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
|
Malak M, Qian C, James J, Nair S, Grantham J, Ericson MB. Insights into metabolic changes during epidermal differentiation as revealed by multiphoton microscopy with fluorescence lifetime imaging. Sci Rep 2025; 15:6377. [PMID: 39984626 PMCID: PMC11845624 DOI: 10.1038/s41598-025-90101-4] [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: 06/11/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
Rapid developments in the field of organotypic cultures have generated a growing need for effective and non-invasive methods for quality control during tissue development. In this study, we correlate metabolic changes with epidermal differentiation and demonstrate that multiphoton microscopy with fluorescence lifetime imaging (MPM-FLIM) can be applied to monitor epidermal differentiation of keratinocytes with respect to proliferative and differentiated states. In a 2D keratinocyte tissue culture model, increased expression of differentiation markers keratin-1 and keratin-10 was induced with calcium supplementation. An accompanying shift from glycolysis to mitochondrial respiration was detected in metabolic flux assays. Analysis of MPM-FLIM images acquired at 750 nm and 900 nm excitation revealed a decreased relative fraction of intracellular NADH and FAD after high calcium treatment, consistent with increased oxidative phosphorylation. Epidermal differentiation could be monitored over a 96 h period. Discrimination analysis based on k-means clustering generated clusters that correlated well with the duration of high Ca2+ treatment, suggesting that MPM-FLIM can provide useful parameters for monitoring keratinocyte differentiation.
Collapse
Affiliation(s)
- Monika Malak
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Chen Qian
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden.
| | - Jeemol James
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Syam Nair
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
- Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 416 85, Sweden
| | - Julie Grantham
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Marica B Ericson
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden.
| |
Collapse
|
3
|
Bhar B, Das E, Manikumar K, Mandal BB. 3D Bioprinted Human Skin Model Recapitulating Native-Like Tissue Maturation and Immunocompetence as an Advanced Platform for Skin Sensitization Assessment. Adv Healthc Mater 2024; 13:e2303312. [PMID: 38478847 DOI: 10.1002/adhm.202303312] [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: 09/28/2023] [Revised: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Physiologically-relevant in vitro skin models hold the utmost importance for efficacy assessments of pharmaceutical and cosmeceutical formulations, offering valuable alternatives to animal testing. Here, an advanced immunocompetent 3D bioprinted human skin model is presented to assess skin sensitization. Initially, a photopolymerizable bioink is formulated using silk fibroin methacrylate, gelatin methacrylate, and photoactivated human platelet releasate. The developed bioink shows desirable physicochemical and rheological attributes for microextrusion bioprinting. The tunable physical and mechanical properties of bioink are modulated through variable photocuring time for optimization. Thereafter, the bioink is utilized to 3D bioprint "sandwich type" skin construct where an artificial basement membrane supports a biomimetic epidermal layer on one side and a printed pre-vascularized dermal layer on the other side within a transwell system. The printed construct is further cultured in the air-liquid interface for maturation. Immunofluorescence staining demonstrated a differentiated keratinocyte layer and dermal extracellular matrix (ECM)-remodeling by fibroblasts and endothelial cells. The biochemical estimations and gene-expression analysis validate the maturation of the printed model. The incorporation of macrophages further enhances the physiological relevance of the model. This model effectively classifies skin irritative and non-irritative substances, thus establishing itself as a suitable pre-clinical screening platform for sensitization tests.
Collapse
Affiliation(s)
- Bibrita Bhar
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Eshani Das
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Kodieswaran Manikumar
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| |
Collapse
|
4
|
Reed JM, Wolfe BE, Romero LM. Is resilience a unifying concept for the biological sciences? iScience 2024; 27:109478. [PMID: 38660410 PMCID: PMC11039332 DOI: 10.1016/j.isci.2024.109478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
There is increasing interest in applying resilience concepts at different scales of biological organization to address major interdisciplinary challenges from cancer to climate change. It is unclear, however, whether resilience can be a unifying concept consistently applied across the breadth of the biological sciences, or whether there is limited capacity for integration. In this review, we draw on literature from molecular biology to community ecology to ascertain commonalities and shortcomings in how resilience is measured and interpreted. Resilience is studied at all levels of biological organization, although the term is often not used. There is a suite of resilience mechanisms conserved across biological scales, and there are tradeoffs that affect resilience. Resilience is conceptually useful to help diverse researchers think about how biological systems respond to perturbations, but we need a richer lexicon to describe the diversity of perturbations, and we lack widely applicable metrics of resilience.
Collapse
Affiliation(s)
- J. Michael Reed
- Department of Biology, Tufts University, Medford 02155, MA, USA
| | | | | |
Collapse
|
5
|
Kang DS, Moriarty A, Wang YJ, Thomas A, Hao J, Unger BA, Klotz R, Ahmmed S, Amzaleg Y, Martin S, Vanapalli S, Xu K, Smith A, Shen K, Yu M. Ectopic Expression of a Truncated Isoform of Hair Keratin 81 in Breast Cancer Alters Biophysical Characteristics to Promote Metastatic Propensity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2300509. [PMID: 37949677 PMCID: PMC10837353 DOI: 10.1002/advs.202300509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/28/2023] [Indexed: 11/12/2023]
Abstract
Keratins are an integral part of cell structure and function. Here, it is shown that ectopic expression of a truncated isoform of keratin 81 (tKRT81) in breast cancer is upregulated in metastatic lesions compared to primary tumors and patient-derived circulating tumor cells, and is associated with more aggressive subtypes. tKRT81 physically interacts with keratin 18 (KRT18) and leads to changes in the cytosolic keratin intermediate filament network and desmosomal plaque formation. These structural changes are associated with a softer, more elastically deformable cancer cell with enhanced adhesion and clustering ability leading to greater in vivo lung metastatic burden. This work describes a novel biomechanical mechanism by which tKRT81 promotes metastasis, highlighting the importance of the biophysical characteristics of tumor cells.
Collapse
Affiliation(s)
- Diane S. Kang
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
| | - Aidan Moriarty
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Yiru Jess Wang
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Amal Thomas
- Department of Molecular and Computational BiologyUSC David and Dana Dornsife College of LettersArts and SciencesUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Jia Hao
- Department of Biomedical EngineeringViterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Bret A. Unger
- Department of ChemistryUniversity of California at BerkeleyBerkeleyCA94720USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Shamim Ahmmed
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| | - Yonatan Amzaleg
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
| | - Stuart Martin
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Siva Vanapalli
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| | - Ke Xu
- Department of ChemistryUniversity of California at BerkeleyBerkeleyCA94720USA
| | - Andrew Smith
- Department of Molecular and Computational BiologyUSC David and Dana Dornsife College of LettersArts and SciencesUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Keyue Shen
- Department of Biomedical EngineeringViterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| |
Collapse
|
6
|
Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, Rossi AB. Type 2 Inflammation Contributes to Skin Barrier Dysfunction in Atopic Dermatitis. JID INNOVATIONS 2022; 2:100131. [PMID: 36059592 PMCID: PMC9428921 DOI: 10.1016/j.xjidi.2022.100131] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
Skin barrier dysfunction, a defining feature of atopic dermatitis (AD), arises from multiple interacting systems. In AD, skin inflammation is caused by host-environment interactions involving keratinocytes as well as tissue-resident immune cells such as type 2 innate lymphoid cells, basophils, mast cells, and T helper type 2 cells, which produce type 2 cytokines, including IL-4, IL-5, IL-13, and IL-31. Type 2 inflammation broadly impacts the expression of genes relevant for barrier function, such as intracellular structural proteins, extracellular lipids, and junctional proteins, and enhances Staphylococcus aureus skin colonization. Systemic anti‒type 2 inflammation therapies may improve dysfunctional skin barrier in AD.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- CLDN, claudin
- FFA, free fatty acid
- ILC2, type 2 innate lymphoid cell
- Jaki, Jak inhibitor
- K, keratin
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NMF, natural moisturizing factor
- PAR, protease-activated receptor
- PDE-4, phosphodiesterase-4
- SC, stratum corneum
- SG, stratum granulosum
- TCI, topical calcineurin inhibitor
- TCS, topical corticosteroid
- TEWL, transepidermal water loss
- TJ, tight junction
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor alpha
- TYK, tyrosine kinase
- Th, T helper
- ZO, zona occludens
- hBD, human β-defensin
Collapse
Affiliation(s)
- Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA,Correspondence: Lisa A. Beck, Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, New York 14642, USA.
| | - Michael J. Cork
- Sheffield Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease (IICD), The University of Sheffield, The Medical School, Sheffield, United Kingdom
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
7
|
Weber MS, Eibauer M, Sivagurunathan S, Magin TM, Goldman RD, Medalia O. Structural heterogeneity of cellular K5/K14 filaments as revealed by cryo-electron microscopy. eLife 2021; 10:70307. [PMID: 34323216 PMCID: PMC8360650 DOI: 10.7554/elife.70307] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Keratin intermediate filaments are an essential and major component of the cytoskeleton in epithelial cells. They form a stable yet dynamic filamentous network extending from the nucleus to the cell periphery, which provides resistance to mechanical stresses. Mutations in keratin genes are related to a variety of epithelial tissue diseases. Despite their importance, the molecular structure of keratin filaments remains largely unknown. In this study, we analyzed the structure of keratin 5/keratin 14 filaments within ghost mouse keratinocytes by cryo-electron microscopy and cryo-electron tomography. By averaging a large number of keratin segments, we have gained insights into the helical architecture of the filaments. Two-dimensional classification revealed profound variations in the diameter of keratin filaments and their subunit organization. Computational reconstitution of filaments of substantial length uncovered a high degree of internal heterogeneity along single filaments, which can contain regions of helical symmetry, regions with less symmetry and regions with significant diameter fluctuations. Cross-section views of filaments revealed that keratins form hollow cylinders consisting of multiple protofilaments, with an electron dense core located in the center of the filament. These findings shed light on the complex and remarkable heterogenic architecture of keratin filaments, suggesting that they are highly flexible, dynamic cytoskeletal structures.
