1
|
Arruda VM, Azevedo GT, Granato MJMG, Matos ACP, Araújo TG, Guerra JFDC. Oxidative Stress and Annexin A2 Differential Expression in Free Fatty Acids-Induced Non-Alcoholic Fatty Liver Disease in HepG2 Cells. Int J Mol Sci 2024; 25:9591. [PMID: 39273539 PMCID: PMC11395542 DOI: 10.3390/ijms25179591] [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: 07/03/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a rising global burden, affecting one in four adults. Despite the increasing prevalence of NAFLD, the exact cellular and molecular mechanisms remain unclear, and effective therapeutic strategies are still limited. In vitro models of NAFLD are critical to understanding the pathogenesis and searching for effective therapies; thus, we evaluated the effects of free fatty acids (FFAs) on NAFLD hallmarks and their association with the modulation of Annexin A2 (ANXA2) and Keratin 17 (KRT17) in HepG2 cells. Our results show that oleic and palmitic acids can differentially induce intracellular lipid accumulation, cell death, and promote oxidative stress by increasing lipid peroxidation, protein carbonylation, and antioxidant defense depletion. Moreover, a markedly increased expression of inflammatory cytokines demonstrated the activation of inflammation pathways associated with lipotoxicity and oxidative stress. ANXA2 overexpression and KRT17 nuclear translocation were also observed, supporting the role of both molecules in the progression of liver disease. Taken together, these data provide insights into the interplay between ANXA2 and KRT17 in NAFLD, paving the way for understanding molecular mechanisms involved with the disease and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Vinícius Marques Arruda
- Laboratory of Metabolic Biochemistry and Redox Processes, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (V.M.A.); (G.T.A.); (M.J.M.G.G.)
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (A.C.P.M.); (T.G.A.)
| | - Gabriela Tolentino Azevedo
- Laboratory of Metabolic Biochemistry and Redox Processes, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (V.M.A.); (G.T.A.); (M.J.M.G.G.)
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (A.C.P.M.); (T.G.A.)
| | - Maria Júlia Maia Gonçalves Granato
- Laboratory of Metabolic Biochemistry and Redox Processes, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (V.M.A.); (G.T.A.); (M.J.M.G.G.)
| | - André Carlos Pereira Matos
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (A.C.P.M.); (T.G.A.)
| | - Thaise Gonçalves Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (A.C.P.M.); (T.G.A.)
| | - Joyce Ferreira da Costa Guerra
- Laboratory of Metabolic Biochemistry and Redox Processes, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas 38700-002, Brazil; (V.M.A.); (G.T.A.); (M.J.M.G.G.)
| |
Collapse
|
2
|
Romashin DD, Tolstova TV, Varshaver AM, Kozhin PM, Rusanov AL, Luzgina NG. Keratins 6, 16, and 17 in Health and Disease: A Summary of Recent Findings. Curr Issues Mol Biol 2024; 46:8627-8641. [PMID: 39194725 DOI: 10.3390/cimb46080508] [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: 06/05/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Keratins 6, 16, and 17 occupy unique positions within the keratin family. These proteins are not commonly found in the healthy, intact epidermis, but their expression increases in response to damage, inflammation, and hereditary skin conditions, as well as cancerous cell transformations and tumor growth. As a result, there is an active investigation into the potential use of these proteins as biomarkers for different pathologies. Recent studies have revealed the role of these keratins in regulating keratinocyte migration, proliferation, and growth, and more recently, their nuclear functions, including their role in maintaining nuclear structure and responding to DNA damage, have also been identified. This review aims to summarize the latest research on keratins 6, 16, and 17, their regulation in the epidermis, and their potential use as biomarkers in various skin conditions.
Collapse
Affiliation(s)
| | | | | | - Peter M Kozhin
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | | | | |
Collapse
|
3
|
Romashin D, Rusanov A, Tolstova T, Varshaver A, Netrusov A, Kozhin P, Luzgina N. Loss of mutant p53 in HaCaT keratinocytes promotes cadmium-induced keratin 17 expression and cell death. Biochem Biophys Res Commun 2024; 709:149834. [PMID: 38547608 DOI: 10.1016/j.bbrc.2024.149834] [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: 01/17/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Cadmium exposure induces dermatotoxicity and epidermal barrier disruption and leads to the development of various pathologies. HaCaT cells are immortalized human keratinocytes that are widely used as alternatives to primary human keratinocytes, particularly for evaluating cadmium toxicity. HaCaT cells bear two gain-of-function (GOF) mutations in the TP53 gene, which strongly affect p53 function. Mutant forms of p53 are known to correlate with increased resistance to various stimuli, including exposure to cytotoxic substances. In addition, keratin 17 (KRT17) was recently shown to be highly expressed in HaCaT cells in response to genotoxic stress. Moreover, p53 is a direct transcriptional repressor of KRT17. However, the impact of TP53 mutations in HaCaT cells on the regulation of cell death and keratin 17 expression is unclear. In this study, we aimed to evaluate the impact of p53 on the response to Cd-induced cytotoxicity. METHODS AND RESULTS Employing the MTT assay and Annexin V/propidium iodide staining, we demonstrated that knockout of TP53 leads to a decrease in the sensitivity of HaCaT cells to the cytotoxic effects of cadmium. Specifically, HaCaT cells with TP53 knockout (TP53 KO HaCaT) exhibited cell death at a cadmium concentration of 10 μM or higher, whereas wild-type cells displayed cell death at a concentration of 30 μM. Furthermore, apoptotic cells were consistently detected in TP53 KO HaCaT cells upon exposure to low concentrations of cadmium (10 and 20 μM) but not in wild-type cells. Our findings also indicate that cadmium cytotoxicity is mediated by reactive oxygen species (ROS), which were significantly increased only in TP53 knockout cells treated with 30 μM cadmium. An examination of proteomic data revealed that TP53 knockout in HaCaT cells resulted in the upregulation of proteins involved in the regulation of apoptosis, redox systems, and DNA repair. Moreover, RT‒qPCR and immunoblotting showed that cadmium toxicity leads to dose-dependent induction of keratin 17 in p53-deficient cells but not in wild-type cells. CONCLUSIONS The connection between mutant p53 in HaCaT keratinocytes and increased resistance to cadmium toxicity was demonstrated for the first time. Proteomic profiling revealed that TP53 knockout in HaCaT cells led to the activation of apoptosis regulatory circuits, redox systems, and DNA repair. In addition, our data support the involvement of keratin 17 in the regulation of DNA repair and cell death. Apparently, the induction of keratin 17 is p53-independent but may be inhibited by mutant p53.
