1
|
Redmond CJ, Steiner SN, Cohen E, Johnson CN, Özlü N, Coulombe PA. Keratin 15 promotes a progenitor cell state in basal keratinocytes of skin epidermis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640633. [PMID: 40060679 PMCID: PMC11888442 DOI: 10.1101/2025.02.27.640633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
The type I intermediate filament proteins keratin 14 (K14) and keratin 15 (K15) are common to all complex epithelia. K14 is highly expressed by progenitor keratinocytes, in which it provides essential mechanical integrity and gates keratinocyte entry into differentiation by sequestering YAP1, a transcriptional effector of Hippo signaling, to the cytoplasm. K15 has long been used as a marker of hair bulge stem cells though its specific role in skin epithelia is unknown. Here we show that the lack of two biochemical determinants, a cysteine residue within the stutter motif of the central rod domain and a 14-3-3 binding site in the N-terminal head domain, renders K15 unable to effectively sequester YAP1 in the cytoplasm. We combine insight obtained from cell culture and transgenic mouse models with computational analyses of transcriptomics data and propose a model in which the K15:K14 ratio promotes a progenitor state and antagonizes differentiation in keratinocytes of the epidermis.
Collapse
|
2
|
Benito-Martínez S, Salavessa L, Macé AS, Lardier N, Fraisier V, Sirés-Campos J, Jani RA, Romao M, Gayrard C, Plessis M, Hurbain I, Nait-Meddour C, Morel E, Boniotto M, Manneville JB, Bernerd F, Duval C, Raposo G, Delevoye C. Keratin intermediate filaments mechanically position melanin pigments for genome photoprotection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.632531. [PMID: 39868182 PMCID: PMC11761041 DOI: 10.1101/2025.01.15.632531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Melanin pigments block genotoxic agents by positioning on the sun-exposed side of human skin keratinocytes' nucleus. How this position is regulated and its role in genome photoprotection remains unknown. By developing a model of human keratinocytes internalizing extracellular melanin into pigment organelles, we show that keratin 5/14 intermediate filaments mechanically control the 3D perinuclear position of pigments, shielding DNA from photodamage. Imaging and microrheology in human disease-related model identify structural keratin cages surrounding pigment organelles to stiffen their microenvironment and maintain their 3D position. Optimum pigment spatialization is required for DNA photoprotection and rely on the interplay between intermediate filaments and microtubules bridged by plectin cytolinkers. Thus, the mechanically-driven proximity of pigment organelles to the nucleus is a key photoprotective parameter. Uncovering how human skin counteracts solar radiation by positioning the melanin microparasol next to the genome anticipates that dynamic spatialization of organelles is a physiological UV stress response.
Collapse
Affiliation(s)
- Silvia Benito-Martínez
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
| | - Laura Salavessa
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Anne-Sophie Macé
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Nathan Lardier
- Institut Curie, PSL Research University, CNRS, UMR144, Molecular Mechanisms of Intracellular Transport, 75005 Paris, France
| | - Vincent Fraisier
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Julia Sirés-Campos
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
| | - Riddhi Atul Jani
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
| | - Maryse Romao
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | | | - Marion Plessis
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Ilse Hurbain
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Cécile Nait-Meddour
- Univ Paris Est Creteil, INSERM, IMRB, Translational Neuropsychiatry, F-94010 Creteil, France
| | - Etienne Morel
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Michele Boniotto
- Univ Paris Est Creteil, INSERM, IMRB, Translational Neuropsychiatry, F-94010 Creteil, France
| | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR144, Molecular Mechanisms of Intracellular Transport, 75005 Paris, France
- Laboratoire Matières et Systèmes Complexes (MSC), Université Paris Cité, CNRS, UMR7057, 10 rue Alice Domon et Léonie Duquet, F-75013, Paris, France
| | | | | | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| |
Collapse
|
3
|
Outla Z, Prechova M, Korelova K, Gemperle J, Gregor M. Mechanics of cell sheets: plectin as an integrator of cytoskeletal networks. Open Biol 2025; 15:240208. [PMID: 39875099 PMCID: PMC11774597 DOI: 10.1098/rsob.240208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Epithelia are multicellular sheets that form barriers defining the internal and external environments. The constant stresses acting at this interface require that epithelial sheets are mechanically robust and provide a selective barrier to the hostile exterior. These properties are mediated by cellular junctions which are physically linked with heavily crosslinked cytoskeletal networks. Such hardwiring is facilitated by plakins, a family of giant modular proteins which serve as 'molecular bridges' between different cytoskeletal filaments and multiprotein adhesion complexes. Dysfunction of cytoskeletal crosslinking compromises epithelial biomechanics and structural integrity. Subsequent loss of barrier function leads to disturbed tissue homeostasis and pathological consequences such as skin blistering or intestinal inflammation. In this article, we highlight the importance of the cytolinker protein plectin for the functional organization of epithelial cytoskeletal networks. In particular, we focus on the ability of plectin to act as an integrator of the epithelial cytoarchitecture that defines the biomechanics of the whole tissue. Finally, we also discuss the role of cytoskeletal crosslinking in emerging aspects of epithelial mechanobiology that are critical for the maintenance of epithelial homeostasis.
Collapse
Affiliation(s)
- Zuzana Outla
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magdalena Prechova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Gemperle
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
4
|
A Kaleidoscope of Keratin Gene Expression and the Mosaic of Its Regulatory Mechanisms. Int J Mol Sci 2023; 24:ijms24065603. [PMID: 36982676 PMCID: PMC10052683 DOI: 10.3390/ijms24065603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Keratins are a family of intermediate filament-forming proteins highly specific to epithelial cells. A combination of expressed keratin genes is a defining property of the epithelium belonging to a certain type, organ/tissue, cell differentiation potential, and at normal or pathological conditions. In a variety of processes such as differentiation and maturation, as well as during acute or chronic injury and malignant transformation, keratin expression undergoes switching: an initial keratin profile changes accordingly to changed cell functions and location within a tissue as well as other parameters of cellular phenotype and physiology. Tight control of keratin expression implies the presence of complex regulatory landscapes within the keratin gene loci. Here, we highlight patterns of keratin expression in different biological conditions and summarize disparate data on mechanisms controlling keratin expression at the level of genomic regulatory elements, transcription factors (TFs), and chromatin spatial structure.
Collapse
|
5
|
Ievlev V, Lynch TJ, Freischlag KW, Gries CB, Shah A, Pai AC, Ahlers BA, Park S, Engelhardt JF, Parekh KR. Krt14 and Krt15 differentially regulate regenerative properties and differentiation potential of airway basal cells. JCI Insight 2023; 8:e162041. [PMID: 36512409 PMCID: PMC9977304 DOI: 10.1172/jci.insight.162041] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Keratin expression dynamically changes in airway basal cells (BCs) after acute and chronic injury, yet the functional consequences of these changes on BC behavior remain unknown. In bronchiolitis obliterans (BO) after lung transplantation, BC clonogenicity declines, which is associated with a switch from keratin15 (Krt15) to keratin14 (Krt14). We investigated these keratins' roles using Crispr-KO in vitro and in vivo and found that Krt14-KO and Krt15-KO produce contrasting phenotypes in terms of differentiation and clonogenicity. Primary mouse Krt14-KO BCs did not differentiate into club and ciliated cells but had enhanced clonogenicity. By contrast, Krt15-KO did not alter BC differentiation but impaired clonogenicity in vitro and reduced the number of label-retaining BCs in vivo after injury. Krt14, but not Krt15, bound the tumor suppressor stratifin (Sfn). Disruption of Krt14, but not of Krt15, reduced Sfn protein abundance and increased expression of the oncogene dNp63a during BC differentiation, whereas dNp63a levels were reduced in Krt15-KO BCs. Overall, the phenotype of Krt15-KO BCs contrasts with Krt14-KO phenotype and resembles the phenotype in BO with decreased clonogenicity, increased Krt14, and decreased dNp63a expression. This work demonstrates that Krt14 and Krt15 functionally regulate BC behavior, which is relevant in chronic disease states like BO.
