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
|
WAKITANI S, KAWABATA R, SHIRATAKI S, YASUDA M. Distribution of keratin subtypes in the thymus of Japanese black calves during acute thymic involution. J Vet Med Sci 2025; 87:131-134. [PMID: 39631889 PMCID: PMC11830441 DOI: 10.1292/jvms.24-0413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Immunohistochemistry for keratins 5, 8, 14, and 18 was performed on Japanese Black calf thymuses at various stages of acute thymic involution. Keratins 5 and 14 were predominantly localized in the thymic medulla, while keratins 8 and 18 were broadly distributed throughout the parenchyma. Despite thymic involution, the distribution patterns of these keratins remained consistent. The cortical area, assessed by keratin 5 staining, progressively decreased with involution but retained approximately 40% of the total parenchyma even at the most severe stage. These results suggest that the thymic cortex shrinks but does not completely disappear during acute thymic involution in calves.
Collapse
Affiliation(s)
- Shoichi WAKITANI
- Laboratory of Veterinary Anatomy, Faculty of Agriculture,
University of Miyazaki, Miyazaki, Japan
| | - Risako KAWABATA
- Laboratory of Veterinary Anatomy, Faculty of Agriculture,
University of Miyazaki, Miyazaki, Japan
| | - Sora SHIRATAKI
- Laboratory of Veterinary Anatomy, Faculty of Agriculture,
University of Miyazaki, Miyazaki, Japan
| | - Masahiro YASUDA
- Laboratory of Veterinary Anatomy, Faculty of Agriculture,
University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
3
|
Outla Z, Prechova M, Korelova K, Gemperle J, Gregor M. Mechanics of cell sheets: plectin as an integrator of cytoskeletal networks. Open Biol 2025; 15:240208. [PMID: 39875099 PMCID: PMC11774597 DOI: 10.1098/rsob.240208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Epithelia are multicellular sheets that form barriers defining the internal and external environments. The constant stresses acting at this interface require that epithelial sheets are mechanically robust and provide a selective barrier to the hostile exterior. These properties are mediated by cellular junctions which are physically linked with heavily crosslinked cytoskeletal networks. Such hardwiring is facilitated by plakins, a family of giant modular proteins which serve as 'molecular bridges' between different cytoskeletal filaments and multiprotein adhesion complexes. Dysfunction of cytoskeletal crosslinking compromises epithelial biomechanics and structural integrity. Subsequent loss of barrier function leads to disturbed tissue homeostasis and pathological consequences such as skin blistering or intestinal inflammation. In this article, we highlight the importance of the cytolinker protein plectin for the functional organization of epithelial cytoskeletal networks. In particular, we focus on the ability of plectin to act as an integrator of the epithelial cytoarchitecture that defines the biomechanics of the whole tissue. Finally, we also discuss the role of cytoskeletal crosslinking in emerging aspects of epithelial mechanobiology that are critical for the maintenance of epithelial homeostasis.
Collapse
Affiliation(s)
- Zuzana Outla
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magdalena Prechova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Gemperle
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
4
|
Li J, Li S, Zhang Q, Liang M, Chen X, Feng Y, Pan Z, Hu T, Wu Q, Chen G, Zouboulis CC, Mo X, Ju Q. Apocrine Gland Damage and the Release of Specific Keratins in Early Stage Indicate the Crucial Involvement of Apocrine Glands in Hidradenitis Suppurativa. J Invest Dermatol 2024:S0022-202X(24)02893-8. [PMID: 39547394 DOI: 10.1016/j.jid.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 11/17/2024]
Abstract
The apocrine glands (AGs) are not considered to be primarily involved in hidradenitis suppurativa (HS). This study investigated the potential role of AGs in HS pathogenesis using immunohistochemistry and single-cell sequencing of nonlesional skin and early lesional skin (LS) from patients with HS (n = 12) and healthy controls (n = 8). AG cell destruction was more frequent, and AG size was significantly reduced in the nonlesional skin and LS. Barrier-related genes (eg, CLDN1 and CDH1) were downregulated in the AGs of the nonlesional skin and LS. Damaged AGs in the LS primarily recruited and activated neutrophils through the CXCL-CXCR and SAA1-FPR2 pathways. Elevated levels of specific keratins (keratin 18 and keratin 19) released from damaged AGs were observed on the skin surface of patients and were associated with disease severity. Keratin 19 was also detected in the dermis of the nonlesional skin and LS and was surrounded by neutrophils and macrophages. Moreover, serum keratin 19 levels in patients (N = 20) were significantly negatively correlated with the age at HS onset. Collectively, our findings provide previously unreported evidence that the AGs are damaged and release specific keratins in early HS lesions, indicating a crucial role of the AGs in HS pathogenesis.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Sitong Li
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Qiujing Zhang
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Mengchen Liang
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Xiang Chen
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yibo Feng
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Zhanyan Pan
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Tingting Hu
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Qiong Wu
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Christos C Zouboulis
- Department of Dermatology, Venereology, Allergology and Immunology, Staedtisches Klinikum Dessau, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Dessau, Germany
| | - Xiaohui Mo
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| | - Qiang Ju
- Department of Dermatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Cohen E, Johnson CN, Wasikowski R, Billi AC, Tsoi LC, Kahlenberg JM, Gudjonsson JE, Coulombe PA. Significance of stress keratin expression in normal and diseased epithelia. iScience 2024; 27:108805. [PMID: 38299111 PMCID: PMC10828818 DOI: 10.1016/j.isci.2024.108805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
A group of keratin intermediate filament genes, the type II KRT6A-C and type I KRT16 and KRT17, are deemed stress responsive as they are induced in keratinocytes of surface epithelia in response to environmental stressors, in skin disorders (e.g., psoriasis) and in carcinomas. Monitoring stress keratins is widely used to identify keratinocytes in an activated state. Here, we analyze single-cell transcriptomic data from healthy and diseased human skin to explore the properties of stress keratins. Relative to keratins occurring in healthy skin, stress-induced keratins are expressed at lower levels and show lesser type I-type II pairwise regulation. Stress keratins do not "replace" the keratins expressed during normal differentiation nor reflect cellular proliferation. Instead, stress keratins are consistently co-regulated with genes with roles in differentiation, inflammation, and/or activation of innate immunity at the single-cell level. These findings provide a roadmap toward explaining the broad diversity and contextual regulation of keratins.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Craig N. Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Allison C. Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pierre A. Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
6
|
Ober-Reynolds B, Wang C, Ko JM, Rios EJ, Aasi SZ, Davis MM, Oro AE, Greenleaf WJ. Integrated single-cell chromatin and transcriptomic analyses of human scalp identify gene-regulatory programs and critical cell types for hair and skin diseases. Nat Genet 2023; 55:1288-1300. [PMID: 37500727 PMCID: PMC11190942 DOI: 10.1038/s41588-023-01445-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 06/17/2023] [Indexed: 07/29/2023]
Abstract
Genome-wide association studies have identified many loci associated with hair and skin disease, but identification of causal variants requires deciphering of gene-regulatory networks in relevant cell types. We generated matched single-cell chromatin profiles and transcriptomes from scalp tissue from healthy controls and patients with alopecia areata, identifying diverse cell types of the hair follicle niche. By interrogating these datasets at multiple levels of cellular resolution, we infer 50-100% more enhancer-gene links than previous approaches and show that aggregate enhancer accessibility for highly regulated genes predicts expression. We use these gene-regulatory maps to prioritize cell types, genes and causal variants implicated in the pathobiology of androgenetic alopecia (AGA), eczema and other complex traits. AGA genome-wide association studies signals are enriched in dermal papilla regulatory regions, supporting the role of these cells as drivers of AGA pathogenesis. Finally, we train machine learning models to nominate single-nucleotide polymorphisms that affect gene expression through disruption of transcription factor binding, predicting candidate functional single-nucleotide polymorphism for AGA and eczema.
