1
|
Pera EM, Nilsson-De Moura J, Pomeshchik Y, Roybon L, Milas I. Inhibition of the serine protease HtrA1 by SerpinE2 suggests an extracellular proteolytic pathway in the control of neural crest migration. eLife 2024; 12:RP91864. [PMID: 38634469 PMCID: PMC11026092 DOI: 10.7554/elife.91864] [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] [Indexed: 04/19/2024] Open
Abstract
We previously showed that SerpinE2 and the serine protease HtrA1 modulate fibroblast growth factor (FGF) signaling in germ layer specification and head-to-tail development of Xenopus embryos. Here, we present an extracellular proteolytic mechanism involving this serpin-protease system in the developing neural crest (NC). Knockdown of SerpinE2 by injected antisense morpholino oligonucleotides did not affect the specification of NC progenitors but instead inhibited the migration of NC cells, causing defects in dorsal fin, melanocyte, and craniofacial cartilage formation. Similarly, overexpression of the HtrA1 protease impaired NC cell migration and the formation of NC-derived structures. The phenotype of SerpinE2 knockdown was overcome by concomitant downregulation of HtrA1, indicating that SerpinE2 stimulates NC migration by inhibiting endogenous HtrA1 activity. SerpinE2 binds to HtrA1, and the HtrA1 protease triggers degradation of the cell surface proteoglycan Syndecan-4 (Sdc4). Microinjection of Sdc4 mRNA partially rescued NC migration defects induced by both HtrA1 upregulation and SerpinE2 downregulation. These epistatic experiments suggest a proteolytic pathway by a double inhibition mechanism. SerpinE2 ┤HtrA1 protease ┤Syndecan-4 → NC cell migration.
Collapse
Affiliation(s)
- Edgar M Pera
- Vertebrate Developmental Biology Laboratory, Department of Laboratory Medicine, Lund Stem Cell Center, University of LundLundSweden
| | - Josefine Nilsson-De Moura
- Vertebrate Developmental Biology Laboratory, Department of Laboratory Medicine, Lund Stem Cell Center, University of LundLundSweden
| | - Yuriy Pomeshchik
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund Stem Cell Center, Strategic Research Area MultiPark, Lund UniversityLundSweden
| | - Laurent Roybon
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund Stem Cell Center, Strategic Research Area MultiPark, Lund UniversityLundSweden
| | - Ivana Milas
- Vertebrate Developmental Biology Laboratory, Department of Laboratory Medicine, Lund Stem Cell Center, University of LundLundSweden
| |
Collapse
|
2
|
Overview of Human HtrA Family Proteases and Their Distinctive Physiological Roles and Unique Involvement in Diseases, Especially Cancer and Pregnancy Complications. Int J Mol Sci 2021; 22:ijms221910756. [PMID: 34639128 PMCID: PMC8509474 DOI: 10.3390/ijms221910756] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 12/25/2022] Open
Abstract
The mammalian high temperature requirement A (HtrA) proteins are a family of evolutionarily conserved serine proteases, consisting of four homologs (HtrA1-4) that are involved in many cellular processes such as growth, unfolded protein stress response and programmed cell death. In humans, while HtrA1, 2 and 3 are widely expressed in multiple tissues with variable levels, HtrA4 expression is largely restricted to the placenta with the protein released into maternal circulation during pregnancy. This limited expression sets HtrA4 apart from the rest of the family. All four HtrAs are active proteases, and their specific cellular and physiological roles depend on tissue type. The dysregulation of HtrAs has been implicated in many human diseases such as cancer, arthritis, neurogenerative ailments and reproductive disorders. This review first discusses HtrAs broadly and then focuses on the current knowledge of key molecular characteristics of individual human HtrAs, their similarities and differences and their reported physiological functions. HtrAs in other species are also briefly mentioned in the context of understanding the human HtrAs. It then reviews the distinctive involvement of each HtrA in various human diseases, especially cancer and pregnancy complications. It is noteworthy that HtrA4 expression has not yet been reported in any primary tumour samples, suggesting an unlikely involvement of this HtrA in cancer. Collectively, we accentuate that a better understanding of tissue-specific regulation and distinctive physiological and pathological roles of each HtrA will improve our knowledge of many processes that are critical for human health.
Collapse
|
3
|
Scarth JA, Patterson MR, Morgan EL, Macdonald A. The human papillomavirus oncoproteins: a review of the host pathways targeted on the road to transformation. J Gen Virol 2021; 102:001540. [PMID: 33427604 PMCID: PMC8148304 DOI: 10.1099/jgv.0.001540] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
Persistent infection with high-risk human papillomaviruses (HR-HPVs) is the causal factor in over 99 % of cervical cancer cases, and a significant proportion of oropharyngeal and anogenital cancers. The key drivers of HPV-mediated transformation are the oncoproteins E5, E6 and E7. Together, they act to prolong cell-cycle progression, delay differentiation and inhibit apoptosis in the host keratinocyte cell in order to generate an environment permissive for viral replication. The oncoproteins also have key roles in mediating evasion of the host immune response, enabling infection to persist. Moreover, prolonged infection within the cellular environment established by the HR-HPV oncoproteins can lead to the acquisition of host genetic mutations, eventually culminating in transformation to malignancy. In this review, we outline the many ways in which the HR-HPV oncoproteins manipulate the host cellular environment, focusing on how these activities can contribute to carcinogenesis.
Collapse
Affiliation(s)
- James A. Scarth
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Molly R. Patterson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Ethan L. Morgan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Present address: Tumour Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institute of Health, Bethesda, MD 20892, USA
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| |
Collapse
|
4
|
Tavarna T, Phillips PL, Wu XJ, Reyes L. Fetal growth restriction is a host specific response to infection with an impaired spiral artery remodeling-inducing strain of Porphyromonas gingivalis. Sci Rep 2020; 10:14606. [PMID: 32884071 PMCID: PMC7471333 DOI: 10.1038/s41598-020-71762-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/13/2020] [Indexed: 01/03/2023] Open
Abstract
Porphyromonas gingivalis is a periodontal pathogen implicated in a range of pregnancy disorders that involve impaired spiral artery remodeling (ISAR) with or without fetal growth restriction (FGR). Using a rodent periodontitis model, we assessed the ability of P. gingivalis to produce ISAR and FGR in Sprague Dawley (SD) and Wistar (WIS) rats. Both infected SD and WIS rats developed ISAR, but only WIS rats developed FGR despite both rat strains having equivalent microbial loads within the placenta. Neither maternal systemic inflammation nor placental (fetal) inflammation was a feature of FGR in WIS rats. Unique to infected WIS rats, was loss of trophoblast cell density within the junctional zone of the placenta that was not present in SD tissues. In addition, infected WIS rats had a higher proportion of junctional zone trophoblast cells positive for cytoplasmic high temperature requirement A1 (Htra1), a marker of cellular oxidative stress. Our results show a novel phenomenon present in P. gingivalis-induced FGR, with relevance to human disease since dysregulation of placental Htra1 and placental oxidative stress are features of preeclamptic placentas and preeclampsia with FGR.