Collapse
Affiliation(s)
- Vitaly Ievlev
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Thomas J. Lynch
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Kyle W. Freischlag
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Caitlyn B. Gries
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Anit Shah
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Albert C. Pai
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Bethany A. Ahlers
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Soo Park
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Kalpaj R. Parekh
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
6
|
Human iPSC-derived-keratinocytes, a useful model to identify and explore pathological phenotype of Epidermolysis Bullosa Simplex. J Invest Dermatol 2022; 142:2695-2705.e11. [PMID: 35490743 DOI: 10.1016/j.jid.2022.04.009] [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: 07/05/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022]
Abstract
Epidermolysis Bullosa Simplex (EBS), an autosomal dominant skin disorder, is characterized by skin fragility. Genetically, majority of cases are related to missense mutations in two keratin genes, KRT5 or KRT14, leading to cytolysis of basal keratinocytes and intraepidermal blistering. Progress towards identification of treatments have been hampered by incomplete understanding of the mechanisms underlying this disease, and availability of relevant and reliable in vitro models recapitulating the physiopathological mechanisms. Recent advances in stem cell field have fueled the prospect that these limitations could be overcome thanks to the availability of disease-specific human induced pluripotent stem cells (hiPSC). Here, we generated hiPSC-derived keratinocytes from patients carrying KRT5 dominant mutations and compared them to non-affected hiPSC-derived keratinocytes as well as their primary counterparts. Our results demonstrated that EBS hiPSC-derived keratinocytes displayed proliferative defects, increased capacity to migrate, alteration of ERK signaling pathway and cytoplasmic keratin filament aggregates as observed in primary EBS keratinocytes. Of interest, EBS hiPSC-derived keratinocytes exhibited a downregulation of hemidesmosomal proteins revealing the different effects of KRT5 mutations on keratin cytoskeletal organization. Combination of culture miniaturization and treatment with the chaperone molecule 4-PBA, our results demonstrated that hiPSC-derived keratinocytes represent a suitable model for identifying novel therapies for EBS.
Collapse
|
7
|
Evtushenko NA, Beilin AK, Kosykh AV, Vorotelyak EA, Gurskaya NG. Keratins as an Inflammation Trigger Point in Epidermolysis Bullosa Simplex. Int J Mol Sci 2021; 22:ijms222212446. [PMID: 34830328 PMCID: PMC8624175 DOI: 10.3390/ijms222212446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/21/2022] Open
Abstract
Epidermolysis bullosa simplex (EBS) is a group of inherited keratinopathies that, in most cases, arise due to mutations in keratins and lead to intraepidermal ruptures. The cellular pathology of most EBS subtypes is associated with the fragility of the intermediate filament network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Mutations in keratins 5/14 or in other genes that encode associated proteins induce structural disarrangements of different strengths depending on their locations in the genes. Keratin aggregates display impaired dynamics of assembly and diminished solubility and appear to be the trigger for endoplasmic reticulum (ER) stress upon being phosphorylated by MAPKs. Global changes in cellular signaling mainly occur in cases of severe dominant EBS mutations. The spectrum of changes initiated by phosphorylation includes the inhibition of proteasome degradation, TNF-α signaling activation, deregulated proliferation, abnormal cell migration, and impaired adherence of keratinocytes. ER stress also leads to the release of proinflammatory danger-associated molecular pattern (DAMP) molecules, which enhance avalanche-like inflammation. Many instances of positive feedback in the course of cellular stress and the development of sterile inflammation led to systemic chronic inflammation in EBS. This highlights the role of keratin in the maintenance of epidermal and immune homeostasis.
Collapse
Affiliation(s)
- Nadezhda A. Evtushenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
| | - Arkadii K. Beilin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova 26, 119334 Moscow, Russia;
| | - Anastasiya V. Kosykh
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
| | - Ekaterina A. Vorotelyak
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova 26, 119334 Moscow, Russia;
| | - Nadya G. Gurskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Correspondence:
| |
Collapse
|
8
|
Seleit A, Gross K, Onistschenko J, Hoang OP, Theelke J, Centanin L. Local tissue interactions govern pLL patterning in medaka. Dev Biol 2021; 481:1-13. [PMID: 34517003 DOI: 10.1016/j.ydbio.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/12/2021] [Accepted: 09/03/2021] [Indexed: 11/03/2022]
Abstract
Vertebrate organs are arranged in a stereotypic, species-specific position along the animal body plan. Substantial morphological variation exists between related species, especially so in the vastly diversified teleost clade. It is still unclear how tissues, organs and systems can accommodate such diverse scaffolds. Here, we use the distinctive arrangement of neuromasts in the posterior lateral line (pLL) system of medaka fish to address the tissue-interactions defining a pattern. We show that patterning in this peripheral nervous system is established by autonomous organ precursors independent of neuronal wiring. In addition, we target the keratin 15 gene to generate stuck-in-the-midline (siml) mutants, which display epithelial lesions and a disrupted pLL patterning. By using siml/wt chimeras, we determine that the aberrant siml pLL pattern depends on the mutant epithelium, since a wild type epithelium can rescue the siml phenotype. Inducing epithelial lesions by 2-photon laser ablation during pLL morphogenesis phenocopies siml genetic mutants and reveals that epithelial integrity defines the final position of the embryonic pLL neuromasts. Our results using the medaka pLL disentangle intrinsic from extrinsic properties during the establishment of a sensory system. We speculate that intrinsic programs guarantee proper organ morphogenesis, while instructive interactions from surrounding tissues facilitates the accommodation of sensory organs to the diverse body plans found among teleosts.
Collapse
Affiliation(s)
- Ali Seleit
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany; Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Karen Gross
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany; Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Jasmin Onistschenko
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany; Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Oi Pui Hoang
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany
| | - Jonas Theelke
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany
| | - Lázaro Centanin
- Laboratory of Clonal Analysis of Post-Embryonic Stem Cells, Centre for Organismal Studies (COS) Heidelberg, Im Neuenheimer Feld 230, Universität Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
9
|
Deng Z, Cangkrama M, Butt T, Jane SM, Carpinelli MR. Grainyhead-like transcription factors: guardians of the skin barrier. Vet Dermatol 2021; 32:553-e152. [PMID: 33843098 DOI: 10.1111/vde.12956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023]
Abstract
There has been selective pressure to maintain a skin barrier since terrestrial animals evolved 360 million years ago. These animals acquired an unique integumentary system with a keratinized, stratified, squamous epithelium surface barrier. The barrier protects against dehydration and entry of microbes and toxins. The skin barrier centres on the stratum corneum layer of the epidermis and consists of cornified envelopes cemented by the intercorneocyte lipid matrix. Multiple components of the barrier undergo cross-linking by transglutaminase (TGM) enzymes, while keratins provide additional mechanical strength. Cellular tight junctions also are crucial for barrier integrity. The grainyhead-like (GRHL) transcription factors regulate the formation and maintenance of the integument in diverse species. GRHL3 is essential for formation of the skin barrier during embryonic development, whereas GRHL1 maintains the skin barrier postnatally. This is achieved by transactivation of Tgm1 and Tgm5, respectively. In addition to its barrier function, GRHL3 plays key roles in wound repair and as an epidermal tumour suppressor. In its former role, GRHL3 activates the planar cell polarity signalling pathway to mediate wound healing by providing directional migration cues. In squamous epithelium, GRHL3 regulates the balance between proliferation and differentiation, and its loss induces squamous cell carcinoma (SCC). In the skin, this is mediated through increased expression of MIR21, which reduces the expression levels of GRHL3 and its direct target, PTEN, leading to activation of the PI3K-AKT signalling pathway. These data position the GRHL family as master regulators of epidermal homeostasis across a vast gulf of evolutionary history.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Michael Cangkrama
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Tariq Butt
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen M Jane
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Marina R Carpinelli
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
10
|
Makita E, Matsuzaki Y, Fukui T, Matsui A, Minakawa S, Nakano H, Ito K, Kijima H, Sawamura D. Autoantibodies to BPAG1e Trigger Experimental Bullous Pemphigoid in Mice. J Invest Dermatol 2020; 141:1167-1176.e3. [PMID: 33069726 DOI: 10.1016/j.jid.2020.08.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022]
Abstract
Bullous pemphigoid (BP) is an autoimmune blistering disease that targets the hemidesmosomal proteins BP180 and BP230/BPAG1e. Whereas the role of anti-BP180 antibodies has been extensively characterized, the pathogenicity of anti-BPAG1e antibodies remains unclear. The purpose of this study is to elucidate the role of antibodies to BPAG1e in the experimental bullous pemphigoid models. We generated Bpag1 conditional knockout mice, where the knockout of Bpag1 is restricted to keratin 5-expressing epithelial cells. Bpag1 conditional knockout mice were immunized with the C-terminal portion of BPAG1e, and the splenocytes were injected into Rag2-/- mice intravenously. The recipient mice presented with erosion on the feet and tails. Microscopic examination showed subepidermal blisters and a linear deposition of IgG at the dermal-epidermal junction. To assess the potential role of trauma on BP development, we inflicted surface wounds on the dorsum of the Rag2-/- recipient mice after adoptive transfer. The wounded Rag2-/- mice had increased morbidity and severity of BP-like symptoms. Moreover, the depletion of B cells from splenocytes abolished a subepidermal blistering phenotype in vivo. These findings demonstrate that antibodies to BPAG1e might play a pathogenic role in causing subepidermal blistering, and external factors, including trauma, might be a trigger for BP development.
