1
|
Pagano S, Somm E, Juillard C, Liaudet N, Ino F, Ferrari J, Braunersreuther V, Jornayvaz FR, Vuilleumier N. Linking Antibodies Against Apolipoprotein A-1 to Metabolic Dysfunction-Associated Steatohepatitis in Mice. Int J Mol Sci 2024; 25:11875. [PMID: 39595946 PMCID: PMC11594174 DOI: 10.3390/ijms252211875] [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: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MASLD) is a common liver and health issue associated with heightened cardiovascular disease (CVD) risk, with Cytokeratin 18 (CK-18) as a marker of liver injury across the MASLD to cirrhosis spectrum. Autoantibodies against apolipoprotein A-1 (AAA-1s) predict increased CVD risk, promoting atherosclerosis and liver steatosis in apoE-/- mice, though their impact on liver inflammation and fibrosis remains unclear. This study examined AAA-1s' impact on low-grade inflammation, liver steatosis, and fibrosis using a MASLD mouse model exposed to AAA-1s passive immunization (PI). Ten-week-old male C57BL/6J mice under a high-fat diet underwent PI with AAA-1s or control antibodies for ten days. Compared to controls, AAA-1-immunized mice showed higher plasma CK-18 (5.3 vs. 2.1 pg/mL, p = 0.031), IL-6 (13 vs. 6.9 pg/mL, p = 0.035), IL-10 (27.3 vs. 9.8 pg/mL, p = 0.007), TNF-α (32.1 vs. 24.2 pg/mL, p = 0.032), and liver steatosis (93.4% vs. 73.8%, p = 0.007). Transcriptomic analyses revealed hepatic upregulation of pro-fibrotic mRNAs in AAA-1-recipient mice, though histological changes were absent. In conclusion, short-term AAA-1 PI exacerbated liver steatosis, inflammation, and pro-fibrotic gene expression, suggesting that AAA-1s may play a role in MASLD progression. Further research with prolonged AAA-1 exposure is warranted to clarify their potential role in liver fibrosis and associated complications.
Collapse
Affiliation(s)
- Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland;
- Department of Medicine, Medical Faculty, Geneva University, 1211 Geneva, Switzerland;
| | - Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (E.S.); (F.I.); (F.R.J.)
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, the Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Catherine Juillard
- Department of Medicine, Medical Faculty, Geneva University, 1211 Geneva, Switzerland;
| | - Nicolas Liaudet
- Bioimaging Core Facility, Medical Faculty, University of Geneva, 1211 Geneva, Switzerland;
| | - Frédérique Ino
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (E.S.); (F.I.); (F.R.J.)
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, the Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Johan Ferrari
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; (J.F.); (V.B.)
| | - Vincent Braunersreuther
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; (J.F.); (V.B.)
| | - François R. Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (E.S.); (F.I.); (F.R.J.)
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, the Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland;
- Department of Medicine, Medical Faculty, Geneva University, 1211 Geneva, Switzerland;
| |
Collapse
|
2
|
Aydın N, Ketani MA, Sağsöz H. The expression of intermediate filaments in the abomasum of ruminants: A comparative study. Anat Histol Embryol 2024; 53:e13088. [PMID: 38979752 DOI: 10.1111/ahe.13088] [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: 03/25/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Intermediate filaments (IFs) are key molecular factors of the cell and have been reported to play an important role in maintaining the structural integrity and functionality of the abomasum. This study was designed to determine the regional distribution, cellular localization and expression of several IFs, including CK8, CK18, CK19, vimentin, desmin, peripherin and nestin, as well as the connective tissue component laminin, in the bovine, ovine and caprine abomasa. Immunohistochemical analyses demonstrated varying levels of expression of CK8, CK18, CK19, vimentin, desmin, nestin, peripherin and laminin in the bovine, ovine and caprine abomasa. CK8 immunoreactions were particularly evident in the luminal and glandular epithelia of the glands found in the abomasal cardia, fundus and pylorus in all three species. In the bovine abomasum, CK18 immunoreactions were stronger in the parietal cells, compared to the chief cells. In the abomasum of all three species, the smooth muscle as well as the smooth muscle cells of the vascular media in the cardiac, fundic and pyloric regions showed strong immunoreactivity. In all three species, the cardiac, fundic and pyloric regions of the abomasum showed strong peripherin and nestin immunoreactions in the luminal and glandular epithelial cells, stromal and smooth muscle cells, nervous plexuses and blood vessels. The expression patterns of IFs and laminin in the ruminant abomasum suggest that these proteins play a structural role in the cytoskeleton and are effective in maintaining abomasal tissue integrity and stability.
Collapse
Affiliation(s)
- Nurşin Aydın
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey
| | - M Aydın Ketani
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey
| | - Hakan Sağsöz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
3
|
Gao X, Wang G, Zhang M, Zhang X, Zhang S, Long H. LINC01485 contributes to colorectal cancer progression by targeting miR-383-5p/KRT80 axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:398-408. [PMID: 37782686 DOI: 10.1002/tox.23983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/22/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
Long non-coding RNAs (lncRNAs) are important in tumorigenesis and the development of multiple malignant human tumors, including colorectal cancer (CRC). We aimed to determine the regulatory mechanism of LINC01485 and its biological function in CRC. We estimated the expression of miR-383-5p, KRT80, and LINC01485 in CRC cells and tissues using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting. The results were confirmed using RNA immunoprecipitation (RIP) and dual-luciferase assays. Binding relationships among miR-383-5p, LINC01485, and KRT80 were assessed. We explored the molecular mechanisms and functions of the LINC01485/miR-383-5p/KRT80 axis using CCK-8 and colony formation assays. Expression of the apoptotic markers Bcl-2 and Bax was quantified by western blotting, and the effects of LINC01485 on tumor development in vivo were investigated using xenograft tumors. Both LINC01485 and KRT80 were upregulated, whereas miR-383-5p was downregulated in CRC cells and tissues. Knockdown of LINC01485 attenuated CRC cell growth and xenograft tumor formation in vivo, whereas LINC01485 enhanced the proliferative capacity of CRC cells but inhibited apoptosis by sponging miR-383-5p to increase KRT80 expression in CRC cells. The regulatory molecular mechanism of the LINC01485/miR-383-5p/KRT80 axis plays a crucial role in CRC progression. Our findings highlight novel pathways and promising biomarkers for diagnostic and therapeutic application to patients with CRC.
Collapse
Affiliation(s)
- Xia Gao
- Department of Oncology, Wuhan Asia General Hospital, Wuhan, China
| | - Guangxin Wang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, China
| | - Min Zhang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, China
| | - Xinxin Zhang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, China
| | - Shuosheng Zhang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, China
| | - Haocheng Long
- Department of General Surgery, Wuhan Third Hospital, Wuhan, China
| |
Collapse
|
4
|
Goralska J, Razny U, Gruca A, Zdzienicka A, Micek A, Dembinska-Kiec A, Solnica B, Malczewska-Malec M. Plasma Cytokeratin-18 Fragment Level Reflects the Metabolic Phenotype in Obesity. Biomolecules 2023; 13:biom13040675. [PMID: 37189422 DOI: 10.3390/biom13040675] [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: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
There is growing interest in the non-invasive identification and monitoring of the outcome of liver damage in obese patients. Plasma cytokeratin-18 (CK-18) fragment levels correlate with the magnitude of hepatocyte apoptosis and have recently been proposed to independently predict the presence of non-alcoholic steatohepatitis (NASH). The aim of the study was to analyze the associations of CK-18 with obesity and related complications: insulin resistance, impaired lipid metabolism and the secretion of hepatokines, adipokines and pro-inflammatory cytokines. The study involved 151 overweight and obese patients (BMI 25-40), without diabetes, dyslipidemia or apparent liver disease. Liver function was assessed based on alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT) and the fatty liver index (FLI). CK-18 M30 plasma levels, FGF-21, FGF-19 and cytokines were determined by ELISA. CK-18 values >150 U/l were accompanied by high ALT, GGT and FLI, insulin resistance, postprandial hypertriglyceridemia, elevated FGF-21 and MCP-1 and decreased adiponectin. ALT activity was the strongest independent factor influencing high CK-18 plasma levels, even after an adjustment for age, sex and BMI [β coefficient (95%CI): 0.40 (0.19-0.61)]. In conclusion, the applied CK-18 cut-off point at 150 U/l allows to distinguish between two metabolic phenotypes in obesity.
Collapse
Affiliation(s)
- Joanna Goralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Anna Gruca
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Anna Zdzienicka
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Agnieszka Micek
- Institute of Nursing and Midwifery, Jagiellonian University Medical College; Michałowskiego 12, 31-126 Krakow, Poland
| | - Aldona Dembinska-Kiec
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Bogdan Solnica
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Malgorzata Malczewska-Malec
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| |
Collapse
|
5
|
Keratin 80 Promotes Migration and Invasion of Non-Small Cell Lung Cancer Cells by Regulating the TGF-β/SMAD Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2630351. [PMID: 36248424 PMCID: PMC9553464 DOI: 10.1155/2022/2630351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 06/04/2022] [Indexed: 12/04/2022]
Abstract
Upregulation of keratin 80 (KRT80) expression levels and carcinogenic function has been found in several types of tumors. However, its contribution and mechanism in NSCLC remain to be outlined. In this study, bioinformatic investigation from the TCGA dataset revealed that KRT80 was confirmed to be elevated in human NSCLC tissues. The results of qRT-PCR and Western blot assays disclosed that KRT80 was uplifted in NSCLC cells. Data from CCK-8 and colony formation assays exhibited that depletion of KRT80 restrained NSCLC cell proliferation. Findings from Transwell and Western blot assays illustrated that downregulation of KRT80 inhibited NSCLC cell migration, invasion, and EMT. Further mechanism exploration implied that KRT80 may be included within the regulation of EMT of NSCLC cells by affecting the TGF-β/SMAD pathway. Moreover, depletion of KRT80 attenuated xenograft tumor growth and the expressions of KRT80, Ki-67, and TGFBR1. In conclusion, depletion of KRT80 repressed NSCLC cell proliferation, invasion, and EMT, possibly mediated by the TGF-β/SMAD signaling pathway, indicating that KRT80 may be a potentially useful target for NSCLC.