Collapse
Affiliation(s)
- Tanvi Tavarna
- Department of Pathobiological Sciences, University of Wisconsin - Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, USA
| | - Priscilla L Phillips
- Microbiology and Immunology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Xiao-Jun Wu
- Department of Pathobiological Sciences, University of Wisconsin - Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, USA
| | - Leticia Reyes
- Department of Pathobiological Sciences, University of Wisconsin - Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, USA.
| |
Collapse
|
5
|
Li Y, Yuan J, Rothzerg E, Wu X, Xu H, Zhu S, Xu J. Molecular structure and the role of high-temperature requirement protein 1 in skeletal disorders and cancers. Cell Prolif 2019; 53:e12746. [PMID: 31867863 PMCID: PMC7048211 DOI: 10.1111/cpr.12746] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 01/09/2023] Open
Abstract
Human high‐temperature requirement protein 1 (HTRA1) is a member of serine proteases and consists of four well‐defined domains—an IGFBP domain, a Kazal domain, a protease domain and a PDZ domain. HTRA1 is a secretory protein and also present intracellularly and associated with microtubules. HTRA1 regulates a broad range of physiological processes via its proteolytic activity. This review examines the role of HTRA1 in bone biology, osteoarthritis, intervertebral disc (IVD) degeneration and tumorigenesis. HTRA1 mediates diverse pathological processes via a variety of signalling pathways, such as TGF‐β and NF‐κB. The expression of HTRA1 is increased in arthritis and IVD degeneration, suggesting that HTRA1 protein is attributed to cartilage degeneration and disease progression. Emerging evidence also suggests that HTRA1 has a role in tumorigenesis. Further understanding the mechanisms by which HTRA1 displays as an extrinsic and intrinsic regulator in a cell type–specific manner will be important for the development of HTRA1 as a therapeutic target.
Collapse
Affiliation(s)
- Yihe Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jinbo Yuan
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Emel Rothzerg
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Xinghuo Wu
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
6
|
Wright AA, Todorovic M, Murtaza M, St John JA, Ekberg JA. Macrophage migration inhibitory factor and its binding partner HTRA1 are expressed by olfactory ensheathing cells. Mol Cell Neurosci 2019; 102:103450. [PMID: 31794879 DOI: 10.1016/j.mcn.2019.103450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/10/2019] [Accepted: 11/27/2019] [Indexed: 01/10/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is an important regulator of innate immunity with key roles in neural regeneration and responses to pathogens, amongst a multitude of other functions. The expression of MIF and its binding partners has been characterised throughout the nervous system, with one key exception: the primary olfactory nervous system. Here, we showed in young mice (postnatal day 10) that MIF is expressed in the olfactory nerve by olfactory ensheathing glial cells (OECs) and by olfactory nerve fibroblasts. We also examined the expression of potential binding partners for MIF, and found that the serine protease HTRA1, known to be inhibited by MIF, was also expressed at high levels by OECs and olfactory fibroblasts in vivo and in vitro. We also demonstrated that MIF mediated segregation between OECs and J774a.1 cells (a monocyte/macrophage cell line) in co-culture, which suggests that MIF contributes to the fact that macrophages are largely absent from olfactory nerve fascicles. Phagocytosis assays of axonal debris demonstrated that MIF strongly stimulates phagocytosis by OECs, which indicates that MIF may play a role in the response of OECs to the continual turnover of olfactory axons that occurs throughout life.
Collapse
Affiliation(s)
- A A Wright
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - M Todorovic
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; School of Nursing and Midwifery, Griffith University, Nathan, Queensland, Australia
| | - M Murtaza
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - J A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - J A Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
7
|
Wenta T, Rychlowski M, Jarzab M, Lipinska B. HtrA4 Protease Promotes Chemotherapeutic-Dependent Cancer Cell Death. Cells 2019; 8:cells8101112. [PMID: 31546993 PMCID: PMC6829446 DOI: 10.3390/cells8101112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 12/26/2022] Open
Abstract
The HtrA4 human protease is crucial in placentation and embryo implantation, and its altered level is connected with pre-eclampsia. The meta-analyses of microarray assays revealed that the HtrA4 level is changed in brain tumors and breast and prostate cancers, which suggests its involvement in oncogenesis. In spite of the HtrA4 involvement in important physiological and pathological processes, its function in the cell is poorly understood. In this work, using lung and breast cancer cell lines, we showed for the first time that the full-length HtrA4 and its N-terminally deleted variant promote cancer cell death induced by chemotherapeutic drugs by enhancing apoptosis. The effect is dependent on the HtrA4 proteolytic activity, and the N-terminally deleted HtrA4 is more efficient in the cell death stimulation. Furthermore, HtrA4 increases the effect of chemotherapeutics on the clonogenic potential and motility of cancer cells, and it increases cell cycle arrest at the G2/M phase. HtrA4 may modulate cell death by degrading the anti-apoptotic XIAP protein and also by proteolysis of the executioner pro-caspase 7 and cytoskeletal proteins, actin and β-tubulin. These findings provide new insight into the mechanism of the HtrA4 protease function in cell death and oncogenesis, and they may help to develop new anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | - Michal Rychlowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| |
Collapse
|
8
|
Gagné A, Têtu B, Orain M, Turcotte S, Plante M, Grégoire J, Renaud MC, Bairati I, Trueel D. HtrA1 expression and the prognosis of high-grade serous ovarian carcinoma: a cohort study using digital analysis. Diagn Pathol 2018; 13:57. [PMID: 30131069 PMCID: PMC6104006 DOI: 10.1186/s13000-018-0736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/14/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The expression of high temperature requirement factor A1 (Htra1) has been reported to be decreased in ovarian carcinoma, but its prognostic effect remains undetermined. METHODS We evaluated the impact of HtrA1 downregulation in tumoral tissues on cancer progression and death in women with serous ovarian carcinoma. HtrA1 staining was performed on tissue microarrays (TMA) comprised of tumor samples from a cohort of 106 women who were diagnosed with primary high-grade serous ovarian carcinoma and receiving standard treatment at the Québec University Hospital between 1993 and 2006. HtrA1 expression was assessed visually (percentage of positive nuclei) and by digital image analysis (percentage of positive area). Cox regression multivariate models included standard prognostic factors and were used to estimate adjusted hazard ratios (aHR) for progression or death in the cohort. RESULTS By visual analysis, a low percentage of HtrA1-positive nuclei (< 10% vs ≥10%) tend to be associated with a lower risk of progression (aHR = 0.71; 95% Confidence interval (CI) = 0.46-1.09; P = 0.11) and mortality (aHR = 0.65; 95% CI = 0.41-1.04; P = 0.07). Low nuclear HtrA1 expression assessed by digital image analysis (< median % vs ≥ median %) showed a significant association with lower risk of progression (aHR = 0.62; 95% CI = 0.40-0.95; p = 0.03) and death (aHR = 0.60; 95% CI = 0.38-0.95; p = 0.03). CONCLUSION Altogether, our results demonstrate that nuclear downregulation of HtrA1 is associated with a better prognosis in women with high grade serous ovarian carcinoma.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/analysis
- Cell Nucleus/chemistry
- Cell Nucleus/pathology
- Cohort Studies
- Down-Regulation
- Female
- High-Temperature Requirement A Serine Peptidase 1/analysis
- Humans
- Image Interpretation, Computer-Assisted
- Immunohistochemistry
- Middle Aged
- Neoplasm Grading
- Neoplasms, Cystic, Mucinous, and Serous/chemistry
- Neoplasms, Cystic, Mucinous, and Serous/mortality
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Ovarian Neoplasms/chemistry
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- Predictive Value of Tests
- Risk Assessment
- Risk Factors
- Time Factors
- Tissue Array Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- Andréanne Gagné