Collapse
Affiliation(s)
- Eiko Makita
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasushi Matsuzaki
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| | - Tomohisa Fukui
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Akinobu Matsui
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Satoko Minakawa
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hajime Nakano
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Ito
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Hiroshi Kijima
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Daisuke Sawamura
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
11
|
Kao WWY. Keratin expression by corneal and limbal stem cells during development. Exp Eye Res 2020; 200:108206. [PMID: 32882212 DOI: 10.1016/j.exer.2020.108206] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Keratins are the forming units of intermediate filaments (IF) that provide mechanical support, and formation of desmosomes between cells and hemi desmosomes with basement membranes for epithelium integrity. Keratin IF are polymers of obligate heterodimer consisting one type I keratin and one type II keratin molecules. There are 54 functional keratin genes in human genome, which are classified into three major groups, i.e., epithelial keratins, hair follicle cell-specific epithelial keratins and hair keratins. Their expression is cell type-specific and developmentally regulated. Corneal epithelium expresses a subgroup of keratins similar to those of epidermal epithelium. Limbal basal stem cells express K5/K14, and K8/K18 and K8/K19 IF suggesting that there probably are two populations of limbal stem cells (LSCs). In human, LSCs at limbal basal layer can directly stratify and differentiate to limbal suprabasal cells that express K3/K12 IF, or centripetally migrate then differentiate to corneal basal transient amplifying cells (TAC) that co-express both K3/K12 and K5/K14 prior to moving upward and assuming suprabasal cells phenotype of only K3/K12 expression that signifies corneal type epithelium differentiation. In rodent, the differentiated cornea epithelial cells express K5/K12 in lieu of K3/K12, because K3 allele exists as a pseudogene and does not encode a functional K3 protein. The basal corneal cells of new-born mice originate from surface ectoderm during embryonic development slowly commit to differentiation of becoming TAC co-expressing K5/K12 and K5/K14 IF. However, the centripetal migration may still occur at a slower rate in young mice, which is accelerated during wound healing. In this review, we will discuss and compare the cornea-specific keratins expression patterns between corneal and epidermal epithelial cells during mouse development, and between human and mouse during development and homeostasis in adult, and pathology caused by a mutation of keratins.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Departments of Ophthalmology, University of Cincinnati, Cincinnati, OH, 45267-0838, USA.
| |
Collapse
|
12
|
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
|
13
|
Johnson AL, Peterson SM, Terry MML, Ferguson B, Colgin LM, Lewis AD. Spontaneous KRT5 Gene Mutation in Rhesus Macaques ( Macaca mulatta): A Novel Nonhuman Primate Model of Epidermolysis Bullosa Simplex. Vet Pathol 2020; 57:344-348. [PMID: 32096448 DOI: 10.1177/0300985819900354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Epidermolysis bullosa simplex (EBS) is an inherited skin disorder characterized by increased skin and mucous membrane fragility. Most cases are caused by mutations in keratin 5 (KRT5) and keratin 14 (KRT14). Mutations of these genes result in cytoskeletal disruption of the basal keratinocytes. Gross and histopathologic findings of 2 clinically affected homozygous rhesus macaques with an insertion variant mutation in KRT5 are described and compared with 6 deceased phenotypically normal animals that were heterozygous for the KRT5 insertion variant. Animals that were homozygous for the KRT5 insertion variant were stillborn and had widespread loss of the epidermis. Microscopic examination confirmed severe ulceration and basal cell vacuolation with basilar vesicle formation in the remaining intact epidermis. Immunohistochemistry for cytokeratin 5 demonstrated lack of epidermal immunoreactivity in homozygotes. DNA sequencing identified a 34-base pair insertion variant in exon 5 of the KRT5 gene. To our knowledge, this is the first report of epidermolysis bullosa in rhesus macaques.
Collapse
Affiliation(s)
- Amanda L Johnson
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Samuel M Peterson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Margaret M L Terry
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Betsy Ferguson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Lois M Colgin
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Anne D Lewis
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
14
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
15
|
Tryon RK, Tolar J, Preusser SM, Riddle MJ, Keene DR, Bower M, Thyagarajan B, Ebens CL. A homozygous frameshift variant in the KRT5 gene is compatible with life and results in severe recessive epidermolysis bullosa simplex. JAAD Case Rep 2019; 5:576-579. [PMID: 31312705 PMCID: PMC6610641 DOI: 10.1016/j.jdcr.2019.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Rebecca K Tryon
- Department of Genetics, University of Minnesota, Minneapolis, Minnesota.,Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Sarah M Preusser
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Megan J Riddle
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota
| | | | - Matthew Bower
- Department of Genetics, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Christen L Ebens
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
16
|
Dmello C, Srivastava SS, Tiwari R, Chaudhari PR, Sawant S, Vaidya MM. Multifaceted role of keratins in epithelial cell differentiation and transformation. J Biosci 2019; 44:33. [PMID: 31180046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Keratins, the epithelial-predominant members of the intermediate filament superfamily, are expressed in a pairwise, tissuespecific and differentiation-dependent manner. There are 28 type I and 26 type II keratins, which share a common structure comprising a central coiled coil α-helical rod domain flanked by two nonhelical head and tail domains. These domains harbor sites for major posttranslational modifications like phosphorylation and glycosylation, which govern keratin function and dynamics. Apart from providing structural support, keratins regulate various signaling machinery involved in cell growth, motility, apoptosis etc. However, tissue-specific functions of keratins in relation to cell proliferation and differentiation are still emerging. Altered keratin expression pattern during and after malignant transformation is reported to modulate different signaling pathways involved in tumor progression in a context-dependent fashion. The current review focuses on the literature related to the role of keratins in the regulation of cell proliferation, differentiation and transformation in different types of epithelia.
Collapse
Affiliation(s)
- Crismita Dmello
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
| | | | | | | | | | | |
Collapse
|
17
|
Bhattacharjee O, Ayyangar U, Kurbet AS, Ashok D, Raghavan S. Unraveling the ECM-Immune Cell Crosstalk in Skin Diseases. Front Cell Dev Biol 2019; 7:68. [PMID: 31134198 PMCID: PMC6514232 DOI: 10.3389/fcell.2019.00068] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/09/2019] [Indexed: 01/06/2023] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and proteoglycans secreted by keratinocytes, fibroblasts and immune cells. The function of the skin ECM has expanded from being a scaffold that provides structural integrity, to a more dynamic entity that is constantly remodeled to maintain tissue homeostasis. The ECM functions as ligands for cell surface receptors such as integrins, dystroglycans, and toll-like receptors (TLRs) and regulate cellular signaling and immune cell dynamics. The ECM also acts as a sink for growth factors and cytokines, providing critical cues during epithelial morphogenesis. Dysregulation in the organization and deposition of ECMs lead to a plethora of pathophysiological conditions that are exacerbated by aberrant ECM-immune cell interactions. In this review, we focus on the interplay between ECM and immune cells in the context of skin diseases and also discuss state of the art therapies that target the key molecular players involved.
Collapse
Affiliation(s)
- Oindrila Bhattacharjee
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Uttkarsh Ayyangar
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Ambika S. Kurbet
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Driti Ashok
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Srikala Raghavan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| |
Collapse
|
18
|
The maintenance of an oral epithelial barrier. Life Sci 2019; 227:129-136. [PMID: 31002922 DOI: 10.1016/j.lfs.2019.04.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/10/2019] [Accepted: 04/14/2019] [Indexed: 02/05/2023]
Abstract
Oral epithelial barrier consists of closely controlled structure of the stratified squamous epithelium, which is the gateway to human bodies and encounters a huge burden of microbial, airborne and dietary antigens, as well as masticatory damage. Once this barrier is destroyed, it will trigger bone loss, tissue damage and microbial dysbiosis and lead to diseases, such as periodontitis, oral mucosal diseases and oral cancer. Recently, increasing evidences showed that different factors including microorganism, saliva, proteins and immune components have been considered to play a critical role in the disruption of oral epithelial barrier. Herein, we discussed mechanisms governing the maintenance of oral epithelial barrier. Besides, the role of oral epithelial barrier failure in oral carcinogenesis will also be talked about.