Collapse
|
6
|
Yang HC, Xing ZK, Shao H, Tan XW, Wang EQ, Liao Y, Chen HJ, Wu XW, Chen XL, Zhang SJ. The expression of cytokeratin and apoptosis-related molecules in echinococcosis related liver injury. Mol Biochem Parasitol 2022; 248:111455. [PMID: 35016896 DOI: 10.1016/j.molbiopara.2022.111455] [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: 09/22/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 10/25/2022]
Abstract
The study aimed to investigate the expression of cytokeratin and apoptosis-related molecules in the livers of two types of hepatic echinococcosis mice models and to preliminarily explore the relationship between the expression of cytokeratin and apoptosis in echinococcosis related liver injury. We established a mouse model infected by Echinococcus granulosus and Echinococcus multilocularis and observed the expression of cytokeratin and apoptosis related proteins in the two types of hepatic echinococcosis tissues during different stages by immunohistochemical staining. A co-culture model was established using normal hepatocytes and different concentrations of E. granulosus and E. multilocularis protoscoleces. Cell Counting Kit-8 was used to detect cell proliferation, flow cytometry was used to detect hepatocyte apoptosis, and western blot was used to quantify cytokeratin and apoptosis-related proteins, such as caspase3, caspase9, Bcl-2, and Bax. Surgical specimens were obtained from patients with hepatic echinococcosis to analyze the expressions of cytokeratin, caspase3, caspase9, Bcl-2, and Bax by western blot. The expressions of cytokeratin and caspase3 were analyzed by immunohistochemistry. The qRT-PCR method was used to determine the expression of CK8 and CK18 in the liver tissues. In vivo experiments showed that compared to that in the control group, the cytokeratin and caspase3 proteins in the liver tissues of the two types of hepatic echinococcosis were strongly expressed around the lesions of liver echinococcosis; there was a difference between cytokeratin expression of the two different echinococcosis parasites in the liver. Echinococcus granulosus and Echinococcus multilocularis in the co-culture model in vitro could promote the expression of CK, caspase3, caspase9, and Bax protein, decrease the expression of Bcl-2, promote hepatocyte apoptosis, and inhibit cell proliferation; in clinical samples, we found that compared with that in the normal tissues, the expression of cytokeratin, caspase3, caspase9, and Bax in echinococcus tissues was high, but that in Bcl-2 was low. Furthermore, the expression of CK8 and CK18 mRNA were higher in echinococcus tissues than that in the normal tissues and immunohistochemistry analysis also showed that cytokeratin and caspase3 levels were higher in echinococcus tissues than that in the normal tissues. The expression of cytokeratin and apoptosis-related molecules, reflecting liver damage, is high in the liver and is caused due to hepatic echinococcosis. This study provides the first evidence of cytokeratin could be useful for evaluating liver tissue damage caused by echinococcus infection.
Collapse
Affiliation(s)
- H C Yang
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China; Department of Hepatobiliary Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - Z K Xing
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China; Department of Hepatobiliary Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - H Shao
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - X W Tan
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - E Q Wang
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - Y Liao
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - H J Chen
- School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - X W Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China
| | - X L Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, 832002, Xinjiang, China
| | - S J Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, Xinjiang, China.
| |
Collapse
|
7
|
Liu O, Wang C, Wang S, Hu Y, Gou R, Dong H, Li S, Li X, Lin B. Keratin 80 regulated by miR-206/ETS1 promotes tumor progression via the MEK/ERK pathway in ovarian cancer. J Cancer 2021; 12:6835-6850. [PMID: 34659572 PMCID: PMC8517993 DOI: 10.7150/jca.64031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/18/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction: Keratin 80 (KRT80) is a type II epithelial keratin protein that plays an important role in cell differentiation and tumor progression. However, its role and mechanisms in ovarian cancer remain unclear. Methods: The effect of KRT80 on the survival and prognosis of patients with ovarian cancer was determined using immunohistochemistry. Cell lines overexpressing KRT80 and with KRT80 knockdown were established to study its effect on the malignant behavior of ovarian cancer cells. Western blotting was used to detect changes in related molecules, and in the MEK/ERK signal transduction pathway. ChIP assay was used to confirm that ETS1 regulates KRT80 at the transcriptional level. A double luciferase assay was used to confirm the target of miR-206. Results: The expression levels of KRT80 were high in ovarian cancer tissue, and were related to survival and prognosis. KRT80 expression is an independent prognostic factor in patients with ovarian cancer. KRT80 overexpression promotes the proliferation of ovarian cancer cells, the transition from G1 phase to S phase, invasion, and migration. KRT80 overexpression increased the expression of BCL2/BAX, CyclinD1, MMP2, MMP9, and N-cadherin, decreased the expression of E-cadherin, and increased the phosphorylation of MEK and ERK. ETS1 binds to the upstream promoter sequence of KRT80 and regulates KRT80 expression at the transcriptional level. ETS1 is a direct target of miR-206 in ovarian cancer cells. Conclusion: KRT80 regulated by miR-206/ETS1 promotes tumor progression via the MEK/ERK pathway in ovarian cancer, and KRT80 may have applications as a screening biomarker and potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Ouxuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Caixia Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuang Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Yuexin Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Rui Gou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Hui Dong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Siting Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| |
Collapse
|
8
|
Yu B, Kong D, Cheng C, Xiang D, Cao L, Liu Y, He Y. Assembly and recognition of keratins: A structural perspective. Semin Cell Dev Biol 2021; 128:80-89. [PMID: 34654627 DOI: 10.1016/j.semcdb.2021.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022]
Abstract
Keratins are one of the major components of cytoskeletal network and assemble into fibrous structures named intermediate filaments (IFs), which are important for maintaining the mechanical properties of cells and tissues. Over the past decades, evidence has shown that the functions of keratins go beyond providing mechanical support for cells, they interact with multiple cellular components and are widely involved in the pathways of cell proliferation, differentiation, motility and death. However, the structural details of keratins and IFs are largely missing and many questions remain regarding the mechanisms of keratin assembly and recognition. Here we briefly review the current structural models and assembly of keratins as well as the interactions of keratins with the binding partners, which may provide a structural view for understanding the mechanisms of keratins in the biological activities and the related diseases.
Collapse
Affiliation(s)
- Bowen Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Dandan Kong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Cheng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longxing Cao
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yingbin Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongning He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Lim Y, Ku NO. Revealing the Roles of Keratin 8/18-Associated Signaling Proteins Involved in the Development of Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:6401. [PMID: 34203895 PMCID: PMC8232640 DOI: 10.3390/ijms22126401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 02/08/2023] Open
Abstract
Although hepatocellular carcinoma (HCC) is developed with various etiologies, protection of hepatocytes seems basically essential to prevent the incidence of HCC. Keratin 8 and keratin 18 (K8/K18) are cytoskeletal intermediate filament proteins that are expressed in hepatocytes. They maintain the cell shape and protect cells under stress conditions. Their protective roles in liver damage have been described in studies of mouse models, and K8/K18 mutation frequency in liver patients. Interestingly, K8/K18 bind to signaling proteins such as transcription factors and protein kinases involved in HCC development. Since K8/K18 are abundant cytoskeletal proteins, K8/K18 binding with the signaling factors can alter the availability of the factors. Herein, we discuss the potential roles of K8/K18 in HCC development.
Collapse
Affiliation(s)
- Younglan Lim
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
- Department of Bio-Convergence ISED, Underwood International College, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
10
|
Valdivia A, Ortega KJ, Bhattacharya SK, Cray C. Capillary Electrophoresis Assessment of Plasma Protein Changes in an African Penguin ( Spheniscus demersus) With Aspergillosis. ACS OMEGA 2020; 5:33280-33289. [PMID: 33403290 PMCID: PMC7774288 DOI: 10.1021/acsomega.0c04983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/03/2020] [Indexed: 05/12/2023]
Abstract
A decrease of avian biodiversity in the African continent has been the result of anthropogenic pressure in the region. This has resulted in the African penguin (Spheniscus demersus) being placed on the endangered species list and requires conservation efforts to maintain its free-ranging population and placement under managed care. In the latter environment, infection by Aspergillus fumigatus can be common. The diagnosis and treatment of this fungal disease in birds has presented with many difficulties, largely due to the diversity and limited knowledge that exists about this species. In this study, we implement a high-resolution capillary electrophoresis system for the fractionation of African penguin plasma, followed by mass spectrometry analysis for the identification of proteins associated with aspergillosis. Several protein differences were revealed, including changes in acute phase proteins and lipid metabolism. In addition, our results demonstrated that fibrinogen β chain is a protein largely present during the inflammatory process in an African penguin infected with A. fumigatus. These findings present a new avenue for the measurement of plasma proteins as a potential method for identifying important biomarkers to aid in monitoring African penguin health.
Collapse
Affiliation(s)
- Anddre
Osmar Valdivia
- Bascom
Palmer Eye Institute, University of Miami, Miami, Florida 33136, United States
- Neuroscience
Graduate Program, University of Miami, Miami, Florida 33136, United States
- Miami
Integrative Metabolomics Research Center, University of Miami, Miami, Florida 33136, United States
| | - Kristen Jasmin Ortega
- Bascom
Palmer Eye Institute, University of Miami, Miami, Florida 33136, United States
- Miami
Integrative Metabolomics Research Center, University of Miami, Miami, Florida 33136, United States
| | - Sanjoy K. Bhattacharya
- Bascom
Palmer Eye Institute, University of Miami, Miami, Florida 33136, United States
- Miami
Integrative Metabolomics Research Center, University of Miami, Miami, Florida 33136, United States
| | - Carolyn Cray
- Miami
Integrative Metabolomics Research Center, University of Miami, Miami, Florida 33136, United States
- Division
of Comparative Pathology, Department of Pathology & Laboratory
Medicine, University of Miami, Miami, Florida 33136, United States
- . Tel.: (305) 243-6700. Fax: (305) 243-5662
| |
Collapse
|
11
|
Lin J, Fan X, Chen J, Xie X, Yu H. Small interfering RNA-mediated knockdown of KRT80 suppresses colorectal cancer proliferation. Exp Ther Med 2020; 20:176. [PMID: 33101466 PMCID: PMC7579811 DOI: 10.3892/etm.2020.9306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 04/17/2020] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world and its development is associated with oncogenic dysfunction. Therefore, the present study aimed to identify differentially expressed genes (DEGs) in CRC tissues and to determine the role of keratin 80 (KRT80) in CRC cell proliferation. DEGs were initially screened in 32 paired CRC tissues and matched adjacent normal tissues from RNA-Seq datasets in The Cancer Genome Atlas database using the limma package in R software. In total, 2,114 DEGs were identified, of which KRT80 was discovered to be the most upregulated in CRC tissues. Moreover, increased KRT80 expression levels were confirmed in tissues collected from 50 patients with CRC using reverse transcription-quantitative PCR, and its increased expression levels were significantly associated with increased lymph node and distant metastasis and a higher pathological stage. Furthermore, KRT80 knockdown using siRNA decreased the viability and proliferation of CRC cells. Finally, pathway analysis revealed that the proteins co-expressed with KRT80 in CRC were enriched in the cell cycle, DNA replication, immune system, metabolism of protein and RNA, signal transduction and other cellular processes. Among them, the cell cycle and DNA replication pathways contained the highest number of the proteins identified. In conclusion, the findings of the present study suggested that KRT80 may be overexpressed in CRC tissues. Furthermore, KRT80 may be involved in the proliferation of CRC cells, which is likely through its ability to regulate the cell cycle and DNA replication pathways, thus it may serve as a potential therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Jiatian Lin
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiaoqin Fan
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junhui Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xina Xie
- Guangdong Key Laboratory of Systems and Synthetic Biology for Urogenital Tumors, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hongjian Yu
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- Correspondence to: Dr Hongjian Yu, Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
12
|
Molecular Modeling of Pathogenic Mutations in the Keratin 1B Domain. Int J Mol Sci 2020; 21:ijms21186641. [PMID: 32927888 PMCID: PMC7555247 DOI: 10.3390/ijms21186641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/29/2022] Open
Abstract
Keratin intermediate filaments constitute the primary cytoskeletal component of epithelial cells. Numerous human disease phenotypes related to keratin mutation remain mechanistically elusive. Our recent crystal structures of the helix 1B heterotetramer from keratin 1/10 enabled further investigation of the effect of pathologic 1B domain mutations on keratin structure. We used our highest resolution keratin 1B structure as a template for homology-modeling the 1B heterotetramers of keratin 5/14 (associated with blistering skin disorders), keratin 8/18 (associated with liver disease), and keratin 74/28 (associated with hair disorder). Each structure was examined for the molecular alterations caused by incorporating pathogenic 1B keratin mutations. Structural modeling indicated keratin 1B mutations can harm the heterodimer interface (R265PK5, L311RK5, R211PK14, I150VK18), the tetramer interface (F231LK1, F274SK74), or higher-order interactions needed for mature filament formation (S233LK1, L311RK5, Q169EK8, H128LK18). The biochemical changes included altered hydrophobic and electrostatic interactions, and altered surface charge, hydrophobicity or contour. Together, these findings advance the genotype-structurotype-phenotype correlation for keratin-based human diseases.
