1
|
Life B, Bettio LE, Gantois I, Christie BR, Leavitt BR. Progranulin is an FMRP target that influences macroorchidism but not behaviour in a mouse model of Fragile X Syndrome. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100094. [PMID: 37416094 PMCID: PMC10319828 DOI: 10.1016/j.crneur.2023.100094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
A growing body of evidence has implicated progranulin in neurodevelopment and indicated that aberrant progranulin expression may be involved in neurodevelopmental disease. Specifically, increased progranulin expression in the prefrontal cortex has been suggested to be pathologically relevant in male Fmr1 knockout (Fmr1 KO) mice, a mouse model of Fragile X Syndrome (FXS). Further investigation into the role of progranulin in FXS is warranted to determine if therapies that reduce progranulin expression represent a viable strategy for treating patients with FXS. Several key knowledge gaps remain. The mechanism of increased progranulin expression in Fmr1 KO mice is poorly understood and the extent of progranulin's involvement in FXS-like phenotypes in Fmr1 KO mice has been incompletely explored. To this end, we have performed a thorough characterization of progranulin expression in Fmr1 KO mice. We find that the phenomenon of increased progranulin expression is post-translational and tissue-specific. We also demonstrate for the first time an association between progranulin mRNA and FMRP, suggesting that progranulin mRNA is an FMRP target. Subsequently, we show that progranulin over-expression in Fmr1 wild-type mice causes reduced repetitive behaviour engagement in females and mild hyperactivity in males but is largely insufficient to recapitulate FXS-associated behavioural, morphological, and electrophysiological abnormalities. Lastly, we determine that genetic reduction of progranulin expression on an Fmr1 KO background reduces macroorchidism but does not alter other FXS-associated behaviours or biochemical phenotypes.
Collapse
Affiliation(s)
- Benjamin Life
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 0B3, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
| | - Luis E.B. Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Ilse Gantois
- Department of Biochemistry, McGill University, Montreal, H3A 2T5, Quebec, Canada
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, H3A 2T5, Quebec, Canada
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Island Medical Program, University of British Columbia, Victoria, BC, V8P 5C2, Canada
- Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Blair R. Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 0B3, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver, BC, V6T 2B5, Canada
- Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
2
|
Zecca C, Tortelli R, Carrera P, Dell'Abate MT, Logroscino G, Ferrari M. Genotype-phenotype correlation in the spectrum of frontotemporal dementia-parkinsonian syndromes and advanced diagnostic approaches. Crit Rev Clin Lab Sci 2022; 60:171-188. [PMID: 36510705 DOI: 10.1080/10408363.2022.2150833] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The term frontotemporal dementia (FTD) refers to a group of progressive neurodegenerative disorders characterized mainly by atrophy of the frontal and anterior temporal lobes. Based on clinical presentation, three main clinical syndromes have traditionally been described: behavioral variant frontotemporal dementia (bvFTD), non-fluent/agrammatic primary progressive aphasia (nfPPA), and semantic variant PPA (svPPA). However, over the last 20 years, it has been recognized that cognitive phenotypes often overlap with motor phenotypes, either motor neuron diseases or parkinsonian signs and/or syndromes like progressive supranuclear palsy (PSP) and cortico-basal syndrome (CBS). Furthermore, FTD-related genes are characterized by genetic pleiotropy and can cause, even in the same family, pure motor phenotypes, findings that underlie the clinical continuum of the spectrum, which has pure cognitive and pure motor phenotypes as the extremes. The genotype-phenotype correlation of the spectrum, FTD-motor neuron disease, has been well defined and extensively investigated, while the continuum, FTD-parkinsonism, lacks a comprehensive review. In this narrative review, we describe the current knowledge about the genotype-phenotype correlation of the spectrum, FTD-parkinsonism, focusing on the phenotypes that are less frequent than bvFTD, namely nfPPA, svPPA, PSP, CBS, and cognitive-motor overlapping phenotypes (i.e. PPA + PSP). From a pathological point of view, they are characterized mainly by the presence of phosphorylated-tau inclusions, either 4 R or 3 R. The genetic correlate of the spectrum can be heterogeneous, although some variants seem to lead preferentially to specific clinical syndromes. Furthermore, we critically review the contribution of genome-wide association studies (GWAS) and next-generation sequencing (NGS) in disentangling the complex heritability of the FTD-parkinsonism spectrum and in defining the genotype-phenotype correlation of the entire clinical scenario, owing to the ability of these techniques to test multiple genes, and so to allow detailed investigations of the overlapping phenotypes. Finally, we conclude with the importance of a detailed genetic characterization and we offer to patients and families the chance to be included in future randomized clinical trials focused on autosomal dominant forms of FTLD.