Collapse
Affiliation(s)
- Miriam S Weber
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Thomas M Magin
- Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
A unique mode of keratinocyte death requires intracellular acidification. Proc Natl Acad Sci U S A 2021; 118:2020722118. [PMID: 33893234 DOI: 10.1073/pnas.2020722118] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The stratum corneum (SC), the outermost epidermal layer, consists of nonviable anuclear keratinocytes, called corneocytes, which function as a protective barrier. The exact modes of cell death executed by keratinocytes of the upper stratum granulosum (SG1 cells) remain largely unknown. Here, using intravital imaging combined with intracellular Ca2+- and pH-responsive fluorescent probes, we aimed to dissect the SG1 death process in vivo. We found that SG1 cell death was preceded by prolonged (∼60 min) Ca2+ elevation and rapid induction of intracellular acidification. Once such intracellular ionic changes were initiated, they became sustained, irreversibly committing the SG1 cells to corneocyte conversion. Time-lapse imaging of isolated murine SG1 cells revealed that intracellular acidification was essential for the degradation of keratohyalin granules and nuclear DNA, phenomena specific to SC corneocyte formation. Furthermore, intravital imaging showed that the number of SG1 cells exhibiting Ca2+ elevation and the timing of intracellular acidification were both tightly regulated by the transient receptor potential cation channel V3. The functional activity of this protein was confirmed in isolated SG1 cells using whole-cell patch-clamp analysis. These findings provide a theoretical framework for improved understanding of the unique molecular mechanisms underlying keratinocyte-specific death mode, namely corneoptosis.
Collapse
|
9
|
Kocher T, Koller U. Advances in gene editing strategies for epidermolysis bullosa. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:81-109. [PMID: 34175052 DOI: 10.1016/bs.pmbts.2020.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Epidermolysis bullosa represents a monogenetic disease comprising a variety of heterogeneous mutations in at least 16 genes encoding structural proteins crucial for skin integrity. Due to well-defined mutations but still lacking causal treatment options for the disease, epidermolysis bullosa represents an ideal candidate for gene therapeutic interventions. Recent developments and improvements in the genome editing field have paved the way for the translation of various gene repair strategies into the clinic. With the ability to accurately predict and monitor targeting events within the human genome, the translation might soon be possible. Here, we describe current advancements in the genome editing field for epidermolysis bullosa, along with a discussion of aspects and strategies for precise and personalized gene editing-based medicine, in order to develop efficient and safe ex vivo as well as in vivo genome editing therapies for epidermolysis bullosa patients in the future.
Collapse
Affiliation(s)
- Thomas Kocher
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria.
| |
Collapse
|
10
|
Tenorio-Mina A, Cortés D, Esquivel-Estudillo J, López-Ornelas A, Cabrera-Wrooman A, Lara-Rodarte R, Escobedo-Avila I, Vargas-Romero F, Toledo-Hernández D, Estudillo E, Acevedo-Fernández JJ, Tapia JSO, Velasco I. Human Keratinocytes Adopt Neuronal Fates After In Utero Transplantation in the Developing Rat Brain. Cell Transplant 2021; 30:963689720978219. [PMID: 33435710 PMCID: PMC7809298 DOI: 10.1177/0963689720978219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 11/30/2022] Open
Abstract
Human skin contains keratinocytes in the epidermis. Such cells share their ectodermal origin with the central nervous system (CNS). Recent studies have demonstrated that terminally differentiated somatic cells can adopt a pluripotent state, or can directly convert its phenotype to neurons, after ectopic expression of transcription factors. In this article we tested the hypothesis that human keratinocytes can adopt neural fates after culturing them in suspension with a neural medium. Initially, keratinocytes expressed Keratins and Vimentin. After neural induction, transcriptional upregulation of NESTIN, SOX2, VIMENTIN, SOX1, and MUSASHI1 was observed, concomitant with significant increases in NESTIN detected by immunostaining. However, in vitro differentiation did not yield the expression of neuronal or astrocytic markers. We tested the differentiation potential of control and neural-induced keratinocytes by grafting them in the developing CNS of rats, through ultrasound-guided injection. For this purpose, keratinocytes were transduced with lentivirus that contained the coding sequence of green fluorescent protein. Cell sorting was employed to select cells with high fluorescence. Unexpectedly, 4 days after grafting these cells in the ventricles, both control and neural-induced cells expressed green fluorescent protein together with the neuronal proteins βIII-Tubulin and Microtubule-Associated Protein 2. These results support the notion that in vivo environment provides appropriate signals to evaluate the neuronal differentiation potential of keratinocytes or other non-neural cell populations.
Collapse
Affiliation(s)
- Andrea Tenorio-Mina
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Daniel Cortés
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Joel Esquivel-Estudillo
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
| | - Adolfo López-Ornelas
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- División de Investigación, Hospital Juárez de México, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
- Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Rolando Lara-Rodarte
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Itzel Escobedo-Avila
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Fernanda Vargas-Romero
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Diana Toledo-Hernández
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
- Centro de Investigación en Dinámica Celular, Instituto de Ciencias, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | | | - Jesús Santa-Olalla Tapia
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| |
Collapse
|
11
|
Ye J, Wu Y, Li M, Gong X, Zhong B. Keratin 8 Mutations Were Associated With Susceptibility to Chronic Hepatitis B and Related Progression. J Infect Dis 2020; 221:464-473. [PMID: 31515557 DOI: 10.1093/infdis/jiz467] [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] [Received: 06/07/2019] [Accepted: 09/10/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Keratin 8 and 18 (K8/K18) are the exclusively expressed keratins intermediate filaments pair in hepatocytes that protect against liver injuries and viral infection. We aimed to explore the genetic link between keratin variants and chronic hepatitis B virus (CHB) infection in a large cohort from a high-epidemic area. METHODS Genomic deoxyribonucleic acid was isolated from patients, and Sanger sequencing was applied to analyze variations in exon regions of K8/18. Biochemical and functional analysis of novel mutations was also performed. RESULTS The 713 participants comprised 173 healthy controls and 540 patients, which covered chronic hepatitis (n = 174), decompensated cirrhosis (n = 192), and primary liver carcinoma (n = 174). The frequency of mutations in K8/18 was significantly higher among patients than among controls (8.15% vs 0.58%, P < .001). Significant differences were found between the chronic hepatitis subgroup and controls in multiple comparisons (6.32% vs 0.58%, P = .006). All 21 missense mutations (3.89%) were detected in the keratin 8 (K8), including 4 novel conserved missense variants (R469C, R469H, A447V, and K483T). Multivariate logistic regression analysis demonstrated a higher risk of acute-on-chronic liver failure (ACLF) and missense variants (odds ratio = 4.38, P = .035). Transfection of these variants caused keratin network disruption in vivo. CONCLUSIONS Novel K8 cytoskeleton-disrupting variants predispose toward ACLF in CHB.
Collapse
Affiliation(s)
- Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Yanqin Wu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Minrui Li
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Xiaorong Gong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| |
Collapse
|
12
|
Addressing the Molecular Mechanism of Longitudinal Lamin Assembly Using Chimeric Fusions. Cells 2020; 9:cells9071633. [PMID: 32645958 PMCID: PMC7407374 DOI: 10.3390/cells9071633] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/28/2022] Open
Abstract
The molecular architecture and assembly mechanism of intermediate filaments have been enigmatic for decades. Among those, lamin filaments are of particular interest due to their universal role in cell nucleus and numerous disease-related mutations. Filament assembly is driven by specific interactions of the elementary dimers, which consist of the central coiled-coil rod domain flanked by non-helical head and tail domains. We aimed to investigate the longitudinal 'head-to-tail' interaction of lamin dimers (the so-called ACN interaction), which is crucial for filament assembly. To this end, we prepared a series of recombinant fragments of human lamin A centred around the N- and C-termini of the rod. The fragments were stabilized by fusions to heterologous capping motifs which provide for a correct formation of parallel, in-register coiled-coil dimers. As a result, we established crystal structures of two N-terminal fragments one of which highlights the propensity of the coiled-coil to open up, and one C-terminal rod fragment. Additional studies highlighted the capacity of such N- and C-terminal fragments to form specific complexes in solution, which were further characterized using chemical cross-linking. These data yielded a molecular model of the ACN complex which features a 6.5 nm overlap of the rod ends.