Collapse
|
13
|
Xia B, Zhang H, Yang M, Du S, Wei J, Ding L. Leukamenin E Induces K8/18 Phosphorylation and Blocks the Assembly of Keratin Filament Networks Through ERK Activation. Int J Mol Sci 2020; 21:ijms21093164. [PMID: 32365802 PMCID: PMC7246489 DOI: 10.3390/ijms21093164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022] Open
Abstract
Leukamenin E is a natural ent-kaurane diterpenoid isolated from Isodon racemosa (Hemsl) Hara that has been found to be a novel and potential keratin filament inhibitor, but its underlying mechanisms remain largely unknown. Here, we show that leukamenin E induces keratin filaments (KFs) depolymerization, largely independently of microfilament (MFs) and microtubules (MTs) in well-spread cells and inhibition of KFs assembly in spreading cells. These effects are accompanied by keratin phosphorylation at K8-Ser73/Ser431 and K18-Ser52 via the by extracellular signal-regulated kinases (ERK) pathway in primary liver carcinoma cells (PLC) and human umbilical vein endothelial cells (HUVECs). Moreover, leukamenin E increases soluble pK8-Ser73/Ser431, pK18-Ser52, and pan-keratin in the cytoplasmic supernatant by immunofluorescence imaging and Western blotting assay. Accordingly, leukamenin E inhibits the spreading and migration of cells. We propose that leukamenin E-induced keratin phosphorylation may interfere with the initiation of KFs assembly and block the formation of a new KFs network, leading to the inhibition of cell spreading. Leukamenin E is a potential target drug for inhibition of KFs assembly.
Collapse
|
14
|
Schreurs O, Karatsaidis A, Balta MG, Grung B, Hals EKB, Schenck K. Expression of keratins 8, 18, and 19 in epithelia of atrophic oral lichen planus. Eur J Oral Sci 2020; 128:7-17. [PMID: 31994252 DOI: 10.1111/eos.12666] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2019] [Indexed: 12/20/2022]
Abstract
Keratins form intermediate filaments of the cytoskeleton in keratinocytes and have roles in cell structure, signaling, intracellular transport, and cell death. Oral lichen planus (OLP) is an oral inflammatory disease with derangements in basal keratinocytes and disruption of the basal membrane. Here, we focused on epithelial expression of keratins 8, 18, and 19 because these proteins are known to modulate cell death. Biopsies were taken from buccal oral mucosa of persons with normal oral mucosa (n = 10) or atrophic OLP (n = 10). Cultured normal oral keratinocytes (n = 4) showed expression of mRNA and protein for keratins 8, 18, and 19. Immunohistochemistry showed consistent staining for keratins 8 and 18 in basal keratinocytes of normal oral mucosa. In OLP, staining for keratin (K)8 was mostly negative and staining for K18 was weak. Keratin 19 was expressed irregularly in most biopsies of normal oral mucosa and not at all in OLP. Several mononuclear leukocytes in the cellular infiltrate showed membrane staining for K8 and K18. Positive staining for K16 confirmed partial collapse of the basal cell layer in OLP. The basal cell niche in OLP therefore appeared to be partly populated with keratinocytes demonstrating a higher degree of differentiation (K8- K18- K19- K16+ ); consequently, such areas may be more susceptible to the action of cell death factors released from the cell infiltrate as a result of lacking the protective, normal keratin present in the basal epithelial cell layer of normal oral mucosa.
Collapse
Affiliation(s)
- Olav Schreurs
- The CrossTalk Group, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Andreas Karatsaidis
- The CrossTalk Group, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Maria G Balta
- The CrossTalk Group, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | | | - Else K B Hals
- TannSpes and Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Karl Schenck
- The CrossTalk Group, Institute of Oral Biology, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Armstrong C, Cassimeris L, Da Silva Santos C, Micoogullari Y, Wagner B, Babasyan S, Brooks S, Galantino-Homer H. The expression of equine keratins K42 and K124 is restricted to the hoof epidermal lamellae of Equus caballus. PLoS One 2019; 14:e0219234. [PMID: 31550264 PMCID: PMC6759161 DOI: 10.1371/journal.pone.0219234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/11/2019] [Indexed: 01/30/2023] Open
Abstract
The equine hoof inner epithelium is folded into primary and secondary epidermal lamellae which increase the dermo-epidermal junction surface area of the hoof and can be affected by laminitis, a common disease of equids. Two keratin proteins (K), K42 and K124, are the most abundant keratins in the hoof lamellar tissue of Equus caballus. We hypothesize that these keratins are lamellar tissue-specific and could serve as differentiation- and disease-specific markers. Our objective was to characterize the expression of K42 and K124 in equine stratified epithelia and to generate monoclonal antibodies against K42 and K124. By RT-PCR analysis, keratin gene (KRT) KRT42 and KRT124 expression was present in lamellar tissue, but not cornea, haired skin, or hoof coronet. In situ hybridization studies showed that KRT124 localized to the suprabasal and, to a lesser extent, basal cells of the lamellae, was absent from haired skin and hoof coronet, and abruptly transitions from KRT124-negative coronet to KRT124-positive proximal lamellae. A monoclonal antibody generated against full-length recombinant equine K42 detected a lamellar keratin of the appropriate size, but also cross-reacted with other epidermal keratins. Three monoclonal antibodies generated against N- and C-terminal K124 peptides detected a band of the appropriate size in lamellar tissue and did not cross-react with proteins from haired skin, corneal limbus, hoof coronet, tongue, glabrous skin, oral mucosa, or chestnut on immunoblots. K124 localized to lamellar cells by indirect immunofluorescence. This is the first study to demonstrate the localization and expression of a hoof lamellar-specific keratin, K124, and to validate anti-K124 monoclonal antibodies.
Collapse
Affiliation(s)
- Caitlin Armstrong
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Lynne Cassimeris
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Claire Da Silva Santos
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Yagmur Micoogullari
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Susanna Babasyan
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Samantha Brooks
- Department of Animal Sciences and University of Florida Genetics institute, University of Florida, Gainesville, Florida, United States of America
| | - Hannah Galantino-Homer
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Jang KH, Yoon HN, Lee J, Yi H, Park SY, Lee SY, Lim Y, Lee HJ, Cho JW, Paik YK, Hancock WS, Ku NO. Liver disease-associated keratin 8 and 18 mutations modulate keratin acetylation and methylation. FASEB J 2019; 33:9030-9043. [PMID: 31199680 DOI: 10.1096/fj.201800263rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Keratin 8 (K8) and keratin 18 (K18) are the intermediate filament proteins whose phosphorylation/transamidation associate with their aggregation in Mallory-Denk bodies found in patients with various liver diseases. However, the functions of other post-translational modifications in keratins related to liver diseases have not been fully elucidated. Here, using a site-specific mutation assay combined with nano-liquid chromatography-tandem mass spectrometry, we identified K8-Lys108 and K18-Lys187/426 as acetylation sites, and K8-Arg47 and K18-Arg55 as methylation sites. Keratin mutation (Arg-to-Lys/Ala) at the methylation sites, but not the acetylation sites, led to decreased stability of the keratin protein. We compared keratin acetylation/methylation in liver disease-associated keratin variants. The acetylation of K8 variants increased or decreased to various extents, whereas the methylation of K18-del65-72 and K18-I150V variants increased. Notably, the highly acetylated/methylated K18-I150V variant was less soluble and exhibited unusually prolonged protein stability, which suggests that additional acetylation of highly methylated keratins has a synergistic effect on prolonged stability. Therefore, the different levels of acetylation/methylation of the liver disease-associated variants regulate keratin protein stability. These findings extend our understanding of how disease-associated mutations in keratins modulate keratin acetylation and methylation, which may contribute to disease pathogenesis.-Jang, K.-H., Yoon, H.-N., Lee, J., Yi, H., Park, S.-Y., Lee, S.-Y., Lim, Y., Lee, H.-J., Cho, J.-W., Paik, Y.-K., Hancock, W. S., Ku, N.-O. Liver disease-associated keratin 8 and 18 mutations modulate keratin acetylation and methylation.