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Bernard Têtu
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Anatomic Pathology and Cytology Department, Hôpital du St-Sacrement, Centre Hospitalier Universitaire (CHU) de Québec, Laval University, 1050 Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
- Department of Pathology, Hôpital du St-Sacrement, Centre Hospitalier Universitaire de Québec, 1050, Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
| | - Michèle Orain
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Anatomic Pathology and Cytology Department, Hôpital du St-Sacrement, Centre Hospitalier Universitaire (CHU) de Québec, Laval University, 1050 Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
| | - Stéphane Turcotte
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Marie Plante
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Jean Grégoire
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Marie-Claude Renaud
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Isabelle Bairati
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Dominique Trueel
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Department of Pathology, Hôpital Saint-Luc, Centre Hospitalier Universitaire de Montréal, 058, rue Saint-Denis, Montréal, Québec H2X 3J4 Canada
- The Research Centre of the University of Montreal Teaching Hospital (CR-CHUM)/Montreal Cancer Institute, 900 Rue St-Denis, Montreal, Quebec H2X 0A9 Canada
- Department of Pathology and Cellular Biology, University of Montreal, 2900, boulevard Édouard-Montpetit, Montreal, Quebec H3T 1J4 Canada
| |
Collapse
|
9
|
Xiao S, Tang YS, Kusumanchi P, Stabler SP, Zhang Y, Antony AC. Folate Deficiency Facilitates Genomic Integration of Human Papillomavirus Type 16 DNA In Vivo in a Novel Mouse Model for Rapid Oncogenic Transformation of Human Keratinocytes. J Nutr 2018; 148:389-400. [PMID: 29546304 DOI: 10.1093/jn/nxx060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Background Epidemiologic and in vitro studies suggest independent linkages between poor folate and/or vitamin B-12 nutrition, genomic human papillomavirus (HPV) type 16 viral integration, and cancer. However, there is no direct evidence in vivo to support the causative role of poor folate nutrition in HPV16 integration into the cellular genome. Objective We tested the hypothesis that folate deficiency enables the integration of HPV16 into the genome of HPV16-harboring keratinocytes, and could thereby influence earlier transformation of these cells to cancer in an animal model. Methods HPV16-harboring human keratinocytes [(HPV16)BC-1-Ep/SL] were differentiated into 3-dimensional HPV16-organotypic rafts under either folate-replete or folate-deficient conditions in vitro. These were then subcutaneously implanted in severely immunocompromised female Beige Nude XID (Hsd: NIHS-LystbgFoxn1nuBtkxid) mice (4-6 wk old, 16-18 g) fed either a folate-replete diet (1200 nmol folate/kg diet) or a progressively folate-deficient diet (600 or 400 nmol folate/kg diet) for 2 mo prior to raft-implantation surgery, and indefinitely thereafter. The tumors that subsequently developed were characterized for onset, pattern of growth, morphology, HPV16 oncogene expression, and HPV16-genomic integration. Results All HPV16-organotypic rafts developed in either folate-replete or physiologic low-folate media in vitro and subsequently implanted in folate-replete mice eventually transformed into aggressive malignancies within weeks. When compared to HPV16-high folate-organotypic raft-derived tumors from mice fed either a 1200 or 600 nmol folate/kg diet, those raft-derived cancers that developed in mice fed a 400 nmol folate/kg diet expressed significantly more HPV16 E6 (1.8-fold more) and E7 (2.8-fold more) oncogenic proteins (P = 0.001), and revealed significantly more HPV16-integration sites in genomic DNA (2-fold more), either directly into, or in the vicinity of, cellular genes (P < 0.05). Conclusions This unprecedented animal model for the consistent rapid transformation of differentiated (HPV16)BC-1-Ep/SL-derived organotypic raft-keratinocytes to cancer in Beige Nude XID mice confirms that dietary folate deficiency can profoundly influence and modulate events leading to HPV16-induced carcinogenesis, and facilitates genomic integration of HPV16 DNA in vivo.
Collapse
Affiliation(s)
- Suhong Xiao
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Ying-Sheng Tang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | | | - Sally P Stabler
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ying Zhang
- Department of Biostatistics, Indiana University Fairbanks School of Public Health, Indianapolis, IN
| | - Asok C Antony
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN.,Richard L Roudebush Veterans Affairs Medical Center, Indianapolis, IN
| |
Collapse
|
10
|
Fex Svenningsen Å, Löring S, Sørensen AL, Huynh HUB, Hjæresen S, Martin N, Moeller JB, Elkjær ML, Holmskov U, Illes Z, Andersson M, Nielsen SB, Benedikz E. Macrophage migration inhibitory factor (MIF) modulates trophic signaling through interaction with serine protease HTRA1. Cell Mol Life Sci 2017; 74:4561-4572. [PMID: 28726057 PMCID: PMC5663815 DOI: 10.1007/s00018-017-2592-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF), a small conserved protein, is abundant in the immune- and central nervous system (CNS). MIF has several receptors and binding partners that can modulate its action on a cellular level. It is upregulated in neurodegenerative diseases and cancer although its function is far from clear. Here, we report the finding of a new binding partner to MIF, the serine protease HTRA1. This enzyme cleaves several growth factors, extracellular matrix molecules and is implicated in some of the same diseases as MIF. We show that the function of the binding between MIF and HTRA1 is to inhibit the proteolytic activity of HTRA1, modulating the availability of molecules that can change cell growth and differentiation. MIF is therefore the first endogenous inhibitor ever found for HTRA1. It was found that both molecules were present in astrocytes and that the functional binding has the ability to modulate astrocytic activities important in development and disease of the CNS.
Collapse
Affiliation(s)
- Åsa Fex Svenningsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark.
| | - Svenja Löring
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands
| | - Anna Lahn Sørensen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Ha Uyen Buu Huynh
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Simone Hjæresen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Nellie Martin
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Jesper Bonnet Moeller
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Weill Cornell Medicine, Cornell University, 413 East 69th Street, New York, 10021, USA
| | - Maria Louise Elkjær
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Malin Andersson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 59, 751 24, Uppsala, Sweden
| | - Solveig Beck Nielsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Eirikur Benedikz
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Faculty of Health, University College Zealand, Parkvej 190, 4700, Næstved, Denmark
| |
Collapse
|
11
|
Zurawa-Janicka D, Wenta T, Jarzab M, Skorko-Glonek J, Glaza P, Gieldon A, Ciarkowski J, Lipinska B. Structural insights into the activation mechanisms of human HtrA serine proteases. Arch Biochem Biophys 2017; 621:6-23. [PMID: 28396256 DOI: 10.1016/j.abb.2017.04.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
Abstract
Human HtrA1-4 proteins belong to the HtrA family of evolutionarily conserved serine proteases and function as important modulators of many physiological processes, including maintenance of mitochondrial homeostasis, cell signaling and apoptosis. Disturbances in their action are linked to severe diseases, including oncogenesis and neurodegeneration. The HtrA1-4 proteins share structural and functional features of other members of the HtrA protein family, however there are several significant differences in structural architecture and mechanisms of action which makes each of them unique. Our goal is to present recent studies regarding human HtrAs. We focus on their physiological functions, structure and regulation, and describe current models of activation mechanisms. Knowledge of molecular basis of the human HtrAs' action is a subject of great interest; it is crucial for understanding their relevance in cellular physiology and pathogenesis as well as for using them as targets in future therapies of diseases such as neurodegenerative disorders and cancer.