Collapse
|
19
|
Dmello C, Srivastava SS, Tiwari R, Chaudhari PR, Sawant S, Vaidya MM. Multifaceted role of keratins in epithelial cell differentiation and transformation. J Biosci 2019. [DOI: 10.1007/s12038-019-9864-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Khani P, Ghazi F, Zekri A, Nasri F, Behrangi E, Aghdam AM, Mirzaei H. Keratins and epidermolysis bullosa simplex. J Cell Physiol 2018; 234:289-297. [PMID: 30078200 DOI: 10.1002/jcp.26898] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/12/2018] [Indexed: 11/10/2022]
Abstract
Keratin intermediate filaments play an important role in maintaining the integrity of the skin structure. Understanding the importance of this subject is possible with the investigation of keratin defects in epidermolysis bullosa simplex (EBS). Nowadays, in addition to clinical criteria, new molecular diagnostic methods, such as next generation sequencing, can help to distinguish the subgroups of EBS more precisely. Because the most important and most commonly occurring molecular defects in these patients are the defects of keratins 5 and14 (KRT5 and KRT14), comprehending the nature structure of these proteins and their involved processes can be very effective in understanding the pathophysiology of this disease and providing new and effective therapeutic platforms to treat it. Here, we summarized the various aspects of the presence of KRT5 and KRT14 in the epidermis, their relation to the incidence and severity of EBS phenotypes, and the processes with which these proteins can affect them.
Collapse
Affiliation(s)
- Pouria Khani
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Farideh Ghazi
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ali Zekri
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Farzad Nasri
- Department of Medical Immunology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Elham Behrangi
- Department of Dermatology and Laser Surgery, Clinical Research Center, Rasoul-e-Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arad Mobasher Aghdam
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Emmerson E, Knox SM. Salivary gland stem cells: A review of development, regeneration and cancer. Genesis 2018; 56:e23211. [PMID: 29663717 PMCID: PMC5980780 DOI: 10.1002/dvg.23211] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/13/2022]
Abstract
Salivary glands are responsible for maintaining the health of the oral cavity and are routinely damaged by therapeutic radiation for head and neck cancer as well as by autoimmune diseases such as Sjögren's syndrome. Regenerative approaches based on the reactivation of endogenous stem cells or the transplant of exogenous stem cells hold substantial promise in restoring the structure and function of these organs to improve patient quality of life. However, these approaches have been hampered by a lack of knowledge on the identity of salivary stem cell populations and their regulators. In this review we discuss our current knowledge on salivary stem cells and their regulators during organ development, homeostasis and regeneration. As increasing evidence in other systems suggests that progenitor cells may be a source of cancer, we also review whether these same salivary stem cells may also be cancer initiating cells.
Collapse
Affiliation(s)
- Elaine Emmerson
- The MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sarah M. Knox
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
22
|
Gullmets J, Torvaldson E, Lindqvist J, Imanishi SY, Taimen P, Meinander A, Eriksson JE. Internal epithelia in Drosophila display rudimentary competence to form cytoplasmic networks of transgenic human vimentin. FASEB J 2017; 31:5332-5341. [PMID: 28778974 DOI: 10.1096/fj.201700332r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/25/2017] [Indexed: 11/11/2022]
Abstract
Cytoplasmic intermediate filaments (cIFs) are found in all eumetazoans, except arthropods. To investigate the compatibility of cIFs in arthropods, we expressed human vimentin (hVim), a cIF with filament-forming capacity in vertebrate cells and tissues, transgenically in Drosophila Transgenic hVim could be recovered from whole-fly lysates by using a standard procedure for intermediate filament (IF) extraction. When this procedure was used to test for the possible presence of IF-like proteins in flies, only lamins and tropomyosin were observed in IF-enriched extracts, thereby providing biochemical reinforcement to the paradigm that arthropods lack cIFs. In Drosophila, transgenic hVim was unable to form filament networks in S2 cells and mesenchymal tissues; however, cage-like vimentin structures could be observed around the nuclei in internal epithelia, which suggests that Drosophila retains selective competence for filament formation. Taken together, our results imply that although the filament network formation competence is partially lost in Drosophila, a rudimentary filament network formation ability remains in epithelial cells. As a result of the observed selective competence for cIF assembly in Drosophila, we hypothesize that internal epithelial cIFs were the last cIFs to disappear from arthropods.-Gullmets, J., Torvaldson, E., Lindqvist, J., Imanishi, S. Y., Taimen, P., Meinander, A., Eriksson, J. E. Internal epithelia in Drosophila display rudimentary competence to form cytoplasmic networks of transgenic human vimentin.
Collapse
Affiliation(s)
- Josef Gullmets
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, Turku, Finland
| | - Elin Torvaldson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Julia Lindqvist
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Susumu Y Imanishi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Pekka Taimen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, Turku, Finland
| | - Annika Meinander
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - John E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; .,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
23
|
Hatzfeld M, Keil R, Magin TM. Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics. Cold Spring Harb Perspect Biol 2017; 9:a029157. [PMID: 28096266 PMCID: PMC5453391 DOI: 10.1101/cshperspect.a029157] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adherens junctions (AJs) and desmosomes connect the actin and keratin filament networks of adjacent cells into a mechanical unit. Whereas AJs function in mechanosensing and in transducing mechanical forces between the plasma membrane and the actomyosin cytoskeleton, desmosomes and intermediate filaments (IFs) provide mechanical stability required to maintain tissue architecture and integrity when the tissues are exposed to mechanical stress. Desmosomes are essential for stable intercellular cohesion, whereas keratins determine cell mechanics but are not involved in generating tension. Here, we summarize the current knowledge of the role of IFs and desmosomes in tissue mechanics and discuss whether the desmosome-keratin scaffold might be actively involved in mechanosensing and in the conversion of chemical signals into mechanical strength.
Collapse
Affiliation(s)
- Mechthild Hatzfeld
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - René Keil
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology and Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
24
|
Tanikawa C, Zhang YZ, Yamamoto R, Tsuda Y, Tanaka M, Funauchi Y, Mori J, Imoto S, Yamaguchi R, Nakamura Y, Miyano S, Nakagawa H, Matsuda K. The Transcriptional Landscape of p53 Signalling Pathway. EBioMedicine 2017; 20:109-119. [PMID: 28558959 PMCID: PMC5478243 DOI: 10.1016/j.ebiom.2017.05.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023] Open
Abstract
Although recent cancer genomics studies have identified a large number of genes that were mutated in human cancers, p53 remains as the most frequently mutated gene. To further elucidate the p53-signalling network, we performed transcriptome analysis on 24 tissues in p53+/+ or p53−/− mice after whole-body X-ray irradiation. Here we found transactivation of a total of 3551 genes in one or more of the 24 tissues only in p53+/+ mice, while 2576 genes were downregulated. p53 mRNA expression level in each tissue was significantly associated with the number of genes upregulated by irradiation. Annotation using TCGA (The Cancer Genome Atlas) database revealed that p53 negatively regulated mRNA expression of several cancer therapeutic targets or pathways such as BTK, SYK, and CTLA4 in breast cancer tissues. In addition, stomach exhibited the induction of Krt6, Krt16, and Krt17 as well as loricrin, an epidermal differentiation marker, after the X-ray irradiation only in p53+/+ mice, implying a mechanism to protect damaged tissues by rapid induction of differentiation. Our comprehensive transcriptome analysis elucidated tissue specific roles of p53 and its signalling networks in DNA-damage response that will enhance our understanding of cancer biology. Transcriptome analysis across 24 mouse tissues identified a total of 3551 p53-induced genes and 2576 p53-repressed genes. p53 mRNA in each tissue was associated with the numbers of p53-induced/repressed genes. p53 would protect gastric epithelial cells from X-ray irradiation through the induction of differentiation and keratinization
p53 is the most frequently mutated tumor-suppressor that functions as a transcription factor. Transcriptome analysis of 280 samples from 24 tissues identified 3551 p53-induced and 2576 p53-repressed genes. p53 negatively regulated several cancer therapeutic targets, BTK, SYK, and CTLA4 in breast cancer tissues. Majority of X-ray responsive genes were regulated by p53 and p53 mRNA expression in each tissue was associated with the numbers of p53-induced and repressed genes, demonstrating its crucial roles in damage response. The whole-body transcriptome analysis revealed that p53 would protect gastric epithelial cells from X-ray irradiation through the induction of differentiation and keratinization.