Collapse
Affiliation(s)
- Kwi-Hoon Jang
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Han-Na Yoon
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Jongeun Lee
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Hayan Yi
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Sang-Yoon Park
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - So-Young Lee
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Younglan Lim
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Hyoung-Joo Lee
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Jin-Won Cho
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Young-Ki Paik
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea
| | - Williams S Hancock
- Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Bio-Convergence Integrated Science and Engineering Division, Underwood International College, Yonsei University, Seoul, Korea
| |
Collapse
|
17
|
Intermediate Filaments as Effectors of Cancer Development and Metastasis: A Focus on Keratins, Vimentin, and Nestin. Cells 2019; 8:cells8050497. [PMID: 31126068 PMCID: PMC6562751 DOI: 10.3390/cells8050497] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Intermediate filament (IF) proteins make up the largest family of cytoskeletal proteins in metazoans, and are traditionally known for their roles in fostering structural integrity in cells and tissues. Remarkably, individual IF genes are tightly regulated in a fashion that reflects the type of tissue, its developmental and differentiation stages, and biological context. In cancer, IF proteins serve as diagnostic markers, as tumor cells partially retain their original signature expression of IF proteins. However, there are also characteristic alterations in IF gene expression and protein regulation. The use of high throughput analytics suggests that tumor-associated alterations in IF gene expression have prognostic value. Parallel research is also showing that IF proteins directly and significantly impact several key cellular properties, including proliferation, death, migration, and invasiveness, with a demonstrated impact on the development, progression, and characteristics of various tumors. In this review, we draw from recent studies focused on three IF proteins most associated with cancer (keratins, vimentin, and nestin) to highlight how several “hallmarks of cancer” described by Hanahan and Weinberg are impacted by IF proteins. The evidence already in hand establishes that IF proteins function beyond their classical roles as markers and serve as effectors of tumorigenesis.
Collapse
|
18
|
Schumacher-Petersen C, Christoffersen BØ, Kirk RK, Ludvigsen TP, Zois NE, Pedersen HD, Vyberg M, Olsen LH. Experimental non-alcoholic steatohepatitis in Göttingen Minipigs: consequences of high fat-fructose-cholesterol diet and diabetes. J Transl Med 2019; 17:110. [PMID: 30943987 PMCID: PMC6448276 DOI: 10.1186/s12967-019-1854-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in humans, and ranges from steatosis to non-alcoholic steatohepatitis (NASH), the latter with risk of progression to cirrhosis. The Göttingen Minipig has been used in studies of obesity and diabetes, but liver changes have not been described. The aim of this study was to characterize hepatic changes in Göttingen Minipigs with or without diabetes, fed a diet high in fat, fructose, and cholesterol to see if liver alterations resemble features of human NAFLD/NASH. METHODS Fifty-four male castrated minipigs (age 6 to 7 months) were distributed into four groups and diet-fed for 13 months. Groups were: lean controls fed standard diet (SD, n = 8), a group fed high fat/fructose/cholesterol diet (FFC, n = 16), a group fed high fat/fructose/cholesterol diet but changed to standard diet after 7 months (diet normalization, FFC/SD, n = 16), and a streptozotocin-induced diabetic group fed high fat/fructose/cholesterol diet (FFCDIA, n = 14). At termination, blood samples for analyses of circulating biomarkers and liver tissue for histopathological assessment and analyses of lipids and glycogen content were collected. RESULTS In comparison with SD and FFC/SD, FFC and FFCDIA pigs developed hepatomegaly with increased content of cholesterol, whereas no difference in triglyceride content was found. FFC and FFCDIA groups had increased values of circulating total cholesterol and triglycerides and the hepatic circulating markers alkaline phosphatase and glutamate dehydrogenase. In the histopathological evaluation, fibrosis (mainly located periportally) and inflammation along with cytoplasmic alterations (characterized by hepatocytes with pale, granulated cytoplasm) were found in FFC and FFCDIA groups compared to SD and FFC/SD. Interestingly, FFC/SD also had fibrosis, a feature not seen in SD. Only two FFC and three FFCDIA pigs had > 5% steatosis, and no hepatocellular ballooning or Mallory-Denk bodies were found in any of the pigs. CONCLUSIONS Fibrosis, inflammation and cytoplasmic alterations were characteristic features in the livers of FCC and FFCDIA pigs. Overall, diabetes did not exacerbate the hepatic changes compared to FFC. The limited presence of the key human-relevant pathological hepatic findings of steatosis and hepatocellular ballooning and the variation in the model, limits its use in preclinical research without further optimisation.
Collapse
Affiliation(s)
- Camilla Schumacher-Petersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 2., 1870, Frederiksberg, Denmark
| | | | - Rikke Kaae Kirk
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Trine Pagh Ludvigsen
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Nora Elisabeth Zois
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark.,In Vivo Pharmacology, Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Henrik Duelund Pedersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 2., 1870, Frederiksberg, Denmark.,Ellegaard Göttingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Mogens Vyberg
- Institute of Pathology, Aalborg University Hospital, Ladegaardsgade 3, 9000, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Soendre Skovvej 15, 9000, Aalborg, Denmark
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 2., 1870, Frederiksberg, Denmark.
| |
Collapse
|
19
|
Corneal keratin aggresome (CKAGG) formation and clearance by proteasome activation. Heliyon 2018; 4:e01012. [PMID: 30619956 PMCID: PMC6313837 DOI: 10.1016/j.heliyon.2018.e01012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/19/2018] [Accepted: 12/03/2018] [Indexed: 01/27/2023] Open
Abstract
Purpose To understand the mechanism of corneal keratin expression and clearance in corneal epithelium with Limbal Stem Cell Deficiency (LSCD). The hypothesis is that LSCD-induced proteasome dysfunction is a contributing factor to keratin aggregation, causing corneal keratin aggresome (CKAGG) formation. Method LSCD was surgically induced in rabbit corneas. LSCD corneal epithelial cells (D-CEC) were collected to investigate keratin K4 and K13 expression and CKAGG formation. Oral mucosal epithelial cells (OMECS) were isolated and cultured to study K4 and K13 expression. Cultured cells were treated with proteasome inhibitor to induce CKAGG formation. Results K4 and K13 were strongly expressed in D-CEC, with additional higher molecular weight bands of K4 and K13, suggesting CKAGG formation. Double staining of K4/K13 and ubiquitin showed co-localization of these keratins with ubiquitin in D-CEC. Proteasome inhibition also showed K4/K13 modification and accumulation in cultured OMECS, similar to D-CEC. Proteasome activation was then performed in cultured OMEC. There was no accumulation of keratins, and levels of unmodified keratins were found significantly reduced. Conclusion Results showed an abnormal expression of K4 and K13 after LSCD-induced proteasome dysfunction, which coalesce to form CKAGG in Corneal Epithelial Cells (CEC). We propose that CKAGG formation may be one of the causative factors of morphological alterations in the injured corneal epithelium, and that CKAGG could potentially be cleared by enhancing proteasome activity.
Collapse
|
20
|
Ramia E, Chiaravalli AM, Bou Nasser Eddine F, Tedeschi A, Sessa F, Accolla RS, Forlani G. CIITA-related block of HLA class II expression, upregulation of HLA class I, and heterogeneous expression of immune checkpoints in hepatocarcinomas: implications for new therapeutic approaches. Oncoimmunology 2018; 8:1548243. [PMID: 30723578 DOI: 10.1080/2162402x.2018.1548243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/31/2018] [Accepted: 11/11/2018] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second cause of death for cancer worldwide, justifying the urgent need for novel therapeutic approaches. Immunotherapeutic strategies based on triggering and/or rescuing tumor antigen-specific T cells may be promising particularly if combined together. As preliminary step toward this goal, we have investigated the expression of antigen presenting molecules (HLA class I and class II) and immune checkpoints (PD-1 and PD-L1) in 43 HCC samples from distinct patients and in HCC cell lines. While normal hepatocytes did not express HLA class I and II, HCC cells strongly upregulated HLA class I while remaining negative for HLA class II. The absence of HLA class II expression in HCC cell lines correlated with lack of expression of the HLA class II transactivator, CIITA, which could not be rescued even after interferon-gamma treatment. This was due to high methylation levels of interferon-gamma-sensitive CIITA promoter IV strongly suggesting a biologically relevant developmental silencing of HLA-II expression in liver cell lineage. HCC tumor tissues showed a variable degree of leukocyte infiltration. Infiltrating lymphocytes expressed PD-1, while PD-L1 was expressed in cells with monocyte-macrophage morphology mostly localized at the tumor margin, but not in tumor cells. De novo expression of HLA class I, instrumental for presenting tumor antigens to cytotoxic T lymphocytes, and the correct characterization of the cells expressing checkpoint inhibitors in the tumor tissue should be the ground for setting novel strategies of combined approaches of immunotherapy in HCC based on tumor peptide vaccines and anti-checkpoint inhibitor antibodies.
Collapse
Affiliation(s)
- Elise Ramia
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Anna Maria Chiaravalli
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Farah Bou Nasser Eddine
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Alessandra Tedeschi
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Roberto S Accolla
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Greta Forlani
- Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| |
Collapse
|
21
|
Alam CM, Silvander JSG, Helenius TO, Toivola DM. Decreased levels of keratin 8 sensitize mice to streptozotocin-induced diabetes. Acta Physiol (Oxf) 2018; 224:e13085. [PMID: 29719117 PMCID: PMC6175344 DOI: 10.1111/apha.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/18/2018] [Accepted: 04/23/2018] [Indexed: 01/10/2023]
Abstract
AIM Diabetes is a result of an interplay between genetic, environmental and lifestyle factors. Keratin intermediate filaments are stress proteins in epithelial cells, and keratin mutations predispose to several human diseases. However, the involvement of keratins in diabetes is not well known. K8 and its partner K18 are the main β-cell keratins, and knockout of K8 (K8-/- ) in mice causes mislocalization of glucose transporter 2, mitochondrial defects, reduced insulin content and altered systemic glucose/insulin control. We hypothesize that K8/K18 offer protection during β-cell stress and that decreased K8 levels contribute to diabetes susceptibility. METHODS K8-heterozygous knockout (K8+/- ) and wild-type (K8+/+ ) mice were used to evaluate the influence of keratin levels on endocrine pancreatic function and diabetes development under basal conditions and after T1D streptozotocin (STZ)-induced β-cell stress and T2D high-fat diet (HFD). RESULTS Murine K8+/- endocrine islets express ~50% less K8/K18 compared with K8+/+ . The decreased keratin levels have little impact on basal systemic glucose/insulin regulation, β-cell health or insulin levels. Diabetes incidence and blood glucose levels are significantly higher in K8+/- mice after low-dose/chronic STZ treatment, and STZ causes more β-cell damage and polyuria in K8+/- compared with K8+/+ . K8 appears upregulated 5 weeks after STZ treatment in K8+/+ islets but not in K8+/- . K8+/- mice showed no major susceptibility risk to HFD compared to K8+/+ . CONCLUSION Partial K8 deficiency reduces β-cell stress tolerance and aggravates diabetes development in response to STZ, while there is no major susceptibility to HFD.