Collapse
Affiliation(s)
| | - Chen Wang
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Dermatology, Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- Institute of Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Justin M Ko
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Eon J Rios
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Dermatology, Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Sumaira Z Aasi
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Mark M Davis
- Institute of Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, USA
| | - Anthony E Oro
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
7
|
Quadri M, Tiso N, Musmeci F, Morasso MI, Brooks SR, Bonetti LR, Panini R, Lotti R, Marconi A, Pincelli C, Palazzo E. CD271 activation prevents low to high-risk progression of cutaneous squamous cell carcinoma and improves therapy outcomes. J Exp Clin Cancer Res 2023; 42:167. [PMID: 37443031 DOI: 10.1186/s13046-023-02737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (cSCC) is the second most prevalent form of skin cancer, showing a rapid increasing incidence worldwide. Although most cSCC can be cured by surgery, a sizeable number of cases are diagnosed at advanced stages, with local invasion and distant metastatic lesions. In the skin, neurotrophins (NTs) and their receptors (CD271 and Trk) form a complex network regulating epidermal homeostasis. Recently, several works suggested a significant implication of NT receptors in cancer. However, CD271 functions in epithelial tumors are controversial and its precise role in cSCC is still to be defined. METHODS Spheroids from cSCC patients with low-risk (In situ or Well-Differentiated cSCC) or high-risk tumors (Moderately/Poorly Differentiated cSCC), were established to explore histological features, proliferation, invasion abilities, and molecular pathways modulated in response to CD271 overexpression or activation in vitro. The effect of CD271 activities on the response to therapeutics was also investigated. The impact on the metastatic process and inflammation was explored in vivo and in vitro, by using zebrafish xenograft and 2D/3D models. RESULTS Our data proved that CD271 is upregulated in Well-Differentiated tumors as compared to the more aggressive Moderately/Poorly Differentiated cSCC, both in vivo and in vitro. We demonstrated that CD271 activities reduce proliferation and malignancy marker expression in patient-derived cSCC spheroids at each tumor grade, by increasing neoplastic cell differentiation. CD271 overexpression significantly increases cSCC spheroid mass density, while it reduces their weight and diameter, and promotes a major fold-enrichment in differentiation and keratinization genes. Moreover, both CD271 overexpression and activation decrease cSCC cell invasiveness in vitro. A significant inhibition of the metastatic process by CD271 was observed in a newly established zebrafish cSCC model. We found that the recruitment of leucocytes by CD271-overexpressing cells directly correlates with tumor killing and this finding was further highlighted by monocyte infiltration in a THP-1-SCC13 3D model. Finally, CD271 activity synergizes with Trk receptor inhibition, by reducing spheroid viability, and significantly improves the outcome of photodynamic therapy (PTD) or chemotherapy in spheroids and zebrafish. CONCLUSION Our study provides evidence that CD271 could prevent the switch between low to high-risk cSCC tumors. Because CD271 contributes to maintaining active differentiative paths and favors the response to therapies, it might be a promising target for future pharmaceutical development.
Collapse
Affiliation(s)
- Marika Quadri
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Natascia Tiso
- Laboratory of Developmental Genetics, Department of Biology, University of Padova, Padova, Italy
| | | | - Maria I Morasso
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Luca Reggiani Bonetti
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossana Panini
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Lotti
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Alessandra Marconi
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Carlo Pincelli
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy
| | - Elisabetta Palazzo
- DermoLAB, Department of Surgical, Medical, Dental and Morphological Science, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
8
|
Cohen B, Cadesky A, Jaggi S. Dermatologic manifestations of thyroid disease: a literature review. Front Endocrinol (Lausanne) 2023; 14:1167890. [PMID: 37251685 PMCID: PMC10214500 DOI: 10.3389/fendo.2023.1167890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/07/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Thyroid hormone is considered one of the key regulatory hormones for skin homeostasis. Multiple organs are affected by the release of peripheral thyroid hormones (T4 and T3) further regulating various functions at a cellular level. Specifically, skin is considered an important target organ in which the thyroid hormone has a significant impact. Multiple skin diseases are associated with thyroid hormone dysregulation. However, other striking dermatologic manifestations are seen in nails and hair as well. Hypothyroidism, hyperthyroidism, and thyroid cancer can have an array of cutaneous manifestations, and we present the recent updates in this field. Methods A PubMed search was performed for updates in any new skin disease findings and treatments between 2010 and 2022. Research published in the past decade and previously known foundational skin findings associated with thyroid disease were presented in this review. Conclusion Cutaneous manifestations of thyroid disease is one of the first notable signs of thyroid hormone dysregulation. This article reviews the recent updates on the thyroid and skin interplay, and it further discusses overt visible findings and various available treatment modalities.
Collapse
|
9
|
A Kaleidoscope of Keratin Gene Expression and the Mosaic of Its Regulatory Mechanisms. Int J Mol Sci 2023; 24:ijms24065603. [PMID: 36982676 PMCID: PMC10052683 DOI: 10.3390/ijms24065603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Keratins are a family of intermediate filament-forming proteins highly specific to epithelial cells. A combination of expressed keratin genes is a defining property of the epithelium belonging to a certain type, organ/tissue, cell differentiation potential, and at normal or pathological conditions. In a variety of processes such as differentiation and maturation, as well as during acute or chronic injury and malignant transformation, keratin expression undergoes switching: an initial keratin profile changes accordingly to changed cell functions and location within a tissue as well as other parameters of cellular phenotype and physiology. Tight control of keratin expression implies the presence of complex regulatory landscapes within the keratin gene loci. Here, we highlight patterns of keratin expression in different biological conditions and summarize disparate data on mechanisms controlling keratin expression at the level of genomic regulatory elements, transcription factors (TFs), and chromatin spatial structure.
Collapse
|
10
|
Ievlev V, Lynch TJ, Freischlag KW, Gries CB, Shah A, Pai AC, Ahlers BA, Park S, Engelhardt JF, Parekh KR. Krt14 and Krt15 differentially regulate regenerative properties and differentiation potential of airway basal cells. JCI Insight 2023; 8:162041. [PMID: 36512409 PMCID: PMC9977304 DOI: 10.1172/jci.insight.162041] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Keratin expression dynamically changes in airway basal cells (BCs) after acute and chronic injury, yet the functional consequences of these changes on BC behavior remain unknown. In bronchiolitis obliterans (BO) after lung transplantation, BC clonogenicity declines, which is associated with a switch from keratin15 (Krt15) to keratin14 (Krt14). We investigated these keratins' roles using Crispr-KO in vitro and in vivo and found that Krt14-KO and Krt15-KO produce contrasting phenotypes in terms of differentiation and clonogenicity. Primary mouse Krt14-KO BCs did not differentiate into club and ciliated cells but had enhanced clonogenicity. By contrast, Krt15-KO did not alter BC differentiation but impaired clonogenicity in vitro and reduced the number of label-retaining BCs in vivo after injury. Krt14, but not Krt15, bound the tumor suppressor stratifin (Sfn). Disruption of Krt14, but not of Krt15, reduced Sfn protein abundance and increased expression of the oncogene dNp63a during BC differentiation, whereas dNp63a levels were reduced in Krt15-KO BCs. Overall, the phenotype of Krt15-KO BCs contrasts with Krt14-KO phenotype and resembles the phenotype in BO with decreased clonogenicity, increased Krt14, and decreased dNp63a expression. This work demonstrates that Krt14 and Krt15 functionally regulate BC behavior, which is relevant in chronic disease states like BO.
Collapse
Affiliation(s)
- Vitaly Ievlev
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Thomas J. Lynch
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Kyle W. Freischlag
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Caitlyn B. Gries
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Anit Shah
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Albert C. Pai
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Bethany A. Ahlers
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| | - Soo Park
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Kalpaj R. Parekh
- Department of Cardiothoracic Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
11
|
Cohen E, Johnson C, Redmond CJ, Nair RR, Coulombe PA. Revisiting the significance of keratin expression in complex epithelia. J Cell Sci 2022; 135:jcs260594. [PMID: 36285538 PMCID: PMC10658788 DOI: 10.1242/jcs.260594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 03/17/2023] Open
Abstract
A large group of keratin genes (n=54 in the human genome) code for intermediate filament (IF)-forming proteins and show differential regulation in epithelial cells and tissues. Keratin expression can be highly informative about the type of epithelial tissue, differentiation status of constituent cells and biological context (e.g. normal versus diseased settings). The foundational principles underlying the use of keratin expression to gain insight about epithelial cells and tissues primarily originated in pioneering studies conducted in the 1980s. The recent emergence of single cell transcriptomics provides an opportunity to revisit these principles and gain new insight into epithelial biology. Re-analysis of single-cell RNAseq data collected from human and mouse skin has confirmed long-held views regarding the quantitative importance and pairwise regulation of specific keratin genes in keratinocytes of surface epithelia. Furthermore, such analyses confirm and extend the notion that changes in keratin gene expression occur gradually as progenitor keratinocytes commit to and undergo differentiation, and challenge the prevailing assumption that specific keratin combinations reflect a mitotic versus a post-mitotic differentiating state. Our findings provide a blueprint for similar analyses in other tissues, and warrant a more nuanced approach in the use of keratin genes as biomarkers in epithelia.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Catherine J. Redmond
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Raji R. Nair
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pierre A. Coulombe
- 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
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Kashyap MP, Khan J, Sinha R, Jin L, Atigadda V, Deshane JS, Ahmed AR, Kilic A, Raman C, Mukhtar MS, Elmets CA, Athar M. Advances in molecular pathogenesis of hidradenitis suppurativa: Dysregulated keratins and ECM signaling. Semin Cell Dev Biol 2022; 128:120-129. [PMID: 35131152 PMCID: PMC9232849 DOI: 10.1016/j.semcdb.2022.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022]
Abstract
Hidradenitis suppurativa (HS) is characterized by deep-seated, highly inflamed, and painful lumps/abscesses, fistulae, and sinus tracts that grow extensively deep in the dermis and are highly immunogenic in nature. In about one-third of the HS patients there is strong evidence for the role of γ-secretase mutations along with dysregulated Notch signaling. However, the contribution of dysregulated Notch signaling in HS pathogenesis in relation to hair follicle alterations and hyper-activation of the immune system remains undefined. A genome-wide association study (GWAS), proteomic data and functional investigations of identified sequence variants in HS pathology are not fully revealing. The disease initiation or progression may involve bacterial infection besides intrinsic functional defects in keratinocytes, which may be key to further exacerbate immune cell infiltration and cytokine production in and around the lesional tissue. The absence of a suitable animal model that could fully recapitulate the pathogenesis of HS is a major impediment for proper understanding the underlying mechanisms and development of effective treatments. The presence of extracellular matrix (ECM) degradation products along with dysregulation in keratinocytes and, dermal fibroblasts ultimately affect immune regulation and are various components of HS pathogenesis. Bacterial infection further exacerbates the complexity of the disease progression. While anti-TNFα therapy shows partial efficacy, treatment to cure HS is absent. Multiple clinical trials targeting various cytokines, complement C5a and ECM products are in progress. This review provides state-of-the-art information on these aspects with a focus on dysregulated keratinocyte and immune cells; and role of ECM, and Keratin functions in this regard.