Collapse
Affiliation(s)
- Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | - Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Joanna Skorko-Glonek
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Przemyslaw Glaza
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Artur Gieldon
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Jerzy Ciarkowski
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| |
Collapse
|
12
|
Ng TK, Liang XY, Lu F, Liu DT, Yam GH, Ma L, Tam PO, Chen H, Cen LP, Chen LJ, Yang Z, Pang CP. Protective effects of an HTRA1 insertion-deletion variant against age-related macular degeneration in the Chinese populations. J Transl Med 2017; 97:43-52. [PMID: 27841854 DOI: 10.1038/labinvest.2016.117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 11/09/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment and irreversible blindness in most developed countries, affecting about 50 million elderly people worldwide. Retinal pigment epithelial (RPE) cell degeneration is the pathophysiological cause of AMD, leading to geographic atrophy and choroidal neovascularization. We and others have previously identified several polymorphisms on chromosome 10q26 (HTRA1 rs11200638 as well as LOC387715 rs10490924 and c.372_815del443ins54) associated with AMD. In this study, we confirmed the association of our previously identified HTRA1 insertion-deletion (indel) variant (c.34delCinsTCCT) in 195 exudative AMD patients and 390 controls from the Hong Kong Chinese cohort with additional 168 patients and 210 controls from the Chengdu Chinese cohort and followed by studying its biological functions in RPE cells. Genetic analysis verified the higher prevalence of c.34delCinsTCCT allele in control subjects (8.0%) than in AMD patients (1.9%; P=7.87 × 10-5, odds ratio=0.229). This protective effect was validated as the haplotype of the c.34delCinsTCCT allele existed independent of the risk haplotype (P=1.17 × 10-5). In vitro studies showed that recombinant HTRA1 c.34delCinsTCCT variant protein was more localized in the endoplasmic reticulum of RPE cells compared with the wild-type protein, and its secretion was delayed. Moreover, ARPE-19 cells expressing HTRA1 c.34delCinsTCCT variant had higher cell viability, lower cell apoptosis and were less responsive to anoikis, supporting its protective role. We revealed a protective AMD-associated HTRA1 variant in Chinese populations and the biological role of HTRA1 in RPE cell degeneration, indicating its involvement in AMD pathogenesis.
Collapse
Affiliation(s)
- Tsz Kin Ng
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Xiao Ying Liang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Fang Lu
- The Sichuan Key Laboratory for Human Disease Gene Study, Chengdu, China
- The Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - David Tl Liu
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Gary Hf Yam
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Li Ma
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Pancy Os Tam
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Haoyu Chen
- The Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Ling Ping Cen
- The Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Li Jia Chen
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Zhenglin Yang
- The Sichuan Key Laboratory for Human Disease Gene Study, Chengdu, China
- The Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Chi Pui Pang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
13
|
Yamamoto Y, Ihara M. Disruption of transforming growth factor-β superfamily signaling: A shared mechanism underlying hereditary cerebral small vessel disease. Neurochem Int 2016; 107:211-218. [PMID: 28034724 DOI: 10.1016/j.neuint.2016.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022]
Abstract
Cerebral small vessel disease (SVD) is not only one of the leading causes of cognitive impairment but also an important contributory factor in Alzheimer's disease. SVD and related white matter changes are common in the elderly, but the underlying pathogenic mechanism remains unclear. The end-stage pathology of SVD often involves replacement of vascular smooth muscle cells with collagenous or other nontensile fibrillary material. Recent studies on hereditary SVD have revealed a close relationship between small vessel pathology and disruption of transforming growth factor-β (TGF-β) superfamily signaling. TGF-β superfamily members, such as TGF-β and bone morphogenetic proteins, are multifunctional proteins that regulate production of extracellular matrix proteins, which in turn control the bioavailability of TGF-β superfamily members and modulate their signaling activities. This article reviews hereditary disorders with small vessel pathology and their relation to TGF-β superfamily signaling.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan.
| |
Collapse
|
14
|
Stuqui B, Conceição ALG, Termini L, Sichero L, Villa LL, Rahal P, Calmon MDF. The differential role of HTRA1 in HPV-positive and HPV-negative cervical cell line proliferation. BMC Cancer 2016; 16:840. [PMID: 27809811 PMCID: PMC5095955 DOI: 10.1186/s12885-016-2873-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND High-risk human papillomaviruses (HPVs) are strongly associated with the development of some malignancies. The E6 and E7 viral oncoproteins are the primary proteins responsible for cell homeostasis alteration and immortalization. Furthermore, the E6 protein from high-risk HPVs can interact with the PDZ (PSD-90/Dlg/ZO-1) domains of cellular proteins, triggering cell transformation. One protein that is associated with pathological conditions and has a PDZ domain is the protease HTRA1 (high temperature requirement 1). This protein is poorly expressed in some cancers, suggesting a tumor suppressor role. The aim of this study was to evaluate the effect of HTRA1 overexpression in HPV16-positive (CasKi) and HPV-negative (C33) cervical cell lines. METHODS The cells were transfected with a vector containing the HTRA1 ORF or an empty vector. HTRA1 overexpression was confirmed by qRT-PCR. The cells were subjected to cell proliferation, colony formation, apoptosis and cell cycle assays. RESULTS C33 cells expressing HTRA1 grew significantly fewer colonies and showed less proliferation than cells without HTRA1 expression. In contrast, in the CasKi cells overexpressing HTRA1, there was an increase in the cell growth rate and in the colonies density compared to cells expressing low levels of HTRA1. An apoptosis assay showed that HTRA1 does not interfere with the apoptosis rate in these cells. A cell cycle immunofluorescence assay revealed more CasKi cells overexpressing HTRA1 in the S phase and more C33 HTRA1-transfected cells in the G0/G1 phase, suggesting that HTRA1 plays different roles in the cell cycle progression of these cells. CONCLUSIONS HTRA1 overexpression prevents cell proliferation in the HPV-negative cell line and increases cell proliferation in the HPV-positive cell line. Although the E6/HTRA1 interaction has already been described in the literature, more studies are required to confirm whether the present functional findings are a result of this interaction.