Collapse
Affiliation(s)
- C. M. Alam
- Department of Biosciences, Cell Biology; Faculty of Science and Engineering; Åbo Akademi University; Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; Turku Finland
| | - J. S. G. Silvander
- Department of Biosciences, Cell Biology; Faculty of Science and Engineering; Åbo Akademi University; Turku Finland
| | - T. O. Helenius
- Department of Biosciences, Cell Biology; Faculty of Science and Engineering; Åbo Akademi University; Turku Finland
| | - D. M. Toivola
- Department of Biosciences, Cell Biology; Faculty of Science and Engineering; Åbo Akademi University; Turku Finland
- Turku Center for Disease Modeling; University of Turku; Turku Finland
| |
Collapse
|
22
|
Keratin 80 promotes migration and invasion of colorectal carcinoma by interacting with PRKDC via activating the AKT pathway. Cell Death Dis 2018; 9:1009. [PMID: 30262880 PMCID: PMC6160410 DOI: 10.1038/s41419-018-1030-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Abstract
Little is known about the function of Keratin 80 (KRT80), an epithelial keratin, in cancer. This study investigated the role of KRT80 in the prognosis of colorectal carcinoma (CRC) and the underlying mechanisms involved in CRC migration and invasion. We analyzed the expression of KRT80 using The Cancer Genome Atlas and Oncomine databases. Higher expression of KRT80 was found to be significantly associated with multiple pathological parameters, lower disease-free survival, and overall survival in CRC patients. Also, KRT80 was an independent prognostic indicator for CRC. Furthermore, altered KRT80 expression impacted migration and invasion of CRC cells, as well as the expression of epithelial–mesenchymal transition (EMT)-related markers and cell morphology via the AKT pathway. Inhibiting the expression of AKT could reverse these phenomena. Liquid Chromatograph Mass Spectrometer/Mass Spectromete, Co-immunoprecipitation, and laser scanning confocal microscopy techniques showed that KRT80 could interact with protein kinase, DNA-activated, catalytic polypeptide (PRKDC). Suppressing PRKDC could inhibit the expression of AKT and EMT, as well as the migration and invasion of CRC cells. Taken together, these results demonstrated that KRT80 was an independent prognostic biomarker for CRC and promoted CRC migration and invasion by interacting with PRKDC via activation of the AKT pathway.
Collapse
|
23
|
Li R, Liao XH, Ye JZ, Li MR, Wu YQ, Hu X, Zhong BH. Association of keratin 8/18 variants with non-alcoholic fatty liver disease and insulin resistance in Chinese patients: A case-control study. World J Gastroenterol 2017; 23:4047-4053. [PMID: 28652657 PMCID: PMC5473123 DOI: 10.3748/wjg.v23.i22.4047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/10/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To test the hypothesis that K8/K18 variants predispose humans to non-alcoholic fatty liver disease (NAFLD) progression and its metabolic phenotypes.
METHODS We selected a total of 373 unrelated adult subjects from our Physical Examination Department, including 200 unrelated NAFLD patients and 173 controls of both genders and different ages. Diagnoses of NAFLD were established according to ultrasonic signs of fatty liver. All subjects were tested for population characteristics, lipid profile, liver tests, as well as glucose tests. Genomic DNA was obtained from peripheral blood with a DNeasy Tissue Kit. K8/K18 coding regions were analyzed, including 15 exons and exon-intron boundaries.
RESULTS Among 200 NAFLD patients, 10 (5%) heterozygous carriers of keratin variants were identified. There were 5 amino-acid-altering heterozygous variants and 6 non-coding heterozygous variants. One novel amino-acid-altering heterozygous variant (K18 N193S) and three novel non-coding variants were observed (K8 IVS5-9A→G, K8 IVS6+19G→A, K18 T195T). A total of 9 patients had a single variant and 1 patient had compound variants (K18 N193S+K8 IVS3-15C→G). Only one R341H variant was found in the control group (1 of 173, 0.58%). The frequency of keratin variants in NAFLD patients was significantly higher than that in the control group (5% vs 0.58%, P = 0.015). Notably, the keratin variants were significantly associated with insulin resistance (IR) in NAFLD patients (8.86% in NAFLD patients with IR vs 2.5% in NAFLD patients without IR, P = 0.043).
CONCLUSION K8/K18 variants are overrepresented in Chinese NAFLD patients and might accelerate liver fat storage through IR.
Collapse
|
24
|
Guldiken N, Kobazi Ensari G, Lahiri P, Couchy G, Preisinger C, Liedtke C, Zimmermann HW, Ziol M, Boor P, Zucman-Rossi J, Trautwein C, Strnad P. Keratin 23 is a stress-inducible marker of mouse and human ductular reaction in liver disease. J Hepatol 2016; 65:552-9. [PMID: 27151178 DOI: 10.1016/j.jhep.2016.04.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/06/2016] [Accepted: 04/23/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Keratins (K) constitute the epithelial intermediate filaments. Among them, K7/K19 are widely used markers of the regenerative liver response termed ductular reaction (DR) that consists of activated biliary epithelial cells (BECs) and hepatic progenitor cells (HPCs) and correlates with liver disease severity. In the present study we aimed to characterize K23 in the liver. METHODS We analyzed the expression and localization of K23 in the digestive system under basal conditions as well as in various human and mouse liver diseases/stress models. Cell culture studies were used to study factors regulating K23 expression. RESULTS In untreated mice, K23 was restricted to biliary epithelia. It was (together with K7/K19) markedly upregulated in three different DR/cholestatic injury models, i.e., multidrug resistance protein 2 (Mdr2) knockouts, animals treated with 3,5-diethoxycarbonyl-1,4-dihydrocollidine or subjected to bile duct ligation. K23 levels correlated with the DR marker Fn14 and immunofluorescence staining showed a distinct co-localization with K7/K19. In chronic human liver disease, K23 expression increased in patients with a more prominent inflammation/fibrosis. A dramatic upregulation (>200times) was observed in patients with acute liver failure (ALF) and end-stage primary biliary cholangitis (PBC). Patients with alcoholic liver cirrhosis displayed increased K23 serum levels. In primary hepatocytes as well as hepatobiliary cell lines, treatment with TNF-related weak inducer of apoptosis (TWEAK), and the type I acute phase inducer interleukin (IL)-1β but not the type II inducer IL-6 elevated K23 expression. CONCLUSIONS K23 represents a specific, stress-inducible DR marker, whose levels correlate with liver disease severity. K23 may represent a useful non-invasive DR marker. LAY SUMMARY Ductular reaction represents a basic response to liver injury and correlates with liver disease severity. Our study identifies K23 as a novel ductular reaction marker in mice and humans.
Collapse
Affiliation(s)
- Nurdan Guldiken
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany
| | - Gokce Kobazi Ensari
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | - Pooja Lahiri
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gabrielle Couchy
- Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, Equipe Labellisée Ligue Contre le Cancer, Paris F-75010, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, F-75010 Paris, France; Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Université Paris Diderot, IUH, F-75010 Paris, France
| | - Christian Preisinger
- Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany
| | - Christian Liedtke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | | | - Marianne Ziol
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Pathology Department, GH Paris-Seine-Saint-Denis, APHP, Bondy, France; Centre de ressources biologiques BB-0033-00027 - Hôpital Jean Verdier, GH Paris-Seine-Saint-Denis, APHP, Bondy, France
| | - Peter Boor
- Department of Nephrology and Institute of Pathology, RWTH University Hospital Aachen, Germany
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, Equipe Labellisée Ligue Contre le Cancer, Paris F-75010, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, F-75010 Paris, France; Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France; Université Paris Diderot, IUH, F-75010 Paris, France
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH University Hospital Aachen, Germany.
| |
Collapse
|
25
|
Ku NO, Strnad P, Bantel H, Omary MB. Keratins: Biomarkers and modulators of apoptotic and necrotic cell death in the liver. Hepatology 2016; 64:966-76. [PMID: 26853542 PMCID: PMC4977204 DOI: 10.1002/hep.28493] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Keratins, formerly known as cytokeratins, are the major epithelial-specific subgroup of intermediate filament proteins. Adult hepatocytes express keratin polypeptides 8 and 18 (K8/K18), whereas cholangiocytes express K8/K18 and keratins 7 and 19 (K7/K19). Keratins function primarily to protect hepatocytes from apoptosis and necrosis, which was revealed using several genetic mouse models. This cytoprotective function was further clarified by the identification of natural human keratin variants that are normally silent, but become pathogenic by predisposing their carriers to apoptosis during acute or chronic liver injury mediated by toxins, virus infection, or metabolic stress. During apoptosis, caspases cleave K18 and K19 at conserved aspartates (human K18/K19: (235) Val-Glu-Val-Asp(↓) ) and K18 at a unique aspartate (human K18: (394) Asp-Ala-Leu-Asp(↓) ), with the latter exposed epitope becoming recognized by the M30 antibody in blood and tissues. Additional K18-containing protein backbone epitopes are detected using the M6 and M5 (termed M65) antibodies. Intact K18 and its associated fragments, which are released into blood during apoptosis and necrosis in various diseases, have been analyzed by enzyme-linked immunosorbent assay using the M30/M65 antibodies or their signal ratios. Furthermore, M30/M65 levels have been used as diagnostic and prognostic biomarkers in acute and chronic liver diseases, including nonalcoholic steatohepatitis and acute liver failure. Other keratin biomarkers include K8/K18/K19-related tissue polypeptide antigen, K18-related tissue polypeptide-specific antigen, and K19-related CYFRA-21-1, which have been evaluated mostly in patients with epithelial tumors. CONCLUSION Keratins and their fragments are released into blood during liver and other epithelial tissue injury. The epithelial specificity of K18/K19, epitope unmasking upon caspase digestion, keratin abundance, and relative keratin stability render them useful biomarkers for hepatocyte and cholangiocyte apoptosis and necrosis. However, the precise biochemical nature and release mechanism of circulating keratins remain unknown. (Hepatology 2016;64:966-976).
Collapse
Affiliation(s)
- Nam-On Ku
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622,Department of Medicine, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622; and VA Ann Arbor Health Care System, Ann Arbor MI, 48105
| |
Collapse
|
26
|
Bigaud E, Corrales FJ. Methylthioadenosine (MTA) Regulates Liver Cells Proteome and Methylproteome: Implications in Liver Biology and Disease. Mol Cell Proteomics 2016; 15:1498-1510. [PMID: 26819315 PMCID: PMC4858935 DOI: 10.1074/mcp.m115.055772] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/22/2016] [Indexed: 12/21/2022] Open
Abstract
Methylthioadenosine phosphorylase (MTAP), a key enzyme in the adenine and methionine salvage pathways, catalyzes the hydrolysis of methylthioadenosine (MTA), a compound suggested to affect pivotal cellular processes in part through the regulation of protein methylation. MTAP is expressed in a wide range of cell types and tissues, and its deletion is common to cancer cells and in liver injury. The aim of this study was to investigate the proteome and methyl proteome alterations triggered by MTAP deficiency in liver cells to define novel regulatory mechanisms that may explain the pathogenic processes of liver diseases. iTRAQ analysis resulted in the identification of 216 differential proteins (p < 0.05) that suggest deregulation of cellular pathways as those mediated by ERK or NFκB. R-methyl proteome analysis led to the identification of 74 differentially methylated proteins between SK-Hep1 and SK-Hep1+ cells, including 116 new methylation sites. Restoring normal MTA levels in SK-Hep1+ cells parallels the specific methylation of 56 proteins, including KRT8, TGF, and CTF8A, which provides a novel regulatory mechanism of their activity with potential implications in carcinogenesis. Inhibition of RNA-binding proteins methylation is especially relevant upon accumulation of MTA. As an example, methylation of quaking protein in Arg(242) and Arg(256) in SK-Hep1+ cells may play a pivotal role in the regulation of its activity as indicated by the up-regulation of its target protein p27(kip1) The phenotype associated with a MTAP deficiency was further verified in the liver of MTAP± mice. Our data support that MTAP deficiency leads to MTA accumulation and deregulation of central cellular pathways, increasing proliferation and decreasing the susceptibility to chemotherapeutic drugs, which involves differential protein methylation. Data are available via ProteomeXchange with identifier PXD002957 (http://www.ebi.ac.uk/pride/archive/projects/PXD002957).