Collapse
Affiliation(s)
- Chiara Zecca
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Rosanna Tortelli
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis and Clinical Molecular Biology Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Teresa Dell'Abate
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy.,Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | | |
Collapse
|
3
|
Alyahya AM. The role of progranulin in ischemic heart disease and its related risk factors. Eur J Pharm Sci 2022; 175:106215. [DOI: 10.1016/j.ejps.2022.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/23/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022]
|
4
|
Serrero G. Progranulin/GP88, A Complex and Multifaceted Player of Tumor Growth by Direct Action and via the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:475-498. [PMID: 34664252 DOI: 10.1007/978-3-030-73119-9_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Investigation of the role of progranulin/GP88 on the proliferation and survival of a wide variety of cells has been steadily increasing. Several human diseases stem from progranulin dysregulation either through its overexpression in cancer or its absence as in the case of null mutations in some form of frontotemporal dementia. The present review focuses on the role of progranulin/GP88 in cancer development, progression, and drug resistance. Various aspects of progranulin identification, biology, and signaling pathways will be described. Information will be provided about its direct role as an autocrine growth and survival factor and its paracrine effect as a systemic factor as well as via interaction with extracellular matrix proteins and with components of the tumor microenvironment to influence drug resistance, migration, angiogenesis, inflammation, and immune modulation. This chapter will also describe studies examining progranulin/GP88 tumor tissue expression as well as circulating level as a prognostic factor for several cancers. Due to the wealth of publications in progranulin, this review does not attempt to be exhaustive but rather provide a thread to lead the readers toward more in-depth exploration of this fascinating and unique protein.
Collapse
|
5
|
Luo Q, He X, Zheng Y, Ning P, Xu Y, Yang D, Shang Y, Gao Z. Elevated progranulin as a novel biomarker to predict poor prognosis in community-acquired pneumonia. J Infect 2019; 80:167-173. [PMID: 31837341 DOI: 10.1016/j.jinf.2019.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 01/20/2023]
Abstract
PURPOSE Prognostic biomarkers help triage initial patients and inform targeted therapy selection. Here, we explored the role of progranulin (PGRN)-implicated in processes ranging from inflammation to neurodegeneration-in patients with community-acquired pneumonia (CAP). METHODS A prospective observational cohort study was conducted during 2017. Patients who required invasive mechanical ventilation and/or had septic shock and were discharged from the hospital were cohort II. Those who died at the hospital were cohort III. Remaining patients discharged from the hospital were cohort I. The primary endpoint was that patients progressed to served as cohort II; the secondary endpoint was that patients progressed to served as cohort III. Serum PGRN levels were detected by ELISA. RESULTS A total of 280 patients constituted the study cohort. 194 (69.3%) were categorized into cohort I, 61 (21.8%) were categorized into cohort II, and 25 (8.9%) were categorized into cohort III. Serum PGRN levels were increased in CAP patients, independently of etiology. Adjusting for clinical parameters, the odds ratios (95%CI) of cohort III and combined cohort II-III were 34.968 (3.743-326.692) and 3.741 (1.496-9.351), respectively, comparing lowest-to-highest quartile PGRN levels. PGRN exhibited high accuracy in predicting 30-day mortality, with AUC 0.862. PGRN combined with CURB-65 or PSI significantly improved prediction performance. Cox proportional regression analysis showed PGRN was an independent predictor for 30-day mortality risk. Cox survival curves confirmed PGRN ≥89.51 ng/mL had a significantly higher mortality rate than PGRN <89.51 ng/mL. CONCLUSION Higher PGRN levels at admission were associated with higher odds of poor prognosis. PGRN can improve the prognostic power of CURB-65 or PSI, so PGRN could be apparently a prognostic biomarker for assisting triage of CAP patients.