Collapse
Affiliation(s)
- Daniil Romashin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Rusanov
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia.
| | - Tatiana Tolstova
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexandra Varshaver
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Netrusov
- Lomonosov Moscow State University, GSP-1, Leninskie Gory, Moscow, 119991, Russia
| | - Peter Kozhin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Nataliya Luzgina
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| |
Collapse
|
4
|
Toivola DM, Polari L, Schwerd T, Schlegel N, Strnad P. The keratin-desmosome scaffold of internal epithelia in health and disease - The plot is thickening. Curr Opin Cell Biol 2024; 86:102282. [PMID: 38000362 DOI: 10.1016/j.ceb.2023.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/11/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023]
Abstract
Keratin (K) intermediate filaments are attached to desmosomes and constitute the orchestrators of epithelial cell and tissue architecture. While their relevance in the epidermis is well recognized, our review focuses on their emerging importance in internal epithelia. The significance of keratin-desmosome scaffolds (KDSs) in the intestine is highlighted by transgenic mouse models and individuals with inflammatory bowel disease who display profound KDS alterations. In lung, high K8 expression defines a transitional cell subset during regeneration, and K8 variants are associated with idiopathic pulmonary fibrosis. Inherited variants in desmosomal proteins are overrepresented in idiopathic lung fibrosis, and familiar eosinophilic esophagitis. K18 serum fragments are established hepatocellular injury markers that correlate with the extent of histological inflammation. K17 expression is modified in multiple tumors, and K17 levels might be of prognostic relevance. These data should spur further studies on biological roles of these versatile tissue protectors and efforts on their therapeutic targeting.
Collapse
Affiliation(s)
- Diana M Toivola
- Cell Biology, Biosciences and InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland.
| | - Lauri Polari
- Cell Biology, Biosciences and InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| | - Tobias Schwerd
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
5
|
Coulombe PA, Pineda CM, Jacob JT, Nair RR. Nuclear roles for non-lamin intermediate filament proteins. Curr Opin Cell Biol 2024; 86:102303. [PMID: 38113712 PMCID: PMC11056187 DOI: 10.1016/j.ceb.2023.102303] [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: 09/27/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 12/21/2023]
Abstract
The nuclear-localized lamins have long been thought to be the only intermediate filaments (IFs) with an impact on the architecture, properties, and functions of the nucleus. Recent studies, however, uncovered significant roles for IFs other than lamins (here referred to as "non-lamin IFs") in regulating key properties of the nucleus in various cell types and biological settings. In the cytoplasm, IFs often occur in the perinuclear space where they contribute to local stiffness and impact the shape and/or the integrity of the nucleus, particularly in cells under stress. In addition, selective non-lamin IF proteins can occur inside the nucleus where they partake in fundamental processes including nuclear architecture and chromatin organization, regulation of gene expression, cell cycle progression, and the repair of DNA damage. This text reviews the evidence supporting a role for non-lamin IF proteins in regulating various properties of the nucleus and highlights opportunities for further study.
Collapse
Affiliation(s)
- 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, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Christopher M Pineda
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Justin T Jacob
- Public Health Laboratory Division, District of Columbia Department of Forensic Sciences, Washington, DC 20024, USA
| | - Raji R Nair
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Nuwayhid R, Schulz T, Siemers F, Schreiter J, Kobbe P, Hofmann G, Langer S, Kurow O. A Platform for Testing the Biocompatibility of Implants: Silicone Induces a Proinflammatory Response in a 3D Skin Equivalent. Biomedicines 2024; 12:224. [PMID: 38275396 PMCID: PMC10813245 DOI: 10.3390/biomedicines12010224] [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: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Biocompatibility testing of materials is carried out in 2D cell cultures or animal models despite serious limitations. 3D skin equivalents are advanced in vitro models for human skin. Silicone has been shown to be noncytotoxic but capable of eliciting an immune response. Our aim was to (1) establish a 3D skin equivalent to (2) assess the proinflammatory properties of silicone. We developed a coculture of keratinocytes and fibroblasts resulting in a 3D skin equivalent with an implant using samples from a breast implant. Samples with and without the silicone implant were studied histologically and immunohistochemically in comparison to native human skin samples. Cytotoxicity was assessed via LDH-assay, and cytokine response was assessed via ELISA. Histologically, our 3D skin equivalents had a four-layered epidermal and a dermal component. The presence of tight junctions was demonstrated in immunofluorescence. The only difference in 3D skin equivalents with implants was an epidermal thinning. Implanting the silicone samples did not cause more cell death, however, an inflammatory cytokine response was triggered. We were able to establish an organotypical 3D skin equivalent with an implant, which can be utilised for studies on biocompatibility of materials. This first integration of silicone into a 3D skin equivalent confirmed previous findings on silicone being non-cell-toxic but capable of exerting a proinflammatory effect.