Collapse
|
13
|
Lee CH, Kim MS, Li S, Leahy DJ, Coulombe PA. Structure-Function Analyses of a Keratin Heterotypic Complex Identify Specific Keratin Regions Involved in Intermediate Filament Assembly. Structure 2020; 28:355-362.e4. [PMID: 31995743 DOI: 10.1016/j.str.2020.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/20/2019] [Accepted: 01/07/2020] [Indexed: 11/29/2022]
Abstract
Intermediate filaments (IFs) provide vital mechanical support in a broad array of cell types. Interference with this role causes cell fragility and accounts for a large number of human diseases. Gaining an understanding of the structure of IFs is paramount to understanding their function and designing therapeutic agents for relevant diseases. Here, we report the 2.6-Å resolution crystal structure of a complex of interacting 2B domains of keratin 5 (K5) and K14. K5 and K14 form a long-range, left-handed coiled coil, with participating α helices aligned in parallel and in register. Follow-up mutagenesis revealed that specific contacts between interacting 2B domains play a crucial role during 10-nm IF assembly, likely at the step of octamer-octamer association. The resulting structural model represents an atomic-resolution visualization of 2B-2B interactions important to filament assembly and provides insight into the defects introduced by mutations in IF genes associated with human skin diseases.
Collapse
Affiliation(s)
- Chang-Hun Lee
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Min-Sung Kim
- Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Shuang Li
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Daniel J Leahy
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
14
|
March OP, Kocher T, Koller U. Context-Dependent Strategies for Enhanced Genome Editing of Genodermatoses. Cells 2020; 9:E112. [PMID: 31906492 PMCID: PMC7016731 DOI: 10.3390/cells9010112] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/27/2019] [Accepted: 12/31/2019] [Indexed: 12/17/2022] Open
Abstract
The skin provides direct protection to the human body from assault by the harsh external environment. The crucial function of this organ is significantly disrupted in genodermatoses patients. Genodermatoses comprise a heterogeneous group of largely monogenetic skin disorders, typically involving mutations in genes encoding structural proteins. Therapeutic options for this debilitating group of diseases, including epidermolysis bullosa, primarily consist of wound management. Genome editing approaches co-opt double-strand break repair pathways to introduce desired sequence alterations at specific loci. Rapid advances in genome editing technologies have the potential to propel novel genetic therapies into the clinic. However, the associated phenotypes of many mutations may be treated via several genome editing strategies. Therefore, for potential clinical applications, implementation of efficient approaches based upon mutation, gene and disease context is necessary. Here, we describe current genome editing approaches for the treatment of genodermatoses, along with a discussion of the optimal strategy for each genetic context, in order to achieve enhanced genome editing approaches.
Collapse
Affiliation(s)
| | | | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (O.P.M.); (T.K.)
| |
Collapse
|
15
|
Armstrong C, Cassimeris L, Da Silva Santos C, Micoogullari Y, Wagner B, Babasyan S, Brooks S, Galantino-Homer H. The expression of equine keratins K42 and K124 is restricted to the hoof epidermal lamellae of Equus caballus. PLoS One 2019; 14:e0219234. [PMID: 31550264 PMCID: PMC6759161 DOI: 10.1371/journal.pone.0219234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/11/2019] [Indexed: 01/30/2023] Open
Abstract
The equine hoof inner epithelium is folded into primary and secondary epidermal lamellae which increase the dermo-epidermal junction surface area of the hoof and can be affected by laminitis, a common disease of equids. Two keratin proteins (K), K42 and K124, are the most abundant keratins in the hoof lamellar tissue of Equus caballus. We hypothesize that these keratins are lamellar tissue-specific and could serve as differentiation- and disease-specific markers. Our objective was to characterize the expression of K42 and K124 in equine stratified epithelia and to generate monoclonal antibodies against K42 and K124. By RT-PCR analysis, keratin gene (KRT) KRT42 and KRT124 expression was present in lamellar tissue, but not cornea, haired skin, or hoof coronet. In situ hybridization studies showed that KRT124 localized to the suprabasal and, to a lesser extent, basal cells of the lamellae, was absent from haired skin and hoof coronet, and abruptly transitions from KRT124-negative coronet to KRT124-positive proximal lamellae. A monoclonal antibody generated against full-length recombinant equine K42 detected a lamellar keratin of the appropriate size, but also cross-reacted with other epidermal keratins. Three monoclonal antibodies generated against N- and C-terminal K124 peptides detected a band of the appropriate size in lamellar tissue and did not cross-react with proteins from haired skin, corneal limbus, hoof coronet, tongue, glabrous skin, oral mucosa, or chestnut on immunoblots. K124 localized to lamellar cells by indirect immunofluorescence. This is the first study to demonstrate the localization and expression of a hoof lamellar-specific keratin, K124, and to validate anti-K124 monoclonal antibodies.
Collapse
Affiliation(s)
- Caitlin Armstrong
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Lynne Cassimeris
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Claire Da Silva Santos
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Yagmur Micoogullari
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Susanna Babasyan
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Samantha Brooks
- Department of Animal Sciences and University of Florida Genetics institute, University of Florida, Gainesville, Florida, United States of America
| | - Hannah Galantino-Homer
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Spörrer M, Prochnicki A, Tölle RC, Nyström A, Esser PR, Homberg M, Athanasiou I, Zingkou E, Schilling A, Gerum R, Thievessen I, Winter L, Bruckner-Tuderman L, Fabry B, Magin TM, Dengjel J, Schröder R, Kiritsi D. Treatment of keratinocytes with 4-phenylbutyrate in epidermolysis bullosa: Lessons for therapies in keratin disorders. EBioMedicine 2019; 44:502-515. [PMID: 31078522 PMCID: PMC6603805 DOI: 10.1016/j.ebiom.2019.04.062] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/18/2022] Open
Abstract
Background Missense mutations in keratin 5 and 14 genes cause the severe skin fragility disorder epidermolysis bullosa simplex (EBS) by collapsing of the keratin cytoskeleton into cytoplasmic protein aggregates. Despite intense efforts, no molecular therapies are available, mostly due to the complex phenotype of EBS, comprising cell fragility, diminished adhesion, skin inflammation and itch. Methods We extensively characterized KRT5 and KRT14 mutant keratinocytes from patients with severe generalized EBS following exposure to the chemical chaperone 4-phenylbutyrate (4-PBA). Findings 4-PBA diminished keratin aggregates within EBS cells and ameliorated their inflammatory phenotype. Chemoproteomics of 4-PBA-treated and untreated EBS cells revealed reduced IL1β expression- but also showed activation of Wnt/β-catenin and NF-kB pathways. The abundance of extracellular matrix and cytoskeletal proteins was significantly altered, coinciding with diminished keratinocyte adhesion and migration in a 4-PBA dose-dependent manner. Interpretation Together, our study reveals a complex interplay of benefits and disadvantages that challenge the use of 4-PBA in skin fragility disorders.
Collapse
Affiliation(s)
- Marina Spörrer
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Ania Prochnicki
- Institute of Neuropathology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Regine C Tölle
- Department of Biology, University of Fribourg, Switzerland
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp R Esser
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Homberg
- Institute of Biology and SIKT, University of Leipzig, Leipzig, Germany
| | - Ioannis Athanasiou
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eleni Zingkou
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Achim Schilling
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; Experimental Otolaryngology, ENT Hospital, Head and Neck Surgery, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Richard Gerum
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Ingo Thievessen
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Lilli Winter
- Institute of Neuropathology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ben Fabry
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas M Magin
- Institute of Biology and SIKT, University of Leipzig, Leipzig, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Switzerland; Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rolf Schröder
- Institute of Neuropathology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Dimitra Kiritsi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Wang F, Chen S, Liu HB, Parent CA, Coulombe PA. Keratin 6 regulates collective keratinocyte migration by altering cell-cell and cell-matrix adhesion. J Cell Biol 2018; 217:4314-4330. [PMID: 30389720 PMCID: PMC6279382 DOI: 10.1083/jcb.201712130] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 09/18/2018] [Accepted: 10/16/2018] [Indexed: 01/21/2023] Open
Abstract
Keratin 6 (K6) isoforms are induced in wound-proximal keratinocytes after injury to skin. Paradoxically, absence of K6 isoforms leads to faster directional cell migration. Wang et al. report that K6 promotes collective keratinocyte migration by interacting with desmoplakin and myosin IIA and stabilizing cell adhesion. The a and b isoforms of keratin 6 (K6), a type II intermediate filament (IF) protein, are robustly induced upon injury to interfollicular epidermis. We previously showed that complete loss of K6a/K6b stimulates keratinocyte migration, correlating with enhanced Src activity. In this study, we demonstrate that this property is cell autonomous, depends on the ECM, and results from elevated speed, enhanced directionality, and an increased rate of focal adhesion disassembly. We show that myosin IIA interacts with K6a/K6b, that its levels are markedly reduced in Krt6a/Krt6b-null keratinocytes, and that inhibiting myosin ATPase activity normalizes the enhanced migration potential of Krt6a/Krt6b-null cells. Desmoplakin, which mediates attachment of IFs to desmosomes, is also expressed at reduced levels and is mislocalized to the nucleus in Krt6a/Krt6b-null cells, correlating with defects in cell adhesion. These findings reveal that K6a/K6b modulate keratinocyte migration by regulating cell–matrix and cell–cell adhesion and highlight a role for keratins in collective cell migration.