Collapse
Affiliation(s)
- Bruna Stuqui
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas - IBILCE/UNESP, Rua Cristóvão Colombo n° 2265, Jardim Nazareth, CEP 15054-000 São José do Rio Preto, SP Brazil
| | - André Luis Giacometti Conceição
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas - IBILCE/UNESP, Rua Cristóvão Colombo n° 2265, Jardim Nazareth, CEP 15054-000 São José do Rio Preto, SP Brazil
| | - Lara Termini
- Center for Translational Investigation in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 251, 8° andar, Bairro Cerqueira César, CEP 01246-000 São Paulo Brazil
| | - Laura Sichero
- Center for Translational Investigation in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 251, 8° andar, Bairro Cerqueira César, CEP 01246-000 São Paulo Brazil
| | - Luisa Lina Villa
- Center for Translational Investigation in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 251, 8° andar, Bairro Cerqueira César, CEP 01246-000 São Paulo Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 251, 8° andar, Bairro Cerqueira César, CEP 01246-000 São Paulo Brazil
| | - Paula Rahal
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas - IBILCE/UNESP, Rua Cristóvão Colombo n° 2265, Jardim Nazareth, CEP 15054-000 São José do Rio Preto, SP Brazil
| | - Marília de Freitas Calmon
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas - IBILCE/UNESP, Rua Cristóvão Colombo n° 2265, Jardim Nazareth, CEP 15054-000 São José do Rio Preto, SP Brazil
| |
Collapse
|
15
|
Ramasamy VS, Islam MI, Haque MA, Shin SY, Park IS. β-Amyloid induces nuclear protease-mediated lamin fragmentation independent of caspase activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1189-99. [PMID: 26876308 DOI: 10.1016/j.bbamcr.2016.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/18/2016] [Accepted: 02/09/2016] [Indexed: 01/22/2023]
Abstract
β-Amyloid (Aβ), a hallmark peptide of Alzheimer's disease, induces both caspase-dependent apoptosis and non-apoptotic cell death. In this study, we examined caspase-independent non-apoptotic cell death preceding caspase activation in Aβ42-treated cells. We first determined the optimal treatment conditions for inducing cell death without caspase activation and selected a double-treatment method involving the incubation of cells with Aβ42 for 4 and 6 h (4+6 h sample). We observed that levels of lamin A (LA) and lamin B (LB) were reduced in the 4+6 h samples. This reduction was decreased by treatment with suc-AAPF-CMK, an inhibitor of nuclear scaffold (NS) protease, but not by treatment with z-VAD-FMK, a pan-caspase inhibitor. In addition, suc-AAPF-CMK decreased the changes in nuclear morphology observed in cells in the 4+6 h samples, which were different from nuclear fragmentation observed in STS-treated cells. Furthermore, suc-AAPF-CMK inhibited cell death in the 4+6 h samples. LA and LB fragmentation occurred in the isolated nuclei and was also inhibited by suc-AAPF-CMK. Together, these data indicated that the fragmentation of LA and LB in the Aβ42-treated cells was induced by an NS protease, whose identity is not clearly determined yet. A correlation between Aβ42 toxicity and the lamin fragmentation by NS protease suggests that inhibition of the protease could be an effective method for controlling the pathological process of AD.
Collapse
Affiliation(s)
- Vijay Sankar Ramasamy
- Department of Bio-Materials Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Md Imamul Islam
- Department of Bio-Materials Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Md Aminul Haque
- Department of Bio-Materials Engineering, Chosun University, Gwangju 501-759, Republic of Korea
| | - Song Yub Shin
- Department of Bio-Materials Engineering, Chosun University, Gwangju 501-759, Republic of Korea; Cellular and Molecular Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Il-Seon Park
- Department of Bio-Materials Engineering, Chosun University, Gwangju 501-759, Republic of Korea; Cellular and Molecular Medicine, Chosun University, Gwangju 501-759, Republic of Korea.
| |
Collapse
|
16
|
Poepsel S, Sprengel A, Sacca B, Kaschani F, Kaiser M, Gatsogiannis C, Raunser S, Clausen T, Ehrmann M. Determinants of amyloid fibril degradation by the PDZ protease HTRA1. Nat Chem Biol 2015; 11:862-9. [PMID: 26436840 DOI: 10.1038/nchembio.1931] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/09/2015] [Indexed: 01/28/2023]
Abstract
Excessive aggregation of proteins has a major impact on cell fate and is a hallmark of amyloid diseases in humans. To resolve insoluble deposits and to maintain protein homeostasis, all cells use dedicated protein disaggregation, protein folding and protein degradation factors. Despite intense recent research, the underlying mechanisms controlling this key metabolic event are not well understood. Here, we analyzed how a single factor, the highly conserved serine protease HTRA1, degrades amyloid fibrils in an ATP-independent manner. This PDZ protease solubilizes protein fibrils and disintegrates the fibrillar core structure, allowing productive interaction of aggregated polypeptides with the active site for rapid degradation. The aggregate burden in a cellular model of cytoplasmic tau aggregation is thus reduced. Mechanistic aspects of ATP-independent proteolysis and its implications in amyloid diseases are discussed.
Collapse
Affiliation(s)
- Simon Poepsel
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Andreas Sprengel
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Barbara Sacca
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Farnusch Kaschani
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Markus Kaiser
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Christos Gatsogiannis
- Department of Structural Biochemistry, Max Planck Institute Molecular Physiology, Dortmund, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute Molecular Physiology, Dortmund, Germany
| | - Tim Clausen
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Michael Ehrmann
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
17
|
ALTOBELLI EMMA, MARZIONI DANIELA, LATTANZI AMEDEO, ANGELETTI PAOLOMATTEO. HtrA1: Its future potential as a novel biomarker for cancer. Oncol Rep 2015; 34:555-66. [PMID: 26035313 PMCID: PMC4487665 DOI: 10.3892/or.2015.4016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/16/2015] [Indexed: 12/23/2022] Open
Abstract
HtrA1 appears to be involved in several physiological processes as well as in the pathogenesis of conditions such as Alzheimer's disease and osteoarthritis. It has also been hypothesized to play a role as a tumor suppressor. This manuscript reviews the current cancer-related HtrA1 research from the methodological and clinical standpoints including studies regarding its potential role as a tumor marker and/or prognostic factor. PRISMA method was used for study selection. The articles thus collected were examined and selected by two independent reviewers; any disagreement was resolved by a methodologist. A laboratory researcher reviewed the methods and laboratory techniques. Fifteen studies met the inclusion criteria and concerned the following cancer sites: the nervous system, bladder, breast, esophagus, stomach, liver, endometrium, thyroid, ovaries, pleura, lung and skin. Most articles described in vivo studies using a morphological approach and immunohistochemistry, whereas protein expression was quantified as staining intensity scored by two raters. Often the results were not comparable due to the different rating scales and study design. Current research on HtrA1 does not conclusively support its role as a tumor suppressor.
Collapse
Affiliation(s)
- EMMA ALTOBELLI
- Department of Life, Health and Environmental Sciences, Epidemiology and Biostatistics Unit, AUSL Teramo, University of L’Aquila, L’Aquila, Italy
| | - DANIELA MARZIONI
- Department of Experimental and Clinical Medicine, University of Ancona, Ancona, Italy
| | - AMEDEO LATTANZI
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - PAOLO MATTEO ANGELETTI
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
18
|
Risør MW, Poulsen ET, Thomsen LR, Dyrlund T, Nielsen TA, Nielsen NC, Sanggaard KW, Enghild JJ. The autolysis of human HtrA1 is governed by the redox state of its N-terminal domain. Biochemistry 2014; 53:3851-7. [PMID: 24846539 PMCID: PMC4067144 DOI: 10.1021/bi401633w] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 05/20/2014] [Indexed: 01/07/2023]
Abstract
Human HtrA1 (high-temperature requirement protein A1) belongs to a conserved family of serine proteases involved in protein quality control and cell fate. The homotrimeric ubiquitously expressed protease has chymotrypsin-like specificity and primarily targets hydrophobic stretches in selected or misfolded substrate proteins. In addition, the enzyme is capable of exerting autolytic activity by removing the N-terminal insulin-like growth factor binding protein (IGFBP)/Kazal-like tandem motif without affecting the protease activity. In this study, we have addressed the mechanism governing the autolytic activity and find that it depends on the integrity of the disulfide bonds in the N-terminal IGFBP/Kazal-like domain. The specificity of the autolytic cleavage reveals a strong preference for cysteine in the P1 position of HtrA1, explaining the lack of autolysis prior to disulfide reduction. Significantly, the disulfides were reduced by thioredoxin, suggesting that autolysis of HtrA1 in vivo is linked to the endogenous redox balance and that the N-terminal domain acts as a redox-sensing switch.