Collapse
Affiliation(s)
- Qiongzhen Luo
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China; Department of Respiratory & Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xinwei He
- Department of Internal Medicine, Xicheng District Zhanlanlu Hospital, Beijing, China
| | - Yali Zheng
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Pu Ning
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yu Xu
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Donghong Yang
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Ying Shang
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Zhancheng Gao
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
6
|
Abstract
Cancer cells have defects in regulatory mechanisms that usually control cell proliferation and homeostasis. Different cancer cells share crucial alterations in cell physiology, which lead to malignant growth. Tumorigenesis or tumor growth requires a series of events that include constant cell proliferation, promotion of metastasis and invasion, stimulation of angiogenesis, evasion of tumor suppressor factors, and avoidance of cell death pathways. All these events in tumor progression may be regulated by growth factors produced by normal or malignant cells. The growth factor progranulin has significant biological effects in different types of cancer. This protein is a regulator of tumorigenesis because it stimulates cell proliferation, migration, invasion, angiogenesis, malignant transformation, resistance to anticancer drugs, and immune evasion. This review focuses on the biological effects of progranulin in several cancer models and provides evidence that this growth factor should be considered as a potential biomarker and target in cancer treatment.
Collapse
|
7
|
Nicholson AM, Finch NA, Almeida M, Perkerson RB, van Blitterswijk M, Wojtas A, Cenik B, Rotondo S, Inskeep V, Almasy L, Dyer T, Peralta J, Jun G, Wood AR, Frayling TM, Fuchsberger C, Fowler S, Teslovich TM, Manning AK, Kumar S, Curran J, Lehman D, Abecasis G, Duggirala R, Pottier C, Zahir HA, Crook JE, Karydas A, Mitic L, Sun Y, Dickson DW, Bu G, Herz J, Yu G, Miller BL, Ferguson S, Petersen RC, Graff-Radford N, Blangero J, Rademakers R. Prosaposin is a regulator of progranulin levels and oligomerization. Nat Commun 2016; 7:11992. [PMID: 27356620 PMCID: PMC4931318 DOI: 10.1038/ncomms11992] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 05/19/2016] [Indexed: 01/09/2023] Open
Abstract
Progranulin (GRN) loss-of-function mutations leading to progranulin protein (PGRN) haploinsufficiency are prevalent genetic causes of frontotemporal dementia. Reports also indicated PGRN-mediated neuroprotection in models of Alzheimer's and Parkinson's disease; thus, increasing PGRN levels is a promising therapeutic for multiple disorders. To uncover novel PGRN regulators, we linked whole-genome sequence data from 920 individuals with plasma PGRN levels and identified the prosaposin (PSAP) locus as a new locus significantly associated with plasma PGRN levels. Here we show that both PSAP reduction and overexpression lead to significantly elevated extracellular PGRN levels. Intriguingly, PSAP knockdown increases PGRN monomers, whereas PSAP overexpression increases PGRN oligomers, partly through a protein–protein interaction. PSAP-induced changes in PGRN levels and oligomerization replicate in human-derived fibroblasts obtained from a GRN mutation carrier, further supporting PSAP as a potential PGRN-related therapeutic target. Future studies should focus on addressing the relevance and cellular mechanism by which PGRN oligomeric species provide neuroprotection. Increasing progranulin (PGRN) levels is a promising approach for treating frontotemporal dementia and other neurodegenerative diseases. Here Nicholson et al. show that the prosaposin (PSAP) locus is associated with plasma PGRN levels and demonstrate that PSAP can alter PGRN levels and its oligomerization.