Collapse
Affiliation(s)
- Mahendra Pratap Kashyap
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Jasim Khan
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Rajesh Sinha
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Lin Jin
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Venkatram Atigadda
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Jessy S Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Ayesha R Ahmed
- Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Ali Kilic
- Division of Plastic Surgery, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Chander Raman
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - M Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Craig A Elmets
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA
| | - Mohammad Athar
- UAB Research Center of Excellence in Arsenicals, Department of Dermatology, University of Alabama at Birmingham, Birmingham AL35294, USA.
| |
Collapse
|
13
|
Warrick E, Duval C, Nouveau S, Piffaut V, Bourreau E, Bastien P, de Lacharrière O, Morita A, Bernerd F. Actinic lentigines from Japanese and European volunteers share similar impaired biological functions. J Dermatol Sci 2022; 107:8-16. [DOI: 10.1016/j.jdermsci.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/09/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022]
|
14
|
Rai A, Ferrão R, Marta D, Vilaça A, Lino M, Rondão T, Ji J, Paiva A, Ferreira L. Antimicrobial Peptide-Tether Dressing Able to Enhance Wound Healing by Tissue Contact. ACS APPLIED MATERIALS & INTERFACES 2022; 14:24213-24228. [PMID: 35584375 DOI: 10.1021/acsami.2c06601] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
No effective therapeutic dressings are currently available in the market that can prevent bacterial infection and simultaneously promote skin regeneration in diabetic patients. The lack of re-epithelization, prevalence of inflammation, and high risk of infection are hallmarks of non-healing wounds. Here, we have evaluated the antimicrobial and pro-regenerative effect of a relatively non-leaching LL37 peptide immobilized in polyurethane (PU)-based wound dressings (PU-adhesive-LL37 dressing). The PU-adhesive-LL37 (63 μg LL37NPs/cm2) dressing killed Gram-positive and Gram-negative bacteria in human serum without inducing bacterial resistance after 16 antimicrobial test cycles in contrast to commercially available dressings with the capacity to release antimicrobial Ag ions. Importantly, type II diabetic mice (db/db mice) treated with the PU-adhesive-LL37 dressing for different periods of time (6 or 14 days) showed enhanced wound healing and re-epithelialization (i.e., high keratin 14/5 levels) and lower macrophage infiltration in the wounds compared to animals treated with PU. The wounds treated with PU-adhesive-LL37 dressings showed also low expression of pro-inflammatory cytokines such as TNF-α and IL6 after 6 days of treatment, indicating that they act as an anti-inflammatory dressing. Additionally, PU-adhesive-LL37 dressings do not induce acute inflammatory responses in the peripheral blood mononuclear cells (PBMCs) after 3 days of exposure, in contrast to controls. Taken together, PU-adhesive-LL37NP dressings might prevent the bacterial infections and facilitate wound healing by tissue contact, inducing re-epithelialization and anti-inflammatory processes in diabetic conditions.
Collapse
Affiliation(s)
- Akhilesh Rai
- Faculty of Medicine, University of Coimbra, Coimbra 3000-354, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Rafaela Ferrão
- Faculty of Medicine, University of Coimbra, Coimbra 3000-354, Portugal
| | - Denise Marta
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Andreia Vilaça
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Miguel Lino
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Tiago Rondão
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Artur Paiva
- Unidade de Gestão Operacional de Citometria, Serviço de Patologia Clínica, Centro Hospitalar e Universitário de Coimbra, Coimbra 3001-301, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculdade de Medicina, Universidade de Coimbra, Polo III-Health Sciences Campus, Coimbra 3000-548, Portugal
- ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, Coimbra 3040-854, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra 3000-354, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| |
Collapse
|
15
|
Human iPSC-derived-keratinocytes, a useful model to identify and explore pathological phenotype of Epidermolysis Bullosa Simplex. J Invest Dermatol 2022; 142:2695-2705.e11. [PMID: 35490743 DOI: 10.1016/j.jid.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022]
Abstract
Epidermolysis Bullosa Simplex (EBS), an autosomal dominant skin disorder, is characterized by skin fragility. Genetically, majority of cases are related to missense mutations in two keratin genes, KRT5 or KRT14, leading to cytolysis of basal keratinocytes and intraepidermal blistering. Progress towards identification of treatments have been hampered by incomplete understanding of the mechanisms underlying this disease, and availability of relevant and reliable in vitro models recapitulating the physiopathological mechanisms. Recent advances in stem cell field have fueled the prospect that these limitations could be overcome thanks to the availability of disease-specific human induced pluripotent stem cells (hiPSC). Here, we generated hiPSC-derived keratinocytes from patients carrying KRT5 dominant mutations and compared them to non-affected hiPSC-derived keratinocytes as well as their primary counterparts. Our results demonstrated that EBS hiPSC-derived keratinocytes displayed proliferative defects, increased capacity to migrate, alteration of ERK signaling pathway and cytoplasmic keratin filament aggregates as observed in primary EBS keratinocytes. Of interest, EBS hiPSC-derived keratinocytes exhibited a downregulation of hemidesmosomal proteins revealing the different effects of KRT5 mutations on keratin cytoskeletal organization. Combination of culture miniaturization and treatment with the chaperone molecule 4-PBA, our results demonstrated that hiPSC-derived keratinocytes represent a suitable model for identifying novel therapies for EBS.
Collapse
|
16
|
Evtushenko NA, Beilin AK, Kosykh AV, Vorotelyak EA, Gurskaya NG. Keratins as an Inflammation Trigger Point in Epidermolysis Bullosa Simplex. Int J Mol Sci 2021; 22:ijms222212446. [PMID: 34830328 PMCID: PMC8624175 DOI: 10.3390/ijms222212446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/21/2022] Open
Abstract
Epidermolysis bullosa simplex (EBS) is a group of inherited keratinopathies that, in most cases, arise due to mutations in keratins and lead to intraepidermal ruptures. The cellular pathology of most EBS subtypes is associated with the fragility of the intermediate filament network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Mutations in keratins 5/14 or in other genes that encode associated proteins induce structural disarrangements of different strengths depending on their locations in the genes. Keratin aggregates display impaired dynamics of assembly and diminished solubility and appear to be the trigger for endoplasmic reticulum (ER) stress upon being phosphorylated by MAPKs. Global changes in cellular signaling mainly occur in cases of severe dominant EBS mutations. The spectrum of changes initiated by phosphorylation includes the inhibition of proteasome degradation, TNF-α signaling activation, deregulated proliferation, abnormal cell migration, and impaired adherence of keratinocytes. ER stress also leads to the release of proinflammatory danger-associated molecular pattern (DAMP) molecules, which enhance avalanche-like inflammation. Many instances of positive feedback in the course of cellular stress and the development of sterile inflammation led to systemic chronic inflammation in EBS. This highlights the role of keratin in the maintenance of epidermal and immune homeostasis.
Collapse
Affiliation(s)
- Nadezhda A. Evtushenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
| | - Arkadii K. Beilin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova 26, 119334 Moscow, Russia;
| | - Anastasiya V. Kosykh
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
| | - Ekaterina A. Vorotelyak
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova 26, 119334 Moscow, Russia;
| | - Nadya G. Gurskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117997 Moscow, Russia; (N.A.E.); (A.K.B.); (A.V.K.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Correspondence:
| |
Collapse
|
17
|
Mancino G, Miro C, Di Cicco E, Dentice M. Thyroid hormone action in epidermal development and homeostasis and its implications in the pathophysiology of the skin. J Endocrinol Invest 2021; 44:1571-1579. [PMID: 33683663 PMCID: PMC8285348 DOI: 10.1007/s40618-020-01492-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Thyroid hormones (THs) are key endocrine regulators of tissue development and homeostasis. They are constantly released into the bloodstream and help to regulate many cell functions. The principal products released by the follicular epithelial cells are T3 and T4. T4, which is the less active form of TH, is produced in greater amounts than T3, which is the most active form of TH. This mechanism highlights the importance of the peripheral regulation of TH levels that goes beyond the central axis. Skin, muscle, liver, bone and heart are finely regulated by TH. In particular, skin is among the target organs most influenced by TH, which is essential for skin homeostasis. Accordingly, skin diseases are associated with an altered thyroid status. Alopecia, dermatitis and vitiligo are associated with thyroiditis and alopecia and eczema are frequently correlated with the Graves' disease. However, only in recent decades have studies started to clarify the molecular mechanisms underlying the effects of TH in epidermal homeostasis. Herein, we summarize the most frequent clinical epidermal alterations linked to thyroid diseases and review the principal mechanisms involved in TH control of keratinocyte proliferation and functional differentiation. Our aim is to define the open questions in this field that are beginning to be elucidated thanks to the advent of mouse models of altered TH metabolism and to obtain novel insights into the physiopathological consequences of TH metabolism on the skin.