Collapse
Affiliation(s)
- Emilie Bigaud
- From the §Department of Hepatology, Proteomics Laboratory, CIMA, University of Navarra; CIBERehd; IDISNA, Pamplona, 31008 Spain
| | - Fernando J Corrales
- From the §Department of Hepatology, Proteomics Laboratory, CIMA, University of Navarra; CIBERehd; IDISNA, Pamplona, 31008 Spain
| |
Collapse
|
27
|
Djudjaj S, Papasotiriou M, Bülow RD, Wagnerova A, Lindenmeyer MT, Cohen CD, Strnad P, Goumenos DS, Floege J, Boor P. Keratins are novel markers of renal epithelial cell injury. Kidney Int 2016; 89:792-808. [PMID: 26924053 DOI: 10.1016/j.kint.2015.10.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 09/25/2015] [Accepted: 10/22/2015] [Indexed: 12/14/2022]
Abstract
Keratins, the intermediate filaments of the epithelial cell cytoskeleton, are up-regulated and post-translationally modified in stress situations. Renal tubular epithelial cell stress is a common finding in progressive kidney diseases, but little is known about keratin expression and phosphorylation. Here, we comprehensively describe keratin expression in healthy and diseased kidneys. In healthy mice, the major renal keratins, K7, K8, K18, and K19, were expressed in the collecting ducts and K8, K18 in the glomerular parietal epithelial cells. Tubular expression of all 4 keratins increased by 20- to 40-fold in 5 different models of renal tubular injury as assessed by immunohistochemistry, Western blot, and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). The up-regulation became significant early after disease induction, increased with disease progression, was found de novo in distal tubules and was accompanied by altered subcellular localization. Phosphorylation of K8 and K18 increased under stress. In humans, injured tubules also exhibited increased keratin expression. Urinary K18 was only detected in mice and patients with tubular cell injury. Keratins labeled glomerular parietal epithelial cells forming crescents in patients and animals. Thus, all 4 major renal keratins are significantly, early, and progressively up-regulated upon tubular injury regardless of the underlying disease and may be novel sensitive markers of renal tubular cell stress.
Collapse
Affiliation(s)
- Sonja Djudjaj
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Institute of Pathology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Marios Papasotiriou
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Department of Nephrology, University Hospital of Patras, Patras, Greece
| | - Roman D Bülow
- Institute of Pathology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Alexandra Wagnerova
- Institute of Pathology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - Maja T Lindenmeyer
- Division of Nephrology and Institute of Physiology, University Zürich, Zürich, Switzerland
| | - Clemens D Cohen
- Division of Nephrology and Institute of Physiology, University Zürich, Zürich, Switzerland
| | - Pavel Strnad
- Department of Internal Medicine 3 and Interdisziplinäres Zentrum für Klinische Forschung, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | | | - Jürgen Floege
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Peter Boor
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Institute of Pathology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
28
|
Chen Y, Guldiken N, Spurny M, Mohammed HHA, Haybaeck J, Pollheimer MJ, Fickert P, Gassler N, Jeon MK, Trautwein C, Strnad P. Loss of keratin 19 favours the development of cholestatic liver disease through decreased ductular reaction. J Pathol 2015; 237:343-354. [PMID: 26108453 DOI: 10.1002/path.4580] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 06/21/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
Keratins (K) are cytoprotective proteins and keratin mutations predispose to the development of multiple human diseases. K19 represents the most widely used marker of biliary and hepatic progenitor cells as well as a marker of ductular reaction that constitutes the basic regenerative response to chronic liver injury. In the present study, we investigated the role of K19 in biliary and hepatic progenitor cells and its importance for ductular reaction. K19 wild-type (WT) and knockout (KO) mice were fed: (a) 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC); (b) cholic acid (CA); (c) a choline-deficient, ethionine-supplemented (CDE) diet; or (d) were subjected to common bile duct ligation (CBDL). The bile composition, liver damage, bile duct proliferation, oval cell content and biliary fibrosis were analysed. In untreated animals, loss of K19 led to redistribution of the K network in biliary epithelial cells (BECs) but to no obvious biliary phenotype. After DDC feeding, K19 KO mice exhibited (compared to WTs): (a) increased cholestasis; (b) less pronounced ductular reaction with reduced ductular proliferation and fewer oval cells; (c) impaired Notch 2 signalling in BECs; (d) lower biliary fibrosis score and biliary bicarbonate concentration. An attenuated oval cell proliferation in K19 KOs was also found after feeding with the CDE diet. K19 KOs subjected to CBDL displayed lower BEC proliferation, oval cell content and less prominent Notch 2 signal. K19 deficiency did not change the extent of CA- or CBDL-induced liver injury and fibrosis. Our results demonstrate that K19 plays an important role in the ductular reaction and might be of importance in multiple chronic liver disorders that frequently display a ductular reaction.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Chemical and Drug Induced Liver Injury/etiology
- Chemical and Drug Induced Liver Injury/genetics
- Chemical and Drug Induced Liver Injury/metabolism
- Chemical and Drug Induced Liver Injury/pathology
- Cholangitis, Sclerosing/chemically induced
- Cholangitis, Sclerosing/genetics
- Cholangitis, Sclerosing/metabolism
- Cholangitis, Sclerosing/pathology
- Cholestasis, Extrahepatic/etiology
- Cholestasis, Extrahepatic/genetics
- Cholestasis, Extrahepatic/metabolism
- Cholestasis, Extrahepatic/pathology
- Cholic Acid
- Choline Deficiency/complications
- Common Bile Duct/metabolism
- Common Bile Duct/pathology
- Common Bile Duct/surgery
- Disease Models, Animal
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Ethionine
- Keratin-19/deficiency
- Keratin-19/genetics
- Ligation
- Liver/metabolism
- Liver/pathology
- Liver Cirrhosis, Biliary/chemically induced
- Liver Cirrhosis, Biliary/genetics
- Liver Cirrhosis, Biliary/metabolism
- Liver Cirrhosis, Biliary/pathology
- Liver Regeneration
- Male
- Mice, Knockout
- Phenotype
- Pyridines
- Signal Transduction
- Stem Cells/metabolism
- Stem Cells/pathology
- Time Factors
Collapse
Affiliation(s)
- Yu Chen
- Department of Internal Medicine III and IZKF, RWTH Aachen, Germany
- Department of Internal Medicine I, University Medical Centre Ulm, Germany
| | - Nurdan Guldiken
- Department of Internal Medicine III and IZKF, RWTH Aachen, Germany
- Department of Internal Medicine I, University Medical Centre Ulm, Germany
| | - Manuela Spurny
- Department of Internal Medicine I, University Medical Centre Ulm, Germany
| | | | | | - Marion J Pollheimer
- Institute of Pathology, Medical University Graz, Austria
- Department of Internal Medicine, Medical University Graz, Austria
| | - Peter Fickert
- Institute of Pathology, Medical University Graz, Austria
- Department of Internal Medicine, Medical University Graz, Austria
| | - Nikolaus Gassler
- Institute of Pathology, University Hospital Aachen, RWTH Aachen, Germany
| | - Min Kyung Jeon
- Institute of Pathology, University Hospital Aachen, RWTH Aachen, Germany
| | | | - Pavel Strnad
- Department of Internal Medicine III and IZKF, RWTH Aachen, Germany
- Department of Internal Medicine I, University Medical Centre Ulm, Germany
| |
Collapse
|
29
|
Wang X, Chen P, Cui J, Yang C, Du H. Keratin 8 is a novel autoantigen of rheumatoid arthritis. Biochem Biophys Res Commun 2015; 465:665-9. [DOI: 10.1016/j.bbrc.2015.07.161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 11/29/2022]
|
30
|
Toivola DM, Habtezion A, Misiorek JO, Zhang L, Nyström JH, Sharpe O, Robinson WH, Kwan R, Omary MB. Absence of keratin 8 or 18 promotes antimitochondrial autoantibody formation in aging male mice. FASEB J 2015; 29:5081-9. [PMID: 26399787 DOI: 10.1096/fj.14-269795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 09/08/2015] [Indexed: 12/16/2022]
Abstract
Human mutations in keratin 8 (K8) and keratin 18 (K18), the intermediate filament proteins of hepatocytes, predispose to several liver diseases. K8-null mice develop chronic liver injury and fragile hepatocytes, dysfunctional mitochondria, and Th2-type colitis. We tested the hypothesis that autoantibody formation accompanies the liver damage that associates with K8/K18 absence. Sera from wild-type control, K8-null, and K18-null mice were analyzed by immunoblotting and immunofluorescence staining of cell and mouse tissue homogenates. Autoantibodies to several antigens were identified in 81% of K8-null male mice 8 mo or older. Similar autoantibodies were detected in aging K18-null male mice that had a related liver phenotype but normal colon compared with K8-null mice, suggesting that the autoantibodies are linked to liver rather than colonic disease. However, these autoantibodies were not observed in nontransgenic mice subjected to 4 chronic injury models. The autoantigens are ubiquitous and partition with mitochondria. Mass spectrometry and purified protein analysis identified, mitochondrial HMG-CoA synthase, aldehyde dehydrogenase, and catalase as the primary autoantigens, and glutamate dehydrogenase and epoxide hydrolase-2 as additional autoantigens. Therefore, absence of the hepatocyte keratins results in production of anti-mitochondrial autoantibodies (AMA) that recognize proteins involved in energy metabolism and oxidative stress, raising the possibility that AMA may be found in patients with keratin mutations that associate with liver and other diseases.