Collapse
Affiliation(s)
- Chizu Tanikawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yao-Zhong Zhang
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ryuta Yamamoto
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yusuke Tsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masami Tanaka
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yuki Funauchi
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Jinichi Mori
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Laboratory of Clinical Genome Sequence, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Rui Yamaguchi
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yusuke Nakamura
- Department of Medicine, Center for Personalized Therapeutics, The University of Chicago, USA; Department of Surgery, Center for Personalized Therapeutics, The University of Chicago, USA
| | - Satoru Miyano
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hidewaki Nakagawa
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Laboratory of Clinical Genome Sequence, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
25
|
Lee B, Watanabe K, Haensel D, Sui JY, Dai X. Overexpression of Transcription Factor Ovol2 in Epidermal Progenitor Cells Results in Skin Blistering. J Invest Dermatol 2017; 137:1805-1808. [PMID: 28457910 DOI: 10.1016/j.jid.2017.02.985] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Briana Lee
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Kazuhide Watanabe
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Jennifer Y Sui
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA.
| |
Collapse
|
26
|
|
27
|
Coulombe PA. Discovery of keratin function and role in genetic diseases: the year that 1991 was. Mol Biol Cell 2016; 27:2807-10. [PMID: 27634744 PMCID: PMC5025267 DOI: 10.1091/mbc.e15-09-0625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 01/27/2023] Open
Abstract
In 1991, a set of transgenic mouse studies took the fields of cell biology and dermatology by storm in providing the first credible evidence that keratin intermediate filaments play a unique and essential role in the structural and mechanical support in keratinocytes of the epidermis. Moreover, these studies intimated that mutations altering the primary structure and function of keratin filaments underlie genetic diseases typified by cellular fragility. This Retrospective on how these studies came to be is offered as a means to highlight the 25th anniversary of these discoveries.
Collapse
Affiliation(s)
- Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Departments of Biological Chemistry, Oncology, and Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
28
|
Allen EHA, Courtney DG, Atkinson SD, Moore JE, Mairs L, Poulsen ET, Schiroli D, Maurizi E, Cole C, Hickerson RP, James J, Murgatroyd H, Smith FJD, MacEwen C, Enghild JJ, Nesbit MA, Leslie Pedrioli DM, McLean WHI, Moore CBT. Keratin 12 missense mutation induces the unfolded protein response and apoptosis in Meesmann epithelial corneal dystrophy. Hum Mol Genet 2016; 25:1176-91. [PMID: 26758872 PMCID: PMC4764196 DOI: 10.1093/hmg/ddw001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/23/2022] Open
Abstract
Meesmann epithelial corneal dystrophy (MECD) is a rare autosomal dominant disorder caused by dominant-negative mutations within the KRT3 or KRT12 genes, which encode the cytoskeletal protein keratins K3 and K12, respectively. To investigate the pathomechanism of this disease, we generated and phenotypically characterized a novel knock-in humanized mouse model carrying the severe, MECD-associated, K12-Leu132Pro mutation. Although no overt changes in corneal opacity were detected by slit-lamp examination, the corneas of homozygous mutant mice exhibited histological and ultrastructural epithelial cell fragility phenotypes. An altered keratin expression profile was observed in the cornea of mutant mice, confirmed by western blot, RNA-seq and quantitative real-time polymerase chain reaction. Mass spectrometry (MS) and immunohistochemistry demonstrated a similarly altered keratin profile in corneal tissue from a K12-Leu132Pro MECD patient. The K12-Leu132Pro mutation results in cytoplasmic keratin aggregates. RNA-seq analysis revealed increased chaperone gene expression, and apoptotic unfolded protein response (UPR) markers, CHOP and Caspase 12, were also increased in the MECD mice. Corneal epithelial cell apoptosis was increased 17-fold in the mutant cornea, compared with the wild-type (P < 0.001). This elevation of UPR marker expression was also observed in the human MECD cornea. This is the first reporting of a mouse model for MECD that recapitulates the human disease and is a valuable resource in understanding the pathomechanism of the disease. Although the most severe phenotype is observed in the homozygous mice, this model will still provide a test-bed for therapies not only for corneal dystrophies but also for other keratinopathies caused by similar mutations.
Collapse
Affiliation(s)
- Edwin H A Allen
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK, Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - David G Courtney
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | - Sarah D Atkinson
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | - Johnny E Moore
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK, Cathedral Eye Clinic, Academy Street, Belfast BT15 1ED, UK
| | - Laura Mairs
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | | | - Davide Schiroli
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | - Eleonora Maurizi
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | - Christian Cole
- Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - Robyn P Hickerson
- Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - John James
- Microscopy Facility, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Helen Murgatroyd
- Department of Ophthalmology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Frances J D Smith
- Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - Carrie MacEwen
- Department of Ophthalmology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Science Park, Aarhus University, Aarhus, Denmark and
| | - M Andrew Nesbit
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK
| | - Deena M Leslie Pedrioli
- Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - W H Irwin McLean
- Centre for Dermatology and Genetic Medicine, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK,
| | - C B Tara Moore
- School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, Northern Ireland, UK,
| |
Collapse
|
29
|
Tan TS, Ng YZ, Badowski C, Dang T, Common JEA, Lacina L, Szeverényi I, Lane EB. Assays to Study Consequences of Cytoplasmic Intermediate Filament Mutations: The Case of Epidermal Keratins. Methods Enzymol 2016; 568:219-53. [PMID: 26795473 DOI: 10.1016/bs.mie.2015.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
The discovery of the causative link between keratin mutations and a growing number of human diseases opened the way for a better understanding of the function of the whole intermediate filament families of cytoskeleton proteins. This chapter describes analytical approaches to identification and interpretation of the consequences of keratin mutations, from the clinical and diagnostic level to cells in tissue culture. Intermediate filament pathologies can be accurately diagnosed from skin biopsies and DNA samples. The Human Intermediate Filament Database collates reported mutations in intermediate filament genes and their diseases, and can help clinicians to establish accurate diagnoses, leading to disease stratification for genetic counseling, optimal care delivery, and future mutation-aligned new therapies. Looking at the best-studied keratinopathy, epidermolysis bullosa simplex, the generation of cell lines mimicking keratinopathies is described, in which tagged mutant keratins facilitate live-cell imaging to make use of today's powerful enhanced light microscopy modalities. Cell stress assays such as cell spreading and cell migration in scratch wound assays can interrogate the consequences of the compromised cytoskeletal network. Application of extrinsic stresses, such as heat, osmotic, or mechanical stress, can enhance the differentiation of mutant keratin cells from wild-type cells. To bring the experiments to the next level, 3D organotypic human cultures can be generated, and even grafted onto the backs of immunodeficient mice for greater in vivo relevance. While development of these assays has focused on mutant K5/K14 cells, the approaches are often applicable to mutations in other intermediate filaments, reinforcing fundamental commonalities in spite of diverse clinical pathologies.
Collapse
Affiliation(s)
| | | | | | - Tram Dang
- Institute of Medical Biology, Singapore
| | | | | | | | | |
Collapse
|
30
|
The p.Glu477Lys Mutation in Keratin 5 Is Strongly Associated with Mortality in Generalized Severe Epidermolysis Bullosa Simplex. J Invest Dermatol 2015; 136:719-721. [PMID: 26743602 DOI: 10.1016/j.jid.2015.11.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/17/2015] [Accepted: 11/30/2015] [Indexed: 11/23/2022]
|
31
|
Alvarado DM, Coulombe PA. Directed expression of a chimeric type II keratin partially rescues keratin 5-null mice. J Biol Chem 2014; 289:19435-47. [PMID: 24867950 DOI: 10.1074/jbc.m114.553867] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The crucial role of structural support fulfilled by keratin intermediate filaments (IFs) in surface epithelia likely requires that they be organized into cross-linked networks. For IFs comprised of keratins 5 and 14 (K5 and K14), which occur in basal keratinocytes of the epidermis, formation of cross-linked bundles is, in part, self-driven through cis-acting determinants. Here, we targeted the expression of a bundling-competent KRT5/KRT8 chimeric cDNA (KRT8bc) or bundling-deficient wild type KRT8 as a control to the epidermal basal layer of Krt5-null mice to assess the functional importance of keratin IF self-organization in vivo. Such targeted expression of K8bc rescued Krt5-null mice with a 47% frequency, whereas K8 completely failed to do so. This outcome correlated with lower than expected levels of K8bc and especially K8 mRNA and protein in the epidermis of E18.5 replacement embryos. Ex vivo culture of embryonic skin keratinocytes confirmed the ability of K8bc to form IFs in the absence of K5. Additionally, electron microscopy analysis of E18.5 embryonic skin revealed that the striking defects observed in keratin IF bundling, cytoarchitecture, and mitochondria are partially restored by K8bc expression. As young adults, viable KRT8bc replacement mice develop alopecia and chronic skin lesions, indicating that the skin epithelia are not completely normal. These findings are consistent with a contribution of self-mediated organization of keratin IFs to structural support and cytoarchitecture in basal layer keratinocytes of the epidermis and underscore the importance of context-dependent regulation for keratin genes and proteins in vivo.
