1
|
Lei D, Liu D, Zhang J, Zhang L, Man MQ. Benefits of topical natural ingredients in epidermal permeability barrier. Front Physiol 2024; 14:1275506. [PMID: 38239888 PMCID: PMC10794395 DOI: 10.3389/fphys.2023.1275506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
Because of the crucial role of epidermal permeability barrier in regulation of cutaneous and extracutaneous functions, great efforts have been made to identify and develop the regimens that can improve epidermal permeability barrier function. Studies have demonstrated that oral administration of natural ingredients can improve epidermal permeability barrier in various skin conditions, including inflammatory dermatoses and UV-irradiation. Moreover, topical applications of some natural ingredients can also accelerate the repair of epidermal permeability barrier after acute barrier disruption and lower transepidermal water loss in the intact skin. Natural ingredient-induced improvements in epidermal permeability barrier function can be attributable to upregulation of keratinocyte differentiation, lipid production, antioxidant, hyaluronic acid production, expression of aquaporin 3 and sodium-hydrogen exchanger 1. In this review, we summarize the benefits of topical natural ingredients in epidermal permeability barrier in normal skin with or without acute barrier disruption and the underlying mechanisms.
Collapse
Affiliation(s)
- Dongyun Lei
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Dan Liu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Junling Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Mao-Qiang Man
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Dermatology Service, Veterans Affairs Medical Center San Francisco, Department of Dermatology, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
2
|
Sotiropoulou G, Zingkou E, Pampalakis G. Reconstructing the epidermal proteolytic cascades in health and disease. J Pathol 2022; 257:545-560. [PMID: 35218558 DOI: 10.1002/path.5888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 11/08/2022]
Abstract
The epidermis is the outer stratified epithelium of the skin, forming the physical barrier that is indispensable for homeostasis. Epidermal proteolysis, mainly but not exclusively executed by kallikrein-related peptidases (KLKs), is tightly regulated to ensure maintenance of physiological skin renewal and an intact skin barrier. Perturbation of epidermal proteolytic networks is implicated in a wide array of rare and common skin pathologies of diverse genetic backgrounds. Recent studies of monogenic human skin diseases and newly developed animal models have revealed new mechanisms of regulation of proteolytic pathways in epidermal physiology and in disease states. These new data have challenged some accepted views, for example the role of matriptase in epidermal desquamation, which turned out to be restricted to mouse skin. The significance of PAR2 signaling in skin inflammation should also be reconsidered in the face of recent findings. Cumulatively, recent studies necessitate a sophisticated redefinition of the proteolytic and signaling pathways that operate in human skin. We elaborate how epidermal proteolysis is finely regulated at multiple levels, and in a spatial manner that was not taken into consideration so far, in which specific proteases are confined to distinct epidermal sublayers. Of interest, transglutaminases have emerged as regulators of epidermal proteolysis and desquamation by spatially fixing endogenous protease inhibitors, constituting regulatory factors that were not recognized before. Furthermore, new evidence suggests a link between proteolysis and lipid metabolism. By synthesis of established notions and recent discoveries, we provide an up-to-date critical parathesis of current knowledge and the extended complexity of proteolysis regulation and signaling pathways in skin. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, 265 04, Greece
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, 265 04, Greece
| | - Georgios Pampalakis
- Department of Pharmacology-Pharmacognosy, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 541 24, Greece
| |
Collapse
|
3
|
Molday RS, Garces FA, Scortecci JF, Molday LL. Structure and function of ABCA4 and its role in the visual cycle and Stargardt macular degeneration. Prog Retin Eye Res 2021; 89:101036. [PMID: 34954332 DOI: 10.1016/j.preteyeres.2021.101036] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022]
Abstract
ABCA4 is a member of the superfamily of ATP-binding cassette (ABC) transporters that is preferentially localized along the rim region of rod and cone photoreceptor outer segment disc membranes. It uses the energy from ATP binding and hydrolysis to transport N-retinylidene-phosphatidylethanolamine (N-Ret-PE), the Schiff base adduct of retinal and phosphatidylethanolamine, from the lumen to the cytoplasmic leaflet of disc membranes. This ensures that all-trans-retinal and excess 11-cis-retinal are efficiently cleared from photoreceptor cells thereby preventing the accumulation of toxic retinoid compounds. Loss-of-function mutations in the gene encoding ABCA4 cause autosomal recessive Stargardt macular degeneration, also known as Stargardt disease (STGD1), and related autosomal recessive retinopathies characterized by impaired central vision and an accumulation of lipofuscin and bis-retinoid compounds. High resolution structures of ABCA4 in its substrate and nucleotide free state and containing bound N-Ret-PE or ATP have been determined by cryo-electron microscopy providing insight into the molecular architecture of ABCA4 and mechanisms underlying substrate recognition and conformational changes induced by ATP binding. The expression and functional characterization of a large number of disease-causing missense ABCA4 variants have been determined. These studies have shed light into the molecular mechanisms underlying Stargardt disease and a classification that reliably predicts the effect of a specific missense mutation on the severity of the disease. They also provide a framework for developing rational therapeutic treatments for ABCA4-associated diseases.
Collapse
Affiliation(s)
- Robert S Molday
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, B.C., Canada; Department of Ophthalmology & Visual Sciences, University of British Columbia, Vancouver, B.C., Canada.
| | - Fabian A Garces
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, B.C., Canada
| | | | - Laurie L Molday
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, B.C., Canada
| |
Collapse
|
4
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021. [DOI: https://doi.org/10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
5
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021; 192:45-54. [PMID: 34619274 DOI: 10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/22/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
Reprogrammed cell metabolism is a well-accepted hallmark of cancer. Metabolism changes provide energy and precursors for macromolecule biosynthesis to satisfy the survival needs of cancer cells. The specific changes in different aspects of lipid metabolism in cancer cells have been focused in recent years. These changes can affect cell growth, proliferation, differentiation and motility through affecting membranes synthesis, energy homeostasis and cell signaling. The tumor suppressor p53 plays vital roles in the control of cell proliferation, senescence, DNA repair, and cell death in cancer through various transcriptional and non-transcriptional activities. Accumulating evidences indicate that p53 also regulates cellular metabolism, which appears to contribute to its tumor suppressive functions. Particularly the role of p53 in regulating lipid metabolism has gained more and more attention in recent decades. In this review, we summarize recent advances in the function of p53 on lipid metabolism in cancer. Further understanding and research on the role of p53 in lipid metabolism regulation will provide a potential therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Ling-Li Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Wen-Jun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China.
| |
Collapse
|
6
|
The Balance between Differentiation and Terminal Differentiation Maintains Oral Epithelial Homeostasis. Cancers (Basel) 2021; 13:cancers13205123. [PMID: 34680271 PMCID: PMC8534139 DOI: 10.3390/cancers13205123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Oral cancer affecting the oral cavity represents the most common cancer of the head and neck region. Oral cancer develops in a multistep process in which normal cells gradually accumulate genetic and epigenetic modifications to evolve into a malignant disease. Mortality for oral cancer patients is high and morbidity has a significant long-term impact on the health and wellbeing of affected individuals, typically resulting in facial disfigurement and a loss of the ability to speak, chew, taste, and swallow. The limited scope to which current treatments are able to control oral cancer underlines the need for novel therapeutic strategies. This review highlights the molecular differences between oral cell proliferation, differentiation and terminal differentiation, defines terminal differentiation as an important tumour suppressive mechanism and establishes a rationale for clinical investigation of differentiation-paired therapies that may improve outcomes in oral cancer. Abstract The oral epithelium is one of the fastest repairing and continuously renewing tissues. Stem cell activation within the basal layer of the oral epithelium fuels the rapid proliferation of multipotent progenitors. Stem cells first undergo asymmetric cell division that requires tightly controlled and orchestrated differentiation networks to maintain the pool of stem cells while producing progenitors fated for differentiation. Rapidly expanding progenitors subsequently commit to advanced differentiation programs towards terminal differentiation, a process that regulates the structural integrity and homeostasis of the oral epithelium. Therefore, the balance between differentiation and terminal differentiation of stem cells and their progeny ensures progenitors commitment to terminal differentiation and prevents epithelial transformation and oral squamous cell carcinoma (OSCC). A recent comprehensive molecular characterization of OSCC revealed that a disruption of terminal differentiation factors is indeed a common OSCC event and is superior to oncogenic activation. Here, we discuss the role of differentiation and terminal differentiation in maintaining oral epithelial homeostasis and define terminal differentiation as a critical tumour suppressive mechanism. We further highlight factors with crucial terminal differentiation functions and detail the underlying consequences of their loss. Switching on terminal differentiation in differentiated progenitors is likely to represent an extremely promising novel avenue that may improve therapeutic interventions against OSCC.
Collapse
|
7
|
Experimental Models for the Study of Hereditary Cornification Defects. Biomedicines 2021; 9:biomedicines9030238. [PMID: 33652877 PMCID: PMC7996736 DOI: 10.3390/biomedicines9030238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/23/2021] [Indexed: 11/16/2022] Open
Abstract
Ichthyoses comprise a broad spectrum of keratinization disorders due to hereditary defects of cornification. Until now, mutations in more than 50 genes, mostly coding for structural proteins involved in epidermal barrier formation, have been identified as causes for different types of these keratinization disorders. However, due to the high heterogeneity and difficulties in the establishment of valid experimental models, research in this field remains challenging and translation of novel findings to clinical practice is difficult. In this review, we provide an overview of existing models to study hereditary cornification defects with focus on ichthyoses and palmoplantar keratodermas.