Collapse
Affiliation(s)
- G Mancino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - C Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - E Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - M Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini 5, 80131, Naples, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Naples, Italy.
| |
Collapse
|
18
|
Deng Z, Cangkrama M, Butt T, Jane SM, Carpinelli MR. Grainyhead-like transcription factors: guardians of the skin barrier. Vet Dermatol 2021; 32:553-e152. [PMID: 33843098 DOI: 10.1111/vde.12956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023]
Abstract
There has been selective pressure to maintain a skin barrier since terrestrial animals evolved 360 million years ago. These animals acquired an unique integumentary system with a keratinized, stratified, squamous epithelium surface barrier. The barrier protects against dehydration and entry of microbes and toxins. The skin barrier centres on the stratum corneum layer of the epidermis and consists of cornified envelopes cemented by the intercorneocyte lipid matrix. Multiple components of the barrier undergo cross-linking by transglutaminase (TGM) enzymes, while keratins provide additional mechanical strength. Cellular tight junctions also are crucial for barrier integrity. The grainyhead-like (GRHL) transcription factors regulate the formation and maintenance of the integument in diverse species. GRHL3 is essential for formation of the skin barrier during embryonic development, whereas GRHL1 maintains the skin barrier postnatally. This is achieved by transactivation of Tgm1 and Tgm5, respectively. In addition to its barrier function, GRHL3 plays key roles in wound repair and as an epidermal tumour suppressor. In its former role, GRHL3 activates the planar cell polarity signalling pathway to mediate wound healing by providing directional migration cues. In squamous epithelium, GRHL3 regulates the balance between proliferation and differentiation, and its loss induces squamous cell carcinoma (SCC). In the skin, this is mediated through increased expression of MIR21, which reduces the expression levels of GRHL3 and its direct target, PTEN, leading to activation of the PI3K-AKT signalling pathway. These data position the GRHL family as master regulators of epidermal homeostasis across a vast gulf of evolutionary history.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Michael Cangkrama
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Tariq Butt
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen M Jane
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Marina R Carpinelli
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
19
|
Sjöqvist M, Antfolk D, Suarez-Rodriguez F, Sahlgren C. From structural resilience to cell specification - Intermediate filaments as regulators of cell fate. FASEB J 2020; 35:e21182. [PMID: 33205514 PMCID: PMC7839487 DOI: 10.1096/fj.202001627r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
During the last decades intermediate filaments (IFs) have emerged as important regulators of cellular signaling events, ascribing IFs with functions beyond the structural support they provide. The organ and developmental stage‐specific expression of IFs regulate cell differentiation within developing or remodeling tissues. Lack of IFs causes perturbed stem cell differentiation in vasculature, intestine, nervous system, and mammary gland, in transgenic mouse models. The aberrant cell fate decisions are caused by deregulation of different stem cell signaling pathways, such as Notch, Wnt, YAP/TAZ, and TGFβ. Mutations in genes coding for IFs cause an array of different diseases, many related to stem cell dysfunction, but the molecular mechanisms remain unresolved. Here, we provide a comprehensive overview of how IFs interact with and regulate the activity, localization and function of different signaling proteins in stem cells, and how the assembly state and PTM profile of IFs may affect these processes. Identifying when, where and how IFs and cell signaling congregate, will expand our understanding of IF‐linked stem cell dysfunction during development and disease.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Freddy Suarez-Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
20
|
Kao WWY. Keratin expression by corneal and limbal stem cells during development. Exp Eye Res 2020; 200:108206. [PMID: 32882212 DOI: 10.1016/j.exer.2020.108206] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Keratins are the forming units of intermediate filaments (IF) that provide mechanical support, and formation of desmosomes between cells and hemi desmosomes with basement membranes for epithelium integrity. Keratin IF are polymers of obligate heterodimer consisting one type I keratin and one type II keratin molecules. There are 54 functional keratin genes in human genome, which are classified into three major groups, i.e., epithelial keratins, hair follicle cell-specific epithelial keratins and hair keratins. Their expression is cell type-specific and developmentally regulated. Corneal epithelium expresses a subgroup of keratins similar to those of epidermal epithelium. Limbal basal stem cells express K5/K14, and K8/K18 and K8/K19 IF suggesting that there probably are two populations of limbal stem cells (LSCs). In human, LSCs at limbal basal layer can directly stratify and differentiate to limbal suprabasal cells that express K3/K12 IF, or centripetally migrate then differentiate to corneal basal transient amplifying cells (TAC) that co-express both K3/K12 and K5/K14 prior to moving upward and assuming suprabasal cells phenotype of only K3/K12 expression that signifies corneal type epithelium differentiation. In rodent, the differentiated cornea epithelial cells express K5/K12 in lieu of K3/K12, because K3 allele exists as a pseudogene and does not encode a functional K3 protein. The basal corneal cells of new-born mice originate from surface ectoderm during embryonic development slowly commit to differentiation of becoming TAC co-expressing K5/K12 and K5/K14 IF. However, the centripetal migration may still occur at a slower rate in young mice, which is accelerated during wound healing. In this review, we will discuss and compare the cornea-specific keratins expression patterns between corneal and epidermal epithelial cells during mouse development, and between human and mouse during development and homeostasis in adult, and pathology caused by a mutation of keratins.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Departments of Ophthalmology, University of Cincinnati, Cincinnati, OH, 45267-0838, USA.
| |
Collapse
|
21
|
Apprich V, Licka T, Freiler S, Gabriel C. Equine Hoof Canker: Bovine Papillomavirus Infection Is Not Associated With Impaired Keratinocyte Differentiation. Vet Pathol 2020; 57:525-534. [PMID: 32347169 DOI: 10.1177/0300985820921820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Impaired keratinocyte differentiation has recently been suggested as a key event in equine hoof canker development. Koilocytotic appearance of keratinocytes, one of the most characteristic morphological alterations in hoof canker tissue, is also a common marker for papillomavirus (PV) infection, and bovine PV-1 and/or -2 (BPV-1/2) has previously been detected in equine canker patients. Therefore, the present study aimed to correlate the frequency and severity of koilocytotic keratinocytes with BPV detection in hoof canker samples. Hoof tissue of 5/18 canker-affected horses and 2/6 control horses tested positive for BPV-1/2 DNA using polymerase chain reaction. Thus, no association between the presence of BPV-1/2 papillomaviral DNA and koilocytotic appearance was found. Proteins associated with but not specific for PV infection were also investigated. Using immunohistochemistry, specific adhesion molecules (E-cadherin and β-catenin) and intermediate filaments (keratins 6 and 14) important for intact epidermal barrier function and keratinocyte differentiation were documented in control samples (n = 6) and in hoof canker tissue samples (n = 19). Altered expression patterns of intermediate filaments and adhesion molecules were demonstrated in canker tissue, confirming the importance of incomplete keratinocyte differentiation, as well as the crucial role of keratinocyte differentiation in hoof canker.
Collapse
Affiliation(s)
| | - Theresia Licka
- University of Veterinary Medicine, Vienna, Austria.,University of Edinburgh, Midlothian, UK
| | | | | |
Collapse
|
22
|
Serum lipids, retinoic acid and phenol red differentially regulate expression of keratins K1, K10 and K2 in cultured keratinocytes. Sci Rep 2020; 10:4829. [PMID: 32179842 PMCID: PMC7076045 DOI: 10.1038/s41598-020-61640-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/27/2020] [Indexed: 01/30/2023] Open
Abstract
Abnormal keratinocyte differentiation is fundamental to pathologies such as skin cancer and mucosal inflammatory diseases. The ability to grow keratinocytes in vitro allows the study of differentiation however any translational value is limited if keratinocytes get altered by the culture method. Although serum lipids (SLPs) and phenol red (PR) are ubiquitous components of culture media their effect on differentiation is largely unknown. We show for the first time that PR and SLP themselves suppress expression of differentiation-specific keratins K1, K10 and K2 in normal human epidermal keratinocytes (NHEK) and two important cell lines, HaCaT and N/TERT-1. Removal of SLP increased expression of K1, K10 and K2 in 2D and 3D cultures, which was further enhanced in the absence of PR. The effect was reversed for K1 and K10 by adding all-trans retinoic acid (ATRA) but increased for K2 in the absence of PR. Furthermore, retinoid regulation of differentiation-specific keratins involves post-transcriptional mechanisms as we show KRT2 mRNA is stabilised whilst KRT1 and KRT10 mRNAs are destabilised in the presence of ATRA. Taken together, our results indicate that the presence of PR and SLP in cell culture media may significantly impact in vitro studies of keratinocyte differentiation.
Collapse
|
23
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
24
|
Dmello C, Srivastava SS, Tiwari R, Chaudhari PR, Sawant S, Vaidya MM. Multifaceted role of keratins in epithelial cell differentiation and transformation. J Biosci 2019; 44:33. [PMID: 31180046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Keratins, the epithelial-predominant members of the intermediate filament superfamily, are expressed in a pairwise, tissuespecific and differentiation-dependent manner. There are 28 type I and 26 type II keratins, which share a common structure comprising a central coiled coil α-helical rod domain flanked by two nonhelical head and tail domains. These domains harbor sites for major posttranslational modifications like phosphorylation and glycosylation, which govern keratin function and dynamics. Apart from providing structural support, keratins regulate various signaling machinery involved in cell growth, motility, apoptosis etc. However, tissue-specific functions of keratins in relation to cell proliferation and differentiation are still emerging. Altered keratin expression pattern during and after malignant transformation is reported to modulate different signaling pathways involved in tumor progression in a context-dependent fashion. The current review focuses on the literature related to the role of keratins in the regulation of cell proliferation, differentiation and transformation in different types of epithelia.
