Antimicrobial Peptide LL-37 Induce Growth Factor Expression from Keratinocytes and Endothelial Cells. J Biol Chem 2016; 291:11635-46. [PMID: 27048655 DOI: 10.1074/jbc.m116.725317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Indexed: 11/06/2022] Open
Abstract
A critical function for skin is that when damaged it must simultaneously identify the nature of the injury, repair barrier function, and limit the intrusion of pathogenic organisms. These needs are carried out through the detection of damage-associated molecular patterns (DAMPs) and a response that includes secretion of cytokines, chemokines, growth factors, and antimicrobial peptides (AMPs). In this study, we analyzed how non-coding double-stranded RNA (dsRNAs) act as a DAMP in the skin and how the human cathelicidin AMP LL-37 might influence growth factor production in response to this DAMP. dsRNA alone significantly increased the expression of multiple growth factors in keratinocytes, endothelial cells, and fibroblasts. Furthermore, RNA sequencing transcriptome analysis found that multiple growth factors increase when cells are exposed to both LL-37 and dsRNA, a condition that mimics normal wounding. Quantitative PCR and/or ELISA validated that growth factors expressed by keratinocytes in these conditions included, but were not limited to, basic fibroblast growth factor (FGF2), heparin-binding EGF-like growth factor (HBEGF), vascular endothelial growth factor C (VEGFC), betacellulin (BTC), EGF, epiregulin (EREG), and other members of the transforming growth factor β superfamily. These results identify a novel role for DAMPs and AMPs in the stimulation of repair and highlight the complex interactions involved in the wound environment.
Collapse
Affiliation(s)
- Christopher A Adase
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - Andrew W Borkowski
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - Ling-Juan Zhang
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - Michael R Williams
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - Emi Sato
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - James A Sanford
- From the Department of Dermatology, University of California, San Diego, California 92093
| | - Richard L Gallo
- From the Department of Dermatology, University of California, San Diego, California 92093
| |
Collapse
|
27
|
Transcription Factor CTIP2 Maintains Hair Follicle Stem Cell Pool and Contributes to Altered Expression of LHX2 and NFATC1. J Invest Dermatol 2015; 135:2593-2602. [PMID: 26176759 PMCID: PMC4640969 DOI: 10.1038/jid.2015.281] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/11/2015] [Accepted: 06/17/2015] [Indexed: 01/11/2023]
Abstract
Transcription factor CTIP2 (COUP-TF-interacting protein 2), also known as BCL11B, is expressed in hair follicles of embryonic and adult skin. Ctip2-null mice exhibit reduced hair follicle density during embryonic development. In contrast, conditional inactivation of Ctip2 in epidermis (Ctip2ep−/− mice) leads to a shorter telogen and premature entry into anagen during the second phase of hair cycling without a detectable change in the number of hair follicles. Keratinocytes of the bulge stem cells niche of Ctip2ep−/− mice proliferate more and undergo reduced apoptosis than the corresponding cells of wild-type mice. However, premature activation of follicular stem cells in mice lacking CTIP2 leads to the exhaustion of this stem cell compartment in comparison to Ctip2L2/L2 mice, which retained quiescent follicle stem cells. CTIP2 modulates expression of genes encoding EGFR and NOTCH1 during formation of hair follicles, and those encoding NFATC1 and LHX2 during normal hair cycling in adult skin. The expression of most of these genes is disrupted in mice lacking CTIP2 and these alterations may underlie the phenotype of Ctip2-null and Ctip2ep−/− mice. CTIP2 appears to serve as a transcriptional organizer that integrates input from multiple signaling cues during hair follicle morphogenesis and hair cycling.
Collapse
|
28
|
Toufighi K, Yang JS, Luis NM, Aznar Benitah S, Lehner B, Serrano L, Kiel C. Dissecting the calcium-induced differentiation of human primary keratinocytes stem cells by integrative and structural network analyses. PLoS Comput Biol 2015; 11:e1004256. [PMID: 25946651 PMCID: PMC4422705 DOI: 10.1371/journal.pcbi.1004256] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/25/2015] [Indexed: 12/19/2022] Open
Abstract
The molecular details underlying the time-dependent assembly of protein complexes in cellular networks, such as those that occur during differentiation, are largely unexplored. Focusing on the calcium-induced differentiation of primary human keratinocytes as a model system for a major cellular reorganization process, we look at the expression of genes whose products are involved in manually-annotated protein complexes. Clustering analyses revealed only moderate co-expression of functionally related proteins during differentiation. However, when we looked at protein complexes, we found that the majority (55%) are composed of non-dynamic and dynamic gene products ('di-chromatic'), 19% are non-dynamic, and 26% only dynamic. Considering three-dimensional protein structures to predict steric interactions, we found that proteins encoded by dynamic genes frequently interact with a common non-dynamic protein in a mutually exclusive fashion. This suggests that during differentiation, complex assemblies may also change through variation in the abundance of proteins that compete for binding to common proteins as found in some cases for paralogous proteins. Considering the example of the TNF-α/NFκB signaling complex, we suggest that the same core complex can guide signals into diverse context-specific outputs by addition of time specific expressed subunits, while keeping other cellular functions constant. Thus, our analysis provides evidence that complex assembly with stable core components and competition could contribute to cell differentiation.