Collapse
|
8
|
Enjalbert F, Dewan P, Caley MP, Jones EM, Morse MA, Kelsell DP, Enright AJ, O'Toole EA. 3D model of harlequin ichthyosis reveals inflammatory therapeutic targets. J Clin Invest 2021; 130:4798-4810. [PMID: 32544098 PMCID: PMC7456239 DOI: 10.1172/jci132987] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 06/10/2020] [Indexed: 02/04/2023] Open
Abstract
The biology of harlequin ichthyosis (HI), a devastating skin disorder caused by loss-of-function mutations in the gene ABCA12, is poorly understood, and to date, no satisfactory treatment has been developed. We sought to investigate pathomechanisms of HI that could lead to the identification of new treatments for improving patients' quality of life. In this study, RNA-Seq and functional assays were performed to define the effects of loss of ABCA12 using HI patient skin samples and an engineered CRISPR/Cas9 ABCA12 KO cell line. The HI living skin equivalent (3D model) recapitulated the HI skin phenotype. The cytokines IL-36α and IL-36γ were upregulated in HI skin, whereas the innate immune inhibitor IL-37 was strongly downregulated. We also identified STAT1 and its downstream target inducible nitric oxide synthase (NOS2) as being upregulated in the in vitro HI 3D model and HI patient skin samples. Inhibition of NOS2 using the inhibitor 1400W or the JAK inhibitor tofacitinib dramatically improved the in vitro HI phenotype by restoring the lipid barrier in the HI 3D model. Our study has identified dysregulated pathways in HI skin that are feasible therapeutic targets.
Collapse
Affiliation(s)
- Florence Enjalbert
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Priya Dewan
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Matthew P Caley
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Eleri M Jones
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mary A Morse
- Adaptive Immunity Research Unit, GlaxoSmithKline Medicine's Research Centre, Stevenage, United Kingdom
| | - David P Kelsell
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Anton J Enright
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Edel A O'Toole
- Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom.,Department of Dermatology, Royal London Hospital, Barts Health NHS Trust ERN-Skin, London, United Kingdom
| |
Collapse
|
9
|
Cottle DL, Ursino GM, Jones LK, Tham MS, Zylberberg AK, Smyth IM. Topical Aminosalicylic Acid Improves Keratinocyte Differentiation in an Inducible Mouse Model of Harlequin Ichthyosis. CELL REPORTS MEDICINE 2020; 1:100129. [PMID: 33294854 PMCID: PMC7691394 DOI: 10.1016/j.xcrm.2020.100129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/10/2020] [Accepted: 10/06/2020] [Indexed: 12/02/2022]
Abstract
Mutations in the lipid transport protein ABCA12 cause the life-threatening skin condition harlequin ichthyosis (HI), which is characterized by the loss of skin barrier function, inflammation, and dehydration. Inflammatory responses in HI increase disease severity by impairing keratinocyte differentiation, suggesting amelioration of this phenotype as a possible therapy for the condition. Existing treatments for HI are based around the use of retinoids, but their value in treating patients during the neonatal period has been questioned relative to other improved management regimens, and their long-term use is associated with side effects. We have developed a conditional mouse model to demonstrate that topical application of the aminosalicylic acid derivatives 5ASA or 4ASA considerably improves HI keratinocyte differentiation without the undesirable side effects of the retinoid acitretin and salicylic acid (aspirin). Analysis of changes in gene expression shows that 4ASA in particular elicits compensatory upregulation of a large family of barrier function-related genes, many of which are associated with other ichthyoses, identifying this compound as a lead candidate for developing topical treatments for HI. Inflammation impairs keratinocyte differentiation and worsens harlequin ichthyosis Harlequin ichthyosis mice can be used to assess therapies for this disease Aminosalicylic acids may be therapeutic treatments for harlequin ichthyosis 4ASA improves skin differentiation and barrier function in harlequin ichthyosis models
Collapse
Affiliation(s)
- Denny L. Cottle
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
| | - Gloria M.A. Ursino
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Lynelle K. Jones
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
| | - Ming Shen Tham
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
| | - Allara K. Zylberberg
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
| | - Ian M. Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program, Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
- Corresponding author
| |
Collapse
|
10
|
Teramura T, Nomura T. Acute skin barrier disruption alters the secretion of lamellar bodies via the multilayered expression of ABCA12. J Dermatol Sci 2020; 100:50-57. [PMID: 32873425 DOI: 10.1016/j.jdermsci.2020.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/29/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The skin barrier consists of multiple lipid-enriched layers, which are characterized by lamellar repeated structures within the intercellular space. Sodium lauryl sulfate is a well-known substance that can disrupt the skin barrier. The mechanisms underlying the barrier repair process, especially the influence of topical sodium lauryl sulfate treatment on lipid transport in the barrier recovery phase, remain unresolved. OBJECTIVE To understand the process of reconstruction of the intercellular lipid layer of the skin after acute barrier disruption by sodium lauryl sulfate treatment in vivo. METHODS Female hairless mice were treated with 3 % sodium lauryl sulfate. Transepidermal water loss measurement, histopathological analysis, and gene expression analysis were performed from 1 to 288 h after the topical application of sodium lauryl sulfate. Western blot analysis, immunofluorescence staining, and transmission electron microscopy analysis were performed to examine the expression level of ATP-binding cassette, sub-family A, member 12 (ABCA12), and the secretion level of lamellar bodies. RESULTS We observed rapid hyper-keratinization at the stratum corneum and the subsequent concurrent secretion of lamellar bodies into the intercellular space of the stratum corneum during the process of skin barrier recovery. ABCA12 expression associated with lipid transportation into lamellar bodies was transiently upregulated and observed in multiple layers in the upper epidermis, especially in the stratum granulosum. CONCLUSION The skin reacts appropriately to maintain its barrier function by first initiating hyper-keratinization and then increasing lamellar body secretion. Activation of ABCA12 is an essential factor for the recovery of skin barrier function.
Collapse
Affiliation(s)
- Takashi Teramura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshifumi Nomura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| |
Collapse
|
11
|
Wang X, Cao C, Li Y, Hai T, Jia Q, Zhang Y, Zheng Q, Yao J, Qin G, Zhang H, Song R, Wang Y, Shui G, Lam SM, Liu Z, Wei H, Meng A, Zhou Q, Zhao J. A harlequin ichthyosis pig model with a novel ABCA12 mutation can be rescued by acitretin treatment. J Mol Cell Biol 2020; 11:1029-1041. [PMID: 30925591 PMCID: PMC6934153 DOI: 10.1093/jmcb/mjz021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 11/20/2022] Open
Abstract
Harlequin ichthyosis (HI) is a severe genetic skin disorder and caused by mutation in the ATP-binding cassette A12 (ABCA12) gene. The retinoid administration has dramatically improved long-term survival of HI, but improvements are still needed. However, the ABCA12 null mice failed to respond to retinoid treatment, which impedes the development of novel cure strategies for HI. Here we generated an ethylnitrosourea mutagenic HI pig model (named Z9), which carries a novel deep intronic mutation IVS49-727 A>G in the ABCA12 gene, resulting in abnormal mRNA splicing and truncated protein production. Z9 pigs exhibit significant clinical symptom as human patients with HI. Most importantly, systemic retinoid treatment significantly prolonged the life span of the mutant pigs via improving epidermal maturation, decreasing epidermal apoptosis, and triggering the expression of ABCA6. Taken together, this pig model perfectly resembles the clinical symptom and molecular pathology of patients with HI and will be useful for understanding mechanistic insight and developing therapeutic strategies.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Yongshun Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Tang Hai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Qitao Jia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Qiantao Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Jing Yao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Guosong Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Hongyong Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Ruigao Song
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University of China, Harbin 150030, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Anming Meng
- School of Life Sciences, Tsinghua University, Beijing 100084, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Chinese Swine Mutagenesis Consortium, Beijing 100101, China
| |
Collapse
|
12
|
Eager KLM, Conyers LE, Woolley SA, Tammen I, O'Rourke BA. A novel ABCA12 frameshift mutation segregates with ichthyosis fetalis in a Polled Hereford calf. Anim Genet 2020; 51:837-838. [PMID: 32567073 DOI: 10.1111/age.12973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Katie L M Eager
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, Menangle, NSW, 2568, Australia
| | - Liberty E Conyers
- Faculty of Science, School of Life Sciences, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Shernae A Woolley
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, Menangle, NSW, 2568, Australia.,Faculty of Science, Sydney School of Veterinary Science, University of Sydney, Camden, NSW, 2570, Australia
| | - Imke Tammen
- Faculty of Science, Sydney School of Veterinary Science, University of Sydney, Camden, NSW, 2570, Australia
| | - Brendon A O'Rourke
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, Menangle, NSW, 2568, Australia
| |
Collapse
|
13
|
Abstract
The terminal differentiation of the epidermis is a complex physiological process. During the past few decades, medical genetics has shown that defects in the stratum corneum (SC) permeability barrier cause a myriad of pathological conditions, ranging from common dry skin to lethal ichthyoses. Contrarily, molecular phylogenetics has revealed that amniotes have acquired a specialized form of cytoprotection cornification that provides mechanical resilience to the SC. This superior biochemical property, along with desiccation tolerance, is attributable to the proper formation of the macromolecular protein-lipid complex termed cornified cell envelopes (CE). Cornification largely depends on the peculiar biochemical and biophysical properties of loricrin, which is a major CE component. Despite its quantitative significance, loricrin knockout (LKO) mice have revealed it to be dispensable for the SC permeability barrier. Nevertheless, LKO mice have brought us valuable lessons. It is also becoming evident that absent loricrin affects skin homeostasis more profoundly in many more aspects than previously expected. Through an extensive review of aggregate evidence, we discuss herein the functional significance of the thiol-rich protein loricrin from a biochemical, genetic, pathological, metabolic, or immunological aspect with some theoretical and speculative perspectives.