Collapse
Affiliation(s)
| | - NiCole A Finch
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Marcio Almeida
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Ralph B Perkerson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | | | - Aleksandra Wojtas
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Basar Cenik
- Department of Neuroscience, Molecular Genetics, and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Sergio Rotondo
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Venette Inskeep
- Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA
| | - Laura Almasy
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Thomas Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Juan Peralta
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Goo Jun
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Andrew R Wood
- Genetics of Complex Traits, St Luke's Campus, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, St Luke's Campus, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Christian Fuchsberger
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Sharon Fowler
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | - Tanya M Teslovich
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Alisa K Manning
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Satish Kumar
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Joanne Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Donna Lehman
- Department of Medicine/Cardiology and Cellular &Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Goncalo Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ravindranath Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Cyril Pottier
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Haaris A Zahir
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Julia E Crook
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Anna Karydas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94143, USA
| | - Laura Mitic
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94143, USA
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Joachim Herz
- Department of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Gang Yu
- Department of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94143, USA
| | - Shawn Ferguson
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55902, USA
| | | | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas 78520, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| |
Collapse
|
8
|
Progranulin Stimulates Proliferation of Mouse Pancreatic Islet Cells and Is Overexpressed in the Endocrine Pancreatic Tissue of an MEN1 Mouse Model. Pancreas 2016; 45:533-40. [PMID: 26495792 DOI: 10.1097/mpa.0000000000000509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Progranulin (PGRN) promotes cell growth and cell cycle progression in several cell types and contributes to tumorigenesis in diverse cancers. We have recently reported PGRN expression in islets and tumors developed in an MEN1 transgenic mouse. Here we sought to investigate PGRN expression and regulation after exposure to hypoxia as well as its effects on pancreatic islet cells and neuroendocrine tumors (NETs) in MEN1(+/−) mice. METHODS Gene and protein expression were analyzed by quantitative polymerase chain reaction, immunohistochemistry, and Western blot. We also investigated PGRN expression in samples from patients carrying pancreatic NETs associated or not with the multiple endocrine neoplasia 1 syndrome, using enzyme-linked immunosorbent assay and immunohistochemistry analysis. RESULTS Progranulin is upregulated in tumors and islets of the MEN1 mouse as well as in the serum of patients with pancreatic NETs associated with glucagonoma syndrome. In normal mice islets and pancreatic tumors, PGRN expression was strongly potentiated by hypoxia. Progranulin promotes cell proliferation in islet cells and βTC-6 cells, a process paralleled by activation of the mitogen-activated protein kinase signaling cascade. CONCLUSIONS Our findings identify PGRN as an effective inducer of pancreatic islet cell proliferation and a possible important factor for pancreatic endocrine tumor development.
Collapse
|
9
|
Tanimoto R, Lu KG, Xu SQ, Buraschi S, Belfiore A, Iozzo RV, Morrione A. Mechanisms of Progranulin Action and Regulation in Genitourinary Cancers. Front Endocrinol (Lausanne) 2016; 7:100. [PMID: 27512385 PMCID: PMC4961702 DOI: 10.3389/fendo.2016.00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 07/08/2016] [Indexed: 11/13/2022] Open
Abstract
The growth factor progranulin has emerged in recent years as a critical regulator of transformation in several cancer models, including breast cancer, glioblastomas, leukemias, and hepatocellular carcinomas. Several laboratories, including ours, have also demonstrated an important role of progranulin in several genitourinary cancers, including ovarian, endometrial, cervical, prostate, and bladder tumors, where progranulin acts as an autocrine growth factor thereby modulating motility and invasion of transformed cells. In this review, we will focus on the mechanisms of action and regulation of progranulin signaling in genitourinary cancers with a special emphasis on prostate and bladder tumors.