Collapse
Affiliation(s)
- Rima Nuwayhid
- Department of Orthopaedic, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (T.S.); (S.L.)
| | - Torsten Schulz
- Department of Orthopaedic, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (T.S.); (S.L.)
| | - Frank Siemers
- Department of Plastic, Hand Surgery and Burn Care, BG Klinikum Bergmannstrost, 06112 Halle, Germany;
| | | | - Philipp Kobbe
- Department of Trauma and Reconstructive Surgery, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany;
- Department of Trauma and Reconstructive Surgery, BG Klinikum Bergmannstrost, 06112 Halle, Germany
| | - Gunther Hofmann
- Department of Trauma, Plastic and Reconstructive Surgery, University Hospital Jena, 07747 Jena, Germany;
| | - Stefan Langer
- Department of Orthopaedic, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (T.S.); (S.L.)
| | - Olga Kurow
- Department of Orthopaedic, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (T.S.); (S.L.)
| |
Collapse
|
7
|
Hosseinalizadeh H, Hussain QM, Poshtchaman Z, Ahsan M, Amin AH, Naghavi S, Mahabady MK. Emerging insights into keratin 7 roles in tumor progression and metastasis of cancers. Front Oncol 2024; 13:1243871. [PMID: 38260844 PMCID: PMC10800941 DOI: 10.3389/fonc.2023.1243871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/26/2023] [Indexed: 01/24/2024] Open
Abstract
Keratin 7 (KRT7), also known as cytokeratin-7 (CK-7) or K7, constitutes the principal constituent of the intermediate filament cytoskeleton and is primarily expressed in the simple epithelia lining the cavities of the internal organs, glandular ducts, and blood vessels. Various pathological conditions, including cancer, have been linked to the abnormal expression of KRT7. KRT7 overexpression promotes tumor progression and metastasis in different human cancers, although the mechanisms of these processes caused by KRT7 have yet to be established. Studies have indicated that the suppression of KRT7 leads to rapid regression of tumors, highlighting the potential of KRT7 as a novel candidate for therapeutic interventions. This review aims to delineate the various roles played by KRT7 in the progression and metastasis of different human malignancies and to investigate its prognostic significance in cancer treatment. Finally, the differential diagnosis of cancers based on the KRT7 is emphasized.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Zahra Poshtchaman
- Department of Nursing, Esfarayen Faculty of Medical Sciences, Esfarayen, Iran
| | | | - Ali H. Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Soroush Naghavi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Zhou P, Li Y, Zhang S, Chen DX, Gao R, Qin P, Yang C, Li Q. KRT17 From Keratinocytes With High Glucose Stimulation Inhibit Dermal Fibroblasts Migration Through Integrin α11. J Endocr Soc 2024; 8:bvad176. [PMID: 38205163 PMCID: PMC10776312 DOI: 10.1210/jendso/bvad176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Indexed: 01/12/2024] Open
Abstract
Objective To investigate the effects of overexpressed keratin 17 (KRT17) on the biology of human dermal fibroblasts (HDFs) and to explore the mechanism of KRT17 in diabetic wound healing. Methods KRT17 expression was tested in diabetic keratinocytes, animal models, and patient skin tissues (Huazhong University of Science and Technology Ethics Committee, [2022] No. 3110). Subsequently, HDFs were stimulated with different concentrations of KRT17 in vitro. Changes in the proliferation and migration of HDFs were observed. Then, identification of KRT17-induced changes in dermal fibroblast of RNA sequencing-based transcriptome analysis was performed. Results KRT17 expression was upregulated under pathological conditions. In vitro stimulation of HDFs with different concentrations of KRT17 inhibited cell migration. RNA-seq data showed that enriched GO terms were extracellular matrix components and their regulation. KEGG analysis revealed that the highest number of enriched genes was PI3K-Akt, in which integrin alpha-11 (ITGA11) mRNA, a key molecule that regulates cell migration, was significantly downregulated. Decreased ITGA11 expression was observed after stimulation of HDFs with KRT17 in vitro. Conclusion Increased expression of KRT17 in diabetic pathological surroundings inhibits fibroblast migration by downregulating the expression of ITGA11. Thus, KRT17 may be a molecular target for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Shan Zhang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dian-Xi Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ruikang Gao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Peiliang Qin
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
9
|
Yu W, Xu H, Sun Z, Du Y, Sun S, Abudureyimu M, Zhang M, Tao J, Ge J, Ren J, Zhang Y. TBC1D15 deficiency protects against doxorubicin cardiotoxicity via inhibiting DNA-PKcs cytosolic retention and DNA damage. Acta Pharm Sin B 2023; 13:4823-4839. [PMID: 38045047 PMCID: PMC10692480 DOI: 10.1016/j.apsb.2023.09.008] [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: 04/22/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 12/05/2023] Open
Abstract
Clinical application of doxorubicin (DOX) is heavily hindered by DOX cardiotoxicity. Several theories were postulated for DOX cardiotoxicity including DNA damage and DNA damage response (DDR), although the mechanism(s) involved remains to be elucidated. This study evaluated the potential role of TBC domain family member 15 (TBC1D15) in DOX cardiotoxicity. Tamoxifen-induced cardiac-specific Tbc1d15 knockout (Tbc1d15CKO) or Tbc1d15 knockin (Tbc1d15CKI) male mice were challenged with a single dose of DOX prior to cardiac assessment 1 week or 4 weeks following DOX challenge. Adenoviruses encoding TBC1D15 or containing shRNA targeting Tbc1d15 were used for Tbc1d15 overexpression or knockdown in isolated primary mouse cardiomyocytes. Our results revealed that DOX evoked upregulation of TBC1D15 with compromised myocardial function and overt mortality, the effects of which were ameliorated and accentuated by Tbc1d15 deletion and Tbc1d15 overexpression, respectively. DOX overtly evoked apoptotic cell death, the effect of which was alleviated and exacerbated by Tbc1d15 knockout and overexpression, respectively. Meanwhile, DOX provoked mitochondrial membrane potential collapse, oxidative stress and DNA damage, the effects of which were mitigated and exacerbated by Tbc1d15 knockdown and overexpression, respectively. Further scrutiny revealed that TBC1D15 fostered cytosolic accumulation of the cardinal DDR element DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Liquid chromatography-tandem mass spectrometry and co-immunoprecipitation denoted an interaction between TBC1D15 and DNA-PKcs at the segment 594-624 of TBC1D15. Moreover, overexpression of TBC1D15 mutant (∆594-624, deletion of segment 594-624) failed to elicit accentuation of DOX-induced cytosolic retention of DNA-PKcs, DNA damage and cardiomyocyte apoptosis by TBC1D15 wild type. However, Tbc1d15 deletion ameliorated DOX-induced cardiomyocyte contractile anomalies, apoptosis, mitochondrial anomalies, DNA damage and cytosolic DNA-PKcs accumulation, which were canceled off by DNA-PKcs inhibition or ATM activation. Taken together, our findings denoted a pivotal role for TBC1D15 in DOX-induced DNA damage, mitochondrial injury, and apoptosis possibly through binding with DNA-PKcs and thus gate-keeping its cytosolic retention, a route to accentuation of cardiac contractile dysfunction in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wenjun Yu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
| | - Haixia Xu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhe Sun
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yuxin Du
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Shiqun Sun
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Miyesaier Abudureyimu
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200030, China
| | - Mengjiao Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Junbo Ge
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
10
|
Yue Z, Lin J, Lu X, Gao Q, Pan M, Zhang Y, Shen S, Zhu WG, Paus R. Keratin 17 Impacts Global Gene Expression and Controls G2/M Cell Cycle Transition in Ionizing Radiation-Induced Skin Damage. J Invest Dermatol 2023; 143:2436-2446.e13. [PMID: 37414246 DOI: 10.1016/j.jid.2023.02.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 07/08/2023]
Abstract
Keratin 17 (K17) is a cytoskeletal protein that is part of the intermediate filaments in epidermal keratinocytes. In K17-/- mice, ionizing radiation induced more severe hair follicle damage, whereas the epidermal inflammatory response was attenuated compared with that in wild-type mice. Both p53 and K17 have a major impact on global gene expression because over 70% of the differentially expressed genes in the skin of wild-type mice showed no expression change in p53-/- or K17-/- skin after ionizing radiation. K17 does not interfere with the dynamics of p53 activation; rather, global p53 binding in the genome is altered in K17-/- mice. The absence of K17 leads to aberrant cell cycle progression and mitotic catastrophe in epidermal keratinocytes, which is due to nuclear retention, thus reducing the degradation of B-Myb, a key regulator of the G2/M cell cycle transition. These results expand our understanding of the role of K17 in regulating global gene expression and ionizing radiation-induced skin damage.