Collapse
Affiliation(s)
- Michael W. Risør
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
- Centre
for Insoluble Protein Structures (inSPIN), Aarhus University, DK-8000 Aarhus, Denmark
| | - Ebbe Toftgaard Poulsen
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| | - Line R. Thomsen
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| | - Thomas
F. Dyrlund
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| | - Tania A. Nielsen
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| | - Niels Chr. Nielsen
- Centre
for Insoluble Protein Structures (inSPIN), Aarhus University, DK-8000 Aarhus, Denmark
- Department
of Chemistry, Aarhus University, DK-8000 Aarhus, Denmark
| | - Kristian W. Sanggaard
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| | - Jan J. Enghild
- Department
of Molecular Biology and Genetics, Aarhus
University, DK-8000 Aarhus, Denmark
| |
Collapse
|
19
|
Graham JR, Chamberland A, Lin Q, Li XJ, Dai D, Zeng W, Ryan MS, Rivera-Bermúdez MA, Flannery CR, Yang Z. Serine protease HTRA1 antagonizes transforming growth factor-β signaling by cleaving its receptors and loss of HTRA1 in vivo enhances bone formation. PLoS One 2013; 8:e74094. [PMID: 24040176 PMCID: PMC3770692 DOI: 10.1371/journal.pone.0074094] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/26/2013] [Indexed: 11/19/2022] Open
Abstract
HTRA1 is a member of the High Temperature Requirement (HTRA1) family of serine proteases, which play a role in several biological and pathological processes. In part, HTRA1 regulation occurs by inhibiting the TGF-β signaling pathway, however the mechanism of inhibition has not been fully defined. Previous studies have shown that HTRA1 is expressed in a variety of tissues, including sites of skeletal development. HTRA1 has also been implicated in the process of bone formation, although the precise manner of regulation is still unknown. This study investigated how HTRA1 regulates TGF-β signaling and examined the in vivo effects of the loss of HTRA1. We demonstrated that recombinant HTRA1 was capable of cleaving both type II and type III TGF-β receptors (TβRII and TβRIII) in vitro in a dose-dependent manner, but it did not affect the integrity of TβRI or TGF-β. Overexpression of HTRA1 led to decreased levels of both TβRII and III on the cell surface but had no effect on TβRI. Silencing HTRA1 expression significantly increased TGF-β binding to the cell surface and TGF-β responsiveness within the cell. To examine the role of HTRA1 in vivo, we generated mice with a targeted gene deletion of HTRA1. Embryonic fibroblasts isolated from these mice displayed an increase in TGF-β-induced expression of several genes known to promote bone formation. Importantly, the loss of HTRA1 in the knockout mice resulted in a marked increase in trabecular bone mass. This study has identified a novel regulatory mechanism by which HTRA1 antagonizes TGF-β signaling, and has shown that HTRA1 plays a key role in the regulation of bone formation.
Collapse
Affiliation(s)
- Julie R. Graham
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Angela Chamberland
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Qingcong Lin
- Global BioTherapeutic Technologies, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - X. Jian Li
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - David Dai
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Weilan Zeng
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Mark S. Ryan
- Immunoscience Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Moisés A. Rivera-Bermúdez
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Carl R. Flannery
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Zhiyong Yang
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
20
|
Abstract
E7 is an accessory protein that is not encoded by all papillomaviruses. The E7 amino terminus contains two regions of similarity to conserved regions 1 and 2 of the adenovirus E1A protein, which are also conserved in the simian vacuolating virus 40 large tumor antigen. The E7 carboxyl terminus consists of a zinc-binding motif, which is related to similar motifs in E6 proteins. E7 proteins play a central role in the human papillomavirus life cycle, reprogramming the cellular environment to be conducive to viral replication. E7 proteins encoded by the cancer-associated alpha human papillomaviruses have potent transforming activities, which together with E6, are necessary but not sufficient to render their host squamous epithelial cell tumorigenic. This article strives to provide a comprehensive summary of the published research studies on human papillomavirus E7 proteins.
Collapse
|
21
|
Wang N, Eckert KA, Zomorrodi AR, Xin P, Pan W, Shearer DA, Weisz J, Maranus CD, Clawson GA. Down-regulation of HtrA1 activates the epithelial-mesenchymal transition and ATM DNA damage response pathways. PLoS One 2012; 7:e39446. [PMID: 22761798 PMCID: PMC3383700 DOI: 10.1371/journal.pone.0039446] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 05/21/2012] [Indexed: 12/18/2022] Open
Abstract
Expression of the serine protease HtrA1 is decreased or abrogated in a variety of human primary cancers, and higher levels of HtrA1 expression are directly related to better response to chemotherapeutics. However, the precise mechanisms leading to HtrA1 down regulation during malignant transformation are unclear. To investigate HtrA1 gene regulation in breast cancer, we characterized expression in primary breast tissues and seven human breast epithelial cell lines, including two non-tumorigenic cell lines. In human breast tissues, HtrA1 expression was prominent in normal ductal glands. In DCIS and in invasive cancers, HtrA1 expression was greatly reduced or lost entirely. HtrA1 staining was also reduced in all of the human breast cancer cell lines, compared with the normal tissue and non-tumorigenic cell line controls. Loss of HtrA1 gene expression was attributable primarily to epigenetic silencing mechanisms, with different mechanisms operative in the various cell lines. To mechanistically examine the functional consequences of HtrA1 loss, we stably reduced and/or overexpressed HtrA1 in the non-tumorigenic MCF10A cell line. Reduction of HtrA1 levels resulted in the epithelial-to-mesenchymal transition with acquisition of mesenchymal phenotypic characteristics, including increased growth rate, migration, and invasion, as well as expression of mesenchymal biomarkers. A concomitant decrease in expression of epithelial biomarkers and all microRNA 200 family members was also observed. Moreover, reduction of HtrA1 expression resulted in activation of the ATM and DNA damage response, whereas overexpression of HtrA1 prevented this activation. Collectively, these results suggest that HtrA1 may function as a tumor suppressor by controlling the epithelial-to-mesenchymal transition, and may function in chemotherapeutic responsiveness by mediating DNA damage response pathways.