Collapse
Affiliation(s)
- Crismita Dmello
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
| | | | | | | | | | | |
Collapse
|
25
|
Mori K, Tamada K, Kurooka H, Matsui M, Takumi T, Yokota Y. Gene expression profile data of the developing small intestine of Id2-deficient mice. Data Brief 2019; 24:103717. [PMID: 30984807 PMCID: PMC6444121 DOI: 10.1016/j.dib.2019.103717] [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] [Received: 11/14/2018] [Revised: 01/23/2019] [Accepted: 01/23/2019] [Indexed: 11/25/2022] Open
Abstract
This article contains data related to the research article entitled “Id2 determines intestinal identity through repression of the foregut transcription factor, Irx5” [1]. Id2 deficient (Id2−/−) mice developed gastric tumors and heterotopic squamous epithelium in the small intestine. These tumors and heterotopic tissues were derived from ectopic gastric cells and squamous cells formed in the small intestine respectively during development. In this study, microarray data of the developing small intestine of Id2−/− mice was analyzed.
Collapse
Affiliation(s)
- Kentaro Mori
- Department of Neurology, Kanazawa Medical University, 1-1 Uchinada, Ishikawa 920-0293, Japan.,Division of Molecular Genetics, Department of Biochemistry and Bioinformative Sciences, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
| | - Kota Tamada
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Hisanori Kurooka
- Division of Molecular Genetics, Department of Biochemistry and Bioinformative Sciences, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui 910-1193, Japan.,Faculty of Nutritional Science, Department of Nutritional Management, Sagami Women's University, Sagamihara, Kanagawa, 252-0383, Japan
| | - Makoto Matsui
- Department of Neurology, Kanazawa Medical University, 1-1 Uchinada, Ishikawa 920-0293, Japan
| | - Toru Takumi
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Yoshifumi Yokota
- Division of Molecular Genetics, Department of Biochemistry and Bioinformative Sciences, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
| |
Collapse
|
26
|
The maintenance of an oral epithelial barrier. Life Sci 2019; 227:129-136. [PMID: 31002922 DOI: 10.1016/j.lfs.2019.04.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/10/2019] [Accepted: 04/14/2019] [Indexed: 02/05/2023]
Abstract
Oral epithelial barrier consists of closely controlled structure of the stratified squamous epithelium, which is the gateway to human bodies and encounters a huge burden of microbial, airborne and dietary antigens, as well as masticatory damage. Once this barrier is destroyed, it will trigger bone loss, tissue damage and microbial dysbiosis and lead to diseases, such as periodontitis, oral mucosal diseases and oral cancer. Recently, increasing evidences showed that different factors including microorganism, saliva, proteins and immune components have been considered to play a critical role in the disruption of oral epithelial barrier. Herein, we discussed mechanisms governing the maintenance of oral epithelial barrier. Besides, the role of oral epithelial barrier failure in oral carcinogenesis will also be talked about.
Collapse
|
27
|
Dmello C, Srivastava SS, Tiwari R, Chaudhari PR, Sawant S, Vaidya MM. Multifaceted role of keratins in epithelial cell differentiation and transformation. J Biosci 2019. [DOI: 10.1007/s12038-019-9864-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
The monoclonal antibody EPR1614Y against the stem cell biomarker keratin K15 lacks specificity and reacts with other keratins. Sci Rep 2019; 9:1943. [PMID: 30760780 PMCID: PMC6374370 DOI: 10.1038/s41598-018-38163-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022] Open
Abstract
Keratin 15 (K15), a type I keratin, which pairs with K5 in epidermis, has been used extensively as a biomarker for stem cells. Two commercial antibodies, LHK15, a mouse monoclonal and EPR1614Y, a rabbit monoclonal, have been widely employed to study K15 expression. Here we report differential reactivity of these antibodies on epithelial cells and tissue sections. Although the two antibodies specifically recognised K15 on western blot, they reacted differently on skin sections and cell lines. LHK15 reacted in patches, whereas EPR1614Y reacted homogenously with the basal keratinocytes in skin sections. In cultured cells, LHK15 did not react with K15 deficient NEB-1, KEB-11, MCF-7 and SW13 cells expressing only exogenous K8 and K18 but reacted when these cells were transduced with K15. On the other hand, EPR1614Y reacted with these cells even though they were devoid of K15. Taken together these results suggest that EPR1614Y recognises a conformational epitope on keratin filaments which can be reconstituted by other keratins as well as by K15. In conclusion, this report highlights that all commercially available antibodies may not be equally specific in identifying the K15 positive stem cell.
Collapse
|
29
|
Stanley RL, Ohashi T, Gordon J, Mowa CN. A proteomic profile of postpartum cervical repair in mice. J Mol Endocrinol 2018; 60:17-28. [PMID: 29259042 DOI: 10.1530/jme-17-0179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 01/01/2023]
Abstract
A timely and complete uterine cervical tissue repair postpartum is of necessity to prevent obstetrical complications, such as cervicitis, ectropion, hemorrhage, repeated miscarriages or abortions and possibly preterm labor and malignancies. We recently characterized the morphological alterations, as well as changes in angiogenic expression profile in a mice uterine cervix during the immediate postpartum period. Here, we build on this previous study using a proteomic analysis to profile postpartum tissue changes in mice cervix during the same period, the first 48 h of postpartum. The current proteomics data reveal a variable expression of several intermediate filaments, cytoskeletal modulators and proteins with immune and/or wound-healing properties. We conclude that postpartum cervical repair involves a rapid and tightly regulated balance between a host of biological factors, notably between anti- and pro-inflammatory factors, executed by the M1 and M2 macrophage cells, as revealed by proteomics and verified by confocal immunofluorescence. Future studies will assess the suitability of some of the key proteins identified in this study as potential markers for determining the phase of postpartum cervical repair in obstetrical complications, such as cervical lacerations.
Collapse
Affiliation(s)
- Robert Lee Stanley
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | - Takako Ohashi
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | - Jacob Gordon
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | | |
Collapse
|
30
|
Nagosa S, Leesch F, Putin D, Bhattacharya S, Altshuler A, Serror L, Amitai-Lange A, Nasser W, Aberdam E, Rouleau M, Tattikota SG, Poy MN, Aberdam D, Shalom-Feuerstein R. microRNA-184 Induces a Commitment Switch to Epidermal Differentiation. Stem Cell Reports 2017; 9. [PMID: 29198823 PMCID: PMC5785777 DOI: 10.1016/j.stemcr.2017.10.030 10.13039/501100003977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
miR-184 is a highly evolutionary conserved microRNA (miRNA) from fly to human. The importance of miR-184 was underscored by the discovery that point mutations in miR-184 gene led to corneal/lens blinding disease. However, miR-184-related function in vivo remained unclear. Here, we report that the miR-184 knockout mouse model displayed increased p63 expression in line with epidermal hyperplasia, while forced expression of miR-184 by stem/progenitor cells enhanced the Notch pathway and induced epidermal hypoplasia. In line, miR-184 reduced clonogenicity and accelerated differentiation of human epidermal cells. We showed that by directly repressing cytokeratin 15 (K15) and FIH1, miR-184 induces Notch activation and epidermal differentiation. The disease-causing miR-184C57U mutant failed to repress K15 and FIH1 and to induce Notch activation, suggesting a loss-of-function mechanism. Altogether, we propose that, by targeting K15 and FIH1, miR-184 regulates the transition from proliferation to early differentiation, while mis-expression or mutation in miR-184 results in impaired homeostasis.
Collapse
Affiliation(s)
- Sara Nagosa
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Friederike Leesch
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Daria Putin
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Swarnabh Bhattacharya
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anna Altshuler
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Laura Serror
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Aya Amitai-Lange
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Waseem Nasser
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Edith Aberdam
- University Paris Diderot, Sorbonne Paris Cité, Paris 75475, France; INSERM U976, Hôpital St-Louis, Paris 75010, France
| | - Matthieu Rouleau
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France; Université Nice Sophia Antipolis, Nice, France
| | - Sudhir G Tattikota
- Max Delbrueck Center for Molecular Medicine, Robert Roessle Strasse 10, Berlin 13125, Germany
| | - Matthew N Poy
- Max Delbrueck Center for Molecular Medicine, Robert Roessle Strasse 10, Berlin 13125, Germany
| | - Daniel Aberdam
- University Paris Diderot, Sorbonne Paris Cité, Paris 75475, France; INSERM U976, Hôpital St-Louis, Paris 75010, France
| | - Ruby Shalom-Feuerstein
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
31
|
Nagosa S, Leesch F, Putin D, Bhattacharya S, Altshuler A, Serror L, Amitai-Lange A, Nasser W, Aberdam E, Rouleau M, Tattikota SG, Poy MN, Aberdam D, Shalom-Feuerstein R. microRNA-184 Induces a Commitment Switch to Epidermal Differentiation. Stem Cell Reports 2017; 9:1991-2004. [PMID: 29198823 PMCID: PMC5785777 DOI: 10.1016/j.stemcr.2017.10.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 12/28/2022] Open
Abstract
miR-184 is a highly evolutionary conserved microRNA (miRNA) from fly to human. The importance of miR-184 was underscored by the discovery that point mutations in miR-184 gene led to corneal/lens blinding disease. However, miR-184-related function in vivo remained unclear. Here, we report that the miR-184 knockout mouse model displayed increased p63 expression in line with epidermal hyperplasia, while forced expression of miR-184 by stem/progenitor cells enhanced the Notch pathway and induced epidermal hypoplasia. In line, miR-184 reduced clonogenicity and accelerated differentiation of human epidermal cells. We showed that by directly repressing cytokeratin 15 (K15) and FIH1, miR-184 induces Notch activation and epidermal differentiation. The disease-causing miR-184C57U mutant failed to repress K15 and FIH1 and to induce Notch activation, suggesting a loss-of-function mechanism. Altogether, we propose that, by targeting K15 and FIH1, miR-184 regulates the transition from proliferation to early differentiation, while mis-expression or mutation in miR-184 results in impaired homeostasis.