Collapse
Affiliation(s)
- ZhiCao Yue
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China.
| | - JianQiong Lin
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - XiaoPeng Lu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - QingXiang Gao
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - MeiPing Pan
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - YaFei Zhang
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - SiTing Shen
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - Wei-Guo Zhu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Ralf Paus
- Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Center for Dermatology Research, School of Biological Sciences, The University of Manchester and NIHR Biomedical Research Center, Manchester, United Kingdom
| |
Collapse
|
11
|
King MC. Dynamic regulation of LINC complex composition and function across tissues and contexts. FEBS Lett 2023; 597:2823-2832. [PMID: 37846646 DOI: 10.1002/1873-3468.14757] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
The concept of mechanotransduction to the nucleus through a direct force transmission mechanism has fascinated cell biologists for decades. Central to such a mechanism is the linker of nucleoskeleton and cytoskeleton (LINC) complex, which spans the nuclear envelope to couple the cytoplasmic cytoskeleton to the nuclear lamina. In reality, there is not one LINC complex identity, but instead, a family of protein configurations of varied composition that exert both shared and unique functions. Regulated expression of LINC complex components, splice variants, and mechanoresponsive protein turnover mechanisms together shape the complement of LINC complex forms present in a given cell type. Disrupting specific gene(s) encoding LINC complex components therefore gives rise to a range of organismal defects. Moreover, evidence suggests that the mechanical environment remodels LINC complexes, providing a feedback mechanism by which cellular context influences the integration of the nucleus into the cytoskeleton. In particular, evidence for crosstalk between the nuclear and cytoplasmic intermediate filament networks communicated through the LINC complex represents an emerging theme in this active area of ongoing investigation.
Collapse
Affiliation(s)
- Megan C King
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
- Department of Molecular, Cell and Developmental Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
12
|
Zhou P, Feng H, Qin W, Li Q. KRT17 from skin cells with high glucose stimulation promotes keratinocytes proliferation and migration. Front Endocrinol (Lausanne) 2023; 14:1237048. [PMID: 37929023 PMCID: PMC10622786 DOI: 10.3389/fendo.2023.1237048] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/19/2023] [Indexed: 11/07/2023] Open
Abstract
Impaired diabetic wound healing is an important issue in diabetic complications. Proliferation and migration of keratinocytes are major processes of skin wound repair after injury. However, hyperkeratosis can affect the speed of wound healing. Based on the results of preliminary experiments on increased KRT17 expression after high glucose stimulation of human skin tissue cells, a cell model of human immortalized keratinocyte (HaCaT) stimulation with different concentrations of KRT17 was established in vitro, and the promotion in cell proliferation and migration were discovered. KRT17 silencing promoted diabetic wound healing in the db/db diabetic wound model. Transcriptome sequencing (RNA-seq) was performed on HaCaT cells after KRT17 stimulation, and analysis showed significant enrichment in the PI3K-AKT signaling pathway, in which the regulation of cell c-MYB mRNA, a key molecule regulating cell proliferation and migration, was significantly upregulated. In vitro assays showed increased c-MYB expression and enhanced pAKT activity after HaCaT cell stimulation by KRT17. We speculate that KRT17 is upregulated under high glucose and promotes keratinocyte proliferation and migration caused hyperkeratosis, through the c-MYB/PI3K-AKT pathway, contributing to delayed wound healing.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijun Feng
- Department of Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Qin
- Department of Endocrinology, Jingshan Union Hospital of Huazhong University of Science and Technology, Jingshan, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Xu Y, Cohen E, Johnson CN, Parent CA, Coulombe PA. Keratin 17- and PKCα-dependent transient amplification of neutrophil influx after repeated stress to the skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561954. [PMID: 37873256 PMCID: PMC10592713 DOI: 10.1101/2023.10.11.561954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neutrophils contribute to the pathogenesis of chronic inflammatory skin diseases. Little is known about the source and identity of the signals mediating their recruitment in inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin and assess whether keratinocyte-derived signals impact neutrophil recruitment. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12h and resolves within 24h. A second TPA treatment or a UVB challenge, when applied at 24h but not 48h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of neutrophil chemoattractants by stressed keratinocytes. We show that K17 binds RACK1, a scaffold essential for PKCα activity. Finally, analyses of RNAseq data reveal the presence of a transcriptomic signature consistent with TAR and PKCα activation in chronic inflammatory skin diseases. These findings uncover a novel, transient, and keratin-dependent mechanism that amplifies neutrophil recruitment to the skin under stress, with direct implications for inflammatory skin disorders.