Collapse
Affiliation(s)
- Diana M Toivola
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Aida Habtezion
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Julia O Misiorek
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Linxing Zhang
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Joel H Nyström
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Orr Sharpe
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - William H Robinson
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Raymond Kwan
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - M Bishr Omary
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Guldiken N, Zhou Q, Kucukoglu O, Rehm M, Levada K, Gross A, Kwan R, James LP, Trautwein C, Omary MB, Strnad P. Human keratin 8 variants promote mouse acetaminophen hepatotoxicity coupled with c-jun amino-terminal kinase activation and protein adduct formation. Hepatology 2015; 62:876-86. [PMID: 25963979 PMCID: PMC4549164 DOI: 10.1002/hep.27891] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/15/2015] [Accepted: 05/03/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Keratins 8 and 18 (K8/K18) are the intermediate filaments proteins of simple-type digestive epithelia and provide important cytoprotective function. K8/K18 variants predispose humans to chronic liver disease progression and poor outcomes in acute acetaminophen (APAP)-related liver failure. Given that K8 G62C and R341H/R341C are common K8 variants in European and North American populations, we studied their biological significance using transgenic mice. Mice that overexpress the human K8 variants, R341H or R341C, were generated and used together with previously described mice that overexpress wild-type K8 or K8 G62C. Mice were injected with 600 mg/kg of APAP or underwent bile duct ligation (BDL). Livers were evaluated by microarray analysis, quantitative real-time polymerase chain reaction, immunoblotting, histological and immunological staining, and biochemical assays. Under basal conditions, the K8 G62C/R341H/R341C variant-expressing mice did not show an obvious liver phenotype or altered keratin filament distribution, whereas K8 G62C/R341C animals had aberrant disulphide cross-linked keratins. Animals carrying the K8 variants displayed limited gene expression changes, but had lower nicotinamide N-methyl transferase (NNMT) levels and were predisposed to APAP-induced hepatotoxicity. NNMT represents a novel K8/K18-associated protein that becomes up-regulated after K8/K18 transfection. The more pronounced liver damage was accompanied by increased and prolonged JNK activation; elevated APAP protein adducts; K8 hyperphosphorylation at S74/S432 with enhanced keratin solubility; and prominent pericentral keratin network disruption. No differences in APAP serum levels, glutathione, or adenosine triphosphate levels were noted. BDL resulted in similar liver injury and biliary fibrosis in all mouse genotypes. CONCLUSION Expression of human K8 variants G62C, R341H, or R341C in mice predisposes to acute APAP hepatotoxicity, thereby providing direct evidence for the importance of these variants in human acute liver failure.
Collapse
Affiliation(s)
- Nurdan Guldiken
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany,Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Qin Zhou
- Department of Medicine, Palo Alto VA Medical Center, CA; and Stanford University Digestive Disease Center, USA
| | - Ozlem Kucukoglu
- Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Melanie Rehm
- Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Kateryna Levada
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - Annika Gross
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - Raymond Kwan
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, and the VA Ann Arbor Health Care System, Ann Arbor, MI, USA
| | - Laura P. James
- Arkansas Children's Hospital Research Institute and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Christian Trautwein
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, and the VA Ann Arbor Health Care System, Ann Arbor, MI, USA
| | - Pavel Strnad
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany,Department of Internal Medicine I, University Hospital Ulm, Ulm Germany,To whom correspondence should be addressed. Corresponding author: Pavel Strnad, Department of Internal Medicine III and IZKF, University Hospital Aachen, Pauwelsstraße 30, D-52074 Aachen, Tel.: +49(241) 80-35324, Fax: +49(241) 80-82455,
| |
Collapse
|
32
|
Usachov V, Urban TJ, Fontana RJ, Gross A, Iyer S, Omary MB, Strnad P. Prevalence of genetic variants of keratins 8 and 18 in patients with drug-induced liver injury. BMC Med 2015; 13:196. [PMID: 26286715 PMCID: PMC4545365 DOI: 10.1186/s12916-015-0418-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 07/03/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Keratin 8 and 18 (K8/K18) cytoskeletal proteins protect hepatocytes from undergoing apoptosis and their mutations predispose to adverse outcomes in acute liver failure (ALF). All known K8/K18 variants occur at relatively non-conserved residues and do not cause keratin cytoskeleton reorganization, whereas epidermal keratin-conserved residue mutations disrupt the keratin cytoskeleton and cause severe skin disease. The aim of our study was to identify keratin variants in idiosyncratic drug-induced liver injury (DILI). METHODS Genomic DNA was isolated from 800 patients enrolled in an ongoing US multicenter study, with DILI attributed to a wide range of drugs. Specific K8/K18 exonic regions were PCR-amplified and screened by denaturing HPLC followed by DNA sequencing. The functional impact of keratin variants was assessed using cell transfection and immune staining. RESULTS Heterozygous and compound amino acid-altering K8/K18 variants were identified in 86 DILI patients and non-coding variants in 15 subjects. Five novel amino acid-altering (K8 Lys393Arg, K8 Ala351Val, K8 Ala358Val, K8 Ile346Val, K18 Asp89His) and two non-coding variants were observed. Several variants segregated with specific ethnic backgrounds but were found at similar frequencies in DILI subjects and ethnically matched population controls. Notably, variants in highly conserved residues of K8 Lys393Arg (ezetimibe/simvastatin-related) and K18 Asp89His (isoniazid-related) were found in patients with fatal DILI. These novel variants also led to keratin network disruption in transfected cells. CONCLUSIONS Novel K8/K18 cytoskeleton-disrupting variants were identified in two patients and segregated with fatal DILI. Other non-cytoskeleton-disrupting keratin variants did not preferentially associate with DILI.
Collapse
Affiliation(s)
- Valentyn Usachov
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany. .,Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany.
| | - Thomas J Urban
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, UNC Eshelman School of Pharmacy, UNC Hamner Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, NC, USA.
| | - Robert J Fontana
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA.
| | - Annika Gross
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| | - Sapna Iyer
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| | | |
Collapse
|
33
|
Sun DW, Zhang YY, Sun XD, Chen YG, Qiu W, Ji M, Lv GY. Prognostic value of cytokeratin 19 in hepatocellular carcinoma: A meta-analysis. Clin Chim Acta 2015; 448:161-9. [PMID: 26164382 DOI: 10.1016/j.cca.2015.06.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 06/25/2015] [Accepted: 06/27/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Although many studies have investigated the relationship between cytokeratin 19 (CK-19) and hepatocellular carcinoma (HCC), the prognostic value of CK-19 in HCC remains inconclusive. METHODS Eligible studies were sought in PubMed, Embase, Web of Science, Cochrane Library and Wanfang databases. Pooled hazard ratios (HRs) and odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were calculated. RESULTS 17 studies with 2943 patients were included in this meta-analysis. Meta-analysis results showed that CK-19 over-expression was significantly associated with overall survival (OS) (HR=1.60, 95% CI: 1.32-1.93, univariate analysis; HR=2.25, 95% CI: 1.79-2.83, multivariate analysis) and disease-free survival (DFS) (HR=1.68, 95% CI: 1.35-2.10, univariate analysis; HR=1.97, 95% CI: 1.54-2.53, multivariate analysis). Meanwhile, CK-19 over-expression was also correlated with decreased 1-year OS rate (OR=0.32, 95% CI: 0.21-0.50), 5-year OS rate (OR=0.44, 95% CI: 0.14-0.87) and 1-year DFS rate (OR=0.51, 95% CI: 0.34-0.76), but not with 5-year DFS rate (OR=0.62, 95% CI: 0.35-1.10). These results suggested that CK-19 over-expression was significantly associated with poor survival rate and early tumor recurrence rate in HCC patients. CONCLUSIONS CK-19 can serve as an indicator of poor prognosis as well as a novel target for treatment in HCC.
Collapse
Affiliation(s)
- Da-wei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China
| | - Ying-yi Zhang
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao-dong Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China
| | - Yu-guo Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China
| | - Wei Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China
| | - Min Ji
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China
| | - Guo-yue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, Jilin Province, China.
| |
Collapse
|
34
|
Szabo S, Wögenstein KL, Österreicher CH, Guldiken N, Chen Y, Doler C, Wiche G, Boor P, Haybaeck J, Strnad P, Fuchs P. Epiplakin attenuates experimental mouse liver injury by chaperoning keratin reorganization. J Hepatol 2015; 62:1357-66. [PMID: 25617501 PMCID: PMC4451473 DOI: 10.1016/j.jhep.2015.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/08/2014] [Accepted: 01/05/2015] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Epiplakin is a member of the plakin protein family and exclusively expressed in epithelial tissues where it binds to keratins. Epiplakin-deficient (Eppk1(-/-)) mice displayed no obvious spontaneous phenotype, but their keratinocytes showed a faster keratin network breakdown in response to stress. The role of epiplakin in the stressed liver remained to be elucidated. METHODS Wild-type (WT) and Eppk1(-/-) mice were subjected to common bile duct ligation (CBDL) or fed with a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing diet. The importance of epiplakin during keratin reorganization was assessed in primary hepatocytes. RESULTS Our experiments revealed that epiplakin is expressed in hepatocytes and cholangiocytes, and binds to keratin 8 (K8) and K18 via multiple domains. In several liver stress models epiplakin and K8 genes displayed identical expression patterns and transgenic K8 overexpression resulted in elevated hepatic epiplakin levels. After CBDL and DDC treatment, Eppk1(-/-) mice developed a more pronounced liver injury and their livers contained larger amounts of hepatocellular keratin granules, indicating impaired disease-induced keratin network reorganization. In line with these findings, primary Eppk1(-/-) hepatocytes showed increased formation of keratin aggregates after treatment with the phosphatase inhibitor okadaic acid, a phenotype which was rescued by the chemical chaperone trimethylamine N-oxide (TMAO). Finally, transfection experiments revealed that Eppk1(-/-) primary hepatocytes were less able to tolerate forced K8 overexpression and that TMAO treatment rescued this phenotype. CONCLUSION Our data indicate that epiplakin plays a protective role during experimental liver injuries by chaperoning disease-induced keratin reorganization.