Collapse
Affiliation(s)
- Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Dennis R. Roop
- Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
14
|
Ursino GM, Fu Y, Cottle DL, Mukhamedova N, Jones LK, Low H, Tham MS, Gan WJ, Mellett NA, Das PP, Weir JM, Ditiatkovski M, Fynch S, Thorn P, Thomas HE, Meikle PJ, Parkington HC, Smyth IM, Sviridov D. ABCA12 regulates insulin secretion from β-cells. EMBO Rep 2020; 21:e48692. [PMID: 32072744 DOI: 10.15252/embr.201948692] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/12/2019] [Accepted: 01/08/2020] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of lipid homeostasis is intimately associated with defects in insulin secretion, a key feature of type 2 diabetes. Here, we explore the role of the putative lipid transporter ABCA12 in regulating insulin secretion from β-cells. Mice with β-cell-specific deletion of Abca12 display impaired glucose-stimulated insulin secretion and eventual islet inflammation and β-cell death. ABCA12's action in the pancreas is independent of changes in the abundance of two other cholesterol transporters, ABCA1 and ABCG1, or of changes in cellular cholesterol or ceramide content. Instead, loss of ABCA12 results in defects in the genesis and fusion of insulin secretory granules and increases in the abundance of lipid rafts at the cell membrane. These changes are associated with dysregulation of the small GTPase CDC42 and with decreased actin polymerisation. Our findings establish a new, pleiotropic role for ABCA12 in regulating pancreatic lipid homeostasis and insulin secretion.
Collapse
Affiliation(s)
- Gloria M Ursino
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Ying Fu
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | | | - Lynelle K Jones
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Hann Low
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Ming Shen Tham
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Wan Jun Gan
- Charles Perkins Centre, Camperdown, NSW, Australia
| | | | - Partha P Das
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | | | | | - Stacey Fynch
- St Vincent's Institute, Fitzroy, Vic., Australia
| | - Peter Thorn
- Charles Perkins Centre, Camperdown, NSW, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Helena C Parkington
- Department of Physiology, Neuroscience Discovery Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| |
Collapse
|
15
|
Woolley SA, Eager KLM, Häfliger IM, Bauer A, Drögemüller C, Leeb T, O'Rourke BA, Tammen I. An ABCA12 missense variant in a Shorthorn calf with ichthyosis fetalis. Anim Genet 2019; 50:749-752. [PMID: 31568573 DOI: 10.1111/age.12856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Two clinical forms of ichthyosis in cattle have been reported, ichthyosis fetalis and congenital ichthyosis. Ichthyosis poses animal welfare and economic issues and the more severe form, ichthyosis fetalis, is lethal. A Shorthorn calf with ichthyosis fetalis was investigated and a likely causal missense variant on chromosome 2 in the ABCA12 gene (NM_001191294.2:c.6776T>C) was identified by whole genome sequencing. Mutations in the ABCA12 gene are known to cause ichthyosis fetalis in cattle and Harlequin ichthyosis in humans. Sanger sequencing of the affected calf and the dam confirmed the variant was homozygous in the affected calf and heterozygous in the dam. Further genotyping of 130 Shorthorn animals from the same property revealed an estimated allele frequency of 3.8%. The presented findings enable genetic testing for breeding and diagnostics.
Collapse
Affiliation(s)
- S A Woolley
- Faculty of Science, Sydney School of Veterinary Science, University of Sydney, Camden, 2570, NSW, Australia
| | - K L M Eager
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, 2568, NSW, Australia
| | - I M Häfliger
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, 3001, Switzerland
| | - A Bauer
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, 3001, Switzerland
| | - C Drögemüller
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, 3001, Switzerland
| | - T Leeb
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, 3001, Switzerland
| | - B A O'Rourke
- NSW Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Menangle, 2568, NSW, Australia
| | - I Tammen
- Faculty of Science, Sydney School of Veterinary Science, University of Sydney, Camden, 2570, NSW, Australia
| |
Collapse
|
16
|
Trigueros-Motos L, van Capelleveen JC, Torta F, Castaño D, Zhang LH, Chai EC, Kang M, Dimova LG, Schimmel AW, Tietjen I, Radomski C, Tan LJ, Thiam CH, Narayanaswamy P, Wu DH, Dorninger F, Yakala GK, Barhdadi A, Angeli V, Dubé MP, Berger J, Dallinga-Thie GM, Tietge UJ, Wenk MR, Hayden MR, Hovingh GK, Singaraja RR. ABCA8 Regulates Cholesterol Efflux and High-Density Lipoprotein Cholesterol Levels. Arterioscler Thromb Vasc Biol 2017; 37:2147-2155. [DOI: 10.1161/atvbaha.117.309574] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/29/2017] [Indexed: 01/18/2023]
Abstract
Objective—
High-density lipoproteins (HDL) are considered to protect against atherosclerosis in part by facilitating the removal of cholesterol from peripheral tissues. However, factors regulating lipid efflux are incompletely understood. We previously identified a variant in adenosine triphosphate–binding cassette transporter A8 (
ABCA8
) in an individual with low HDL cholesterol (HDLc). Here, we investigate the role of ABCA8 in cholesterol efflux and in regulating HDLc levels.
Approach and Results—
We sequenced
ABCA8
in individuals with low and high HDLc and identified, exclusively in low HDLc probands, 3 predicted deleterious heterozygous
ABCA8
mutations (p.Pro609Arg [P609R], IVS17-2 A>G and p.Thr741Stop [T741X]). HDLc levels were lower in heterozygous mutation carriers compared with first-degree family controls (0.86±0.34 versus 1.17±0.26 mmol/L;
P
=0.005). HDLc levels were significantly decreased by 29% (
P
=0.01) in
Abca8b
−/−
mice on a high-cholesterol diet compared with wild-type mice, whereas hepatic overexpression of human
ABCA8
in mice resulted in significant increases in plasma HDLc and the first steps of macrophage-to-feces reverse cholesterol transport. Overexpression of wild-type but not mutant ABCA8 resulted in a significant increase (1.8-fold;
P
=0.01) of cholesterol efflux to apolipoprotein AI in vitro. ABCA8 colocalizes and interacts with adenosine triphosphate–binding cassette transporter A1 and further potentiates adenosine triphosphate–binding cassette transporter A1–mediated cholesterol efflux.
Conclusions—
ABCA8 facilitates cholesterol efflux and modulates HDLc levels in humans and mice.
Collapse
Affiliation(s)
- Laia Trigueros-Motos
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Julian C. van Capelleveen
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Federico Torta
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - David Castaño
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Lin-Hua Zhang
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Ee Chu Chai
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Martin Kang
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Lidiya G. Dimova
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Alinda W.M. Schimmel
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Ian Tietjen
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Chris Radomski
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Liang Juin Tan
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Chung Hwee Thiam
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Pradeep Narayanaswamy
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Daniel Heqing Wu
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Fabian Dorninger
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Gopala Krishna Yakala
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Amina Barhdadi
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Veronique Angeli
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Marie-Pierre Dubé
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Johannes Berger
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Geesje M. Dallinga-Thie
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Uwe J.F. Tietge
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Markus R. Wenk
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Michael R. Hayden
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - G. Kees Hovingh
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| | - Roshni R. Singaraja
- From the Translational Laboratory in Genetic Medicine, A*STAR Institute, and Yong Loo Lin School of Medicine, National University of Singapore (L.T.-M., D.C., E.C.C., L.J.T., D.H.W., G.K.Y., M.R.H., R.R.S.); Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, The Netherlands (J.C.v.C., A.W.M.S., G.M.D.-T., G.K.H.); Faculty of Health Sciences, Simon Fraser University, Canada (I.T.); Department of Biochemistry, Yong Loo Lin School of
| |
Collapse
|
17
|
Kulkarni H, Mamtani M, Wong G, Weir JM, Barlow CK, Dyer TD, Almasy L, Mahaney MC, Comuzzie AG, Duggirala R, Meikle PJ, Blangero J, Curran JE. Genetic correlation of the plasma lipidome with type 2 diabetes, prediabetes and insulin resistance in Mexican American families. BMC Genet 2017; 18:48. [PMID: 28525987 PMCID: PMC5438505 DOI: 10.1186/s12863-017-0515-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 05/11/2017] [Indexed: 01/15/2023] Open
Abstract
Background Differential plasma concentrations of circulating lipid species are associated with pathogenesis of type 2 diabetes (T2D). Whether the wide inter-individual variability in the plasma lipidome contributes to the genetic basis of T2D is unknown. Here, we investigated the potential overlap in the genetic basis of the plasma lipidome and T2D-related traits. Results We used plasma lipidomic data (1202 pedigreed individuals, 319 lipid species representing 23 lipid classes) from San Antonio Family Heart Study in Mexican Americans. Bivariate trait analyses were used to estimate the genetic and environmental correlation of all lipid species with three T2D-related traits: risk of T2D, presence of prediabetes and homeostatic model of assessment – insulin resistance. We found that 44 lipid species were significantly genetically correlated with one or more of the three T2D-related traits. Majority of these lipid species belonged to the diacylglycerol (DAG, 17 species) and triacylglycerol (TAG, 17 species) classes. Six lipid species (all belonging to the triacylglycerol class and containing palmitate at the first position) were significantly genetically correlated with all the T2D-related traits. Conclusions Our results imply that: a) not all plasma lipid species are genetically informative for T2D pathogenesis; b) the DAG and TAG lipid classes partially share genetic basis of T2D; and c) 1-palmitate containing TAGs may provide additional insights into the genetic basis of T2D. Electronic supplementary material The online version of this article (doi:10.1186/s12863-017-0515-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hemant Kulkarni
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA.