Collapse
Affiliation(s)
- Kiana Toufighi
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jae-Seong Yang
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Nuno Miguel Luis
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Salvador Aznar Benitah
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Institute for Research in Biomedicine, Parc Científic de Barcelona, Barcelona, Spain
- * E-mail: (SAB); (BL); (LS); (CK)
| | - Ben Lehner
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- * E-mail: (SAB); (BL); (LS); (CK)
| | - Luis Serrano
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- * E-mail: (SAB); (BL); (LS); (CK)
| | - Christina Kiel
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- * E-mail: (SAB); (BL); (LS); (CK)
| |
Collapse
|
29
|
Zhang LJ, Guerrero-Juarez CF, Hata T, Bapat SP, Ramos R, Plikus MV, Gallo RL. Innate immunity. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science 2015; 347:67-71. [PMID: 25554785 PMCID: PMC4318537 DOI: 10.1126/science.1260972] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipocytes have been suggested to be immunologically active, but their role in host defense is unclear. We observed rapid proliferation of preadipocytes and expansion of the dermal fat layer after infection of the skin by Staphylococcus aureus. Impaired adipogenesis resulted in increased infection as seen in Zfp423(nur12) mice or in mice given inhibitors of peroxisome proliferator-activated receptor γ. This host defense function was mediated through the production of cathelicidin antimicrobial peptide from adipocytes because cathelicidin expression was decreased by inhibition of adipogenesis, and adipocytes from Camp(-/-) mice lost the capacity to inhibit bacterial growth. Together, these findings show that the production of an antimicrobial peptide by adipocytes is an important element for protection against S. aureus infection of the skin.
Collapse
Affiliation(s)
- Ling-juan Zhang
- Division of Dermatology, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA. Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Tissa Hata
- Division of Dermatology, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Sagar P Bapat
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, San Diego, La Jolla, CA 92037, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA. Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA. Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard L Gallo
- Division of Dermatology, University of California, San Diego (UCSD), La Jolla, CA 92093, USA.
| |
Collapse
|
30
|
BCL11B expression in intramembranous osteogenesis during murine craniofacial suture development. Gene Expr Patterns 2014; 17:16-25. [PMID: 25511173 DOI: 10.1016/j.gep.2014.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 11/21/2022]
Abstract
Sutures, where neighboring craniofacial bones are separated by undifferentiated mesenchyme, are major growth sites during craniofacial development. Pathologic fusion of bones within sutures occurs in a wide variety of craniosynostosis conditions and can result in dysmorphic craniofacial growth and secondary neurologic deficits. Our knowledge of the genes involved in suture formation is poor. Here we describe the novel expression pattern of the BCL11B transcription factor protein during murine embryonic craniofacial bone formation. We examined BCL11B protein expression at E14.5, E16.5, and E18.5 in 14 major craniofacial sutures of C57BL/6J mice. We found BCL11B expression to be associated with all intramembranous craniofacial bones examined. The most striking aspects of BCL11B expression were its high levels in suture mesenchyme and increasingly complementary expression with RUNX2 in differentiating osteoblasts during development. BCL11B was also expressed in mesenchyme at the non-sutural edges of intramembranous bones. No expression was seen in osteoblasts involved in endochondral ossification of the cartilaginous cranial base. BCL11B is expressed to potentially regulate the transition of mesenchymal differentiation and suture formation within craniofacial intramembranous bone.
Collapse
|
31
|
Enkhtuya R, Sato T, Wakasugi M, Tuvshintugs B, Miyata H, Sakurai T, Matsunaga T, Yoshioka K. The scaffold protein JLP plays a key role in regulating ultraviolet B-induced apoptosis in mice. Genes Cells 2014; 19:350-8. [PMID: 24520900 DOI: 10.1111/gtc.12135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/23/2013] [Indexed: 11/27/2022]
Abstract
The ultraviolet B (UVB) component of sunlight can cause severe damage to skin cells and even induce skin cancer. Growing evidence indicates that the UVB-induced signaling network is complex and involves diverse cellular processes. In this study, we investigated the role of c-Jun NH2 -terminal kinase-associated leucine zipper protein (JLP), a scaffold protein for mitogen-activated protein kinase (MAPK) signaling cascades, in UVB-induced apoptosis. We found that UVB-induced skin epidermal apoptosis was prevented in Jlp knockout (KO) as well as in keratinocyte-specific Jlp KO mice. Analysis of the repair of UVB-induced DNA damage over time showed no evidence for the involvement of JLP in this process. In contrast, UVB-stimulated p38 MAPK activation in the skin was impaired in both Jlp KO and keratinocyte-specific Jlp KO mice. Moreover, topical treatment of UVB-irradiated mouse skin with a p38 inhibitor significantly suppressed the epidermal apoptosis in wild-type mice, but not in Jlp KO mice. Our findings suggest that JLP in skin basal keratinocytes plays an important role in UVB-induced apoptosis by modulating p38 MAPK signaling pathways. This is the first study to show a critical role for JLP in an in vivo response to environmental stimulation.