Collapse
Affiliation(s)
- Sandra Szabo
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Karl L Wögenstein
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Christoph H Österreicher
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nurdan Guldiken
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Yu Chen
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Carina Doler
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gerhard Wiche
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Peter Boor
- Division of Nephrology and Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | | | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Peter Fuchs
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Guldiken N, Usachov V, Levada K, Trautwein C, Ziol M, Nahon P, Strnad P. Keratins 8 and 18 are type II acute-phase responsive genes overexpressed in human liver disease. Liver Int 2015; 35:1203-12. [PMID: 24930437 DOI: 10.1111/liv.12608] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 05/18/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Keratins (Ks) 7, 8, 18 and 19 constitute important markers and modifiers of liver disease. In mice, K8 and K18 are stress inducible and a dysregulated K8 > K18 stoichiometry predisposes to formation of Mallory-Denk bodies (MDBs), i.e. aggregates characteristic of chronic liver disorders such as alcoholic liver disease (ALD). In our study, we analyse the expression and the regulation of keratins in context of human liver disease. METHODS K7, K8, K18 and K19 mRNA levels were determined in liver biopsies from patients with ALD, non-alcoholic steatohepatitis (NASH), chronic hepatitis B (HBV), hepatitis C (HCV) and from control subjects. HepG2 and Hep3B cells were treated with IL-1β, IL-6 and TNF-α. Mice were injected with turpentine, an established IL-6 inducer. RESULTS K7, K8 and K18 were 1.5- to 3-fold upregulated in livers of ALD and HCV patients with a more active disease, but not in HBV/NASH subjects, while K19 was significantly elevated in all analysed disorders. K8 and K18 expression displayed a strong correlation (r = 0.89), but dysregulated levels with the K8 > K18 state were seen in ALD. All keratins were overexpressed in subjects with moderate vs. minimal inflammation, while K7, K8 and K18 were upregulated in patients with advanced liver fibrosis. In HepG2/Hep3B cells, IL-6 treatment but not IL-1β or TNF-α significantly increased K8 and K18 expression and elevated K18 levels were seen after turpentine injection. CONCLUSIONS Keratins represent type II acute-phase responsive genes overexpressed in specific human liver disorders. A K8 > K18 state occurs in ALD and predisposes to MDB formation.
Collapse
Affiliation(s)
- Nurdan Guldiken
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany; IZKF and Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Toivola DM, Boor P, Alam C, Strnad P. Keratins in health and disease. Curr Opin Cell Biol 2015; 32:73-81. [PMID: 25599598 DOI: 10.1016/j.ceb.2014.12.008] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/09/2014] [Accepted: 12/19/2014] [Indexed: 02/01/2023]
Abstract
The cytoprotective keratins (K) compose the intermediate filaments of epithelial cells and their inherited and spontaneous mutations give rise to keratinopathies. For example, mutations in K1/K5/K10/K14 cause epidermal skin diseases whereas simple epithelial K8/K18/K19 variants predispose to development of several liver disorders. Due to their abundance, tissue- and context-specific expression, keratins constitute excellent diagnostic markers of both neoplastic and non-neoplastic diseases. During injury and in disease, keratin expression levels, cellular localization or posttranslational modifications are altered. Accumulating evidence suggests that these changes modulate multiple processes including cell migration, tumor growth/metastasis and development of infections. Therefore, our understanding of keratins is shifting from diagnostic markers to active disease modifiers.
Collapse
Affiliation(s)
- Diana M Toivola
- Department of Biosciences, Cell Biology, Åbo Akademi University and Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, RWTH University, Aachen, Germany; Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - Catharina Alam
- Department of Biosciences, Cell Biology, Åbo Akademi University and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pavel Strnad
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany.
| |
Collapse
|
37
|
Der Perng M, Quinlan RA. The Dynamic Duo of Small Heat Proteins and IFs Maintain Cell Homeostasis, Resist Cellular Stress and Enable Evolution in Cells and Tissues. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
The cultural divide: exponential growth in classical 2D and metabolic equilibrium in 3D environments. PLoS One 2014; 9:e106973. [PMID: 25222612 PMCID: PMC4164521 DOI: 10.1371/journal.pone.0106973] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/04/2014] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Cellular metabolism can be considered to have two extremes: one is characterized by exponential growth (in 2D cultures) and the other by a dynamic equilibrium (in 3D cultures). We have analyzed the proteome and cellular architecture at these two extremes and found that they are dramatically different. RESULTS Structurally, actin organization is changed, microtubules are increased and keratins 8 and 18 decreased. Metabolically, glycolysis, fatty acid metabolism and the pentose phosphate shunt are increased while TCA cycle and oxidative phosphorylation is unchanged. Enzymes involved in cholesterol and urea synthesis are increased consistent with the attainment of cholesterol and urea production rates seen in vivo. DNA repair enzymes are increased even though cells are predominantly in Go. Transport around the cell--along the microtubules, through the nuclear pore and in various types of vesicles has been prioritized. There are numerous coherent changes in transcription, splicing, translation, protein folding and degradation. The amount of individual proteins within complexes is shown to be highly coordinated. Typically subunits which initiate a particular function are present in increased amounts compared to other subunits of the same complex. SUMMARY We have previously demonstrated that cells at dynamic equilibrium can match the physiological performance of cells in tissues in vivo. Here we describe the multitude of protein changes necessary to achieve this performance.
Collapse
|
39
|
Snider NT, Omary MB. Post-translational modifications of intermediate filament proteins: mechanisms and functions. Nat Rev Mol Cell Biol 2014; 15:163-77. [PMID: 24556839 PMCID: PMC4079540 DOI: 10.1038/nrm3753] [Citation(s) in RCA: 383] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intermediate filaments (IFs) are cytoskeletal and nucleoskeletal structures that provide mechanical and stress-coping resilience to cells, contribute to subcellular and tissue-specific biological functions, and facilitate intracellular communication. IFs, including nuclear lamins and those in the cytoplasm (keratins, vimentin, desmin, neurofilaments and glial fibrillary acidic protein, among others), are functionally regulated by post-translational modifications (PTMs). Proteomic advances highlight the enormous complexity and regulatory potential of IF protein PTMs, which include phosphorylation, glycosylation, sumoylation, acetylation and prenylation, with novel modifications becoming increasingly appreciated. Future studies will need to characterize their on-off mechanisms, crosstalk and utility as biomarkers and targets for diseases involving the IF cytoskeleton.
Collapse
Affiliation(s)
- Natasha T. Snider
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - M. Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
40
|
Alam CM, Silvander JSG, Daniel EN, Tao GZ, Kvarnström SM, Alam P, Omary MB, Hänninen A, Toivola DM. Keratin 8 modulates β-cell stress responses and normoglycaemia. J Cell Sci 2013; 126:5635-44. [PMID: 24144696 DOI: 10.1242/jcs.132795] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Keratin intermediate filament (IF) proteins are epithelial cell cytoskeletal components that provide structural stability and protection from cell stress, among other cellular and tissue-specific functions. Numerous human diseases are associated with IF gene mutations, but the function of keratins in the endocrine pancreas and their potential significance for glycaemic control are unknown. The impact of keratins on β-cell organisation and systemic glucose control was assessed using keratin 8 (K8) wild-type (K8(+/+)) and K8 knockout (K8(-/-)) mice. Islet β-cell keratins were characterised under basal conditions, in streptozotocin (STZ)-induced diabetes and in non-obese diabetic (NOD) mice. STZ-induced diabetes incidence and islet damage was assessed in K8(+/+) and K8(-/-) mice. K8 and K18 were the predominant keratins in islet β-cells and K8(-/-) mice expressed only remnant K18 and K7. K8 deletion resulted in lower fasting glucose levels, increased glucose tolerance and insulin sensitivity, reduced glucose-stimulated insulin secretion and decreased pancreatic insulin content. GLUT2 localisation and insulin vesicle morphology were disrupted in K8(-/-) β-cells. The increased levels of cytoplasmic GLUT2 correlated with resistance to high-dose STZ-induced injury in K8(-/-) mice. However, K8 deletion conferred no long-term protection from STZ-induced diabetes and prolonged STZ-induced stress caused increased exocrine damage in K8(-/-) mice. β-cell keratin upregulation occurred 2 weeks after treatments with low-dose STZ in K8(+/+) mice and in diabetic NOD mice, suggesting a role for keratins, particularly in non-acute islet stress responses. These results demonstrate previously unrecognised functions for keratins in β-cell intracellular organisation, as well as for systemic blood glucose control under basal conditions and in diabetes-induced stress.
Collapse
Affiliation(s)
- Catharina M Alam
- Department of Biosciences, Cell Biology, Åbo Akademi University, Tykistökatu 6A, FIN-20520 Turku, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee J, Jang KH, Kim H, Lim Y, Kim S, Yoon HN, Chung IK, Roth J, Ku NO. Predisposition to apoptosis in keratin 8-null liver is related to inactivation of NF-κB and SAPKs but not decreased c-Flip. Biol Open 2013; 2:695-702. [PMID: 23862017 PMCID: PMC3711037 DOI: 10.1242/bio.20134606] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/01/2013] [Indexed: 12/28/2022] Open
Abstract
Keratin 8 and 18 (K8/K18) are major intermediate filament proteins of liver hepatocytes. They provide mechanical and nonmechanical stability, thereby protecting cells from stress. Hence, K8-null mice are highly sensitive to Fas-mediated liver cell apoptosis. However, the role of c-Flip protein in K8-null related susceptibility to liver injury is controversial. Here we analyzed c-Flip protein expression in various K8 or K18 null/mutant transgenic livers and show that they are similar in all analyzed transgenic livers and that previously reported c-Flip protein changes are due to antibody cross-reaction with mouse K18. Furthermore, analysis of various apoptosis- or cell survival-related proteins demonstrated that inhibition of phosphorylation of NF-κB and various stress activated protein kinases (SAPKs), such as p38 MAPK, p44/42 MAPK and JNK1/2, is related to the higher sensitivity of K8-null hepatocytes whose nuclear NF-κB is rapidly depleted through Fas-mediated apoptosis. Notably, we found that NF-κB and the studied protein kinases are associated with the K8/K18 complex and are released upon phosphorylation. Therefore, interaction of keratins with cell survival-related protein kinases and transcription factors is another important factor for hepatocyte survival.
Collapse
Affiliation(s)
- Jongeun Lee
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University , Seoul 120-749 , Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Keratin 8 variants are infrequent in patients with alcohol-related liver cirrhosis and do not associate with development of hepatocellular carcinoma. BMC Gastroenterol 2012; 12:147. [PMID: 23078008 PMCID: PMC3527286 DOI: 10.1186/1471-230x-12-147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/08/2012] [Indexed: 12/14/2022] Open
Abstract
Background Keratins 8/18 (K8/K18) are established hepatoprotective proteins and K8/K18 variants predispose to development and adverse outcome of multiple liver disorders. The importance of K8/K18 in alcoholic liver disease as well as in established cirrhosis remains unknown. Methods We analyzed the K8 mutational hot-spots in 261 prospectively followed-up patients with alcoholic cirrhosis (mean follow-up 65 months). PCR-amplified samples were pre-screened by denaturing high-performance liquid chromatography and conspicuous samples were sequenced. Results 67 patients developed hepatocellular carcinoma (HCC) and 133 died. Fourteen patients harbored amino-acid-altering K8 variants (5xG62C, 8xR341H). The presence of K8 variants did not associate with development of HCC (log-rank=0.5) or death (log-rank=0.7) and no significant associations were obtained for the single K8 variants after a correction for multiple testing was performed. Conclusions Keratin variants are expressed in a low percentage of patients with alcoholic cirrhosis and do not influence HCC development. Further studies conducted in larger prospective cohorts are needed to find out whether presence of K8 R341H variant predispose to non-HCC-related liver mortality.
Collapse
|