| | - Manju Mamtani
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Gerard Wong
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jacquelyn M Weir
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Thomas D Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Laura Almasy
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Michael C Mahaney
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ravindranath Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, 78520, USA
| |
Collapse
|
18
|
Ip SCI, Cottle DL, Jones LK, Weir JM, Kelsell DP, O'Toole EA, Meikle PJ, Smyth IM. A profile of lipid dysregulation in harlequin ichthyosis. Br J Dermatol 2017; 177:e217-e219. [PMID: 28493316 DOI: 10.1111/bjd.15642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- S C I Ip
- Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, 3800, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Vic, Australia
| | - D L Cottle
- Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, 3800, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Vic, Australia.,Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - L K Jones
- Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, 3800, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Vic, Australia.,Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - J M Weir
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - D P Kelsell
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, 3800, VIC, Australia
| | - E A O'Toole
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, 3800, VIC, Australia
| | - P J Meikle
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - I M Smyth
- Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, 3800, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, Vic, Australia.,Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Kulkarni H, Mamtani M, Blangero J, Curran JE. Lipidomics in the Study of Hypertension in Metabolic Syndrome. Curr Hypertens Rep 2017; 19:7. [DOI: 10.1007/s11906-017-0705-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Abstract
Copper (Cu) is indispensible for growth and development of human organisms. It is required for such fundamental and ubiquitous processes as respiration and protection against reactive oxygen species. Cu also enables catalytic activity of enzymes that critically contribute to the functional identity of many cells and tissues. Pigmentation, production of norepinephrine by the adrenal gland, the key steps in the formation of connective tissue, neuroendocrine signaling, wound healing - all these processes require Cu and depend on Cu entering the secretory pathway. To reach the Cu-dependent enzymes in a lumen of the trans-Golgi network and various vesicular compartments, Cu undertakes a complex journey crossing the extracellular and intracellular membranes and staying firmly on course while traveling in a cytosol. The proteins that assist Cu in this journey by mediating its entry, distribution, and export, have been identified. The accumulating data also indicate that the current model of cellular Cu homeostasis is still a "skeleton" that has to be fleshed out with many new details. This review summarizes recent data on the mechanisms responsible for Cu transfer to the secretory pathway. The emerging new concepts and gaps in our knowledge are discussed.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, 725 N. Wolfe street, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Defects in Stratum Corneum Desquamation Are the Predominant Effect of Impaired ABCA12 Function in a Novel Mouse Model of Harlequin Ichthyosis. PLoS One 2016; 11:e0161465. [PMID: 27551807 PMCID: PMC4994956 DOI: 10.1371/journal.pone.0161465] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/05/2016] [Indexed: 01/23/2023] Open
Abstract
Harlequin Ichthyosis is a severe skin disease caused by mutations in the human gene encoding ABCA12. Here, we characterize a novel mutation in intron 29 of the mouse Abca12 gene that leads to the loss of a 5' splice donor site and truncation of the Abca12 RNA transcript. Homozygous mutants of this smooth skin or smsk allele die perinatally with shiny translucent skin, typical of animal models of Harlequin Ichthyosis. Characterization of smsk mutant skin showed that the delivery of glucosylceramides and CORNEODESMOSIN was defective, while ultrastructural analysis revealed abnormal lamellar bodies and the absence of lipid lamellae in smsk epidermis. Unexpectedly, mutant stratum corneum remained intact when subjected to harsh chemical dissociation procedures. Moreover, both KALLIKREIN 5 and -7 were drastically decreased, with retention of desmoplakin in mutant SC. In cultured wild type keratinocytes, both KALLIKREIN 5 and -7 colocalized with ceramide metabolites following calcium-induced differentiation. Reducing the intracellular levels of glucosylceramide with a glucosylceramide synthase inhibitor resulted in decreased secretion of KALLIKREIN proteases by wild type keratinocytes, but not by smsk mutant keratinocytes. Together, these findings suggest an essential role for ABCA12 in transferring not only lipids, which are required for the formation of multilamellar structures in the stratum corneum, but also proteolytic enzymes that are required for normal desquamation. Smsk mutant mice recapitulate many of the pathological features of HI and can be used to explore novel topical therapies against a potentially lethal and debilitating neonatal disease.
Collapse
|
22
|
Paller AS, Renert-Yuval Y, Suprun M, Esaki H, Oliva M, Huynh TN, Ungar B, Kunjravia N, Friedland R, Peng X, Zheng X, Estrada YD, Krueger JG, Choate KA, Suárez-Fariñas M, Guttman-Yassky E. An IL-17-dominant immune profile is shared across the major orphan forms of ichthyosis. J Allergy Clin Immunol 2016; 139:152-165. [PMID: 27554821 DOI: 10.1016/j.jaci.2016.07.019] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/18/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND The ichthyoses are rare genetic disorders associated with generalized scaling, erythema, and epidermal barrier impairment. Pathogenesis-based therapy is largely lacking because the underlying molecular basis is poorly understood. OBJECTIVE We sought to characterize molecularly cutaneous inflammation and its correlation with clinical and barrier characteristics. METHODS We analyzed biopsy specimens from 21 genotyped patients with ichthyosis (congenital ichthyosiform erythroderma, n = 6; lamellar ichthyosis, n = 7; epidermolytic ichthyosis, n = 5; and Netherton syndrome, n = 3) using immunohistochemistry and RT-PCR and compared them with specimens from healthy control subjects, patients with atopic dermatitis (AD), and patients with psoriasis. Clinical measures included the Ichthyosis Area Severity Index (IASI), which integrates erythema (IASI-E) and scaling (IASI-S); transepidermal water loss; and pruritus. RESULTS Ichthyosis samples showed increased epidermal hyperplasia (increased thickness and keratin 16 expression) and T-cell and dendritic cell infiltrates. Increases of general inflammatory (IL-2), innate (IL-1β), and some TH1/interferon (IFN-γ) markers in patients with ichthyosis were comparable with those in patients with psoriasis or AD. TNF-α levels in patients with ichthyosis were increased only in those with Netherton syndrome but were much lower than in patients with psoriasis and those with AD. Expression of TH2 cytokines (IL-13 and IL-31) was similar to that seen in control subjects. The striking induction of IL-17-related genes or markers synergistically induced by IL-17 and TNF-α (IL-17A/C, IL-19, CXCL1, PI3, CCL20, and IL36G; P < .05) in patients with ichthyosis was similar to that seen in patients with psoriasis. IASI and IASI-E scores strongly correlated with IL-17A (r = 0.74, P < .001) and IL-17/TNF-synergistic/additive gene expression. These markers also significantly correlated with transepidermal water loss, suggesting a link between the barrier defect and inflammation in patients with ichthyosis. CONCLUSION Our data associate a shared TH17/IL-23 immune fingerprint with the major orphan forms of ichthyosis and raise the possibility of IL-17-targeting strategies.
Collapse
Affiliation(s)
- Amy S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| | - Yael Renert-Yuval
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Maria Suprun
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hitokazu Esaki
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Margeaux Oliva
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thy Nhat Huynh
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Benjamin Ungar
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Norma Kunjravia
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Rivka Friedland
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Xiangyu Peng
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiuzhong Zheng
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Yeriel D Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - James G Krueger
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Keith A Choate
- Department of Dermatology, Yale University School of Medicine, New Haven, Conn
| | - Mayte Suárez-Fariñas
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| |
Collapse
|
23
|
DiTommaso T, Jones LK, Cottle DL, Gerdin AK, Vancollie VE, Watt FM, Ramirez-Solis R, Bradley A, Steel KP, Sundberg JP, White JK, Smyth IM. Identification of genes important for cutaneous function revealed by a large scale reverse genetic screen in the mouse. PLoS Genet 2014; 10:e1004705. [PMID: 25340873 PMCID: PMC4207618 DOI: 10.1371/journal.pgen.1004705] [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: 02/14/2014] [Accepted: 08/26/2014] [Indexed: 12/15/2022] Open
Abstract
The skin is a highly regenerative organ which plays critical roles in protecting the body and sensing its environment. Consequently, morbidity and mortality associated with skin defects represent a significant health issue. To identify genes important in skin development and homeostasis, we have applied a high throughput, multi-parameter phenotype screen to the conditional targeted mutant mice generated by the Wellcome Trust Sanger Institute's Mouse Genetics Project (Sanger-MGP). A total of 562 different mouse lines were subjected to a variety of tests assessing cutaneous expression, macroscopic clinical disease, histological change, hair follicle cycling, and aberrant marker expression. Cutaneous lesions were associated with mutations in 23 different genes. Many of these were not previously associated with skin disease in the organ (Mysm1, Vangl1, Trpc4ap, Nom1, Sparc, Farp2, and Prkab1), while others were ascribed new cutaneous functions on the basis of the screening approach (Krt76, Lrig1, Myo5a, Nsun2, and Nf1). The integration of these skin specific screening protocols into the Sanger-MGP primary phenotyping pipelines marks the largest reported reverse genetic screen undertaken in any organ and defines approaches to maximise the productivity of future projects of this nature, while flagging genes for further characterisation.
Collapse
Affiliation(s)
- Tia DiTommaso
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia
| | - Lynelle K. Jones
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia
| | - Denny L. Cottle
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia
| | | | - Anna-Karin Gerdin
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Valerie E. Vancollie
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Fiona M. Watt
- Centre for Stem Cells and Regenerative Medicine King's College London, Guy's Hospital, London, United Kingdom
| | - Ramiro Ramirez-Solis
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Allan Bradley
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Karen P. Steel
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, United Kingdom
| | - John P. Sundberg
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Jacqueline K. White
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Ian M. Smyth
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia
- * E-mail:
| |
Collapse
|
24
|
DiTommaso T, Cottle DL, Pearson HB, Schlüter H, Kaur P, Humbert PO, Smyth IM. Keratin 76 is required for tight junction function and maintenance of the skin barrier. PLoS Genet 2014; 10:e1004706. [PMID: 25340345 PMCID: PMC4207637 DOI: 10.1371/journal.pgen.1004706] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 08/26/2014] [Indexed: 11/18/2022] Open
Abstract
Keratins are cytoskeletal intermediate filament proteins that are increasingly being recognised for their diverse cellular functions. Here we report the consequences of germ line inactivation of Keratin 76 (Krt76) in mice. Homozygous disruption of this epidermally expressed gene causes neonatal skin flaking, hyperpigmentation, inflammation, impaired wound healing, and death prior to 12 weeks of age. We show that this phenotype is associated with functionally defective tight junctions that are characterised by mislocalization of the integral protein CLDN1. We further demonstrate that KRT76 interacts with CLDN1 and propose that this interaction is necessary to correctly position CLDN1 in tight junctions. The mislocalization of CLDN1 has been associated in various dermopathies, including the inflammatory disease, psoriasis. These observations establish a previously unknown connection between the intermediate filament cytoskeleton network and tight junctions and showcase Krt76 null mice as a possible model to study aberrant tight junction driven skin diseases.