Collapse
Affiliation(s)
- Ning Wang
- Gittlen Cancer Research Institute & Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Kristin A. Eckert
- Gittlen Cancer Research Institute & Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Ali R. Zomorrodi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Ping Xin
- Gittlen Cancer Research Institute & Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Weihua Pan
- Gittlen Cancer Research Institute & Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Debra A. Shearer
- Department of Obstetrics & Gynecology, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Judith Weisz
- Department of Obstetrics & Gynecology, Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Costas D. Maranus
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Gary A. Clawson
- Gittlen Cancer Research Institute & Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States of America
- Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
22
|
Tennstaedt A, Pöpsel S, Truebestein L, Hauske P, Brockmann A, Schmidt N, Irle I, Sacca B, Niemeyer CM, Brandt R, Ksiezak-Reding H, Tirniceriu AL, Egensperger R, Baldi A, Dehmelt L, Kaiser M, Huber R, Clausen T, Ehrmann M. Human high temperature requirement serine protease A1 (HTRA1) degrades tau protein aggregates. J Biol Chem 2012; 287:20931-41. [PMID: 22535953 PMCID: PMC3375517 DOI: 10.1074/jbc.m111.316232] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 04/24/2012] [Indexed: 11/06/2022] Open
Abstract
Protective proteases are key elements of protein quality control pathways that are up-regulated, for example, under various protein folding stresses. These proteases are employed to prevent the accumulation and aggregation of misfolded proteins that can impose severe damage to cells. The high temperature requirement A (HtrA) family of serine proteases has evolved to perform important aspects of ATP-independent protein quality control. So far, however, no HtrA protease is known that degrades protein aggregates. We show here that human HTRA1 degrades aggregated and fibrillar tau, a protein that is critically involved in various neurological disorders. Neuronal cells and patient brains accumulate less tau, neurofibrillary tangles, and neuritic plaques, respectively, when HTRA1 is expressed at elevated levels. Furthermore, HTRA1 mRNA and HTRA1 activity are up-regulated in response to elevated tau concentrations. These data suggest that HTRA1 is performing regulated proteolysis during protein quality control, the implications of which are discussed.
Collapse
Affiliation(s)
- Annette Tennstaedt
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Simon Pöpsel
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Linda Truebestein
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Patrick Hauske
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Anke Brockmann
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Nina Schmidt
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Inga Irle
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Barbara Sacca
- the Fakultät Chemie, Biologisch-Chemische Mikrostrukturtechnik, Technische Universität Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Christof M. Niemeyer
- the Fakultät Chemie, Biologisch-Chemische Mikrostrukturtechnik, Technische Universität Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Roland Brandt
- the Fachbereich Biologie/Chemie, University Osnabrueck, D-49076 Osnabrueck, Germany
| | - Hanna Ksiezak-Reding
- the Department of Neurology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, New York 10029
| | - Anca Laura Tirniceriu
- the Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich, Feodor-Lynen-Strasse 23, 81377 Munich, Germany
| | - Rupert Egensperger
- the Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich, Feodor-Lynen-Strasse 23, 81377 Munich, Germany
| | - Alfonso Baldi
- the Department of Biochemistry and Biophysics, Section of Pathology, the Second University of Naples, 80100 Naples, Italy
| | - Leif Dehmelt
- the Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Markus Kaiser
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
| | - Robert Huber
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
- the Department for Chemical Biology, Technische Universität Dortmund University, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
- the Max-Planck-Institut für Biochemie, Am Klopferspitz 18A, 82152 Martinsried, Germany
- the School of Biosciences, Cardiff University, Cardiff CF10 3US, United Kingdom, and
| | - Tim Clausen
- the Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | - Michael Ehrmann
- From the Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitaetsstrasse, 45141 Essen, Germany
- the School of Biosciences, Cardiff University, Cardiff CF10 3US, United Kingdom, and
| |
Collapse
|
23
|
Ng TK, Liang XY, Pang CP. HTRA1 in Age-Related Macular Degeneration. ASIA-PACIFIC JOURNAL OF OPHTHALMOLOGY (PHILADELPHIA, PA.) 2012; 1:51-63. [PMID: 26107018 DOI: 10.1097/apo.0b013e31823e57fe] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of severe visual impairment and irreversible blindness in most developed countries, affecting more than 50 million of elderly people worldwide. Current treatments, such as intravitreal injection of antiangiogenic agents, mitigate the effect of advanced AMD but cannot completely cure the disease. Comprehensive understanding of the AMD pathological mechanisms is important for the development of new therapies. Previously, we identified a single-nucleotide polymorphism (rs11200638) in the promoter region of the high temperature requirement factor A1 (HTRA1) gene on chromosome 10q26 to be associated with exudative AMD. In further biological studies, we have provided evidence that HTRA1 could be a potential disease-causing gene within the 10q26 locus. In this review, we summarize the pathology of AMD and the molecular function of the HtrA1 protein. Also evaluated are the genetic effects of HTRA1 polymorphism on AMD in different populations and interactions with other AMD-associated genes, especially with the complement factor H (CFH) gene, which was identified for nonexudative AMD. The biological roles of HtrA1 are exhaustively examined on its contribution to the multifactorial pathogenic mechanism of AMD. Although HtrA1 should play a part in AMD pathogenesis, a host of other genetic and environmental factors, known and unknown, is involved and warrants intensive future research.
Collapse
Affiliation(s)
- Tsz Kin Ng
- From the Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, China
| | | | | |
Collapse
|
24
|
Dorai H, Santiago A, Campbell M, Tang QM, Lewis MJ, Wang Y, Lu QZ, Wu SL, Hancock W. Characterization of the proteases involved in the N-terminal clipping of glucagon-like-peptide-1-antibody fusion proteins. Biotechnol Prog 2011; 27:220-31. [DOI: 10.1002/btpr.537] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Indexed: 11/06/2022]
|
25
|
Campioni M, Severino A, Manente L, Tuduce IL, Toldo S, Caraglia M, Crispi S, Ehrmann M, He X, Maguire J, De Falco M, De Luca A, Shridhar V, Baldi A. The serine protease HtrA1 specifically interacts and degrades the tuberous sclerosis complex 2 protein. Mol Cancer Res 2010; 8:1248-60. [PMID: 20671064 DOI: 10.1158/1541-7786.mcr-09-0473] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hamartin and tuberin are products of the tumor suppressor genes TSC1 and TSC2, respectively. Mutations affecting either gene result in the tuberous sclerosis syndrome, a neurologic genetic disorder characterized by the formation of multiple benign tumors or hamartomas. In this study, we report the identification of TSC2, but not TSC1, as a substrate of HtrA1, a member of the human HtrA family proteins of serine proteases. We show the direct interaction and colocalization in the cytoplasm of HtrA1 and TSC2 and that HtrA1 cleaves TSC2 both in vitro and in vivo. Finally, we show that alterations in HtrA1 expression cause modifications in phosphorylation status of two downstream targets of TSC2: 4E-BP1 and S6K. Our data suggest that, under particular physiologic or pathologic conditions, HtrA1 degrades TSC2 and activates the downstream targets. Considering that HtrA1 levels are significantly increased during embryogenesis, we speculate that one of the targets of HtrA1 activity during fetal development is the TSC2-TSC1 pathway.