Collapse
|
14
|
Chuang TD, Ton N, Rysling S, Quintanilla D, Boos D, Gao J, McSwiggin H, Yan W, Khorram O. The Influence of Race/Ethnicity on the Transcriptomic Landscape of Uterine Fibroids. Int J Mol Sci 2023; 24:13441. [PMID: 37686244 PMCID: PMC10487975 DOI: 10.3390/ijms241713441] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The objective of this study was to determine if the aberrant expression of select genes could form the basis for the racial disparity in fibroid characteristics. The next-generation RNA sequencing results were analyzed as fold change [leiomyomas/paired myometrium, also known as differential expression (DF)], comparing specimens from White (n = 7) and Black (n = 12) patients. The analysis indicated that 95 genes were minimally changed in tumors from White (DF ≈ 1) but were significantly altered by more than 1.5-fold (up or down) in Black patients. Twenty-one novel genes were selected for confirmation in 69 paired fibroids by qRT-PCR. Among these 21, coding of transcripts for the differential expression of FRAT2, SOX4, TNFRSF19, ACP7, GRIP1, IRS4, PLEKHG4B, PGR, COL24A1, KRT17, MMP17, SLN, CCDC177, FUT2, MYO5B, MYOG, ZNF703, CDC25A, and CDCA7 was significantly higher, while the expression of DAB2 and CAV2 was significantly lower in tumors from Black or Hispanic patients compared with tumors from White patients. Western blot analysis revealed a greater differential expression of PGR-A and total progesterone (PGR-A and PGR-B) in tumors from Black compared with tumors from White patients. Collectively, we identified a set of genes uniquely expressed in a race/ethnicity-dependent manner, which could form the underlying mechanisms for the racial disparity in fibroids and their associated symptoms.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Derek Quintanilla
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Drake Boos
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Jianjun Gao
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Colonocyte keratin 7 is expressed de novo in inflammatory bowel diseases and associated with pathological changes and drug-resistance. Sci Rep 2022; 12:22213. [PMID: 36564440 PMCID: PMC9789078 DOI: 10.1038/s41598-022-26603-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The clinical course of IBD, characterized by relapses and remissions, is difficult to predict. Initial diagnosis can be challenging, and novel disease markers are needed. Keratin 7 (K7) is a cytoskeletal intermediate filament protein not expressed in the colonic epithelium but has been reported in IBD-associated colorectal tumors. Our aim was to analyze whether K7 is expressed in chronic colonic inflammatory diseases and evaluate its potential as a novel biomarker. K7 was analyzed in two patient cohorts using immunohistochemistry-stained colon samples and single-cell quantitative digital pathology methods. K7 was correlated to pathological changes and clinical patient characteristics. Our data shows that K7 is expressed de novo in the colonic epithelium of ulcerative colitis and Crohn's disease IBD patients, but not in collagenous or lymphocytic colitis. K7 mRNA expression was significantly increased in colons of IBD patients compared to controls when assessed in publicly available datasets. While K7 increased in areas with inflammatory activity, it was not expressed in specific crypt compartments and did not correlate with neutrophils or stool calprotectin. K7 was increased in areas proximal to pathological alterations and was most pronounced in drug-resistant ulcerative colitis. In conclusion, colonic epithelial K7 is neo-expressed selectively in IBD patients and could be investigated for its potential as a disease biomarker.
Collapse
|
16
|
Bernstein MN, Ni Z, Prasad A, Brown J, Mohanty C, Stewart R, Newton MA, Kendziorski C. SpatialCorr identifies gene sets with spatially varying correlation structure. CELL REPORTS METHODS 2022; 2:100369. [PMID: 36590683 PMCID: PMC9795364 DOI: 10.1016/j.crmeth.2022.100369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 09/26/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Recent advances in spatially resolved transcriptomics technologies enable both the measurement of genome-wide gene expression profiles and their mapping to spatial locations within a tissue. A first step in spatial transcriptomics data analysis is identifying genes with expression that varies spatially, and robust statistical methods exist to address this challenge. While useful, these methods do not detect spatial changes in the coordinated expression within a group of genes. To this end, we present SpatialCorr, a method for identifying sets of genes with spatially varying correlation structure. Given a collection of gene sets pre-defined by a user, SpatialCorr tests for spatially induced differences in the correlation of each gene set within tissue regions, as well as between and among regions. An application to cutaneous squamous cell carcinoma demonstrates the power of the approach for revealing biological insights not identified using existing methods.
Collapse
Affiliation(s)
| | - Zijian Ni
- Department of Statistics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aman Prasad
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jared Brown
- Department of Statistics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Michael A. Newton
- Department of Statistics, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, USA
| |
Collapse
|
17
|
Chen P, Quan Z, Song X, Gao Z, Yuan K. MDFI is a novel biomarker for poor prognosis in LUAD. Front Oncol 2022; 12:1005962. [PMID: 36300089 PMCID: PMC9589366 DOI: 10.3389/fonc.2022.1005962] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background Approximately 80% of lung cancers are non-small cell lung cancers (NSCLC). Lung adenocarcinoma (LUAD) is the main subtype of NSCLC. The incidence and mortality of lung cancer are also increasing yearly. Myogenic differentiation family inhibitor (MDFI) as a transcription factor, its role in lung cancer has not yet been clarified. Methods LUAD data were downloaded from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO), analyzed and plotted using the R language. Associations between Clinical information and MDFI expression were assessed using logistic regression analyses to explore the effects of MDFI on LUAD. Two sets of tissue microarrays (TMAs) further confirmed the overexpression of MDFI in LUAD and its impact on prognosis. In addition, we examined the correlation between MDFI and immune infiltration. To investigate the effect of MDFI on the biological behavior of LUAD tumor cells by GSEA and GO/KEGG analysis. The survival status and somatic mutational characteristics of patients according to MDFI levels were depicted and analyzed. Results Expression of high MDFI in LUAD tissues via analyzing TCGA dataset (P <0.001). Kaplan-Meier survival analysis indicated a poor prognosis for those patients with LUAD who had upregulated MDFI expression levels (P <0.001). This was also verified by two groups of TMAs (P=0.024). Using logistic statistics analysis, MDFI was identified as an independent predictive factor and was associated with poor prognosis in LUAD (P <0.001, P =0.021). Assessment of clinical characteristics, tumor mutation burden (TMB), and tumor microenvironment (TME) between high- and low-expression score groups showed lower TMB, richer immune cell infiltration, and better prognosis in the low-risk group. Conclusion This study showed that MDFI was overexpressed in LUAD and was significantly associated with poor prognosis, indicating that MDFI may be used as a potential novel biomarker for the diagnosis and prognosis of LUAD. MDFI is associated with immune infiltration of LUAD and it is reasonable to speculate that it plays an important role in tumor proliferation and spread. In view of the significant differences in MDFI expression between different biological activities, LUAD patients with MDFI overexpression may obtain more precise treatment strategies in the clinic.