Collapse
Affiliation(s)
- Fengrong Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI.,Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Song Chen
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Hans B Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Carole A Parent
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI .,Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD.,Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
18
|
HIF stabilization inhibits renal epithelial cell migration and is associated with cytoskeletal alterations. Sci Rep 2018; 8:9497. [PMID: 29934555 PMCID: PMC6015081 DOI: 10.1038/s41598-018-27918-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/13/2018] [Indexed: 11/23/2022] Open
Abstract
Acute kidney injury (AKI) is a common and potentially lethal complication in the hospitalized patients, with hypoxic injury being as a major cause. The loss of renal tubular epithelial cells (TEC), one of the AKI hallmarks, is potentially followed by tubular regeneration process orchestrated by the remaining uninjured TECs that undergo proliferation and migration. In this study, we used human primary TEC to investigate the initiation of tubular cell migration and associated cytoskeletal alterations in response to pharmacological HIF stabilization which resembles the pathophysiology of hypoxia. Tubular cells have been shown to migrate as cohorts in a wound healing assay. Importantly, cells of distal tubular origin moved faster than those of proximal origin. HIF stabilization impaired TEC migration, which was confirmed by live single cell tracking. HIF stabilization significantly reduced tubular cell migration velocity and promoted cell spreading. In contrast to the control conditions, HIF stabilization induced actin filaments rearrangement and cell adhesion molecules including paxillin and focal adhesion kinase. Condensed bundling of keratin fibers was also observed, while the expression of different types of keratins, phosphorylation of keratin 18, and the microtubule structure were not altered. In summary, HIF stabilization reduced the ability of renal tubular cells to migrate and led to cytoskeleton reorganization. Our data suggested an important involvement of HIF stabilization during the epithelial migration underlying the mechanism of renal regeneration in response to AKI.
Collapse
|
19
|
Abashev TM, Metzler MA, Wright DM, Sandell LL. Retinoic acid signaling regulates Krt5 and Krt14 independently of stem cell markers in submandibular salivary gland epithelium. Dev Dyn 2018; 246:135-147. [PMID: 27884045 DOI: 10.1002/dvdy.24476] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Retinoic acid (RA), the active metabolite of vitamin A, has been demonstrated to be important for growth and branching morphogenesis of mammalian embryonic salivary gland epithelium. However, it is not known whether RA functions directly within epithelial cells or in associated tissues that influence morphogenesis of salivary epithelium. Moreover, downstream targets of RA regulation have not been identified. RESULTS Here, we show that canonical RA signaling occurs in multiple tissues of embryonic mouse salivary glands, including epithelium, associated parasympathetic ganglion neurons, and nonneuronal mesenchyme. By culturing epithelium explants in isolation from other tissues, we demonstrate that RA influences epithelium morphogenesis by direct action in that tissue. Moreover, we demonstrate that inhibition of RA signaling represses cell proliferation and expression of FGF10 signaling targets, and upregulates expression of basal epithelial keratins Krt5 and Krt14. Importantly, we show that the stem cell gene Kit is regulated inversely from Krt5/Krt14 by RA signaling. CONCLUSIONS RA regulates Krt5 and Krt14 expression independently of stem cell character in developing salivary epithelium. RA, or chemical inhibitors of RA signaling, could potentially be used for modulating growth and differentiation of epithelial stem cells for the purpose of re-populating damaged glands or generating bioengineered organs. Developmental Dynamics 246:135-147, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Timur M Abashev
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Melissa A Metzler
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Diana M Wright
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Lisa L Sandell
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| |
Collapse
|
20
|
Aushev M, Koller U, Mussolino C, Cathomen T, Reichelt J. Traceless Targeting and Isolation of Gene-Edited Immortalized Keratinocytes from Epidermolysis Bullosa Simplex Patients. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:112-123. [PMID: 28765827 PMCID: PMC5527154 DOI: 10.1016/j.omtm.2017.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/30/2017] [Indexed: 12/20/2022]
Abstract
Epidermolysis bullosa simplex (EBS) is a blistering skin disease caused by dominant-negative mutations in either KRT5 or KRT14, resulting in impairment of keratin filament structure and epidermal fragility. Currently, nearly 200 mutations distributed across the entire length of these genes are known to cause EBS. Genome editing using programmable nucleases enables the development of ex vivo gene therapies for dominant-negative genetic diseases. A clinically feasible strategy involves the disruption of the mutant allele while leaving the wild-type allele unaffected. Our aim was to develop a traceless approach to efficiently disrupt KRT5 alleles using TALENs displaying unbiased monoallelic disruption events and devise a strategy that allows for subsequent screening and isolation of correctly modified keratinocyte clones without the need for selection markers. Here we report on TALENs that efficiently disrupt the KRT5 locus in immortalized patient-derived EBS keratinocytes. Inactivation of the mutant allele using a TALEN working at sub-optimal levels resulted in restoration of intermediate filament architecture. This approach can be used for the functional inactivation of any mutant keratin allele regardless of the position of the mutation within the gene and is furthermore applicable to the treatment of other inherited skin disorders.
Collapse
Affiliation(s)
- Magomet Aushev
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Biomedicine West Wing, Centre for Life, Times Square, Newcastle upon Tyne NE1 3BZ, UK
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses and Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Claudio Mussolino
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany.,Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstrasse 115, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany.,Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstrasse 115, 79106 Freiburg, Germany.,Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Julia Reichelt
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses and Department of Dermatology, University Hospital of the Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
21
|
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
|
22
|
Three-dimensional Organization of Layered Apical Cytoskeletal Networks Associated with Mouse Airway Tissue Development. Sci Rep 2017; 7:43783. [PMID: 28272499 PMCID: PMC5363704 DOI: 10.1038/srep43783] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/30/2017] [Indexed: 01/11/2023] Open
Abstract
The cytoskeleton is an essential cellular component that enables various sophisticated functions of epithelial cells by forming specialized subcellular compartments. However, the functional and structural roles of cytoskeletons in subcellular compartmentalization are still not fully understood. Here we identified a novel network structure consisting of actin filaments, intermediate filaments, and microtubules directly beneath the apical membrane in mouse airway multiciliated cells and in cultured epithelial cells. Three-dimensional imaging by ultra-high voltage electron microscopy and immunofluorescence revealed that the morphological features of each network depended on the cell type and were spatiotemporally integrated in association with tissue development. Detailed analyses using Odf2 mutant mice, which lack ciliary basal feet and apical microtubules, suggested a novel contribution of the intermediate filaments to coordinated ciliary beating. These findings provide a new perspective for viewing epithelial cell differentiation and tissue morphogenesis through the structure and function of apical cytoskeletal networks.
Collapse
|
23
|
Miniaci MC, Irace C, Capuozzo A, Piccolo M, Di Pascale A, Russo A, Lippiello P, Lepre F, Russo G, Santamaria R. Cysteine Prevents the Reduction in Keratin Synthesis Induced by Iron Deficiency in Human Keratinocytes. J Cell Biochem 2016. [PMID: 26212225 DOI: 10.1002/jcb.25286] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
L-cysteine is currently recognized as a conditionally essential sulphur amino acid. Besides contributing to many biological pathways, cysteine is a key component of the keratin protein by its ability to form disulfide bridges that confer strength and rigidity to the protein. In addition to cysteine, iron represents another critical factor in regulating keratins expression in epidermal tissues, as well as in hair follicle growth and maturation. By focusing on human keratinocytes, the aim of this study was to evaluate the effect of cysteine supplementation as nutraceutical on keratin biosynthesis, as well as to get an insight on the interplay of cysteine availability and cellular iron status in regulating keratins expression in vitro. Herein we demonstrate that cysteine promotes a significant up-regulation of keratins expression as a result of de novo protein synthesis, while the lack of iron impairs keratin expression. Interestingly, cysteine supplementation counteracts the adverse effect of iron deficiency on cellular keratin expression. This effect was likely mediated by the up-regulation of transferrin receptor and ferritin, the main cellular proteins involved in iron homeostasis, at last affecting the labile iron pool. In this manner, cysteine may also enhance the metabolic iron availability for DNA synthesis without creating a detrimental condition of iron overload. To the best of our knowledge, this is one of the first study in an in vitro keratinocyte model providing evidence that cysteine and iron cooperate for keratins expression, indicative of their central role in maintaining healthy epithelia.
Collapse
Affiliation(s)
| | - Carlo Irace
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | | | - Annapina Russo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | - Giulia Russo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Rita Santamaria
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
24
|
Deek J, Hecht F, Rossetti L, Wißmiller K, Bausch AR. Mechanics of soft epithelial keratin networks depend on modular filament assembly kinetics. Acta Biomater 2016; 43:218-229. [PMID: 27403885 DOI: 10.1016/j.actbio.2016.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/04/2016] [Accepted: 07/09/2016] [Indexed: 11/15/2022]
Abstract
UNLABELLED Structural adaptability is a pivotal requirement of cytoskeletal structures, enabling their reorganization to meet the cellular needs. Shear stress, for instance, results in large morphological network changes of the human soft epithelial keratin pair K8:K18, and is accompanied by an increase in keratin phosphorylation levels. Yet the mechanisms responsible for the disruption of the network structure in vivo remain poorly understood. To understand the effect of the stress-related site-specific phosphorylation of the K8:K18 pair, we created phosphomimicry mutants - K8(S431E), K8(S73E), K18(S52E) - in vitro, and investigated the various steps of keratin assembly from monomer to network structure using fluorescence and electron microscopy, and using rheology characterized their network mechanical properties. We find that the addition of a charged group produces networks with depleted intra-connectivity, which translates to a mechanically weaker and more deformable network. This large variation in network structure is achieved by the formation of shorter mutant filaments, which exhibit differing assembly kinetics and a manifestly reduced capacity to form the extended structures characteristic of the wild-type system. The similarity in outcome for all the phosphomimicry mutants explored points to a more general mechanism of structural modulation of intermediate filaments via phosphorylation. Understanding the role of kinetic effects in the construction of these cytoskeletal biopolymer networks is critical to elucidating their structure-function properties, providing new insight for the design of keratin-inspired biomaterials. STATEMENT OF SIGNIFICANCE Structural remodeling of cytoskeletal networks accompanies many cellular processes. Interestingly, levels of phosphorylation of the human soft epithelial keratin pair K8:K18 increase during their stress-related structural remodeling. Our multi-scale study sheds light on the poorly understood mechanism with which site-specific phosphorylation induces disruption of the keratin network structure in vivo. We show how phosphorylation reduces keratin filament length, an effect that propagates through to the mesoscopic structure, resulting in the formation of connectivity-depleted and mechanically weaker networks. We determine that the intrinsically-set filament-to-filament attractions that drive bundle assembly give rise to the structural variability by enabling the formation of kinetically-arrested structures. Overall, our results shed light on how self-assembled intermediate filament structures can be tailored to exhibit different structural functionalities.