Collapse
Affiliation(s)
- Tia DiTommaso
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Denny L. Cottle
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Helen B. Pearson
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Holger Schlüter
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Pritinder Kaur
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Anatomy & Neuroscience, University of Melbourne, Melbourne, Australia
| | - Patrick O. Humbert
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Australia
- Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Ian M. Smyth
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- * E-mail:
| |
Collapse
|
25
|
Cottle DL, Ursino GMA, Ip SCI, Jones LK, Ditommaso T, Hacking DF, Mangan NE, Mellett NA, Henley KJ, Sviridov D, Nold-Petry CA, Nold MF, Meikle PJ, Kile BT, Smyth IM. Fetal inhibition of inflammation improves disease phenotypes in harlequin ichthyosis. Hum Mol Genet 2014; 24:436-49. [PMID: 25209981 DOI: 10.1093/hmg/ddu459] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Harlequin ichthyosis (HI) is a severe skin disease which leads to neonatal death in ∼50% of cases. It is the result of mutations in ABCA12, a protein that transports lipids required to establish the protective skin barrier needed after birth. To better understand the life-threatening newborn HI phenotype, we analysed the developing epidermis for consequences of lipid dysregulation in mouse models. We observed a pro-inflammatory signature which was characterized by chemokine upregulation in embryonic skin which is distinct from that seen in other types of ichthyosis. Inflammation also persisted in grafted HI skin. To examine the contribution of inflammation to disease development, we overexpressed interleukin-37b to globally suppress fetal inflammation, observing considerable improvements in keratinocyte differentiation. These studies highlight inflammation as an unexpected contributor to HI disease development in utero, and suggest that inhibiting inflammation may reduce disease severity.
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas F Hacking
- Department of Anaesthetics, Saint Vincent's Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia Department of Paediatric Intensive Care, The Royal Children's Hospital, Melbourne, VIC, Australia
| | | | - Natalie A Mellett
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Katya J Henley
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Claudia A Nold-Petry
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Marcel F Nold
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Benjamin T Kile
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia and
| | - Ian M Smyth
- Department of Biochemistry and Molecular Biology Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| |
Collapse
|
26
|
Discovery in genetic skin disease: the impact of high throughput genetic technologies. Genes (Basel) 2014; 5:615-34. [PMID: 25093584 PMCID: PMC4198921 DOI: 10.3390/genes5030615] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/07/2014] [Accepted: 07/14/2014] [Indexed: 11/21/2022] Open
Abstract
The last decade has seen considerable advances in our understanding of the genetic basis of skin disease, as a consequence of high throughput sequencing technologies including next generation sequencing and whole exome sequencing. We have now determined the genes underlying several monogenic diseases, such as harlequin ichthyosis, Olmsted syndrome, and exfoliative ichthyosis, which have provided unique insights into the structure and function of the skin. In addition, through genome wide association studies we now have an understanding of how low penetrance variants contribute to inflammatory skin diseases such as psoriasis vulgaris and atopic dermatitis, and how they contribute to underlying pathophysiological disease processes. In this review we discuss strategies used to unravel the genes underlying both monogenic and complex trait skin diseases in the last 10 years and the implications on mechanistic studies, diagnostics, and therapeutics.
Collapse
|
27
|
Abstract
The epidermis functions as a physical barrier to the external environment and works to prevent loss of water from the skin. Numerous factors have been implicated in the formation of epidermal barriers, such as cornified envelopes, corneocytes, lipids, junctional proteins, proteases, protease inhibitors, antimicrobial peptides, and transcription factors. This review illustrates human diseases (ichthyoses) and animal models in which the epidermal barrier is disrupted or dysfunctional at steady state owing to ablation of one or more of the above factors. These diseases and animal models help us to understand the complicated mechanisms of epidermal barrier formation and give further insights on epidermal development.
Collapse
|
28
|
Mamtani M, Meikle PJ, Kulkarni H, Weir JM, Barlow CK, Jowett JB, Bellis C, Dyer TD, Almasy L, Mahaney MC, Duggirala R, Comuzzie AG, Blangero J, Curran JE. Plasma dihydroceramide species associate with waist circumference in Mexican American families. Obesity (Silver Spring) 2014; 22:950-6. [PMID: 23929697 PMCID: PMC3918249 DOI: 10.1002/oby.20598] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/03/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Waist circumference (WC), the clinical marker of central obesity, is gaining popularity as a screening tool for type 2 diabetes (T2D). While there is epidemiologic evidence favoring the WC-T2D association, its biological substantiation is generally weak. Our objective was to determine the independent association of plasma lipid repertoire with WC. METHODS Samples and data from the San Antonio Family Heart Study of 1208 Mexican Americans from 42 extended families were used. Association of plasma lipidomic profiles with the cross-sectionally assessed WC was determined. Plasma lipidomic profiling entailed liquid chromatography with mass spectrometry. Statistical analyses included multivariable polygenic regression models and bivariate trait analyses using the SOLAR software. RESULTS After adjusting for age and sex interactions, body mass index, homeostasis model of assessment-insulin resistance, total cholesterol, triglycerides, high density lipoproteins and use of lipid lowering drugs, dihydroceramides as a class were associated with WC. Dihydroceramide species 18:0, 20:0, 22:0, and 24:1 were significantly associated and genetically correlated with WC. Two sphingomyelin species (31:1 and 41:1) were also associated with WC. CONCLUSIONS Plasma dihydroceramide levels independently associate with WC. Thus, high resolution plasma lipidomic studies can provide further credence to the biological underpinnings of the association of WC with T2D.
Collapse
Affiliation(s)
- Manju Mamtani
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Peter J. Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jacquelyn M. Weir
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | | | - Jeremy B. Jowett
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Claire Bellis
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Thomas D. Dyer
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Laura Almasy
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Michael C. Mahaney
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - Anthony G. Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - John Blangero
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Joanne E. Curran
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
29
|
Haller JF, Cavallaro P, Hernandez NJ, Dolat L, Soscia SJ, Welti R, Grabowski GA, Fitzgerald ML, Freeman MW. Endogenous β-glucocerebrosidase activity in Abca12⁻/⁻epidermis elevates ceramide levels after topical lipid application but does not restore barrier function. J Lipid Res 2013; 55:493-503. [PMID: 24293640 DOI: 10.1194/jlr.m044941] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
ABCA12 mutations disrupt the skin barrier and cause harlequin ichthyosis. We previously showed Abca12(-/-) skin has increased glucosylceramide (GlcCer) and correspondingly lower amounts of ceramide (Cer). To examine why loss of ABCA12 leads to accumulation of GlcCer, de novo sphingolipid synthesis was assayed using [(14)C]serine labeling in ex vivo skin cultures. A defect was found in β-glucocerebrosidase (GCase) processing of newly synthesized GlcCer species. This was not due to a decline in GCase function. Abca12(-/-) epidermis had 5-fold more GCase protein (n = 4, P < 0.01), and a 5-fold increase in GCase activity (n = 3, P < 0.05). As with Abca12(+/+) epidermis, immunostaining in null skin showed a typical interstitial distribution of the GCase protein in the Abca12(-/-) stratum corneum. Hence, we tested whether the block in GlcCer conversion could be circumvented by topically providing GlcCer. This approach restored up to 15% of the lost Cer products of GCase activity in the Abca12(-/-) epidermis. However, this level of barrier ceramide replacement did not significantly reduce trans-epidermal water loss function. Our results indicate loss of ABCA12 function results in a failure of precursor GlcCer substrate to productively interact with an intact GCase enzyme, and they support a model of ABCA12 function that is critical for transporting GlcCer into lamellar bodies.
Collapse
Affiliation(s)
- Jorge F Haller
- Lipid Metabolism Unit and Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Farhadi R, Kazemi SH. Harlequin ichthyosis in a neonate born with assisted reproductive technology: a case report. Med J Islam Repub Iran 2013; 27:229-32. [PMID: 24926185 PMCID: PMC4011414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 05/26/2013] [Accepted: 05/30/2013] [Indexed: 10/26/2022] Open
Abstract
Harlequin ichthyosis is a rare and the most severe form of congenital ichthyosis. Although prenatal diagnosis isdifficult for this disorder, recently, this obstacle has markedly improved with the use of DNA-based prenataldiagnosis. Here in, we presented a neonate with harlequin ichthyosis born by assisted reproductive technology(ART). In this case, the diagnosis of harlequin ichthyosis was not established by conventional prenatal screening.
Collapse
Affiliation(s)
- Roya Farhadi
- 1 Assistant professor of neonatology, Pediatric department of Booali-Sina Hospital, Mazandaran University of Medical science, Sari, Iran.
| | - Seyyed Habib Kazemi
- 2MD., Vice-chancellor for Treatment, Mazandaran University of Medical science, Sari, Iran,
| |
Collapse
|
31
|
Hou M, Sun R, Hupe M, Kim PL, Park K, Crumrine D, Lin TK, Santiago JL, Mauro TM, Elias PM, Man MQ. Topical apigenin improves epidermal permeability barrier homoeostasis in normal murine skin by divergent mechanisms. Exp Dermatol 2013; 22:210-5. [PMID: 23489424 DOI: 10.1111/exd.12102] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2013] [Indexed: 12/18/2022]
Abstract
The beneficial effects of certain herbal medicines on cutaneous function have been appreciated for centuries. Among these agents, chrysanthemum extract, apigenin, has been used for skin care, particularly in China, for millennia. However, the underlying mechanisms by which apigenin benefits the skin are not known. In this study, we first determined whether topical apigenin positively influences permeability barrier homoeostasis, and then the basis thereof. Hairless mice were treated topically with either 0.1% apigenin or vehicle alone twice daily for 9 days. At the end of the treatments, permeability barrier function was assessed with either an electrolytic water analyzer or a Tewameter. Our results show that topical apigenin significantly enhanced permeability barrier homoeostasis after tape stripping, although basal permeability barrier function remained unchanged. Improved barrier function correlated with enhanced filaggrin expression and lamellar body production, which was paralleled by elevated mRNA levels for the epidermal ABCA12. The mRNA levels for key lipid synthetic enzymes also were upregulated by apigenin. Finally, both cathelicidin-related peptide and mouse beta-defensin 3 immunostaining were increased by apigenin. We conclude that topical apigenin improves epidermal permeability barrier function by stimulating epidermal differentiation, lipid synthesis and secretion, as well as cutaneous antimicrobial peptide production. Apigenin could be useful for the prevention and treatment of skin disorders characterized by permeability barrier dysfunction, associated with reduced filaggrin levels and impaired antimicrobial defenses, such as atopic dermatitis.