Collapse
Affiliation(s)
- Mara Campioni
- Department of Biochemistry, Section of Pathology, Second University of Naples, Via L. Armanni 5, 80138 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
He X, Ota T, Liu P, Su C, Chien J, Shridhar V. Downregulation of HtrA1 promotes resistance to anoikis and peritoneal dissemination of ovarian cancer cells. Cancer Res 2010; 70:3109-18. [PMID: 20388781 PMCID: PMC5579484 DOI: 10.1158/0008-5472.can-09-3557] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We previously identified serine protease HtrA1 as a downregulated gene in epithelial ovarian cancer (EOC), but the functional consequence of loss of HtrA1 in EOC remains largely unclear. Here, we report that loss of HtrA1 attenuates anoikis--a critical physiologic barrier for tumor metastasis. In response to loss of anchorage, HtrA1 expression was upregulated in SKOV3 cells, resulting in autocatalytic activation of HtrA1. Stable knockdown of HtrA1 in SKOV3 and TOV21G cells resulted in resistance to anoikis due to enhanced activation of epidermal growth factor receptor (EGFR)/AKT pathway. In suspended SKOV3 cells, enhanced expression of HtrA1 inhibited EGFR/AKT pathway, leading to increased cell death, whereas protease-inactive mutant HtrA1 failed to result in either the inhibition of EGFR/AKT pathway or increased cell death, suggesting the requirement of HtrA1 protease activity in regulating anoikis. Immunoprecipitation and immunofluorescence assays revealed that HtrA1 interacted with EGFR not only on the cell membrane but also in the nucleus. Most importantly, downregulation of HtrA1 significantly enhanced the peritoneal dissemination of SKOV3ip1 cells in nonobese diabetic/severe combined immunodeficient mice, with increased phospho-EGFR level in corresponding tumor nodules compared with that in xenografts originated from the control cells. Taken together, these data reveal for the first time a novel function of HtrA1 in promoting anoikis by attenuating activation of EGFR/AKT pathway that may contribute to its metastasis suppression capacity, thus providing a possible explanation for the aggressive nature of human ovarian tumors with downregulated HtrA1.
Collapse
Affiliation(s)
- Xiaoping He
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Takayo Ota
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Peng Liu
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Changqing Su
- Department of Gene and Viral Therapy, Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, 225 Changhai Rd., Shanghai 200438, China
| | - Jeremy Chien
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Viji Shridhar
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Oncoprotein metastasis and its suppression revisited. J Exp Clin Cancer Res 2010; 29:30. [PMID: 20380702 PMCID: PMC2855699 DOI: 10.1186/1756-9966-29-30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 04/09/2010] [Indexed: 11/16/2022] Open
Abstract
The past two decades have witnessed an increasing appreciation of the role of the tumor microenvironment, of genetic and epigenetic alterations in normal cells adjacent to tumors and of the migration of normal cells with aberrant intrinsic properties in cancer pathophysiology. Aside from these insights, a novel concept termed "oncoprotein metastasis" (OPM) has recently been advanced and proposed to reflect protein-based neoplastic phenomena that might occur even before any modifications relating to the morphology, location or (epi)genetic outfit of cells during the malignant process. Here, evidence is presented that supports the OPM perception and thus should contribute not only to further rethink the definition of a normal cell, but also the treatment of cancer disease in the years to come.
Collapse
|
28
|
Abstract
The importance of microRNAs (miRs) in control of gene expression is now clearly recognized. While individual microRNAs are thought to target hundreds of disparate mRNAs via imperfect base pairing, little is known about the characteristics of miR target sites. Here we show that the miRs can be aligned with empirically identified accessible sites in a target RNA (Cytokeratin 19, KRT), and that some of the aligned miRs functionally down-regulate KRT expression post-transcriptionally. We employed an RNase-H-based random library selection protocol to identify accessible sites in KRT RNA. We then aligned the Sanger Institute database collection of human miRs to KRT mRNA, and also aligned them using the web-based MicroInspector program. Most miRs aligned with the accessible sites identified empirically; those not aligned with the empirically identified sites also functioned effectively in RNase-H-based assays. Similar results were obtained with a second target RNA (Mammoglobin). Transient transfection assays established that some of the miRs which aligned with KRT significantly down-regulated it at the protein level, with no effect on RNA level. The functionally effective miRs aligned within the coding region of KRT, whereas a number of miRs which aligned with the 3'-untranslated region did not produce down-regulation.
Collapse
Affiliation(s)
- Weihua Pan
- Departments of Pathology and Biochemistry & Molecular Biology, Gittlen Cancer Research Foundation, Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania
| | - Ping Xin
- Departments of Pathology and Biochemistry & Molecular Biology, Gittlen Cancer Research Foundation, Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania
| | - Gary A. Clawson
- Departments of Pathology and Biochemistry & Molecular Biology, Gittlen Cancer Research Foundation, Hershey Medical Center, Pennsylvania State University, Hershey, Pennsylvania
- Materials Research Institute, Pennsylvania State University, State College, Pennsylvania
| |
Collapse
|
29
|
Chien J, Ota T, Aletti G, Shridhar R, Boccellino M, Quagliuolo L, Baldi A, Shridhar V. Serine protease HtrA1 associates with microtubules and inhibits cell migration. Mol Cell Biol 2009; 29:4177-87. [PMID: 19470753 PMCID: PMC2715801 DOI: 10.1128/mcb.00035-09] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 02/25/2009] [Accepted: 05/18/2009] [Indexed: 12/20/2022] Open
Abstract
HtrA1 belongs to a family of serine proteases found in organisms ranging from bacteria to humans. Bacterial HtrA1 (DegP) is a heat shock-induced protein that behaves as a chaperone at low temperature and as a protease at high temperature to help remove unfolded proteins during heat shock. In contrast to bacterial HtrA1, little is known about the function of human HtrA1. Here, we report the first evidence that human HtrA1 is a microtubule-associated protein and modulates microtubule stability and cell motility. Intracellular HtrA1 is localized to microtubules in a PDZ (PSD95, Dlg, ZO1) domain-dependent, nocodazole-sensitive manner. During microtubule assembly, intracellular HtrA associates with centrosomes and newly polymerized microtubules. In vitro, purified HtrA1 promotes microtubule assembly. Moreover, HtrA1 cosediments and copurifies with microtubules. Purified HtrA1 associates with purified alpha- and beta-tubulins, and immunoprecipitation of endogenous HtrA1 results in coprecipitation of alpha-, beta-, and gamma-tubulins. Finally, downregulation of HtrA1 promotes cell motility, whereas enhanced expression of HtrA1 attenuates cell motility. These results offer an original identification of HtrA1 as a microtubule-associated protein and provide initial mechanistic insights into the role of HtrA1 in the regulation of cell motility by modulating microtubule stability.
Collapse
Affiliation(s)
- Jeremy Chien
- Experimental Pathology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Chien J, He X, Shridhar V. Identification of tubulins as substrates of serine protease HtrA1 by mixture-based oriented peptide library screening. J Cell Biochem 2009; 107:253-63. [PMID: 19301262 PMCID: PMC2983094 DOI: 10.1002/jcb.22121] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Serine protease HtrA1 belongs to a family of chymotrypsin-like proteases that were first identified in bacteria and later in mammalian systems. These proteases were identified as components of protein quality control in prokaryotic systems and as regulators of diverse signaling pathways in mammalian systems. In particular, HtrA1 is implicated in trophoblast cell migration and invasion, tumor progression, chemotherapy-induced cytotoxicity, osteoarthritis, age-related macular degeneration, and pathogenesis of Alzheimer's disease. However, systematic analysis of its potential substrates in biological system is still lacking. Therefore, we performed a mixture-based oriented peptide library screening to identify putative substrates of HtrA1. We identified [AEGR]-[LAGR]-[IAMLR]-[TVIAL] as consensus residues for P1 to P4 sites. We identified several putative substrates of HtrA1 involved in the pathogenesis of various diseases. In this study, we report on the identification of tubulins as potential substrates of HtrA1, and validated tubulins as in vitro and intracellular substrates of HtrA1. These results provide initial insights into substrate identification and functional characterization of HtrA1 in pathogenesis of various diseases.
Collapse
Affiliation(s)
- Jeremy Chien
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Xiaoping He
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Viji Shridhar
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|