Collapse
Affiliation(s)
- Joanna Deek
- Lehrstuhl für Zellbiophysik E27, Technische Universität München, James-Franck-Straße 1, 85748 Garching, Germany
| | - Fabian Hecht
- Lehrstuhl für Zellbiophysik E27, Technische Universität München, James-Franck-Straße 1, 85748 Garching, Germany
| | - Leone Rossetti
- Lehrstuhl für Zellbiophysik E27, Technische Universität München, James-Franck-Straße 1, 85748 Garching, Germany
| | - Katharina Wißmiller
- Lehrstuhl für Zellbiophysik E27, Technische Universität München, James-Franck-Straße 1, 85748 Garching, Germany
| | - Andreas R Bausch
- Lehrstuhl für Zellbiophysik E27, Technische Universität München, James-Franck-Straße 1, 85748 Garching, Germany.
| |
Collapse
|
25
|
Langbein L, Eckhart L, Fischer H, Rogers MA, Praetzel-Wunder S, Parry DAD, Kittstein W, Schweizer J. Localisation of keratin K78 in the basal layer and first suprabasal layers of stratified epithelia completes expression catalogue of type II keratins and provides new insights into sequential keratin expression. Cell Tissue Res 2016; 363:735-50. [PMID: 26340985 DOI: 10.1007/s00441-015-2278-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/07/2015] [Indexed: 01/21/2023]
Abstract
Among the 26 human type II keratins, K78 is the only one that has not yet been explored with regard to its expression characteristics. Here, we show that, at both the transcriptional and translational levels, K78 is strongly expressed in the basal and parabasal cell layers with decreasing intensity in the lower suprabasal cells of keratinising and non-keratinising squamous epithelia and keratinocyte cultures. The same pattern has been detected at the transcriptional level in the corresponding mouse epithelia. Murine K78 protein, which contains an extraordinary large extension of its tail domain, which is unique among all known keratins, is not detectable by the antibody used. Concomitant studies in human epithelia have confirmed K78 co-expression with the classical basal keratins K5 and K14. Similarly, K78 co-expression with the differentiation-related type I keratins K10 (epidermis) and K13 (non-keratinising epithelia) occurs in the parabasal cell layer, whereas that of the corresponding type II keratins K1 (epidermis) and K4 (non-keratinising epithelia) unequivocally starts subsequent to the respective type I keratins. Our data concerning K78 expression modify the classical concept of keratin pair K5/K14 representing the basal compartment and keratin pairs K1/K10 or K4/K13 defining the differentiating compartment of stratified epithelia. Moreover, the K78 expression pattern and the decoupled K1/K10 and K4/K13 expression define the existence of a hitherto unperceived early differentiation stage in the parabasal layer characterized by K78/K10 or K78/K13 expression.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Animals
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Embryonic Development
- Epidermis/metabolism
- Epithelium/metabolism
- Evolution, Molecular
- Fluorescent Antibody Technique
- Gene Expression Regulation
- Genetic Loci
- Humans
- In Situ Hybridization
- Keratinocytes/metabolism
- Keratins, Type II/chemistry
- Keratins, Type II/genetics
- Keratins, Type II/metabolism
- Mice, Inbred C57BL
- Molecular Sequence Data
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sequence Analysis, Protein
Collapse
Affiliation(s)
- Lutz Langbein
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center, A110, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| | - Leopold Eckhart
- Department of Dermatology, Medical University Vienna, Vienna, Austria
| | - Heinz Fischer
- Department of Dermatology, Medical University Vienna, Vienna, Austria
| | - Michael A Rogers
- Department of Molecular Genetics of the German Cancer Research Center, Heidelberg, Germany
| | - Silke Praetzel-Wunder
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center, A110, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - David A D Parry
- Institute of Fundamental Sciences and Riddet Institute, Massey University, Palmerston North, New Zealand
| | | | | |
Collapse
|
26
|
Abstract
Keratins comprise the type I and type II intermediate filament-forming proteins and occur primarily in epithelial cells. They are encoded by 54 evolutionarily conserved genes (28 type I, 26 type II) and regulated in a pairwise and tissue type-, differentiation-, and context-dependent manner. Keratins serve multiple homeostatic and stress-enhanced mechanical and nonmechanical functions in epithelia, including the maintenance of cellular integrity, regulation of cell growth and migration, and protection from apoptosis. These functions are tightly regulated by posttranslational modifications as well as keratin-associated proteins. Genetically determined alterations in keratin-coding sequences underlie highly penetrant and rare disorders whose pathophysiology reflects cell fragility and/or altered tissue homeostasis. Moreover, keratin mutation or misregulation represents risk factors or genetic modifiers for several acute and chronic diseases. This chapter focuses on keratins that are expressed in skin epithelia, and details a number of basic protocols and assays that have proven useful for analyses being carried out in skin.
Collapse
Affiliation(s)
- Fengrong Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Abigail Zieman
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
27
|
Order and disorder in intermediate filament proteins. FEBS Lett 2015; 589:2464-76. [PMID: 26231765 DOI: 10.1016/j.febslet.2015.07.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 11/20/2022]
Abstract
Intermediate filaments (IFs), important components of the cytoskeleton, provide a versatile, tunable network of self-assembled proteins. IF proteins contain three distinct domains: an α-helical structured rod domain, flanked by intrinsically disordered head and tail domains. Recent studies demonstrated the functional importance of the disordered domains, which differ in length and amino-acid sequence among the 70 different human IF genes. Here, we investigate the biophysical properties of the disordered domains, and review recent findings on the interactions between them. Our analysis highlights key components governing IF functional roles in the cytoskeleton, where the intrinsically disordered domains dictate protein-protein interactions, supramolecular assembly, and macro-scale order.
Collapse
|
28
|
Feng X, Coulombe PA. Complementary roles of specific cysteines in keratin 14 toward the assembly, organization, and dynamics of intermediate filaments in skin keratinocytes. J Biol Chem 2015. [PMID: 26216883 DOI: 10.1074/jbc.m115.654749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently showed that inter-keratin disulfide bonding plays an important role in the assembly, organization, and dynamics of keratin intermediate filaments in skin keratinocytes. In particular, cysteine 367 located in the central α-helical rod domain of keratin 14 is necessary for the formation of a stable perinuclear network of keratin filaments (with type II partner keratin 5) in skin keratinocytes analyzed by static and live cell imaging. Here, we show that two additional cysteine residues located in the non-helical head domain of K14, Cys-4 and Cys-40, also participate in inter-keratin disulfide bonding and tandemly play a key role complementary to that of Cys-367 in the assembly, organization, and dynamics of keratin filaments in skin keratinocytes in primary culture. Analysis of K14 variants with single or multiple substitutions of cysteine residues points to a spatial and temporal hierarchy in how Cys-4/Cys-40 and Cys-367 regulate keratin assembly in vitro and filament dynamics in live keratinocytes in culture. Our findings substantiate the importance and complexity of a novel determinant, namely inter-keratin disulfide bonding, for the regulation of several aspects of keratin filaments in surface epithelia.
Collapse
Affiliation(s)
- Xia Feng
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205 and
| | - Pierre A Coulombe
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205 and the Departments of Biological Chemistry, Dermatology, and Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
29
|
Complete Structure of an Epithelial Keratin Dimer: Implications for Intermediate Filament Assembly. PLoS One 2015; 10:e0132706. [PMID: 26181054 PMCID: PMC4504709 DOI: 10.1371/journal.pone.0132706] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/17/2015] [Indexed: 01/05/2023] Open
Abstract
Keratins are cytoskeletal proteins that hierarchically arrange into filaments, starting with the dimer sub-unit. They are integral to the structural support of cells, in skin, hair and nails. In skin, keratin is thought to play a critical role in conferring the barrier properties and elasticity of skin. In general, the keratin dimer is broadly described by a tri-domain structure: a head, a central rod and a tail. As yet, no atomistic-scale picture of the entire dimer structure exists; this information is pivotal for establishing molecular-level connections between structure and function in intermediate filament proteins. The roles of the head and tail domains in facilitating keratin filament assembly and function remain as open questions. To address these, we report results of molecular dynamics simulations of the entire epithelial human K1/K10 keratin dimer. Our findings comprise: (1) the first three-dimensional structural models of the complete dimer unit, comprising of the head, rod and tail domains; (2) new insights into the chirality of the rod-domain twist gained from analysis of the full domain structure; (3) evidence for tri-subdomain partitioning in the head and tail domains; and, (4) identification of the residue characteristics that mediate non-covalent contact between the chains in the dimer. Our findings are immediately applicable to other epithelial keratins, such as K8/K18 and K5/K14, and to intermediate filament proteins in general.