Collapse
Affiliation(s)
- David M Alvarado
- From the Training Program in Cellular and Molecular Medicine and Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Pierre A Coulombe
- From the Training Program in Cellular and Molecular Medicine and Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland 21205Departments of Biological Chemistry and Dermatology, School of Medicine and
| |
Collapse
|
32
|
Kitagawa N, Inai Y, Higuchi Y, Iida H, Inai T. Inhibition of JNK in HaCaT cells induced tight junction formation with decreased expression of cytokeratin 5, cytokeratin 17 and desmoglein 3. Histochem Cell Biol 2014; 142:389-99. [DOI: 10.1007/s00418-014-1219-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2014] [Indexed: 11/24/2022]
|
33
|
Skin Fragility and Impaired Desmosomal Adhesion in Mice Lacking All Keratins. J Invest Dermatol 2014; 134:1012-1022. [DOI: 10.1038/jid.2013.416] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/27/2013] [Accepted: 09/12/2013] [Indexed: 12/22/2022]
|
34
|
Beyond expectations: novel insights into epidermal keratin function and regulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 311:265-306. [PMID: 24952920 DOI: 10.1016/b978-0-12-800179-0.00007-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The epidermis is a stratified epithelium that relies on its cytoskeleton and cell junctions to protect the body against mechanical injury, dehydration, and infections. Keratin intermediate filament proteins are involved in many of these functions by forming cell-specific cytoskeletal scaffolds crucial for the maintenance of cell and tissue integrity. In response to various stresses, the expression and organization of keratins are altered at transcriptional and posttranslational levels to restore tissue homeostasis. Failure to restore tissue homeostasis in the presence of keratin gene mutations results in acute and chronic skin disorders for which currently no rational therapies are available. Here, we review the recent progress on the role of keratins in cytoarchitecture, adhesion, signaling, and inflammation. By focusing on epidermal keratins, we illustrate the contribution of keratin isotypes to differentiated epithelial functions.
Collapse
|
35
|
Tai G, Ranjzad P, Marriage F, Rehman S, Denley H, Dixon J, Mitchell K, Day PJR, Woolf AS. Cytokeratin 15 marks basal epithelia in developing ureters and is upregulated in a subset of urothelial cell carcinomas. PLoS One 2013; 8:e81167. [PMID: 24260555 PMCID: PMC3832456 DOI: 10.1371/journal.pone.0081167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/09/2013] [Indexed: 11/29/2022] Open
Abstract
The mammalian ureter contains a water-tight epithelium surrounded by smooth muscle. Key molecules have been defined which regulate ureteric bud initiation and drive the differentiation of ureteric mesenchyme into peristaltic smooth muscle. Less is known about mechanisms underlying the developmental patterning of the multilayered epithelium characterising the mature ureter. In skin, which also contains a multilayered epithelium, cytokeratin 15 (CK15), an acidic intermediate filament protein, marks cells whose progeny contribute to epidermal regeneration following wounding. Moreover, CK15+ precursor cells in skin can give rise to basal cell carcinomas. In the current study, using transcriptome microarrays of embryonic wild type mouse ureters, Krt15, coding for CK15, was detected. Quantitative polymerase chain reaction analyses confirmed the initial finding and demonstrated that Krt15 levels increased during the fetal period when the ureteric epithelium becomes multilayered. CK15 protein was undetectable in the ureteric bud, the rudiment from which the ureter grows. Nevertheless, later in fetal development, CK15 was immunodetected in a subset of basal urothelial cells in the ureteric stalk. Superficial epithelial cells, including those positive for the differentiation marker uroplakin III, were CK15-. Transformation-related protein 63 (P63) has been implicated in epithelial differentiation in murine fetal urinary bladders. In wild type fetal ureters, CK15+ cells were positive for P63, and p63 homozygous null mutant ureters lacked CK15+ cells. In these mutant ureters, sections of the urothelium were monolayered versus the uniform multilayering found in wild type littermates. Human urothelial cell carcinomas account for considerable morbidity and mortality. CK15 was upregulated in a subset of invasive ureteric and urinary bladder cancers. Thus, in ureter development, the absence of CK15 is associated with a structurally simplified urothelium whereas, postnatally, increased CK15 levels feature in malignant urothelial overgrowth. CK15 may be a novel marker for urinary tract epithelial precursor cells.
Collapse
Affiliation(s)
- Guangping Tai
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom ; Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Keratins as the main component for the mechanical integrity of keratinocytes. Proc Natl Acad Sci U S A 2013; 110:18513-8. [PMID: 24167246 DOI: 10.1073/pnas.1313491110] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Keratins are major components of the epithelial cytoskeleton and are believed to play a vital role for mechanical integrity at the cellular and tissue level. Keratinocytes as the main cell type of the epidermis express a differentiation-specific set of type I and type II keratins forming a stable network and are major contributors of keratinocyte mechanical properties. However, owing to compensatory keratin expression, the overall contribution of keratins to cell mechanics was difficult to examine in vivo on deletion of single keratin genes. To overcome this problem, we used keratinocytes lacking all keratins. The mechanical properties of these cells were analyzed by atomic force microscopy (AFM) and magnetic tweezers experiments. We found a strong and highly significant softening of keratin-deficient keratinocytes when analyzed by AFM on the cell body and above the nucleus. Magnetic tweezers experiments fully confirmed these results showing, in addition, high viscous contributions to magnetic bead displacement in keratin-lacking cells. Keratin loss neither affected actin or microtubule networks nor their overall protein concentration. Furthermore, depolymerization of actin preserves cell softening in the absence of keratin. On reexpression of the sole basal epidermal keratin pair K5/14, the keratin filament network was reestablished, and mechanical properties were restored almost to WT levels in both experimental setups. The data presented here demonstrate the importance of keratin filaments for mechanical resilience of keratinocytes and indicate that expression of a single keratin pair is sufficient for almost complete reconstitution of their mechanical properties.
Collapse
|
37
|
Bose A, Teh MT, Mackenzie IC, Waseem A. Keratin k15 as a biomarker of epidermal stem cells. Int J Mol Sci 2013; 14:19385-98. [PMID: 24071939 PMCID: PMC3821562 DOI: 10.3390/ijms141019385] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/05/2013] [Accepted: 09/10/2013] [Indexed: 01/21/2023] Open
Abstract
Keratin 15 (K15) is type I keratin protein co-expressed with the K5/K14 pair present in the basal keratinocytes of all stratified epithelia. Although it is a minor component of the cytoskeleton with a variable expression pattern, nonetheless its expression has been reported as a stem cell marker in the bulge of hair follicles. Conversely, suprabasal expression of K15 has also been reported in both normal and diseased tissues, which is inconsistent with its role as a stem cell marker. Our recently published work has given evidence of the molecular pathways that seem to control the expression of K15 in undifferentiated and differentiated cells. In this article, we have critically reviewed the published work to establish the reliability of K15 as an epidermal stem cell marker.
Collapse
Affiliation(s)
- Amrita Bose
- Centre for Clinical and Diagnostic Oral Sciences, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK.
| | | | | | | |
Collapse
|
38
|
Keratin 9 is required for the structural integrity and terminal differentiation of the palmoplantar epidermis. J Invest Dermatol 2013; 134:754-763. [PMID: 23962810 PMCID: PMC3923277 DOI: 10.1038/jid.2013.356] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 06/21/2013] [Accepted: 06/27/2013] [Indexed: 11/12/2022]
Abstract
Keratin 9 (K9) is a type I intermediate filament protein whose expression is confined to the suprabasal layers of the palmoplantar epidermis. Although mutations in the K9 gene are known to cause epidermolytic palmoplantar keratoderma, a rare dominant-negative skin disorder, its functional significance is poorly understood. To gain insight into the physical requirement and importance of K9, we generated K9-deficient (Krt9−/−) mice. Here, we report that adult Krt9−/−mice develop calluses marked by hyperpigmentation that are exclusively localized to the stress-bearing footpads. Histological, immunohistochemical, and immunoblot analyses of these regions revealed hyperproliferation, impaired terminal differentiation, and abnormal expression of keratins K5, K14, and K2. Furthermore, the absence of K9 induces the stress-activated keratins K6 and K16. Importantly, mice heterozygous for the K9-null allele (Krt9+/−) show neither an overt nor histological phenotype, demonstrating that one Krt9 allele is sufficient for the developing normal palmoplantar epidermis. Together, our data demonstrate that complete ablation of K9 is not tolerable in vivo and that K9 is required for terminal differentiation and maintaining the mechanical integrity of palmoplantar epidermis.