Collapse
Affiliation(s)
- Sara Nagosa
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Friederike Leesch
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Daria Putin
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Swarnabh Bhattacharya
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anna Altshuler
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Laura Serror
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Aya Amitai-Lange
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Waseem Nasser
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Edith Aberdam
- University Paris Diderot, Sorbonne Paris Cité, Paris 75475, France; INSERM U976, Hôpital St-Louis, Paris 75010, France
| | - Matthieu Rouleau
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France; Université Nice Sophia Antipolis, Nice, France
| | - Sudhir G Tattikota
- Max Delbrueck Center for Molecular Medicine, Robert Roessle Strasse 10, Berlin 13125, Germany
| | - Matthew N Poy
- Max Delbrueck Center for Molecular Medicine, Robert Roessle Strasse 10, Berlin 13125, Germany
| | - Daniel Aberdam
- University Paris Diderot, Sorbonne Paris Cité, Paris 75475, France; INSERM U976, Hôpital St-Louis, Paris 75010, France
| | - Ruby Shalom-Feuerstein
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
32
|
Mao Y, Xiong L, Li L. Comparison of the proteomes of mouse Skin Derived Precursors (SKPs) and SKP‐derived fibroblasts (SFBs) by iTRAQ. J Cell Biochem 2017; 119:1134-1140. [PMID: 28745444 DOI: 10.1002/jcb.26281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/10/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Yujie Mao
- Department of Dermatology, Sichuan Academy of Science & Sichuan Provincial People's HospitalUniversity of Electronic Science and TechnologyChengduChina
- Department of DermatologyWest China Hospital, Sichuan UniversityChengduChina
| | - Lidan Xiong
- Department of DermatologyWest China Hospital, Sichuan UniversityChengduChina
| | - Li Li
- Department of DermatologyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
33
|
Gullmets J, Torvaldson E, Lindqvist J, Imanishi SY, Taimen P, Meinander A, Eriksson JE. Internal epithelia in Drosophila display rudimentary competence to form cytoplasmic networks of transgenic human vimentin. FASEB J 2017; 31:5332-5341. [PMID: 28778974 DOI: 10.1096/fj.201700332r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/25/2017] [Indexed: 11/11/2022]
Abstract
Cytoplasmic intermediate filaments (cIFs) are found in all eumetazoans, except arthropods. To investigate the compatibility of cIFs in arthropods, we expressed human vimentin (hVim), a cIF with filament-forming capacity in vertebrate cells and tissues, transgenically in Drosophila Transgenic hVim could be recovered from whole-fly lysates by using a standard procedure for intermediate filament (IF) extraction. When this procedure was used to test for the possible presence of IF-like proteins in flies, only lamins and tropomyosin were observed in IF-enriched extracts, thereby providing biochemical reinforcement to the paradigm that arthropods lack cIFs. In Drosophila, transgenic hVim was unable to form filament networks in S2 cells and mesenchymal tissues; however, cage-like vimentin structures could be observed around the nuclei in internal epithelia, which suggests that Drosophila retains selective competence for filament formation. Taken together, our results imply that although the filament network formation competence is partially lost in Drosophila, a rudimentary filament network formation ability remains in epithelial cells. As a result of the observed selective competence for cIF assembly in Drosophila, we hypothesize that internal epithelial cIFs were the last cIFs to disappear from arthropods.-Gullmets, J., Torvaldson, E., Lindqvist, J., Imanishi, S. Y., Taimen, P., Meinander, A., Eriksson, J. E. Internal epithelia in Drosophila display rudimentary competence to form cytoplasmic networks of transgenic human vimentin.
Collapse
Affiliation(s)
- Josef Gullmets
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, Turku, Finland
| | - Elin Torvaldson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Julia Lindqvist
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Susumu Y Imanishi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Pekka Taimen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, Turku, Finland
| | - Annika Meinander
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - John E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; .,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
34
|
Lehmann J, Seebode C, Emmert S. Forschung zu Genodermatosen durch neue Genom-Editing-Methoden. J Dtsch Dermatol Ges 2017; 15:783-790. [PMID: 28763594 DOI: 10.1111/ddg.13270_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/25/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Janin Lehmann
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsmedizin Rostock.,Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Göttingen
| | - Christina Seebode
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsmedizin Rostock
| | - Steffen Emmert
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsmedizin Rostock.,Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Göttingen
| |
Collapse
|
35
|
Peng T, Chanthaphavong RS, Sun S, Trigilio JA, Phasouk K, Jin L, Layton ED, Li AZ, Correnti CE, De van der Schueren W, Vazquez J, O'Day DR, Glass IA, Knipe DM, Wald A, Corey L, Zhu J. Keratinocytes produce IL-17c to protect peripheral nervous systems during human HSV-2 reactivation. J Exp Med 2017; 214:2315-2329. [PMID: 28663436 PMCID: PMC5551564 DOI: 10.1084/jem.20160581] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 03/22/2017] [Accepted: 05/24/2017] [Indexed: 11/04/2022] Open
Abstract
Despite frequent herpes simplex virus (HSV) reactivation, peripheral nerve destruction and sensory anesthesia are rare. We discovered that skin biopsies obtained during asymptomatic human HSV-2 reactivation exhibit a higher density of nerve fibers relative to biopsies during virological and clinical quiescence. We evaluated the effects of HSV infection on keratinocytes, the initial target of HSV replication, to better understand this observation. Keratinocytes produced IL-17c during HSV-2 reactivation, and IL-17RE, an IL-17c-specific receptor, was expressed on nerve fibers in human skin and sensory neurons in dorsal root ganglia. In ex vivo experiments, exogenous human IL-17c provided directional guidance and promoted neurite growth and branching in microfluidic devices. Exogenous murine IL-17c pretreatment reduced apoptosis in HSV-2-infected primary neurons. These results suggest that IL-17c is a neurotrophic cytokine that protects peripheral nerve systems during HSV reactivation. This mechanism could explain the lack of nerve damage from recurrent HSV infection and may provide insight to understanding and treating sensory peripheral neuropathies.
Collapse
Affiliation(s)
- Tao Peng
- Department of Laboratory Medicine, University of Washington, Seattle, WA .,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Sijie Sun
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - James A Trigilio
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Khamsone Phasouk
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lei Jin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Erik D Layton
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Alvason Z Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Colin E Correnti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Julio Vazquez
- Shared Resources Scientific Imaging, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Diana R O'Day
- Laboratory of Developmental Biology, University of Washington, Seattle, WA
| | - Ian A Glass
- Laboratory of Developmental Biology, University of Washington, Seattle, WA
| | - David M Knipe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA
| | - Anna Wald
- Department of Laboratory Medicine, University of Washington, Seattle, WA.,Department of Medicine, University of Washington, Seattle, WA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lawrence Corey
- Department of Laboratory Medicine, University of Washington, Seattle, WA.,Department of Medicine, University of Washington, Seattle, WA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jia Zhu
- Department of Laboratory Medicine, University of Washington, Seattle, WA .,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
36
|
Chee MK, Jo SK, Sohn KC, Kim CD, Lee JH, Lee YH. Effects of Brn2 overexpression on eccrine sweat gland development in the mouse paw. Biochem Biophys Res Commun 2017. [PMID: 28648603 DOI: 10.1016/j.bbrc.2017.06.138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Eccrine sweat glands regulate body temperature by secreting water and electrolytes. In humans, eccrine sweat glands are ubiquitous in the skin, except in the lips and external genitalia. In mice, eccrine sweat glands are present only in the paw pad. Brn2 is a protein belonging to a large family of transcription factors. A few studies have examined Brn2 in melanoma cells and epidermal keratinocytes. This study investigated changes in the skin in the K5-Brn2 transgenic mouse, which overexpresses Brn2 and contains the keratin 5 promotor. Interestingly, the volume of eccrine sweat glands was reduced markedly in the K5-Brn2 transgenic mouse compared with the wild-type, while the expression of aquaporin 5, important molecule in sweat secretion, was increased in each sweat gland cell, probably to compensate for the reduction in gland development. However, sweating response to a pilocarpine injection in the hind paw was significantly decreased in the K5-Brn2 transgenic mouse compared with the wild-type. The paw epidermis was thicker in the K5-Brn2 transgenic mouse compared with the wild-type. Taken together, eccrine sweat gland development and sweat secretion were suppressed markedly in the K5-Brn2 transgenic mouse. These results may be associated with dominant development of the epidermis by Brn2 overexpression in the paw skin.