Collapse
Affiliation(s)
- Maihua Hou
- Dermatology Service, Veterans Affairs Medical Center, and University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sviridov D. Many ABCs make light work with cholesterol. Aging (Albany NY) 2013; 5:637-8. [PMID: 24244949 PMCID: PMC3808694 DOI: 10.18632/aging.100596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
33
|
Akiyama M. The roles of ABCA12 in epidermal lipid barrier formation and keratinocyte differentiation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:435-40. [PMID: 23954554 DOI: 10.1016/j.bbalip.2013.08.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/06/2013] [Accepted: 08/08/2013] [Indexed: 01/01/2023]
Abstract
ATP-binding cassette (ABC) transporters form a large superfamily of transporters that bind and hydrolyze ATP to transport various molecules across limiting membranes or into vesicles. The ABCA subfamily members are thought to transport lipid materials. ABCA12 is a keratinocyte transmembrane lipid transporter protein associated with the transport of lipids via lamellar granules. ABCA12 is considered to transport lipids including ceramides to form extracellular lipid layers in the stratum corneum of the epidermis, which is essential for skin barrier function. ABCA12 mutations are known to underlie the three major types of autosomal recessive congenital ichthyoses: harlequin ichthyosis, lamellar ichthyosis and congenital ichthyosiform erythroderma. ABCA12 mutations result in defective lipid transport via lamellar granules in the keratinocytes, leading to ichthyosis phenotypes from malformation of the stratum corneum lipid barrier. Studies on ABCA12-deficient bioengineered models have revealed that lipid transport by ABCA12 is required for keratinocyte differentiation and epidermal morphogenesis. Defective lipid transport due to loss of ABCA12 function leads to the accumulation of intracellular lipids, including glucosylceramides and gangliosides, in the epidermal keratinocytes. The accumulation of gangliosides seems to result in the apoptosis of Abca12(-/-) keratinocytes. It was reported that AKT activation occurs in Abca12(-/-) granular-layer keratinocytes, which suggests that AKT activation serves to prevent the cell death of Abca12(-/-) keratinocytes. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.
Collapse
Affiliation(s)
- Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Tsurumai-cho 65, Showa-ku, Nagoya 466-8550, Japan.
| |
Collapse
|
34
|
Fu Y, Mukhamedova N, Ip S, D'Souza W, Henley KJ, DiTommaso T, Kesani R, Ditiatkovski M, Jones L, Lane RM, Jennings G, Smyth IM, Kile BT, Sviridov D. ABCA12 regulates ABCA1-dependent cholesterol efflux from macrophages and the development of atherosclerosis. Cell Metab 2013; 18:225-38. [PMID: 23931754 DOI: 10.1016/j.cmet.2013.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 03/05/2013] [Accepted: 07/02/2013] [Indexed: 11/16/2022]
Abstract
ABCA12 is involved in the transport of ceramides in skin, but it may play a wider role in lipid metabolism. We show that, in Abca12-deficient macrophages, cholesterol efflux failed to respond to activation with LXR agonists. Abca12 deficiency caused a reduction in the abundance of Abca1, Abcg1, and Lxrβ. Overexpression of Lxrβ reversed the effects. Mechanistically, Abca12 deficiency did not affect expression of genes involved in cholesterol metabolism. Instead, a physical association between Abca1, Abca12, and Lxrβ proteins was established. Abca12 deficiency enhanced interaction between Abca1 and Lxrβ and the degradation of Abca1. Overexpression of ABCA12 in HeLa-ABCA1 cells increased the abundance and stability of ABCA1. Abca12 deficiency caused an accumulation of cholesterol in macrophages and the formation of foam cells, impaired reverse cholesterol transport in vivo, and increased the development of atherosclerosis in irradiated Apoe(-/-) mice reconstituted with Apoe(-/-)Abca12(-/-) bone marrow. Thus, ABCA12 regulates the cellular cholesterol metabolism via an LXRβ-dependent posttranscriptional mechanism.
Collapse
Affiliation(s)
- Ying Fu
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Radner FPW, Fischer J. The important role of epidermal triacylglycerol metabolism for maintenance of the skin permeability barrier function. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:409-15. [PMID: 23928127 DOI: 10.1016/j.bbalip.2013.07.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 12/29/2022]
Abstract
Survival in a terrestrial, dry environment necessitates a permeability barrier for regulated permeation of water and electrolytes in the cornified layer of the skin (the stratum corneum) to minimize desiccation of the body. This barrier is formed during cornification and involves a cross-linking of corneocyte proteins as well as an extensive remodeling of lipids. The cleavage of precursor lipids from lamellar bodies by various hydrolytic enzymes generates ceramides, cholesterol, and non-esterified fatty acids for the extracellular lipid lamellae in the stratum corneum. However, the important role of epidermal triacylglycerol (TAG) metabolism during formation of a functional permeability barrier in the skin was only recently discovered. Humans with mutations in the ABHD5/CGI-58 (α/β hydrolase domain containing protein 5, also known as comparative gene identification-58, CGI-58) gene suffer from a defect in TAG catabolism that causes neutral lipid storage disease with ichthyosis. In addition, mice with deficiencies in genes involved in TAG catabolism (Abhd5/Cgi-58 knock-out mice) or TAG synthesis (acyl-CoA:diacylglycerol acyltransferase-2, Dgat2 knock-out mice) also develop severe skin permeability barrier dysfunctions and die soon after birth due to increased dehydration. As a result of these defects in epidermal TAG metabolism, humans and mice lack ω-(O)-acylceramides, which leads to malformation of the cornified lipid envelope of the skin. In healthy skin, this epidermal structure provides an interface for the linkage of lamellar membranes with corneocyte proteins to maintain permeability barrier homeostasis. This review focuses on recent advances in the understanding of biochemical mechanisms involved in epidermal neutral lipid metabolism and the generation of a functional skin permeability barrier. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.
Collapse
Affiliation(s)
- Franz P W Radner
- Institute for Human Genetics, University Medical Center Freiburg, Freiburg 79106, Germany.
| | - Judith Fischer
- Institute for Human Genetics, University Medical Center Freiburg, Freiburg 79106, Germany
| |
Collapse
|
36
|
Weir JM, Wong G, Barlow CK, Greeve MA, Kowalczyk A, Almasy L, Comuzzie AG, Mahaney MC, Jowett JBM, Shaw J, Curran JE, Blangero J, Meikle PJ. Plasma lipid profiling in a large population-based cohort. J Lipid Res 2013; 54:2898-908. [PMID: 23868910 DOI: 10.1194/jlr.p035808] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We have performed plasma lipid profiling using liquid chromatography electrospray ionization tandem mass spectrometry on a population cohort of more than 1,000 individuals. From 10 μl of plasma we were able to acquire comparative measures of 312 lipids across 23 lipid classes and subclasses including sphingolipids, phospholipids, glycerolipids, and cholesterol esters (CEs) in 20 min. Using linear and logistic regression, we identified statistically significant associations of lipid classes, subclasses, and individual lipid species with anthropometric and physiological measures. In addition to the expected associations of CEs and triacylglycerol with age, sex, and body mass index (BMI), ceramide was significantly higher in males and was independently associated with age and BMI. Associations were also observed for sphingomyelin with age but this lipid subclass was lower in males. Lysophospholipids were associated with age and higher in males, but showed a strong negative association with BMI. Many of these lipids have previously been associated with chronic diseases including cardiovascular disease and may mediate the interactions of age, sex, and obesity with disease risk.
Collapse
|
37
|
Kulkarni H, Meikle PJ, Mamtani M, Weir JM, Barlow CK, Jowett JB, Bellis C, Dyer TD, Johnson MP, Rainwater DL, Almasy L, Mahaney MC, Comuzzie AG, Blangero J, Curran JE. Plasma lipidomic profile signature of hypertension in Mexican American families: specific role of diacylglycerols. Hypertension 2013; 62:621-6. [PMID: 23798346 DOI: 10.1161/hypertensionaha.113.01396] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both as a component of metabolic syndrome and as an independent entity, hypertension poses a continued challenge with regard to its diagnosis, pathogenesis, and treatment. Previous studies have documented connections between hypertension and indicators of lipid metabolism. Novel technologies, such as plasma lipidomic profiling, promise a better understanding of disorders in which there is a derangement of the lipid metabolism. However, association of plasma lipidomic profiles with hypertension in a high-risk population, such as Mexican Americans, has not been evaluated before. Using the rich data and sample resource from the ongoing San Antonio Family Heart Study, we conducted plasma lipidomic profiling by combining high-performance liquid chromatography with tandem mass spectroscopy to characterize 319 lipid species in 1192 individuals from 42 large and extended Mexican American families. Robust statistical analyses using polygenic regression models, liability threshold models, and bivariate trait analyses implemented in the SOLAR software were conducted after accounting for obesity, insulin resistance, and relative abundance of various lipoprotein fractions. Diacylglycerols, in general, and the DG 16:0/22:5 and DG 16:0/22:6 lipid species, in particular, were significantly associated with systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP), as well as liability of incident hypertension measured during 7140.17 person-years of follow-up. Four lipid species, including the DG 16:0/22:5 and DG 16:0/22:6 species, showed significant genetic correlations with the liability of hypertension in bivariate trait analyses. Our results demonstrate the value of plasma lipidomic profiling in the context of hypertension and identify disturbance of diacylglycerol metabolism as an independent biomarker of hypertension.