Collapse
|
39
|
Pan X, Hobbs RP, Coulombe PA. The expanding significance of keratin intermediate filaments in normal and diseased epithelia. Curr Opin Cell Biol 2013; 25:47-56. [PMID: 23270662 PMCID: PMC3578078 DOI: 10.1016/j.ceb.2012.10.018] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/31/2012] [Accepted: 10/31/2012] [Indexed: 12/17/2022]
Abstract
Intermediate filaments are assembled from a diverse group of evolutionary conserved proteins and are specified in a tissue-dependent, cell type-dependent, and context-dependent fashion in the body. Genetic mutations in intermediate filament proteins account for a large number of diseases, ranging from skin fragility conditions to cardiomyopathies and premature aging. Keratins, the epithelial-specific intermediate filaments, are now recognized as multi-faceted effectors in their native context. In this review, we emphasize the recent progress made in defining the role of keratins towards the regulation of cytoarchitecture, cell growth and proliferation, apoptosis, and cell motility during embryonic development, in normal adult tissues, and in select diseases such as cancer.
Collapse
Affiliation(s)
- Xiaoou Pan
- Dept. of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Ryan P. Hobbs
- Dept. of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Pierre A. Coulombe
- Dept. of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
40
|
Seltmann K, Roth W, Kröger C, Loschke F, Lederer M, Hüttelmaier S, Magin TM. Keratins mediate localization of hemidesmosomes and repress cell motility. J Invest Dermatol 2012; 133:181-90. [PMID: 22895363 DOI: 10.1038/jid.2012.256] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The keratin (K)-hemidesmosome (HD) interaction is crucial for cell-matrix adhesion and migration in several epithelia, including the epidermis. Mutations in constituent proteins cause severe blistering skin disorders by disrupting the adhesion complex. Despite extensive studies, the role of keratins in HD assembly and maintenance is only partially understood. Here we address this issue in keratinocytes in which all keratins are depleted by genome engineering. Unexpectedly, such keratinocytes maintain many characteristics of their normal counterparts. However, the absence of the entire keratin cytoskeleton leads to loss of plectin from the hemidesmosomal plaque and scattering of the HD transmembrane core along the basement membrane zone. To investigate the functional consequences, we performed migration and adhesion assays. These revealed that, in the absence of keratins, keratinocytes adhere much faster to extracellular matrix substrates and migrate approximately two times faster compared with wild-type cells. Reexpression of the single keratin pair K5 and K14 fully reversed the above phenotype. Our data uncover a role of keratins, which to our knowledge is previously unreported, in the maintenance of HDs upstream of plectin, with implications for epidermal homeostasis and pathogenesis. They support the view that the downregulation of keratins observed during epithelial-mesenchymal transition supports the migratory and invasive behavior of tumor cells.
Collapse
|
41
|
Menon GK, Cleary GW, Lane ME. The structure and function of the stratum corneum. Int J Pharm 2012; 435:3-9. [PMID: 22705878 DOI: 10.1016/j.ijpharm.2012.06.005] [Citation(s) in RCA: 254] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 06/01/2012] [Indexed: 01/04/2023]
Abstract
Over the past 150 years the skin's structure and function has been the subject of much investigation by scientists. The stratum corneum (SC), the skin's outermost layer and interface with the outside world is now well recognized as the barrier that prevents unwanted materials from entering, and excessive loss of water from exiting the body. This review summarizes the major advances in our understanding of this formidable membrane. The structure of the SC is outlined as well as techniques to visualize the barrier. The lipid organization and ionic gradients, as well as the metabolic responses and underlying cellular signalling that lead to barrier repair and homeostasis are discussed. Finally, a brief overview of the molecular and genetic factors that determine the development of a competent permeability barrier is provided.
Collapse
|
42
|
Thieu K, Ruiz ME, Owens DM. Cells of origin and tumor-initiating cells for nonmelanoma skin cancers. Cancer Lett 2012; 338:82-8. [PMID: 22579650 DOI: 10.1016/j.canlet.2012.05.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 04/26/2012] [Accepted: 05/03/2012] [Indexed: 01/01/2023]
Abstract
The epidermis of the skin is a multilayered stratified epithelium whose primary function is to provide a barrier against our external environment. As a result, cells in the epidermis are subject to constant assault from environmental pathogens, many of which can cause deleterious mutations. However, most of these mutations do not lead to skin cancer. One explanation is that most genetic hits are sustained by mature or transit cells with limited proliferative capacity and only stem cells that acquire genetic alterations have the potential to propagate a frank tumor. In this mini-review we will discuss recent studies that provide some of the first genetic evidence to support a stem cell origin for a number of skin cancer types.
Collapse
Affiliation(s)
- Khanh Thieu
- Department of Dermatology, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | |
Collapse
|
43
|
Abstract
The intermediate filament keratin 15 (K15) is present in variable amounts in various stratified epithelia, but has also been reported to be a stem cell marker in the hair follicle. Using peptide specific antibodies, we evaluated the temporal and spatial distribution pattern of K15 expression/localization during normal epidermal development and initiation of hair follicle formation, and in the injured mature epidermis (e.g., during acute injury and repair and in tumorigenesis). During development, K15 expression is first localized to a subset of epidermal basal cells and the overlying periderm at E12.5, but its expression is seen throughout the basal layer by E15.5 and beyond. In hair follicle morphogenesis, initial peg formation occurs in a K15-null area at E14.5 and as peg elongation proceeds through to the mature hair follicle, K15 expression follows the leading edge with positive cells restricted to the outer root sheath. In an epidermal injury model, K15 is first up-regulated and associated with both the basal and suprabasal layers of the interfollicular epidermis then expression becomes sporadic and down-regulated before a basal layer-specific association is re-established in the repaired epidermis. During tumorigenesis, K15 is first mis-expressed, and is ultimately down-regulated. Our data suggest that K15 protein expression may reflect not only expression in a stem or progenitor cell subpopulation, but also reflects the activity and responsiveness of basal-like cells to loss of homeostasis of the epidermal differentiation program. Thus, the data suggest caution in using K15 alone to delineate epidermal stem cells, and underscore the need for further investigation of K15 and other markers in epidermal cell subpopulations.
Collapse
Affiliation(s)
- Tammy-Claire Troy
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research at the Ottawa Hospital Research Institute, 501 Smyth Road-CCW5226, Ottawa, Ontario K1Y 8L6, Canada.
| | | | | |
Collapse
|
44
|
Coulombe PA, Lee CH. Defining keratin protein function in skin epithelia: epidermolysis bullosa simplex and its aftermath. J Invest Dermatol 2012; 132:763-75. [PMID: 22277943 PMCID: PMC3279600 DOI: 10.1038/jid.2011.450] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Epidermolysis bullosa simplex (EBS) is a rare genetic condition typified by superficial bullous lesions following incident frictional trauma to the skin. Most cases of EBS are due to dominantly acting mutations in keratin 14 (K14) or K5, the type I and II intermediate filament (IF) proteins that copolymerize to form a pancytoplasmic network of 10 nm filaments in basal keratinocytes of epidermis and related epithelia. Defects in K5-K14 filament network architecture cause basal keratinocytes to become fragile, and account for their rupture upon exposure to mechanical trauma. The discovery of the etiology and pathophysiology of EBS was intimately linked to the quest for an understanding of the properties and function of keratin filaments in skin epithelia. Since then, continued cross-fertilization between basic science efforts and clinical endeavors has highlighted several additional functional roles for keratin proteins in the skin, suggested new avenues for effective therapies for keratin-based diseases, and expanded our understanding of the remarkable properties of the skin as an organ system.
Collapse
Affiliation(s)
- Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
45
|
Wallace L, Roberts-Thompson L, Reichelt J. Deletion of K1/K10 does not impair epidermal stratification but affects desmosomal structure and nuclear integrity. J Cell Sci 2012; 125:1750-8. [PMID: 22375063 DOI: 10.1242/jcs.097139] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Keratins K1 and K10 are the most abundant proteins in the upper epidermis where they polymerize to form intermediate filaments (IFs). In addition to their well-established function in providing epidermal stability, K1/K10 (i.e. the dimer between K1 and K10) IFs are supposed to be important for terminal epidermal differentiation and barrier formation. It was previously shown that the imbalanced deletion of one of the partner keratins, K10, disturbed epidermal homoeostasis, although stability was provided by compensatory upregulation of K5/K14, which formed IFs together with the remaining K1. Here, we show that deletion of both partner keratins, K1 and K10, results in lethal postnatal skin fragility in mice. Krt1(-/-);Krt10(-/-) mice revealed that K1/K10 IFs are unexpectedly dispensable for epidermal stratification. Although the stratum corneum was less compact and cornified envelope differentiation was impaired, a dye exclusion assay showed that the development of a functional water barrier was surprisingly independent from the presence of K1/K10 IFs. The deletion of K1/K10 was not compensated by any other keratin pair such as the basal epidermal keratins K5/K14, and electron microscopy revealed total absence of IFs in the suprabasal epidermis. Although plakoglobin was unchanged, the expression of the desmosomal proteins desmoplakin, desmocollin 1 and desmoglein 1 were altered and suprabasal desmosomes were smaller in Krt1(-/-);Krt10(-/-) than in wild-type epidermis suggesting an involvement of K1/K10 IFs in desmosome dynamics. Furthermore, Krt1(-/-);Krt10(-/-) mice showed premature loss of nuclei during epidermal differentiation and lower levels of emerin, lamin A/C and Sun1, revealing a previously unknown function for IFs in maintaining nuclear integrity in the upper epidermis.