Collapse
Affiliation(s)
- Min Keun Chee
- Department of Anatomy, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seong Kyeong Jo
- Department of Anatomy, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Kyung Cheol Sohn
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chang Deok Kim
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jeung-Hoon Lee
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Young Ho Lee
- Department of Anatomy, College of Medicine, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
37
|
Lehmann J, Seebode C, Emmert S. Research on genodermatoses using novel genome-editing tools. J Dtsch Dermatol Ges 2017. [PMID: 28622433 DOI: 10.1111/ddg.13270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genodermatoses comprise a clinically heterogeneous group of mostly devastating disorders affecting the skin. To date, treatment options have in general been limited to symptom relief. However, the recent technical evolution in genome editing has ushered in a new era in the development of causal therapies for rare monogenetic diseases such as genodermatoses. The present review revisits the advantages and drawbacks of engineered nuclease tools currently available: zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), meganucleases, and - the most innovative - clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) nuclease 9 (CRISPR/Cas9) system. A mechanistic overview of the different modes of action of these programmable nucleases as well as their significance for causal therapy of genodermatoses is presented. Remaining limitations and challenges such as efficient delivery and off-target activity are critically discussed, highlighting both the past and future of gene therapy in dermatology.
Collapse
Affiliation(s)
- Janin Lehmann
- Clinic for Dermatology und Venereology, University Medical Center, Rostock, Germany.,Clinic for Dermatology, Venereology, and Allergology, University Medical Center Goettingen, Germany
| | - Christina Seebode
- Clinic for Dermatology und Venereology, University Medical Center, Rostock, Germany
| | - Steffen Emmert
- Clinic for Dermatology und Venereology, University Medical Center, Rostock, Germany.,Clinic for Dermatology, Venereology, and Allergology, University Medical Center Goettingen, Germany
| |
Collapse
|
38
|
Genome-wide analysis suggests a differential microRNA signature associated with normal and diabetic human corneal limbus. Sci Rep 2017; 7:3448. [PMID: 28615632 PMCID: PMC5471258 DOI: 10.1038/s41598-017-03449-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Small non-coding RNAs, in particular microRNAs (miRNAs), regulate fine-tuning of gene expression and can impact a wide range of biological processes. However, their roles in normal and diseased limbal epithelial stem cells (LESC) remain unknown. Using deep sequencing analysis, we investigated miRNA expression profiles in central and limbal regions of normal and diabetic human corneas. We identified differentially expressed miRNAs in limbus vs. central cornea in normal and diabetic (DM) corneas including both type 1 (T1DM/IDDM) and type 2 (T2DM/NIDDM) diabetes. Some miRNAs such as miR-10b that was upregulated in limbus vs. central cornea and in diabetic vs. normal limbus also showed significant increase in T1DM vs. T2DM limbus. Overexpression of miR-10b increased Ki-67 staining in human organ-cultured corneas and proliferation rate in cultured corneal epithelial cells. MiR-10b transfected human organ-cultured corneas showed downregulation of PAX6 and DKK1 and upregulation of keratin 17 protein expression levels. In summary, we report for the first time differential miRNA signatures of T1DM and T2DM corneal limbus harboring LESC and show that miR-10b could be involved in the LESC maintenance and/or their early differentiation. Furthermore, miR-10b upregulation may be an important mechanism of corneal diabetic alterations especially in the T1DM patients.
Collapse
|
39
|
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
|
40
|
Lee B, Watanabe K, Haensel D, Sui JY, Dai X. Overexpression of Transcription Factor Ovol2 in Epidermal Progenitor Cells Results in Skin Blistering. J Invest Dermatol 2017; 137:1805-1808. [PMID: 28457910 DOI: 10.1016/j.jid.2017.02.985] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Briana Lee
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Kazuhide Watanabe
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Jennifer Y Sui
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California, USA.
| |
Collapse
|
41
|
|
42
|
He Y, Maier K, Leppert J, Hausser I, Schwieger-Briel A, Weibel L, Theiler M, Kiritsi D, Busch H, Boerries M, Hannula-Jouppi K, Heikkilä H, Tasanen K, Castiglia D, Zambruno G, Has C. Monoallelic Mutations in the Translation Initiation Codon of KLHL24 Cause Skin Fragility. Am J Hum Genet 2016; 99:1395-1404. [PMID: 27889062 DOI: 10.1016/j.ajhg.2016.11.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/07/2016] [Indexed: 11/28/2022] Open
Abstract
The genetic basis of epidermolysis bullosa, a group of genetic disorders characterized by the mechanically induced formation of skin blisters, is largely known, but a number of cases still remain genetically unsolved. Here, we used whole-exome and targeted sequencing to identify monoallelic mutations, c.1A>G and c.2T>C, in the translation initiation codon of the gene encoding kelch-like protein 24 (KLHL24) in 14 individuals with a distinct skin-fragility phenotype and skin cleavage within basal keratinocytes. Remarkably, mutation c.1A>G occurred de novo and was recurrent in families originating from different countries. The striking similarities of the clinical features of the affected individuals point to a unique and very specific pathomechanism. We showed that mutations in the translation initiation codon of KLHL24 lead to the usage of a downstream translation initiation site with the same reading frame and formation of a truncated polypeptide. The pathobiology was examined in keratinocytes and fibroblasts of the affected individuals and via expression of mutant KLHL24, and we found mutant KLHL24 to be associated with abnormalities of intermediate filaments in keratinocytes and fibroblasts. In particular, KLHL24 mutations were associated with irregular and fragmented keratin 14. Recombinant overexpression of normal KLHL24 promoted keratin 14 degradation, whereas mutant KLHL24 showed less activity than the normal molecule. These findings identify KLHL24 mutations as a cause of skin fragility and identify a role for KLHL24 in maintaining the balance between intermediate filament stability and degradation required for skin integrity.
Collapse
Affiliation(s)
- Yinghong He
- Department of Dermatology, University Medical Center Freiburg, Freiburg 79104, Germany
| | - Kristin Maier
- Department of Dermatology, University Medical Center Freiburg, Freiburg 79104, Germany
| | - Juna Leppert
- Department of Dermatology, University Medical Center Freiburg, Freiburg 79104, Germany
| | - Ingrid Hausser
- Department of Pathology, University of Heidelberg, Heidelberg 69120, Germany
| | - Agnes Schwieger-Briel
- Department of Paediatric Dermatology, University Children's Hospital Zurich, Zurich 8091, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Lisa Weibel
- Department of Paediatric Dermatology, University Children's Hospital Zurich, Zurich 8091, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Martin Theiler
- Department of Paediatric Dermatology, University Children's Hospital Zurich, Zurich 8091, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Dimitra Kiritsi
- Department of Dermatology, University Medical Center Freiburg, Freiburg 79104, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg 79104, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Comprehensive Cancer Center Freiburg, Freiburg 79106, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg 79104, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Comprehensive Cancer Center Freiburg, Freiburg 79106, Germany
| | - Katariina Hannula-Jouppi
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland; Folkhälsan Institute of Genetics, University of Helsinki, Helsinki 00014, Finland
| | - Hannele Heikkilä
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Kaisa Tasanen
- Department of Dermatology, PEDEGO Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90014, Finland
| | - Daniele Castiglia
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico, Rome 00167, Italy
| | - Giovanna Zambruno
- Genetic and Rare Diseases Research Area, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome 00165, Italy
| | - Cristina Has
- Department of Dermatology, University Medical Center Freiburg, Freiburg 79104, Germany.
| |
Collapse
|
43
|
Lin Z, Li S, Feng C, Yang S, Wang H, Ma D, Zhang J, Gou M, Bu D, Zhang T, Kong X, Wang X, Sarig O, Ren Y, Dai L, Liu H, Zhang J, Li F, Hu Y, Padalon-Brauch G, Vodo D, Zhou F, Chen T, Deng H, Sprecher E, Yang Y, Tan X. Stabilizing mutations of KLHL24 ubiquitin ligase cause loss of keratin 14 and human skin fragility. Nat Genet 2016; 48:1508-1516. [DOI: 10.1038/ng.3701] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/26/2016] [Indexed: 12/30/2022]
|
44
|
Coulombe PA. Discovery of keratin function and role in genetic diseases: the year that 1991 was. Mol Biol Cell 2016; 27:2807-10. [PMID: 27634744 PMCID: PMC5025267 DOI: 10.1091/mbc.e15-09-0625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 01/27/2023] Open
Abstract
In 1991, a set of transgenic mouse studies took the fields of cell biology and dermatology by storm in providing the first credible evidence that keratin intermediate filaments play a unique and essential role in the structural and mechanical support in keratinocytes of the epidermis. Moreover, these studies intimated that mutations altering the primary structure and function of keratin filaments underlie genetic diseases typified by cellular fragility. This Retrospective on how these studies came to be is offered as a means to highlight the 25th anniversary of these discoveries.