Collapse
Affiliation(s)
- Pengyu Chen
- Division of Thoracic Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- School of Medicine, Dalian Medical University, Dalian, China
| | - Zhen Quan
- Division of Thoracic Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- School of Medicine, Dalian Medical University, Dalian, China
| | - Xueyu Song
- Division of Thoracic Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- School of Medicine, Dalian Medical University, Dalian, China
| | - Zhaojia Gao
- Division of Thoracic Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Kai Yuan
- Division of Thoracic Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
18
|
The Adhesion G-Protein-Coupled Receptor GPR115/ADGRF4 Regulates Epidermal Differentiation and Associates with Cytoskeletal KRT1. Cells 2022; 11:cells11193151. [PMID: 36231117 PMCID: PMC9563031 DOI: 10.3390/cells11193151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 11/27/2022] Open
Abstract
Among the 33 human adhesion G-protein-coupled receptors (aGPCRs), a unique subfamily of GPCRs, only ADGRF4, encoding GPR115, shows an obvious skin-dominated transcriptomic profile, but its expression and function in skin is largely unknown. Here, we report that GPR115 is present in a small subset of basal and in most suprabasal, noncornified keratinocytes of the stratified epidermis, supporting epidermal transcriptomic data. In psoriatic skin, characterized by hyperproliferation and delayed differentiation, the expression of GPR115 and KRT1/10, the fundamental suprabasal keratin dimer, is delayed. The deletion of ADGRF4 in HaCaT keratinocytes grown in an organotypic mode abrogates KRT1 and reduces keratinocyte stratification, indicating a role of GPR115 in epidermal differentiation. Unexpectedly, endogenous GPR115, which is not glycosylated and is likely not proteolytically processed, localizes intracellularly along KRT1/10-positive keratin filaments in a regular pattern. Our data demonstrate a hitherto unknown function of GPR115 in the regulation of epidermal differentiation and KRT1.
Collapse
|
19
|
Mohamad J, Sarig O, Beattie P, Malovitski K, Assaf S, O'Toole E, Schwartz J, Evans H, Samuelov L, Sprecher E. A unique skin phenotype resulting from a large heterozygous deletion spanning six keratin genes. Br J Dermatol 2022; 187:773-777. [PMID: 35822506 DOI: 10.1111/bjd.21766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/03/2022] [Accepted: 07/09/2022] [Indexed: 12/01/2022]
Abstract
The phenotypic spectrum of genodermatoses is continuously expanding. Three siblings were referred because of a highly unusual phenotype comprising alopecia, dystrophic nails, palmoplantar keratoderma, and trauma-induced skin blistering. Whole exome sequencing analysis identified a heterozygous large genomic alteration of ~116,0000 bp resulting in the deletion of the KRT9, KRT14, KRT15, KRT16, KRT19 genes as well as part of KRT17. This genomic change leads to the generation of a truncated KRT17 protein composed of the first 3 exons of the gene and part of intron 3. The three patients were found to carry the heterozygous genomic deletion while their healthy parents did not, indicative of germline mosaicism. The genomic alteration was found to result in reduced KRT17 expression in patient skin. More importantly, the abnormal truncated KRT17 was found to exert a deleterious effect on keratinocyte cytoskeleton formation, leading to keratin aggregation. Co-expression of wildtype and truncated KRT17 proteins also caused keratin aggregation, demonstrating that the deletion exerts a dominant negative effect. In conclusion, we are reporting on a novel clinical phenotype that was found to result from germline mosaicism for a large genomic deletion spanning 6 keratin genes, thus expanding the spectrum of clinical manifestations associated with keratin disorders.
Collapse
Affiliation(s)
- Janan Mohamad
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Sarig
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Paula Beattie
- Department of Dermatology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Kiril Malovitski
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sari Assaf
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Edel O'Toole
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Dermatology, The Royal London Hospital, London, United Kingdom
| | | | - Holly Evans
- Pachyonychia Congenita Project, Holladay, Utah, USA
| | - Liat Samuelov
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Tommasi C, Breuer J. The Biology of Varicella-Zoster Virus Replication in the Skin. Viruses 2022; 14:982. [PMID: 35632723 PMCID: PMC9147561 DOI: 10.3390/v14050982] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023] Open
Abstract
The replication of varicella-zoster virus (VZV) in skin is critical to its pathogenesis and spread. Primary infection causes chickenpox, which is characterised by centrally distributed skin blistering lesions that are rich in infectious virus. Cell-free virus in the cutaneous blistering lesions not only spreads to cause further cases, but infects sensory nerve endings, leading to the establishment of lifelong latency in sensory and autonomic ganglia. The reactivation of virus to cause herpes zoster is again characterised by localised painful skin blistering rash containing infectious virus. The development of in vitro and in vivo models of VZV skin replication has revealed aspects of VZV replication and pathogenesis in this important target organ and improved our understanding of the vaccine strain vOKa attenuation. In this review, we outline the current knowledge on VZV interaction with host signalling pathways, the viral association with proteins associated with epidermal terminal differentiation, and how these interconnect with the VZV life cycle to facilitate viral replication and shedding.