Collapse
|
30
|
Feng X, Coulombe PA. A role for disulfide bonding in keratin intermediate filament organization and dynamics in skin keratinocytes. ACTA ACUST UNITED AC 2015; 209:59-72. [PMID: 25869667 PMCID: PMC4395492 DOI: 10.1083/jcb.201408079] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Disulfide bonds involving cysteine 367 in K14 play a crucial role in the assembly, dynamics, and organization of K14-containing filaments in epidermal keratinocytes. We recently reported that a trans-dimer, homotypic disulfide bond involving Cys367 in keratin 14 (K14) occurs in an atomic-resolution structure of the interacting K5/K14 2B domains and in keratinocyte cell lines. Here we show that a sizable fraction of the K14 and K5 protein pools participates in interkeratin disulfide bonding in primary cultures of mouse skin keratinocytes. By comparing the properties of wild-type K14 with a completely cysteine-free variant thereof, we found that K14-dependent disulfide bonding limited filament elongation during polymerization in vitro but was necessary for the genesis of a perinuclear-concentrated network of keratin filaments, normal keratin cycling, and the sessile behavior of the nucleus and whole cell in keratinocytes studied by live imaging. Many of these phenotypes were rescued when analyzing a K14 variant harboring a single Cys residue at position 367. These findings establish disulfide bonding as a novel and important mechanism regulating the assembly, intracellular organization, and dynamics of K14-containing intermediate filaments in skin keratinocytes.
Collapse
Affiliation(s)
- Xia Feng
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health; and Department of Biological Chemistry and Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health; and Department of Biological Chemistry and Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205 Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health; and Department of Biological Chemistry and Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205 Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health; and Department of Biological Chemistry and Department of Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
31
|
A Drosophila Model of Epidermolysis Bullosa Simplex. J Invest Dermatol 2015; 135:2031-2039. [PMID: 25830653 PMCID: PMC4519992 DOI: 10.1038/jid.2015.129] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/16/2015] [Accepted: 03/24/2015] [Indexed: 12/03/2022]
Abstract
The blistering skin disorder Epidermolysis bullosa simplex (EBS) results from dominant mutations in K5 or K14 genes, encoding the intermediate filament network of basal epidermal keratinocytes. The mechanisms governing keratin network formation and collapse due to EBS mutations remain incompletely understood. Drosophila lacks cytoplasmic intermediate filaments, providing a ‚null’ environment to examine the formation of keratin networks and determine mechanisms by which mutant keratins cause pathology. Here, we report that ubiquitous co-expression of transgenes encoding wild-type human K14 and K5 resulted in the formation of extensive keratin networks in Drosophila epithelial and non-epithelial tissues, causing no overt phenotype. Similar to mammalian cells, treatment of transgenic fly tissues with phosphatase inhibitors caused keratin network collapse, validating Drosophila as a genetic model system to investigate keratin dynamics. Co-expression of K5 and a K14R125C mutant that causes the most severe form of EBS resulted in widespread formation of EBS-like cytoplasmic keratin aggregates in epithelial and non-epithelial fly tissues. Expression of K14R125C/K5 caused semi-lethality; adult survivors developed wing blisters and were flightless due to lack of intercellular adhesion during wing heart development. This Drosophila model of EBS is valuable for the identification of pathways altered by mutant keratins and for development of EBS therapies.
Collapse
|
32
|
Florimond C, Sahin A, Vidilaseris K, Dong G, Landrein N, Dacheux D, Albisetti A, Byard EH, Bonhivers M, Robinson DR. BILBO1 is a scaffold protein of the flagellar pocket collar in the pathogen Trypanosoma brucei. PLoS Pathog 2015; 11:e1004654. [PMID: 25822645 PMCID: PMC4379179 DOI: 10.1371/journal.ppat.1004654] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 01/04/2015] [Indexed: 01/23/2023] Open
Abstract
The flagellar pocket (FP) of the pathogen Trypanosoma brucei is an important single copy structure that is formed by the invagination of the pellicular membrane. It is the unique site of endo- and exocytosis and is required for parasite pathogenicity. The FP consists of distinct structural sub-domains with the least explored being the annulus/horseshoe shaped flagellar pocket collar (FPC). To date the only known component of the FPC is the protein BILBO1, a cytoskeleton protein that has a N-terminus that contains an ubiquitin-like fold, two EF-hand domains, plus a large C-terminal coiled-coil domain. BILBO1 has been shown to bind calcium, but in this work we demonstrate that mutating either or both calcium-binding domains prevents calcium binding. The expression of deletion or mutated forms of BILBO1 in trypanosomes and mammalian cells demonstrate that the coiled-coil domain is necessary and sufficient for the formation of BILBO1 polymers. This is supported by Yeast two-hybrid analysis. Expression of full-length BILBO1 in mammalian cells induces the formation of linear polymers with comma and globular shaped termini, whereas mutation of the canonical calcium-binding domain resulted in the formation of helical polymers and mutation in both EF-hand domains prevented the formation of linear polymers. We also demonstrate that in T. brucei the coiled-coil domain is able to target BILBO1 to the FPC and to form polymers whilst the EF-hand domains influence polymers shape. This data indicates that BILBO1 has intrinsic polymer forming properties and that binding calcium can modulate the form of these polymers. We discuss whether these properties can influence the formation of the FPC. Trypanosoma brucei avoids destruction by, in part, changing its surface glycoprotein coat, which is trafficked onto the cell surface via an invagination of the cell surface called the flagellar pocket. The pocket is essential for pathogenicity. The distal membrane of the pocket is anchored to a cytoskeleton structure called the flagellar pocket collar (FPC). The FPC is a ring/horseshoe shaped structure, which itself is attached to the single copy flagellum of the parasite. How the “ring” shape of the collar is formed is not understood. Moreover, the only known protein component of the FPC is the protein BILBO1. BILBO1 is modular and has a distinct N-terminal domain, two EF-hand calcium-binding domains and a large C-terminal coiled-coil domain. Here we demonstrate that mutating the EF hand domains prevent calcium binding and that the coiled-coil domain is not only sufficient to target to the collar, but can also form polymers in mammalian cells. Mutating either or both calcium-binding domains of BILBO1 influences polymer formation and type when expressed in mammalian and trypanosome cells. Our premise is that BILBO1 has intrinsic polymer forming properties that are essential for the flagellar pocket collar making the pocket a target for intervention.
Collapse
Affiliation(s)
- Célia Florimond
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Annelise Sahin
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Keni Vidilaseris
- Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Gang Dong
- CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France; Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Nicolas Landrein
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Denis Dacheux
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France; Institut Polytechnique de Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR-CNRS 5234, Bordeaux, France
| | - Anna Albisetti
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Edward H Byard
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Mélanie Bonhivers
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| | - Derrick R Robinson
- University Bordeaux, Microbiologie Fondamentale et Pathogenicité, Bordeaux, France; CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
| |
Collapse
|
33
|
Choate KA, Lu Y, Zhou J, Elias PM, Zaidi S, Paller AS, Farhi A, Nelson-Williams C, Crumrine D, Milstone LM, Lifton RP. Frequent somatic reversion of KRT1 mutations in ichthyosis with confetti. J Clin Invest 2015; 125:1703-7. [PMID: 25774499 DOI: 10.1172/jci64415] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/05/2015] [Indexed: 01/19/2023] Open
Abstract
Widespread reversion of genetic disease is rare; however, such events are particularly evident in some skin disorders in which normal clones develop on a background of affected skin. We previously demonstrated that mutations in keratin 10 (KRT10) cause ichthyosis with confetti (IWC), a severe dominant disorder that is characterized by progressive development of hundreds of normal skin spots via revertant mosaicism. Here, we report on a clinical and histological IWC subtype in which affected subjects have red, scaly skin at birth, experience worsening palmoplantar keratoderma in childhood, and develop hundreds of normal skin spots, beginning at around 20 years of age, that increase in size and number over time. We identified a causal de novo mutation in keratin 1 (KRT1). Similar to IWC-causing KRT10 mutations, this mutation in KRT1 resulted in a C-terminal frameshift, replacing 22 C-terminal amino acids with an alternate 30-residue peptide. Mutant KRT1 caused partial collapse of the cytoplasmic intermediate filament network and mislocalized to the nucleus. As with KRT10 mutations causing IWC, reversion of KRT1 mutations occurred via mitotic recombination. Because reversion is not observed with other disease-causing keratin mutations, the results of this study implicate KRT1 and KRT10 C-terminal frameshift mutations in the high frequency of revertant mosaicism in IWC.
Collapse
|
34
|
Nolting JF, Möbius W, Köster S. Mechanics of individual keratin bundles in living cells. Biophys J 2014; 107:2693-9. [PMID: 25468348 PMCID: PMC4255224 DOI: 10.1016/j.bpj.2014.10.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 12/04/2022] Open
Abstract
Along with microtubules and microfilaments, intermediate filaments are a major component of the eukaryotic cytoskeleton and play a key role in cell mechanics. In cells, keratin intermediate filaments form networks of bundles that are sparser in structure and have lower connectivity than, for example, actin networks. Because of this, bending and buckling play an important role in these networks. Buckling events, which occur due to compressive intracellular forces and cross-talk between the keratin network and other cytoskeletal components, are measured here in situ. By applying a mechanical model for the bundled filaments, we can access the mechanical properties of both the keratin bundles themselves and the surrounding cytosol. Bundling is characterized by a coupling parameter that describes the strength of the linkage between the individual filaments within a bundle. Our findings suggest that coupling between the filaments is mostly complete, although it becomes weaker for thicker bundles, with some relative movement allowed.