Collapse
Affiliation(s)
- Lee Wallace
- Institute of Cellular Medicine and North East England Stem Cell Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | | |
Collapse
|
46
|
Beriault DR, Haddad O, McCuaig JV, Robinson ZJ, Russell D, Lane EB, Fudge DS. The mechanical behavior of mutant K14-R125P keratin bundles and networks in NEB-1 keratinocytes. PLoS One 2012; 7:e31320. [PMID: 22363617 PMCID: PMC3283645 DOI: 10.1371/journal.pone.0031320] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 01/05/2012] [Indexed: 01/27/2023] Open
Abstract
Epidermolysis bullosa simplex (EBS) is an inherited skin-blistering disease that is caused by dominant mutations in the genes for keratin K5 or K14 proteins. While the link between keratin mutations and keratinocyte fragility in EBS patients is clear, the exact biophysical mechanisms underlying cell fragility are not known. In this study, we tested the hypotheses that mutant K14-R125P filaments and/or networks in human keratinocytes are mechanically defective in their response to large-scale deformations. We found that mutant filaments and networks exhibit no obvious defects when subjected to large uniaxial strains and have no negative effects on the ability of human keratinocytes to survive large strains. We also found that the expression of mutant K14-R125P protein has no effect on the morphology of the F-actin or microtubule networks or their responses to large strains. Disassembly of the F-actin network with Latrunculin A unexpectedly led to a marked decrease in stretch-induced necrosis in both WT and mutant cells. Overall, our results contradict the hypotheses that EBS mutant keratin filaments and/or networks are mechanically defective. We suggest that future studies should test the alternative hypothesis that keratinocytes in EBS cells are fragile because they possess a sparser keratin network.
Collapse
Affiliation(s)
| | - Oualid Haddad
- Department of Integrative Biology, University of Guelph, Guelph, Canada
| | - John V. McCuaig
- Department of Integrative Biology, University of Guelph, Guelph, Canada
| | | | - David Russell
- Cancer Research United Kingdom (UK) Cell Structure Research Group, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - E. Birgitte Lane
- Cancer Research United Kingdom (UK) Cell Structure Research Group, College of Life Sciences, University of Dundee, Dundee, Scotland
- Institute of Medical Biology, Singapore, Singapore
| | - Douglas S. Fudge
- Department of Integrative Biology, University of Guelph, Guelph, Canada
- * E-mail:
| |
Collapse
|
47
|
Alam H, Sehgal L, Kundu ST, Dalal SN, Vaidya MM. Novel function of keratins 5 and 14 in proliferation and differentiation of stratified epithelial cells. Mol Biol Cell 2011; 22:4068-78. [PMID: 21900500 PMCID: PMC3204069 DOI: 10.1091/mbc.e10-08-0703] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Keratin expression in stratified epithelia is tightly regulated during squamous cell differentiation. Keratins 5 and 14 are expressed in mitotically active basal layer cells, but their function is not well defined. Reported here is the possible role of K14 in regulation of cell proliferation/differentiation in stratified epithelial cells. Keratins are cytoplasmic intermediate filament proteins preferentially expressed by epithelial tissues in a site-specific and differentiation-dependent manner. The complex network of keratin filaments in stratified epithelia is tightly regulated during squamous cell differentiation. Keratin 14 (K14) is expressed in mitotically active basal layer cells, along with its partner keratin 5 (K5), and their expression is down-regulated as cells differentiate. Apart from the cytoprotective functions of K14, very little is known about K14 regulatory functions, since the K14 knockout mice show postnatal lethality. In this study, K14 expression was inhibited using RNA interference in cell lines derived from stratified epithelia to study the K14 functions in epithelial homeostasis. The K14 knockdown clones demonstrated substantial decreases in the levels of the K14 partner K5. These cells showed reduction in cell proliferation and delay in cell cycle progression, along with decreased phosphorylated Akt levels. K14 knockdown cells also exhibited enhanced levels of activated Notch1, involucrin, and K1. In addition, K14 knockdown AW13516 cells showed significant reduction in tumorigenicity. Our results suggest that K5 and K14 may have a role in maintenance of cell proliferation potential in the basal layer of stratified epithelia, modulating phosphatidylinositol 3-kinase/Akt–mediated cell proliferation and/or Notch1-dependent cell differentiation.
Collapse
Affiliation(s)
- Hunain Alam
- Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | | | | | | | | |
Collapse
|
48
|
Abstract
The development of multicellular organisms, as well as maintenance of organ architecture and function, requires robust regulation of cell fates. This is in part achieved by conserved signaling pathways through which cells process extracellular information and translate this information into changes in proliferation, differentiation, migration, and cell shape. Gene deletion studies in higher eukaryotes have assigned critical roles for components of the extracellular matrix (ECM) and their cellular receptors in a vast number of developmental processes, indicating that a large proportion of this signaling is regulated by cell-ECM interactions. In addition, genetic alterations in components of this signaling axis play causative roles in several human diseases. This review will discuss what genetic analyses in mice and lower organisms have taught us about adhesion signaling in development and disease.
Collapse
Affiliation(s)
- Sara A Wickström
- Paul Gerson Una Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50937 Cologne, Germany.
| | | | | |
Collapse
|
49
|
Abstract
Keratins are the intermediate filament (IF)-forming proteins of epithelial cells. Since their initial characterization almost 30 years ago, the total number of mammalian keratins has increased to 54, including 28 type I and 26 type II keratins. Keratins are obligate heteropolymers and, similarly to other IFs, they contain a dimeric central α-helical rod domain that is flanked by non-helical head and tail domains. The 10-nm keratin filaments participate in the formation of a proteinaceous structural framework within the cellular cytoplasm and, as such, serve an important role in epithelial cell protection from mechanical and non-mechanical stressors, a property extensively substantiated by the discovery of human keratin mutations predisposing to tissue-specific injury and by studies in keratin knockout and transgenic mice. More recently, keratins have also been recognized as regulators of other cellular properties and functions, including apico-basal polarization, motility, cell size, protein synthesis and membrane traffic and signaling. In cancer, keratins are extensively used as diagnostic tumor markers, as epithelial malignancies largely maintain the specific keratin patterns associated with their respective cells of origin, and, in many occasions, full-length or cleaved keratin expression (or lack there of) in tumors and/or peripheral blood carries prognostic significance for cancer patients. Quite intriguingly, several studies have provided evidence for active keratin involvement in cancer cell invasion and metastasis, as well as in treatment responsiveness, and have set the foundation for further exploration of the role of keratins as multifunctional regulators of epithelial tumorigenesis.
Collapse
Affiliation(s)
- V Karantza
- Department of Medicine, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| |
Collapse
|
50
|
Knox S, Lombaert I, Reed X, Vitale-Cross L, Gutkind J, Hoffman M. Parasympathetic innervation maintains epithelial progenitor cells during salivary organogenesis. Science 2010; 329:1645-7. [PMID: 20929848 PMCID: PMC3376907 DOI: 10.1126/science.1192046] [Citation(s) in RCA: 243] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The maintenance of a progenitor cell population as a reservoir of undifferentiated cells is required for organ development and regeneration. However, the mechanisms by which epithelial progenitor cells are maintained during organogenesis are poorly understood. We report that removal of the parasympathetic ganglion in mouse explant organ culture decreased the number and morphogenesis of keratin 5-positive epithelial progenitor cells. These effects were rescued with an acetylcholine analog. We demonstrate that acetylcholine signaling, via the muscarinic M1 receptor and epidermal growth factor receptor, increased epithelial morphogenesis and proliferation of the keratin 5-positive progenitor cells. Parasympathetic innervation maintained the epithelial progenitor cell population in an undifferentiated state, which was required for organogenesis. This mechanism for epithelial progenitor cell maintenance may be targeted for organ repair or regeneration.
Collapse
Affiliation(s)
| | | | - X. Reed
- Matrix and Morphogenesis Unit, LCDB
| | - L Vitale-Cross
- OPCB, NIDCR, NIH, 30 Convent Dr, Bethesda, MD 20892, USA
| | - J.S. Gutkind
- OPCB, NIDCR, NIH, 30 Convent Dr, Bethesda, MD 20892, USA
| | | |
Collapse
|