Collapse
Affiliation(s)
- Cristina Tommasi
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Judith Breuer
- Department of Infection, Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
21
|
APEX2-Mediated Proximity Labeling Resolves the DDIT4-Interacting Proteome. Int J Mol Sci 2022; 23:ijms23095189. [PMID: 35563580 PMCID: PMC9102673 DOI: 10.3390/ijms23095189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
DNA damage-inducible transcript 4 (DDIT4) is a ubiquitous protein whose expression is transiently increased in response to various stressors. Chronic expression has been linked to various pathologies, including neurodegeneration, inflammation, and cancer. DDIT4 is best recognized for repressing mTORC1, an essential protein complex activated by nutrients and hormones. Accordingly, DDIT4 regulates metabolism, oxidative stress, hypoxic survival, and apoptosis. Despite these well-defined biological functions, little is known about its interacting partners and their unique molecular functions. Here, fusing an enhanced ascorbate peroxidase 2 (APEX2) biotin-labeling enzyme to DDIT4 combined with mass spectrometry, the proteins in the immediate vicinity of DDIT4 in either unstressed or acute stress conditions were identified in situ. The context-dependent interacting proteomes were quantitatively but not functionally distinct. DDIT4 had twice the number of interaction partners during acute stress compared to unstressed conditions, and while the two protein lists had minimal overlap in terms of identity, the proteins’ molecular function and classification were essentially identical. Moonlighting keratins and ribosomal proteins dominated the proteomes in both unstressed and stressed conditions, with many of their members having established non-canonical and indispensable roles during stress. Multiple keratins regulate mTORC1 signaling via the recruitment of 14-3-3 proteins, whereas ribosomal proteins control translation, cell cycle progression, DNA repair, and death by sequestering critical proteins. In summary, two potentially distinct mechanisms of DDIT4 molecular function have been identified, paving the way for additional research to confirm and consolidate these findings.
Collapse
|
22
|
Pang B, Zhu Z, Xiao C, Luo Y, Fang H, Bai Y, Sun Z, Ma J, Dang E, Wang G. Keratin 17 Is Required for Lipid Metabolism in Keratinocytes and Benefits Epidermal Permeability Barrier Homeostasis. Front Cell Dev Biol 2022; 9:779257. [PMID: 35096815 PMCID: PMC8790522 DOI: 10.3389/fcell.2021.779257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022] Open
Abstract
The epidermal barrier refers to the stratum corneum, the uppermost layer of the skin, and constitutes the first line of defense against invasion by potentially harmful pathogens, diminishes trans-epidermal water loss, and plays a crucial role in the maintenance of skin homeostasis. Keratin 17 (K17) is a type I epithelial keratin with multiple functions, including in skin inflammation, epithelial cell growth, protein synthesis, and tumorigenesis. However, the relationship between K17 and the skin barrier has yet to be systematically investigated. In this study, we found that acute disruption of the epidermal permeability barrier led to a rapid increase in epidermal K17 expression in vivo. Krt17 gene deficiency in mice resulted in decreased expression of lipid metabolism-related enzymes and antimicrobial peptides, while also delaying epidermal permeability barrier recovery after acute disruption. Adenovirus-mediated overexpression of K17 enhanced, whereas siRNA-mediated knockdown of Krt17 inhibited, the expression of fatty acid synthase (FASN) and that of the transcription factors SREBP-1 and PPARγ in vitro. We further confirmed that K17 can facilitate the nuclear transportation of SREBP-1 and PPARγ and promote lipid synthesis in keratinocytes. This study demonstrated that K17 contributes to the restoration of the epidermal permeability barrier via stabilizing lipid metabolism in keratinocytes.
Collapse
Affiliation(s)
- Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenlai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chunying Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yixin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhongbin Sun
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyi Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
23
|
Foote AG, Lungova V, Thibeault SL. Piezo1-expressing vocal fold epithelia modulate remodeling via effects on self-renewal and cytokeratin differentiation. Cell Mol Life Sci 2022; 79:591. [PMID: 36376494 PMCID: PMC9663367 DOI: 10.1007/s00018-022-04622-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Mechanoreceptors are implicated as functional afferents within mucosa of the airways and the recent discovery of mechanosensitive channels Piezo1 and Piezo2 has proved essential for cells of various mechanically sensitive tissues. However, the role for Piezo1/2 in vocal fold (VF) mucosal epithelia, a cell that withstands excessive biomechanical insult, remains unknown. The purpose of this study was to test the hypothesis that Piezo1 is required for VF mucosal repair pathways of epithelial cell injury. Utilizing a sonic hedgehog (shh) Cre line for epithelial-specific ablation of Piezo1/2 mechanoreceptors, we investigated 6wk adult VF mucosa following naphthalene exposure for repair strategies at 1, 3, 7 and 14 days post-injury (dpi). PIEZO1 localized to differentiated apical epithelia and was paramount for epithelial remodeling events. Injury to wildtype epithelium was most appreciated at 3 dpi. Shhcre/+; Piezo1loxP/loxP, Piezo2 loxP/+ mutant epithelium exhibited severe cell/nuclear defects compared to injured controls. Conditional ablation of Piezo1 and/or Piezo2 to uninjured VF epithelium did not result in abnormal phenotypes across P0, P15 and 6wk postnatal stages compared to heterozygote and control tissue. Results demonstrate a role for Piezo1-expressing VF epithelia in regulating self-renewal via effects on p63 transcription and YAP subcellular translocation-altering cytokeratin differentiation.