Collapse
Affiliation(s)
- Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78245, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Autosomal recessive congenital ichthyosis. ACTAS DERMO-SIFILIOGRAFICAS 2013; 104:270-84. [PMID: 23562412 DOI: 10.1016/j.adengl.2011.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/13/2011] [Indexed: 12/15/2022] Open
Abstract
The term autosomal recessive congenital ichthyosis (ARCI) refers to a group of rare disorders of keratinization classified as nonsyndromic forms of ichthyosis. This group was traditionally divided into lamellar ichthyosis (LI) and congenital ichthyosiform erythroderma (CIE) but today it also includes harlequin ichthyosis, self-healing collodion baby, acral self-healing collodion baby, and bathing suit ichthyosis. The combined prevalence of LI and CIE has been estimated at 1 case per 138 000 to 300 000 population. In some countries or regions, such as Norway and the coast of Galicia, the prevalence may be higher due to founder effects. ARCI is genetically highly heterogeneous and has been associated with 6 genes to date: TGM1, ALOXE3, ALOX12B, NIPAL4, CYP4F22, and ABCA12. In this article, we review the current knowledge on ARCI, with a focus on clinical, histological, ultrastructural, genetic, molecular, and treatment-related aspects.
Collapse
|
40
|
Kulkarni H, Meikle PJ, Mamtani M, Weir JM, Barlow CK, Jowett JB, Bellis C, Dyer TD, Johnson MP, Rainwater DL, Almasy L, Mahaney MC, Comuzzie AG, Blangero J, Curran JE. Variability in associations of phosphatidylcholine molecular species with metabolic syndrome in Mexican-American families. Lipids 2013; 48:497-503. [PMID: 23494580 DOI: 10.1007/s11745-013-3781-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/19/2013] [Indexed: 12/14/2022]
Abstract
Plasma lipidomic studies using high performance liquid chromatography and mass spectroscopy offer detailed insights into metabolic processes. Taking the example of the most abundant plasma lipid class (phosphatidylcholines) we used the rich phenotypic and lipidomic data from the ongoing San Antonio Family Heart Study of large extended Mexican-American families to assess the variability of association of the plasma phosphatidylcholine species with metabolic syndrome. Using robust statistical analytical methods, our study made two important observations. First, there was a wide variability in the association of phosphatidylcholine species with risk measures of metabolic syndrome. Phosphatidylcholine 40:7 was associated with a low risk while phosphatidylcholines 32:1 and 38:3 were associated with a high risk of metabolic syndrome. Second, all the odd chain phosphatidylcholines were associated with a reduced risk of metabolic syndrome implying that phosphatidylcholines derived from dairy products might be beneficial against metabolic syndrome. Our results demonstrate the value of lipid species-specific information provided by the upcoming array of lipidomic studies and open potential avenues for prevention and control of metabolic syndrome in high prevalence settings.
Collapse
Affiliation(s)
- Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Goldstein I, Rotter V. Regulation of lipid metabolism by p53 - fighting two villains with one sword. Trends Endocrinol Metab 2012; 23:567-75. [PMID: 22819212 DOI: 10.1016/j.tem.2012.06.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 01/01/2023]
Abstract
Both cellular and systemic metabolism of lipids are paramount for homeostasis, and their malfunction leads to devastating pathologies. Recently, exciting findings have linked the p53 tumor suppressor to the regulation of lipid metabolism. Here, we summarize these findings showing a clear role for p53 in enhancing lipid catabolism while inhibiting its anabolism. We also describe the multitude of genes regulated by p53 that participate in or regulate systemic lipid transport. From the compilation of available data a scenario is emerging in which p53 regulates genes involved in lipid metabolism - both in a cancer-preventive effort and, intriguingly, as a means to prevent atherosclerosis. Thus, by regulating lipid metabolism, p53 fights the two major causes of death worldwide - atherosclerosis and cancer.
Collapse
Affiliation(s)
- Ido Goldstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100 Israel
| | | |
Collapse
|
42
|
Coleman JA, Quazi F, Molday RS. Mammalian P4-ATPases and ABC transporters and their role in phospholipid transport. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:555-74. [PMID: 23103747 DOI: 10.1016/j.bbalip.2012.10.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 02/08/2023]
Abstract
Transport of phospholipids across cell membranes plays a key role in a wide variety of biological processes. These include membrane biosynthesis, generation and maintenance of membrane asymmetry, cell and organelle shape determination, phagocytosis, vesicle trafficking, blood coagulation, lipid homeostasis, regulation of membrane protein function, apoptosis, etc. P(4)-ATPases and ATP binding cassette (ABC) transporters are the two principal classes of membrane proteins that actively transport phospholipids across cellular membranes. P(4)-ATPases utilize the energy from ATP hydrolysis to flip aminophospholipids from the exocytoplasmic (extracellular/lumen) to the cytoplasmic leaflet of cell membranes generating membrane lipid asymmetry and lipid imbalance which can induce membrane curvature. Many ABC transporters play crucial roles in lipid homeostasis by actively transporting phospholipids from the cytoplasmic to the exocytoplasmic leaflet of cell membranes or exporting phospholipids to protein acceptors or micelles. Recent studies indicate that some ABC proteins can also transport phospholipids in the opposite direction. The importance of P(4)-ATPases and ABC transporters is evident from the findings that mutations in many of these transporters are responsible for severe human genetic diseases linked to defective phospholipid transport. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.
Collapse
Affiliation(s)
- Jonathan A Coleman
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, B.C., Canada
| | | | | |
Collapse
|
43
|
Brown FC, Scott N, Rank G, Collinge JE, Vadolas J, Vickaryous N, Whitelaw N, Whitelaw E, Kile BT, Jane SM, Curtis DJ. ENU mutagenesis identifies the first mouse mutants reproducing human β-thalassemia at the genomic level. Blood Cells Mol Dis 2012; 50:86-92. [PMID: 23040355 DOI: 10.1016/j.bcmd.2012.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 09/11/2012] [Indexed: 12/24/2022]
Abstract
Forward genetic screens have been performed in many species to identify phenotypes in specific organ systems. We have undertaken a large-scale N-ethyl-N-nitrosourea (ENU) mutagenesis screen to identify dominant mutations that perturb erythropoiesis in mice. Mutant mice that displayed an erythrocyte mean cell volume (MCV) greater than three standard deviations from the population mean were identified. Two of these lines, RBC13 and RBC14, displayed a hypochromic, microcytic anemia, accompanied by a marked reticulocytosis, splenomegaly and diminished red cell survival. Timed pregnancies from heterozygous intercrosses revealed that a quarter of the embryos displayed severe anemia and did not survive beyond embryonic day (E) 18.5, consistent with homozygous β-thalassemia. Genetic complementation studies with a β-thalassemia mouse line reproduced the embryonic lethality in compound heterozygotes and a genomic custom capture array and massively parallel sequencing of the β-globin locus identified the causative mutations. The RBC13 line displayed a nonsense mutation at codon 40 in exon 2 of the β-major gene, invoking parallels with the common β(0)39 thalassemia mutation seen in humans. The RBC14 line exhibited a mutation at the polyadenylation signal of the β-major gene, exactly replicating a human β-thalassemia mutation. The RBC13 and RBC14 lines are the first β-thalassemia mouse models that reproduce human β-thalassemia at the genomic level, and as such highlight the power of ENU mutagenesis screens in generating mouse models of human disease.
Collapse
Affiliation(s)
- Fiona C Brown
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Harlequin ichthyosis: ABCA12 mutations underlie defective lipid transport, reduced protease regulation and skin-barrier dysfunction. Cell Tissue Res 2012; 351:281-8. [PMID: 22864982 DOI: 10.1007/s00441-012-1474-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/21/2012] [Indexed: 01/09/2023]
Abstract
Harlequin ichthyosis (HI) is a devastating autosomal recessive congenital skin disease. It has been vital to elucidate the biological importance of the protein ABCA12 in skin-barrier permeability, following the discovery that ABCA12 gene mutations can result in this rare disease. ATP-binding cassette transporter A12 (ABCA12) is a member of the subfamily of ATP-binding cassette transporters and functions to transport lipid glucosylceramides (GlcCer) to the extracellular space through lamellar granules (LGs). GlcCer are hydrolysed into hydroxyceramides extracellularly and constitute a portion of the extracellular lamellar membrane, lipid envelope and lamellar granules. In HI skin, loss of function of ABCA12 due to null mutations results in impaired lipid lamellar membrane formation in the cornified layer, leading to defective permeability of the skin barrier. In addition, abnormal lamellar granule formation (distorted shape, reduced in number or absent) could further cause aberrant production of LG-associated desquamation enzymes, which are likely to contribute to the impaired skin barrier in HI. This article reviews current opinions on the patho-mechanisms of ABCA12 action in HI and potential therapeutic interventions based on targeted molecular therapy and gene therapy strategies.
Collapse
|
45
|
Barber MN, Risis S, Yang C, Meikle PJ, Staples M, Febbraio MA, Bruce CR. Plasma lysophosphatidylcholine levels are reduced in obesity and type 2 diabetes. PLoS One 2012; 7:e41456. [PMID: 22848500 PMCID: PMC3405068 DOI: 10.1371/journal.pone.0041456] [Citation(s) in RCA: 254] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 06/21/2012] [Indexed: 12/21/2022] Open
Abstract
Background Obesity and type 2 diabetes (T2DM) are associated with increased circulating free fatty acids and triacylglycerols. However, very little is known about specific molecular lipid species associated with these diseases. In order to gain further insight into this, we performed plasma lipidomic analysis in a rodent model of obesity and insulin resistance as well as in lean, obese and obese individuals with T2DM. Methodology/Principal Findings Lipidomic analysis using liquid chromatography coupled to mass spectrometry revealed marked changes in the plasma of 12 week high fat fed mice. Although a number of triacylglycerol and diacylglycerol species were elevated along with of a number of sphingolipids, a particularly interesting finding was the high fat diet (HFD)-induced reduction in lysophosphatidylcholine (LPC) levels. As liver, skeletal muscle and adipose tissue play an important role in metabolism, we next determined whether the HFD altered LPCs in these tissues. In contrast to our findings in plasma, only very modest changes in tissue LPCs were noted. To determine when the change in plasma LPCs occurred in response to the HFD, mice were studied after 1, 3 and 6 weeks of HFD. The HFD caused rapid alterations in plasma LPCs with most changes occurring within the first week. Consistent with our rodent model, data from our small human cohort showed a reduction in a number of LPC species in obese and obese individuals with T2DM. Interestingly, no differences were found between the obese otherwise healthy individuals and the obese T2DM patients. Conclusion Irrespective of species, our lipidomic profiling revealed a generalized decrease in circulating LPC species in states of obesity. Moreover, our data indicate that diet and adiposity, rather than insulin resistance or diabetes per se, play an important role in altering the plasma LPC profile.