Collapse
Affiliation(s)
- Jens-Friedrich Nolting
- Institute for X-Ray Physics, Georg-August-Universität Göttingen, Göttingen, Germany; Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Wiebke Möbius
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany; Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, Georg-August-Universität Göttingen, Göttingen, Germany; Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
35
|
Leube RE, Moch M, Kölsch A, Windoffer R. "Panta rhei": Perpetual cycling of the keratin cytoskeleton. BIOARCHITECTURE 2014; 1:39-44. [PMID: 21866261 DOI: 10.4161/bioa.1.1.14815] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 01/12/2011] [Accepted: 01/12/2011] [Indexed: 01/13/2023]
Abstract
The filamentous cytoskeletal systems fulfil seemingly incompatible functions by maintaining a stable scaffolding to ensure tissue integrity and simultaneously facilitating rapid adaptation to intracellular processes and environmental stimuli. This paradox is particularly obvious for the abundant keratin intermediate filaments in epithelial tissues. The epidermal keratin cytoskeleton, for example, supports the protective and selective barrier function of the skin while enabling rapid growth and remodelling in response to physical, chemical and microbial challenges. We propose that these dynamic properties are linked to the perpetual re-cycling of keratin intermediate filaments that we observe in cultured cells. This cycle of assembly and disassembly is independent of protein biosynthesis and consists of distinct, temporally and spatially defined steps. In this way, the keratin cytoskeleton remains in constant motion but stays intact and is also able to restructure rapidly in response to specific regulatory cues as is needed, e.g., during division, differentiation and wound healing.
Collapse
Affiliation(s)
- Rudolf E Leube
- Institute of Molecular and Cellular Anatomy; RWTH Aachen University; Aachen, Germany
| | | | | | | |
Collapse
|
36
|
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
|
37
|
Kayser J, Haslbeck M, Dempfle L, Krause M, Grashoff C, Buchner J, Herrmann H, Bausch AR. The small heat shock protein Hsp27 affects assembly dynamics and structure of keratin intermediate filament networks. Biophys J 2014; 105:1778-85. [PMID: 24138853 DOI: 10.1016/j.bpj.2013.09.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/02/2013] [Accepted: 09/09/2013] [Indexed: 01/07/2023] Open
Abstract
The mechanical properties of living cells are essential for many processes. They are defined by the cytoskeleton, a composite network of protein fibers. Thus, the precise control of its architecture is of paramount importance. Our knowledge about the molecular and physical mechanisms defining the network structure remains scarce, especially for the intermediate filament cytoskeleton. Here, we investigate the effect of small heat shock proteins on the keratin 8/18 intermediate filament cytoskeleton using a well-controlled model system of reconstituted keratin networks. We demonstrate that Hsp27 severely alters the structure of such networks by changing their assembly dynamics. Furthermore, the C-terminal tail domain of keratin 8 is shown to be essential for this effect. Combining results from fluorescence and electron microscopy with data from analytical ultracentrifugation reveals the crucial role of kinetic trapping in keratin network formation.
Collapse
Affiliation(s)
- Jona Kayser
- Lehrstuhl für Zellbiophysik, Technische Universität München, Garching, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
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: 58] [Impact Index Per Article: 5.3] [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
|
39
|
Kim JS, Lee CH, Su BY, Coulombe PA. Mathematical modeling of the impact of actin and keratin filaments on keratinocyte cell spreading. Biophys J 2013. [PMID: 23199911 DOI: 10.1016/j.bpj.2012.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Keratin intermediate filaments (IFs) form cross-linked arrays to fulfill their structural support function in epithelial cells and tissues subjected to external stress. How the cross-linking of keratin IFs impacts the morphology and differentiation of keratinocytes in the epidermis and related surface epithelia remains an open question. Experimental measurements have established that keratinocyte spreading area is inversely correlated to the extent of keratin IF bundling in two-dimensional culture. In an effort to quantitatively explain this relationship, we developed a mathematical model in which isotropic cell spreading is considered as a first approximation. Relevant physical properties such as actin protrusion, adhesion events, and the corresponding response of lamellum formation at the cell periphery are included in this model. Through optimization with experimental data that relate time-dependent changes in keratinocyte surface area during spreading, our simulation results confirm the notion that the organization and mechanical properties of cross-linked keratin filaments affect cell spreading; in addition, our results provide details of the kinetics of this effect. These in silico findings provide further support for the notion that differentiation-related changes in the density and intracellular organization of keratin IFs affect tissue architecture in epidermis and related stratified epithelia.
Collapse
Affiliation(s)
- Jin Seob Kim
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Rebecca L Haines
- Epithelial Biology Group, Institute of Medical Biology, Immunos, Singapore
| | | |
Collapse
|
41
|
Dynamic gradients of an intermediate filament-like cytoskeleton are recruited by a polarity landmark during apical growth. Proc Natl Acad Sci U S A 2013; 110:E1889-97. [PMID: 23641002 DOI: 10.1073/pnas.1305358110] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Intermediate filament (IF)-like cytoskeleton emerges as a versatile tool for cellular organization in all kingdoms of life, underscoring the importance of mechanistically understanding its diverse manifestations. We showed previously that, in Streptomyces (a bacterium with a mycelial lifestyle similar to that of filamentous fungi, including extreme cell and growth polarity), the IF protein FilP confers rigidity to the hyphae by an unknown mechanism. Here, we provide a possible explanation for the IF-like function of FilP by demonstrating its ability to self-assemble into a cis-interconnected regular network in vitro and its localization into structures consistent with a cytoskeletal network in vivo. Furthermore, we reveal that a spatially restricted interaction between FilP and DivIVA, the main component of the Streptomyces polarisome complex, leads to formation of apical gradients of FilP in hyphae undergoing active tip extension. We propose that the coupling between the mechanism driving polar growth and the assembly of an IF cytoskeleton provides each new hypha with an additional stress-bearing structure at its tip, where the nascent cell wall is inevitably more flexible and compliant while it is being assembled and matured. Our data suggest that recruitment of cytoskeleton around a cell polarity landmark is a broadly conserved strategy in tip-growing cells.
Collapse
|
42
|
Shimada H, Nambu-Niibori A, Wilson-Morifuji M, Mizuguchi S, Araki N, Sumiyoshi H, Sato M, Mezaki Y, Senoo H, Ishikawa K, Hatano Y, Okamoto O, Fujiwara S. Epiplakin modifies the motility of the HeLa cells and accumulates at the outer surfaces of 3-D cell clusters. J Dermatol 2013; 40:249-58. [PMID: 23398049 DOI: 10.1111/1346-8138.12076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/21/2012] [Indexed: 11/27/2022]
Abstract
Elimination of epiplakin (EPPK) by gene targeting in mice results in acceleration of keratinocyte migration during wound healing, suggesting that epithelial cellular EPPK may be important for the regulation of cellular motility. To study the function of EPPK, we developed EPPK knock-down (KD) and EPPK-overexpressing HeLa cells and analyzed cellular phenotypes and motility by fluorescence/differential interference contrast time-lapse microscopy and immunolocalization of actin and vimentin. Cellular motility of EPPK-KD cells was significantly elevated, but that of EPPK-overexpressing cells was obviously depressed. Many spike-like projections were observed on EPPK-KD cells, with fewer such structures on overexpressing cells. By contrast, in EPPK-KD cells, expression of E-cadherin was unchanged but vimentin fibers were thinner and sparser than in controls, and they were more concentrated at the peri-nucleus, as observed in migrating keratinocytes at wound edges in EPPK(-/-) mice. In Matrigel 3-D cultures, EPPK co-localized on the outer surface of cell clusters with zonula occludens-1 (ZO-1), a marker of tight junctions. Our results suggest that EPPK is associated with the machinery for cellular motility and contributes to tissue architecture via the rearrangement of intermediate filaments.
Collapse
Affiliation(s)
- Hiromitsu Shimada
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Makarova G, Bette M, Schmidt A, Jacob R, Cai C, Rodepeter F, Betz T, Sitterberg J, Bakowsky U, Moll R, Neff A, Sesterhenn A, Teymoortash A, Ocker M, Werner JA, Mandic R. Epidermal growth factor-induced modulation of cytokeratin expression levels influences the morphological phenotype of head and neck squamous cell carcinoma cells. Cell Tissue Res 2013; 351:59-72. [PMID: 23111772 DOI: 10.1007/s00441-012-1500-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 09/07/2012] [Indexed: 11/30/2022]
Abstract
The migratory ability of tumor cells requires cytoskeletal rearrangement processes. Epidermal growth factor receptor (EGFR)-signaling tightly correlates with tumor progression in head and neck squamous cell carcinomas (HNSCCs), and has previously been implicated in the regulation of cytokeratin (CK) expression. In this study, HNSCC cell lines were treated with EGF, and CK expression levels were monitored by Western blot analysis. Changes in cellular morphology were documented by fluorescence- and atomic force microscopy. Some of the cell lines demonstrated an EGF-dependent modulation of CK expression levels. Interestingly, regression of some CK subtypes or initial up-regulation followed by downregulation at higher EGF-levels could also be observed in the tested cell lines. Overall, the influence of EGF on CK expression levels appeared variable and cell-type-dependent. Real-time cellular analysis of EGF-treated and -untreated HNSCC cell lines demonstrated a rise over time in cellular impedance. In three of the EGF-treated HNSCC cell lines, this rise was markedly higher than in untreated controls, whereas in one of the cell lines the gain of cellular impedance was paradoxically reduced after EGF treatment, which was found to correlate with changes in cellular morphology rather than with relevant changes in cellular viability or proliferation. After treating HNSCC cells with EGF, CK filaments frequently appeared diffusely distributed throughout the cytoplasm, and in some cases were found in a perinuclear localization, the latter being reminiscent to observations by other groups. In summary, the data points to a possible role of EGFR in modulating HNSCC cell morphology.