Collapse
Affiliation(s)
- Alexander G. Foote
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| | - Vlasta Lungova
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| | - Susan L. Thibeault
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
24
|
Baraks G, Tseng R, Pan CH, Kasliwal S, Leiton CV, Shroyer KR, Escobar-Hoyos LF. Dissecting the Oncogenic Roles of Keratin 17 in the Hallmarks of Cancer. Cancer Res 2021; 82:1159-1166. [PMID: 34921015 PMCID: PMC9016724 DOI: 10.1158/0008-5472.can-21-2522] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Gabriella Baraks
- Undergraduate Program in Biomedical Engineering, Stony Brook University, Stony Brook, New York
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Robert Tseng
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Chun-Hao Pan
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Molecular and Cellular Biology Graduate Program, Stony Brook University, New York
| | - Saumya Kasliwal
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Cindy V. Leiton
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Kenneth R. Shroyer
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Corresponding Authors: Kenneth R. Shroyer, Pathology, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794. Phone: 631-444-3000; E-mail: Kenneth.; and Luisa F. Escobar-Hoyos, 15 York Street PO Box 208040, New Haven, CT 06513. Phone: 203-737-2003; E-mail:
| | - Luisa F. Escobar-Hoyos
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Department of Therapeutic Radiology and Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
- Corresponding Authors: Kenneth R. Shroyer, Pathology, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794. Phone: 631-444-3000; E-mail: Kenneth.; and Luisa F. Escobar-Hoyos, 15 York Street PO Box 208040, New Haven, CT 06513. Phone: 203-737-2003; E-mail:
| |
Collapse
|
25
|
Doorbar J, Zheng K, Aiyenuro A, Yin W, Walker CM, Chen Y, Egawa N, Griffin HM. Principles of epithelial homeostasis control during persistent human papillomavirus infection and its deregulation at the cervical transformation zone. Curr Opin Virol 2021; 51:96-105. [PMID: 34628359 DOI: 10.1016/j.coviro.2021.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022]
Abstract
Human papillomaviruses establish a reservoir of infection in the epithelial basal layer. To do this they limit their gene expression to avoid immune detection and modulate epithelial homeostasis pathways to inhibit the timing of basal cell delamination and differentiation to favour persistence. For low-risk Alpha papillomaviruses, which cause benign self-limiting disease in immunocompetent individuals, it appears that cell competition at the lesion edge restricts expansion. These lesions may be considered as self-regulating homeostatic structures, with epithelial cells of the hair follicles and sweat glands, which are proposed targets of the Beta and Mu papillomaviruses, showing similar restrictions to their expansion across the epithelium as a whole. In the absence of immune control, which facilitates deregulated viral gene expression, such lesions can expand, leading to problematic papillomatosis in afflicted individuals. By contrast, he high-risk Alpha HPV types can undergo deregulated viral gene expression in immunocompetent hosts at a number of body sites, including the cervical transformation zone (TZ) where they can drive the formation of neoplasia. Homeostasis at the TZ is poorly understood, but involves two adjacent epithelial cell population, one of which has the potential to stratify and to produce a multilayed squamous epithelium. This process of metaplasia involves a specialised cell type known as the reserve cell, which has for several decades been considered as the cell of origin of cervical cancer. It is becoming clear that during evolution, HPV gene products have acquired functions directly linked to their requirements to modify the normal processes of epithelial homestasis at their various sites of infection. These protein functions are beginning to provide new insight into homeostasis regulation at different body sites, and are likely to be central to our understanding of HPV epithelial tropisms.
Collapse
Affiliation(s)
- John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom.
| | - Ke Zheng
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Ademola Aiyenuro
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Wen Yin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Caroline M Walker
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Yuwen Chen
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Heather M Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| |
Collapse
|
26
|
Colom B, Herms A, Hall MWJ, Dentro SC, King C, Sood RK, Alcolea MP, Piedrafita G, Fernandez-Antoran D, Ong SH, Fowler JC, Mahbubani KT, Saeb-Parsy K, Gerstung M, Hall BA, Jones PH. Mutant clones in normal epithelium outcompete and eliminate emerging tumours. Nature 2021; 598:510-514. [PMID: 34646013 PMCID: PMC7612642 DOI: 10.1038/s41586-021-03965-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Human epithelial tissues accumulate cancer-driver mutations with age1-9, yet tumour formation remains rare. The positive selection of these mutations suggests that they alter the behaviour and fitness of proliferating cells10-12. Thus, normal adult tissues become a patchwork of mutant clones competing for space and survival, with the fittest clones expanding by eliminating their less competitive neighbours11-14. However, little is known about how such dynamic competition in normal epithelia influences early tumorigenesis. Here we show that the majority of newly formed oesophageal tumours are eliminated through competition with mutant clones in the adjacent normal epithelium. We followed the fate of nascent, microscopic, pre-malignant tumours in a mouse model of oesophageal carcinogenesis and found that most were rapidly lost with no indication of tumour cell death, decreased proliferation or an anti-tumour immune response. However, deep sequencing of ten-day-old and one-year-old tumours showed evidence of selection on the surviving neoplasms. Induction of highly competitive clones in transgenic mice increased early tumour removal, whereas pharmacological inhibition of clonal competition reduced tumour loss. These results support a model in which survival of early neoplasms depends on their competitive fitness relative to that of mutant clones in the surrounding normal tissue. Mutant clones in normal epithelium have an unexpected anti-tumorigenic role in purging early tumours through cell competition, thereby preserving tissue integrity.
Collapse
Affiliation(s)
- B Colom
- Wellcome Sanger Institute, Hinxton, UK
| | - A Herms
- Wellcome Sanger Institute, Hinxton, UK
| | - M W J Hall
- Wellcome Sanger Institute, Hinxton, UK
- MRC Cancer Unit, University of Cambridge, Hutchison-MRC Research Centre, Cambridge, UK
| | - S C Dentro
- Wellcome Sanger Institute, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | - C King
- Wellcome Sanger Institute, Hinxton, UK
| | - R K Sood
- Wellcome Sanger Institute, Hinxton, UK
| | - M P Alcolea
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Hutchison-MRC Research Centre, Cambridge, UK
| | - G Piedrafita
- Wellcome Sanger Institute, Hinxton, UK
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - D Fernandez-Antoran
- Wellcome Sanger Institute, Hinxton, UK
- Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - S H Ong
- Wellcome Sanger Institute, Hinxton, UK
| | | | - K T Mahbubani
- Department of Surgery and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - K Saeb-Parsy
- Department of Surgery and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - M Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - B A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - P H Jones
- Wellcome Sanger Institute, Hinxton, UK.
- MRC Cancer Unit, University of Cambridge, Hutchison-MRC Research Centre, Cambridge, UK.
| |
Collapse
|