Collapse
Affiliation(s)
- Pierre A Coulombe
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Departments of Biological Chemistry, Oncology, and Dermatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
45
|
Kriegbaum MC, Jacobsen B, Füchtbauer A, Hansen GH, Christensen IJ, Rundsten CF, Persson M, Engelholm LH, Madsen AN, Di Meo I, Lund IK, Holst B, Kjaer A, Lærum OD, Füchtbauer EM, Ploug M. C4.4A gene ablation is compatible with normal epidermal development and causes modest overt phenotypes. Sci Rep 2016; 6:25833. [PMID: 27169360 PMCID: PMC4864438 DOI: 10.1038/srep25833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/22/2016] [Indexed: 12/13/2022] Open
Abstract
C4.4A is a modular glycolipid-anchored Ly6/uPAR/α-neurotoxin multidomain protein that exhibits a prominent membrane-associated expression in stratified squamous epithelia. C4.4A is also expressed in various solid cancer lesions, where high expression levels often are correlated to poor prognosis. Circumstantial evidence suggests a role for C4.4A in cell adhesion, migration, and invasion, but a well-defined biological function is currently unknown. In the present study, we have generated and characterized the first C4.4A-deficient mouse line to gain insight into the functional significance of C4.4A in normal physiology and cancer progression. The unchallenged C4.4A-deficient mice were viable, fertile, born in a normal Mendelian distribution and, surprisingly, displayed normal development of squamous epithelia. The C4.4A-deficient mice were, nonetheless, significantly lighter than littermate controls predominantly due to differences in fat mass. Congenital C4.4A deficiency delayed migration of keratinocytes enclosing incisional skin wounds in male mice. In chemically induced bladder carcinomas, C4.4A deficiency attenuated the incidence of invasive lesions despite having no effect on total tumour burden. This new C4.4A-deficient mouse line provides a useful platform for future studies on functional aspects of C4.4A in tumour cell invasion in vivo.
Collapse
Affiliation(s)
- Mette Camilla Kriegbaum
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Jacobsen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Annette Füchtbauer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Gert Helge Hansen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ib Jarle Christensen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Carsten Friis Rundsten
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Morten Persson
- Department of Clinical Physiology, Nuclear Medicine &PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Lars Henning Engelholm
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Ivano Di Meo
- Unit of Molecular Neurogenetics, Foundation IRCCS Neurological Institute "Carlo Besta", Milano, Italy
| | - Ida Katrine Lund
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Deparment of Neuroscience and Pharmacology, University of Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine &PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Ole Didrik Lærum
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, The Gade Laboratory of Pathology, University of Bergen, Norway
| | | | - Michael Ploug
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
Eghtedari Y, Richardson A, Mai K, Heng B, Guillemin GJ, Wakefield D, Di Girolamo N. Keratin 14 Expression in Epithelial Progenitor Cells of the Developing Human Cornea. Stem Cells Dev 2016; 25:699-711. [DOI: 10.1089/scd.2016.0039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yas Eghtedari
- Ocular Diseases Research Group, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Alexander Richardson
- Ocular Diseases Research Group, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Kelly Mai
- Ocular Diseases Research Group, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Benjamin Heng
- Neuroinflammation Group, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, Australia
| | - Gilles J. Guillemin
- Neuroinflammation Group, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, Australia
| | - Denis Wakefield
- Ocular Diseases Research Group, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Nick Di Girolamo
- Ocular Diseases Research Group, School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
47
|
Schmelzer E, Over P, Gridelli B, Gerlach JC. Response of Primary Human Bone Marrow Mesenchymal Stromal Cells and Dermal Keratinocytes to Thermal Printer Materials In Vitro. J Med Biol Eng 2016; 36:153-167. [PMID: 27231463 PMCID: PMC4853461 DOI: 10.1007/s40846-016-0118-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/15/2015] [Indexed: 01/08/2023]
Abstract
Advancement in thermal three-dimensional printing techniques has greatly increased the possible applications of various materials in medical applications and tissue engineering. Yet, potential toxic effects on primary human cells have been rarely investigated. Therefore, we compared four materials commonly used in thermal printing for bioengineering, namely thermally printed acrylonitrile butadiene styrene, MED610, polycarbonate, and polylactic acid, and investigated their effects on primary human adult skin epidermal keratinocytes and bone marrow mesenchymal stromal cells (BM-MSCs) in vitro. We investigated indirect effects on both cell types caused by potential liberation of soluble substances from the materials, and also analyzed BM-MSCs in direct contact with the materials. We found that even in culture without direct contact with the materials, the culture with MED610 (and to a lesser extent acrylonitrile butadiene styrene) significantly affected keratinocytes, reducing cell numbers and proliferation marker Ki67 expression, and increasing glucose consumption, lactate secretion, and expression of differentiation-associated genes. BM-MSCs had decreased metabolic activity, and exhibited increased cell death in direct culture on the materials. MED610 and acrylonitrile butadiene styrene induced the strongest expression of genes associated to differentiation and estrogen receptor activation. In conclusion, we found strong cell-type-specific effects of the materials, suggesting that materials for applications in regenerative medicine should be carefully selected not only based on their mechanical properties but also based on their cell-type-specific biological effects.
Collapse
Affiliation(s)
- Eva Schmelzer
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Suite 216, Pittsburgh, PA 15203 USA
| | - Patrick Over
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Suite 216, Pittsburgh, PA 15203 USA
| | - Bruno Gridelli
- University of Pittsburgh Medical Center, Pittsburgh, PA USA ; Department of Surgery, ISMETT-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, Palermo, Italy
| | - Jörg C Gerlach
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Suite 216, Pittsburgh, PA 15203 USA ; Department of Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pennsylvania, USA
| |
Collapse
|
48
|
Tan TS, Ng YZ, Badowski C, Dang T, Common JEA, Lacina L, Szeverényi I, Lane EB. Assays to Study Consequences of Cytoplasmic Intermediate Filament Mutations: The Case of Epidermal Keratins. Methods Enzymol 2016; 568:219-53. [PMID: 26795473 DOI: 10.1016/bs.mie.2015.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
The discovery of the causative link between keratin mutations and a growing number of human diseases opened the way for a better understanding of the function of the whole intermediate filament families of cytoskeleton proteins. This chapter describes analytical approaches to identification and interpretation of the consequences of keratin mutations, from the clinical and diagnostic level to cells in tissue culture. Intermediate filament pathologies can be accurately diagnosed from skin biopsies and DNA samples. The Human Intermediate Filament Database collates reported mutations in intermediate filament genes and their diseases, and can help clinicians to establish accurate diagnoses, leading to disease stratification for genetic counseling, optimal care delivery, and future mutation-aligned new therapies. Looking at the best-studied keratinopathy, epidermolysis bullosa simplex, the generation of cell lines mimicking keratinopathies is described, in which tagged mutant keratins facilitate live-cell imaging to make use of today's powerful enhanced light microscopy modalities. Cell stress assays such as cell spreading and cell migration in scratch wound assays can interrogate the consequences of the compromised cytoskeletal network. Application of extrinsic stresses, such as heat, osmotic, or mechanical stress, can enhance the differentiation of mutant keratin cells from wild-type cells. To bring the experiments to the next level, 3D organotypic human cultures can be generated, and even grafted onto the backs of immunodeficient mice for greater in vivo relevance. While development of these assays has focused on mutant K5/K14 cells, the approaches are often applicable to mutations in other intermediate filaments, reinforcing fundamental commonalities in spite of diverse clinical pathologies.
Collapse
Affiliation(s)
| | | | | | - Tram Dang
- Institute of Medical Biology, Singapore
| | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Di Girolamo N, Bobba S, Raviraj V, Delic NC, Slapetova I, Nicovich PR, Halliday GM, Wakefield D, Whan R, Lyons JG. Tracing the fate of limbal epithelial progenitor cells in the murine cornea. Stem Cells 2015; 33:157-69. [PMID: 24966117 DOI: 10.1002/stem.1769] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 05/24/2014] [Indexed: 12/15/2022]
Abstract
Stem cell (SC) division, deployment, and differentiation are processes that contribute to corneal epithelial renewal. Until now studying the destiny of these cells in a living mammal has not been possible. However, the advent of inducible multicolor genetic tagging and powerful imaging technologies has rendered this achievable in the translucent and readily accessible murine cornea. K14CreER(T2)-Confetti mice that harbor two copies of the Brainbow 2.1 cassette, yielding up to 10 colors from the stochastic recombination of fluorescent proteins, were used to monitor K-14(+) progenitor cell dynamics within the corneal epithelium in live animals. Multicolored columns of cells emerged from the basal limbal epithelium as they expanded and migrated linearly at a rate of 10.8 µm/day toward the central cornea. Moreover, the permanent expression of fluorophores, passed on from progenitor to progeny, assisted in discriminating individual clones as spectrally distinct streaks containing more than 1,000 cells within the illuminated area. The centripetal clonal expansion is suggestive that a single progenitor cell is responsible for maintaining a narrow corridor of corneal epithelial cells. Our data are in agreement with the limbus as the repository for SC as opposed to SC being distributed throughout the central cornea. This is the first report describing stem/progenitor cell fate determination in the murine cornea using multicolor genetic tracing. This model represents a powerful new resource to monitor SC kinetics and fate choice under homeostatic conditions, and may assist in assessing clonal evolution during corneal development, aging, wound-healing, disease, and following transplantation.
Collapse
Affiliation(s)
- N Di Girolamo
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|