Collapse
Affiliation(s)
- Radnaa Enkhtuya
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kiel C, Ebhardt HA, Burnier J, Portugal C, Sabidó E, Zimmermann T, Aebersold R, Serrano L. Quantification of ErbB network proteins in three cell types using complementary approaches identifies cell-general and cell-type-specific signaling proteins. J Proteome Res 2013; 13:300-13. [PMID: 24313378 DOI: 10.1021/pr400878x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Relating protein concentration to cell-type-specific responses is one of the remaining challenges for obtaining a quantitative systems level understanding of mammalian signaling. Here we used mass-spectrometry (MS)- and antibody-based quantitative proteomic approaches to measure protein abundances for 75% of a hand-curated reconstructed ErbB network of 198 proteins, in two established cell types (HEK293 and MCF-7) and in primary keratinocyte cells. Comparison with other quantitative studies allowed building a set of ErbB network proteins expressed in all cells and another which are cell-specific and could impart specific properties to the network. As a proof-of-concept of the importance of protein concentration, we generated a small simplified mathematical model encompassing ligand binding, followed by receptor dimerization, activation, and degradation. The model predicts ErbB phosphorylation in HEK293, MCF-7, and keratinocyte cells simply by incorporating cell-type-specific ErbB1, ErbB2, and caveolin-1 abundances but otherwise contains similar rate constants. Altogether, the data provide a resource for protein abundances and localization to be included in larger mathematical models, enabling the generation of cell-type-specific computational models. MS data have been deposited to the ProteomeXchange via PRIDE (with identifier PXD000623) and PASSEL (with identifier PASS00372).
Collapse
Affiliation(s)
- Christina Kiel
- EMBL/CRG Systems Biology Research Unit, ‡Proteomics Unit, §Advanced Light Microscopy Core Facility, Centre for Genomic Regulation (CRG) , Dr. Aiguader 88, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Wang Z, Zhang LJ, Guha G, Li S, Kyrylkova K, Kioussi C, Leid M, Ganguli-Indra G, Indra AK. Selective ablation of Ctip2/Bcl11b in epidermal keratinocytes triggers atopic dermatitis-like skin inflammatory responses in adult mice. PLoS One 2012; 7:e51262. [PMID: 23284675 PMCID: PMC3527437 DOI: 10.1371/journal.pone.0051262] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 10/31/2012] [Indexed: 11/18/2022] Open
Abstract
Background Ctip2 is crucial for epidermal homeostasis and protective barrier formation in developing mouse embryos. Selective ablation of Ctip2 in epidermis leads to increased transepidermal water loss (TEWL), impaired epidermal proliferation, terminal differentiation, as well as altered lipid composition during development. However, little is known about the role of Ctip2 in skin homeostasis in adult mice. Methodology/Principal Findings To study the role of Ctip2 in adult skin homeostasis, we utilized Ctip2ep−/− mouse model in which Ctip2 is selectively deleted in epidermal keratinocytes. Measurement of TEWL, followed by histological, immunohistochemical, and RT-qPCR analyses revealed an important role of Ctip2 in barrier maintenance and in regulating adult skin homeostasis. We demonstrated that keratinocytic ablation of Ctip2 leads to atopic dermatitis (AD)-like skin inflammation, characterized by alopecia, pruritus and scaling, as well as extensive infiltration of immune cells including T lymphocytes, mast cells, and eosinophils. We observed increased expression of T-helper 2 (Th2)-type cytokines and chemokines in the mutant skin, as well as systemic immune responses that share similarity with human AD patients. Furthermore, we discovered that thymic stromal lymphopoietin (TSLP) expression was significantly upregulated in the mutant epidermis as early as postnatal day 1 and ChIP assay revealed that TSLP is likely a direct transcriptional target of Ctip2 in epidermal keratinocytes. Conclusions/Significance Our data demonstrated a cell-autonomous role of Ctip2 in barrier maintenance and epidermal homeostasis in adult mice skin. We discovered a crucial non-cell autonomous role of keratinocytic Ctip2 in suppressing skin inflammatory responses by regulating the expression of Th2-type cytokines. It is likely that the epidermal hyperproliferation in the Ctip2-lacking epidermis may be secondary to the compensatory response of the adult epidermis that is defective in barrier functions. Our results establish an initiating role of epidermal TSLP in AD pathogenesis via a novel repressive regulatory mechanism enforced by Ctip2.
Collapse
Affiliation(s)
- Zhixing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Ling-juan Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Gunjan Guha
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Shan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Kateryna Kyrylkova
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
- Molecular Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Mark Leid
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
- Molecular Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Science Center, Oregon State University, Corvallis, Oregon, United States of America
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
- Molecular Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
- Molecular Cell Biology Program, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Science Center, Oregon State University, Corvallis, Oregon, United States of America
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|