Collapse
Affiliation(s)
- Galina Makarova
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Baldingerstrasse, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lee CH, Kim MS, Chung BM, Leahy DJ, Coulombe PA. Structural basis for heteromeric assembly and perinuclear organization of keratin filaments. Nat Struct Mol Biol 2012; 19:707-15. [PMID: 22705788 PMCID: PMC3864793 DOI: 10.1038/nsmb.2330] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 05/18/2012] [Indexed: 01/07/2023]
Abstract
There is as yet no high-resolution data regarding the structure and organization of keratin intermediate filaments, which are obligate heteropolymers providing vital mechanical support in epithelia. We report the crystal structure of interacting 2B regions from the central coiled-coil domains of keratins 5 and 14 (K5 and K14), expressed in progenitor keratinocytes of epidermis. The interface of the K5-K14 coiled-coil heterodimer has asymmetric salt bridges, hydrogen bonds and hydrophobic contacts, and its surface exhibits a notable charge polarization. A trans-dimer homotypic disulfide bond involving Cys367 in K14's stutter region occurs in the crystal and in skin keratinocytes, where it is concentrated in a keratin filament cage enveloping the nucleus. We show that K14-Cys367 impacts nuclear shape in cultured keratinocytes and that mouse epidermal keratinocytes lacking K14 show aberrations in nuclear structure, highlighting a new function for keratin filaments.
Collapse
Affiliation(s)
- Chang-Hun Lee
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Min-Sung Kim
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Byung Min Chung
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel J Leahy
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Rotty JD, Coulombe PA. A wound-induced keratin inhibits Src activity during keratinocyte migration and tissue repair. ACTA ACUST UNITED AC 2012; 197:381-9. [PMID: 22529101 PMCID: PMC3341159 DOI: 10.1083/jcb.201107078] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Keratin 6 negatively regulates Src kinase activity and the migratory potential of skin keratinocytes during wound repair. Injury to the epidermis triggers an elaborate homeostatic response resulting in tissue repair and recovery of the vital barrier function. The type II keratins 6a and 6b (K6a and K6b) are among the genes induced early on in wound-proximal keratinocytes and maintained during reepithelialization. Paradoxically, genetic ablation of K6a and K6b results in enhanced keratinocyte migration. In this paper, we show that this trait results from activation of Src kinase and key Src substrates that promote cell migration. Endogenous Src physically associated with keratin proteins in keratinocytes in a K6-dependent fashion. Purified Src bound K6-containing filaments via its SH2 domain in a novel phosphorylation-independent manner, resulting in kinase inhibition. K6 protein was enriched in the detergent-resistant membrane (DRM), a key site of Src inhibition, and DRMs from K6-null keratinocytes were depleted of both keratin and Src. We conclude that K6 negatively regulates Src kinase activity and the migratory potential of skin keratinocytes during wound repair. Our findings may also be important in related contexts such as cancer.
Collapse
Affiliation(s)
- Jeremy D Rotty
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21202, USA
| | | |
Collapse
|
47
|
Rönnberg T, Jääskeläinen K, Blot G, Parviainen V, Vaheri A, Renkonen R, Bouloy M, Plyusnin A. Searching for cellular partners of hantaviral nonstructural protein NSs: Y2H screening of mouse cDNA library and analysis of cellular interactome. PLoS One 2012; 7:e34307. [PMID: 22506017 PMCID: PMC3323627 DOI: 10.1371/journal.pone.0034307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 02/27/2012] [Indexed: 12/29/2022] Open
Abstract
Hantaviruses (Bunyaviridae) are negative-strand RNA viruses with a tripartite genome. The small (S) segment encodes the nucleocapsid protein and, in some hantaviruses, also the nonstructural protein (NSs). The aim of this study was to find potential cellular partners for the hantaviral NSs protein. Toward this aim, yeast two-hybrid (Y2H) screening of mouse cDNA library was performed followed by a search for potential NSs protein counterparts via analyzing a cellular interactome. The resulting interaction network was shown to form logical, clustered structures. Furthermore, several potential binding partners for the NSs protein, for instance ACBD3, were identified and, to prove the principle, interaction between NSs and ACBD3 proteins was demonstrated biochemically.
Collapse
Affiliation(s)
- Tuomas Rönnberg
- Department of Virology, Haartman Institute, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Windoffer R, Beil M, Magin TM, Leube RE. Cytoskeleton in motion: the dynamics of keratin intermediate filaments in epithelia. ACTA ACUST UNITED AC 2012; 194:669-78. [PMID: 21893596 PMCID: PMC3171125 DOI: 10.1083/jcb.201008095] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epithelia are exposed to multiple forms of stress. Keratin intermediate filaments are abundant in epithelia and form cytoskeletal networks that contribute to cell type–specific functions, such as adhesion, migration, and metabolism. A perpetual keratin filament turnover cycle supports these functions. This multistep process keeps the cytoskeleton in motion, facilitating rapid and protein biosynthesis–independent network remodeling while maintaining an intact network. The current challenge is to unravel the molecular mechanisms underlying the regulation of the keratin cycle in relation to actin and microtubule networks and in the context of epithelial tissue function.
Collapse
Affiliation(s)
- Reinhard Windoffer
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52057 Aachen, Germany
| | | | | | | |
Collapse
|
49
|
Chung BM, Murray CI, Van Eyk JE, Coulombe PA. Identification of novel interaction between annexin A2 and keratin 17: evidence for reciprocal regulation. J Biol Chem 2012; 287:7573-81. [PMID: 22235123 DOI: 10.1074/jbc.m111.301549] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Keratins are cytoplasmic intermediate filament proteins providing crucial structural support in epithelial cells. Keratin expression has diagnostic and even prognostic value in disease settings, and recent studies have uncovered modulatory roles for select keratin proteins in signaling pathways regulating cell growth and cell death. Elevated keratin expression in select cancers is correlated with higher expression of EGF receptor (EGFR), whose overexpression and/or mutation give rise to cancer. To explore the role of keratins in oncogenic signaling pathways, we examined the regulation of epithelial growth-associated keratin 17 (K17) in response to EGFR activation. K17 is specifically up-regulated in detergent-soluble fraction upon EGFR activation, and immunofluorescence analysis revealed alterations in K17-containing filaments. Interestingly, we identified AnxA2 as a novel interacting partner of K17, and this interaction is antagonized by EGFR activation. K17 and AnxA2 proteins show reciprocal regulation. Modulating expression of AnxA2 altered K17 stability, and AnxA2 overexpression delays EGFR-mediated change in K17 detergent solubility. Down-regulation of K17 expression, in turn, results in decreased AnxA2 phosphorylation at Tyr-23. These findings uncover a novel interaction involving K17 and AnxA2 and identify AnxA2 as a potential regulator of keratin filaments.
Collapse
Affiliation(s)
- Byung-Min Chung
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
50
|
Strnad P, Usachov V, Debes C, Gräter F, Parry DAD, Omary MB. Unique amino acid signatures that are evolutionarily conserved distinguish simple-type, epidermal and hair keratins. J Cell Sci 2012; 124:4221-32. [PMID: 22215855 DOI: 10.1242/jcs.089516] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Keratins (Ks) consist of central α-helical rod domains that are flanked by non-α-helical head and tail domains. The cellular abundance of keratins, coupled with their selective cell expression patterns, suggests that they diversified to fulfill tissue-specific functions although the primary structure differences between them have not been comprehensively compared. We analyzed keratin sequences from many species: K1, K2, K5, K9, K10, K14 were studied as representatives of epidermal keratins, and compared with K7, K8, K18, K19, K20 and K31, K35, K81, K85, K86, which represent simple-type (single-layered or glandular) epithelial and hair keratins, respectively. We show that keratin domains have striking differences in their amino acids. There are many cysteines in hair keratins but only a small number in epidermal keratins and rare or none in simple-type keratins. The heads and/or tails of epidermal keratins are glycine and phenylalanine rich but alanine poor, whereas parallel domains of hair keratins are abundant in prolines, and those of simple-type epithelial keratins are enriched in acidic and/or basic residues. The observed differences between simple-type, epidermal and hair keratins are highly conserved throughout evolution. Cysteines and histidines, which are infrequent keratin amino acids, are involved in de novo mutations that are markedly overrepresented in keratins. Hence, keratins have evolutionarily conserved and domain-selectively enriched amino acids including glycine and phenylalanine (epidermal), cysteine and proline (hair), and basic and acidic (simple-type epithelial), which reflect unique functions related to structural flexibility, rigidity and solubility, respectively. Our findings also support the importance of human keratin 'mutation hotspot' residues and their wild-type counterparts.
Collapse
Affiliation(s)
- Pavel Strnad
- Department of Internal Medicine I, Center for Internal Medicine, University Medical Center Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | | | | | | | | | | |
Collapse
|