Collapse
Affiliation(s)
- Melissa N. Barber
- Biobank, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Steve Risis
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Christine Yang
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J. Meikle
- Metabolomics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Margaret Staples
- Department of Epidemiology, Monash University, Melbourne, Victoria, Australia
| | - Mark A. Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Clinton R. Bruce
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
46
|
|
47
|
Prade E, Tobiasch M, Hitkova I, Schäffer I, Lian F, Xing X, Tänzer M, Rauser S, Walch A, Feith M, Post S, Röcken C, Schmid RM, Ebert MPA, Burgermeister E. Bile acids down-regulate caveolin-1 in esophageal epithelial cells through sterol responsive element-binding protein. Mol Endocrinol 2012; 26:819-32. [PMID: 22474125 DOI: 10.1210/me.2011-1140] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bile acids are synthesized from cholesterol and are major risk factors for Barrett adenocarcinoma (BAC) of the esophagus. Caveolin-1 (Cav1), a scaffold protein of membrane caveolae, is transcriptionally regulated by cholesterol via sterol-responsive element-binding protein-1 (SREBP1). Cav1 protects squamous epithelia by controlling cell growth and stabilizing cell junctions and matrix adhesion. Cav1 is frequently down-regulated in human cancers; however, the molecular mechanisms that lead to this event are unknown. We show that the basal layer of the nonneoplastic human esophageal squamous epithelium expressed Cav1 mainly at intercellular junctions. In contrast, Cav1 was lost in 95% of tissue specimens from BAC patients (n = 100). A strong cytoplasmic expression of Cav1 correlated with poor survival in a small subgroup (n = 5) of BAC patients, and stable expression of an oncogenic Cav1 variant (Cav1-P132L) in the human BAC cell line OE19 promoted proliferation. Cav1 was also detectable in immortalized human squamous epithelial, Barrett esophagus (CPC), and squamous cell carcinoma cells (OE21), but was low in BAC cell lines (OE19, OE33). Mechanistically, bile acids down-regulated Cav1 expression by inhibition of the proteolytic cleavage of 125-kDa pre-SREBP1 from the endoplasmic reticulum/Golgi apparatus and nuclear translocation of active 68-kDa SREBP1. This block in SREBP1's posttranslational processing impaired transcriptional activation of SREBP1 response elements in the proximal human Cav1 promoter. Cav1 was also down-regulated in esophagi from C57BL/6 mice on a diet enriched with 1% (wt/wt) chenodeoxycholic acid. Mice deficient for Cav1 or the nuclear bile acid receptor farnesoid X receptor showed hyperplasia and hyperkeratosis of the basal cell layer of esophageal epithelia, respectively. These data indicate that bile acid-mediated down-regulation of Cav1 marks early changes in the squamous epithelium, which may contribute to onset of Barrett esophagus metaplasia and progression to BAC.
Collapse
Affiliation(s)
- Elke Prade
- Department of Chemistry, Klinikum rechts der Isar, Technische Universität München, D-81675 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Cholesterol content of cells must be maintained within the very tight limits, too much or too little cholesterol in a cell results in disruption of cellular membranes, apoptosis and necrosis. Cells can source cholesterol from intracellular synthesis and from plasma lipoproteins, both sources are sufficient to fully satisfy cells' requirements for cholesterol. The processes of cholesterol synthesis and uptake are tightly regulated and deficiencies of cholesterol are rare. Excessive cholesterol is more common problem. With the exception of hepatocytes and to some degree adrenocortical cells, cells are unable to degrade cholesterol. Cells have two options to reduce their cholesterol content: to convert cholesterol into cholesteryl esters, an option with limited capacity as overloading cells with cholesteryl esters is also toxic, and cholesterol efflux, an option with potentially unlimited capacity. Cholesterol efflux is a specific process that is regulated by a number of intracellular transporters, such as ATP binding cassette transporter proteins A1 (ABCA1) and G1 (ABCG1) and scavenger receptor type B1. The natural acceptor of cholesterol in plasma is high density lipoprotein (HDL) and apolipoprotein A-I. The cholesterol efflux assay is designed to quantitate the rate of cholesterol efflux from cultured cells. It measures the capacity of cells to maintain cholesterol efflux and/or the capacity of plasma acceptors to accept cholesterol released from cells. The assay consists of the following steps. Step 1: labelling cellular cholesterol by adding labelled cholesterol to serum-containing medium and incubating with cells for 24-48 h. This step may be combined with loading of cells with cholesterol. Step 2: incubation of cells in serum-free medium to equilibrate labelled cholesterol among all intracellular cholesterol pools. This stage may be combined with activation of cellular cholesterol transporters. Step 3: incubation of cells with extracellular acceptor and quantitation of movement of labelled cholesterol from cells to the acceptor. If cholesterol precursors were used to label newly synthesized cholesterol, a fourth step, purification of cholesterol, may be required. The assay delivers the following information: (i) how a particular treatment (a mutation, a knock-down, an overexpression or a treatment) affects the capacity of cell to efflux cholesterol and (ii) how the capacity of plasma acceptors to accept cholesterol is affected by a disease or a treatment. This method is often used in context of cardiovascular research, metabolic and neurodegenerative disorders, infectious and reproductive diseases.
Collapse
Affiliation(s)
- Hann Low
- Baker IDI Heart and Diabetes Institute
| | | | | |
Collapse
|
49
|
Goldstein I, Ezra O, Rivlin N, Molchadsky A, Madar S, Goldfinger N, Rotter V. p53, a novel regulator of lipid metabolism pathways. J Hepatol 2012; 56:656-62. [PMID: 22037227 DOI: 10.1016/j.jhep.2011.08.022] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 08/03/2011] [Accepted: 08/09/2011] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS In this study we aimed at characterizing the regulation of hepatic metabolic pathways by the p53 transcription factor. METHODS Analysis of gene expression following alteration of p53 status in several human- and mouse-derived cells using microarray analysis, quantitative real-time PCR, chromatin immunoprecipitation, and reporter gene assays. A functional assay was performed to determine lipid transfer activity. RESULTS We identified a novel role for the p53 protein in regulating lipid and lipoprotein metabolism, a process not yet conceived as related to p53, which is known mainly for its tumor suppressive functions. We revealed a group of 341 genes whose expression was induced by p53 in the liver-derived cell line HepG2. Twenty of these genes encode proteins involved in many aspects of lipid homeostasis. The mode of regulation of three representative genes (Pltp, Abca12, and Cel) was further characterized. In addition to HepG2, the genes were induced following activation of p53 in human primary hepatic cells isolated from liver donors. p53-dependent regulation of these genes was evident in other cell types namely Hep3B cells, mouse hepatocytes, and fibroblasts. Furthermore, p53 was found to bind to the genes' promoters in designated p53 responsive elements and thereby increase transcription. Importantly, p53 augmented the activity of secreted PLTP, which plays a major role in lipoprotein biology and atherosclerosis pathology. CONCLUSIONS These findings expose another facet of p53 functions unrelated to tumor suppression and render it a novel regulator of hepatic lipid metabolism and consequently of systemic lipid homeostasis and atherosclerosis development.
Collapse
Affiliation(s)
- Ido Goldstein
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel.
| | | | | | | | | | | | | |
Collapse
|
50
|
Dahlqvist J, Westermark GT, Vahlquist A, Dahl N. Ichthyin/NIPAL4 localizes to keratins and desmosomes in epidermis and Ichthyin mutations affect epidermal lipid metabolism. Arch Dermatol Res 2012; 304:377-86. [PMID: 22258272 DOI: 10.1007/s00403-012-1207-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/01/2012] [Accepted: 01/05/2012] [Indexed: 11/29/2022]
Abstract
Autosomal recessive congenital ichthyosis (ARCI) is a group of disorders characterized by abnormal desquamation of the skin and a disrupted epidermal water barrier. Ichthyin/NIPAL4 gene mutations have been identified in a subgroup of ARCI patients, but the role of ichthyin in epidermis remains elusive. In order to obtain new insights concerning the characteristics of ichthyin and the ARCI pathogenesis, we studied the expression and localization of ichthyin and related epidermal components in cultured keratinocytes and skin sections from patients with Ichthyin mutations and healthy controls. We observed an up-regulation of Ichthyin mRNA levels after in vitro differentiation of keratinocytes from both a patient with Ichthyin mutations and controls. Confocal and electron microscopy analyses of immunolabeled skin sections revealed that ichthyin localizes to desmosomes and keratins in both patients with mutant Ichthyin and controls, with an increased immunolabeling in patients. Nile red lipid analysis of skin sections exposed intra-cellular lipid accumulations in cells of the granular and cornified layers in patients but not in controls, consistent with the pathognomonic lipid membrane structures previously identified in epidermis from patients. Our combined findings indicate that ichthyin is associated with keratins and desmosomes in epidermis and is involved in lipid metabolism, possibly through processing of lamellar bodies. These results provide new clues to the understanding of the epidermal water barrier and the pathogenesis in ARCI.
Collapse
Affiliation(s)
- Johanna Dahlqvist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| | | | | | | |
Collapse
|