Collapse
Affiliation(s)
- Ryuta Tanimoto
- Biology of Prostate Cancer Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kuojung G. Lu
- Biology of Prostate Cancer Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shi-Qiong Xu
- Biology of Prostate Cancer Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Simone Buraschi
- Cancer Cell Biology and Signaling Program, Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Antonino Belfiore
- Department of Health Sciences, Endocrinology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Renato V. Iozzo
- Cancer Cell Biology and Signaling Program, Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea Morrione
- Biology of Prostate Cancer Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- *Correspondence: Andrea Morrione,
| |
Collapse
|
10
|
Sato K, Yamanaka Y, Ishii M, Ishibashi K, Ogura Y, Ohtani-Kaneko R, Nishihara M, Nedachi T. Dual cell protective mechanisms activated by differing levels of oxidative stress in HT22 murine hippocampal cells. Biosci Biotechnol Biochem 2014; 78:1495-503. [DOI: 10.1080/09168451.2014.936343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Abstract
Oxidative stress is recognized as one of the pathogenic mechanisms involved in neurodegenerative disease. However, recent evidence has suggested that regulation of cellular fate in response to oxidative stress appears to be dependent on the stress levels. In this study, using HT22 cells, we attempted to understand how an alteration in the oxidative stress levels would influence neuronal cell fate. HT22 cell viability was reduced with exposure to high levels of oxidative stress, whereas, low levels of oxidative stress promoted cell survival. Erk1/2 activation induced by a low level of oxidative stress played a role in this cell protective effect. Intriguingly, subtoxic level of H2O2 induced expression of a growth factor, progranulin (PGRN), and exogenous PGRN pretreatment attenuated HT22 cell death induced by high concentrations of H2O2 in Erk1/2-dependent manner. Together, our study indicates that two different cell protection mechanisms are activated by differing levels of oxidative stress in HT22 cells.
Collapse
Affiliation(s)
- Kazunori Sato
- Graduate School of Life Sciences, Toyo University, Gunma, Japan
| | - Yuki Yamanaka
- Faculty of Life Sciences, Toyo University, Gunma, Japan
| | - Masaya Ishii
- Faculty of Life Sciences, Toyo University, Gunma, Japan
| | | | - Yurina Ogura
- Graduate School of Life Sciences, Toyo University, Gunma, Japan
| | - Ritsuko Ohtani-Kaneko
- Graduate School of Life Sciences, Toyo University, Gunma, Japan
- Faculty of Life Sciences, Toyo University, Gunma, Japan
- Bio-Nano Electronics Research Centre, Toyo University, Saitama, Japan
| | - Masugi Nishihara
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Taku Nedachi
- Graduate School of Life Sciences, Toyo University, Gunma, Japan
- Faculty of Life Sciences, Toyo University, Gunma, Japan
| |
Collapse
|
11
|
Judy ME, Nakamura A, Huang A, Grant H, McCurdy H, Weiberth KF, Gao F, Coppola G, Kenyon C, Kao AW. A shift to organismal stress resistance in programmed cell death mutants. PLoS Genet 2013; 9:e1003714. [PMID: 24068943 PMCID: PMC3778000 DOI: 10.1371/journal.pgen.1003714] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 06/26/2013] [Indexed: 11/27/2022] Open
Abstract
Animals have many ways of protecting themselves against stress; for example, they can induce animal-wide, stress-protective pathways and they can kill damaged cells via apoptosis. We have discovered an unexpected regulatory relationship between these two types of stress responses. We find that C. elegans mutations blocking the normal course of programmed cell death and clearance confer animal-wide resistance to a specific set of environmental stressors; namely, ER, heat and osmotic stress. Remarkably, this pattern of stress resistance is induced by mutations that affect cell death in different ways, including ced-3 (cell death defective) mutations, which block programmed cell death, ced-1 and ced-2 mutations, which prevent the engulfment of dying cells, and progranulin (pgrn-1) mutations, which accelerate the clearance of apoptotic cells. Stress resistance conferred by ced and pgrn-1 mutations is not additive and these mutants share altered patterns of gene expression, suggesting that they may act within the same pathway to achieve stress resistance. Together, our findings demonstrate that programmed cell death effectors influence the degree to which C. elegans tolerates environmental stress. While the mechanism is not entirely clear, it is intriguing that animals lacking the ability to efficiently and correctly remove dying cells should switch to a more global animal-wide system of stress resistance. As an animal interacts with its environment, it invariably encounters stressful conditions such as extreme temperatures, drought, UV exposure and harmful xenobiotics. Since the ability to respond appropriately to stressful stimuli is paramount to survival, organisms have developed sophisticated stress response programs. Some stressful conditions cause damaged cells to commit suicide (undergo apoptosis), whereas others cause the entire organism to develop mechanisms to resist environmental stress. Studying the small roundworm C. elegans, we find that these two responses are somehow linked: perturbing the mechanisms that allow cells to undergo apoptosis changes the whole animal's response to environmental stress. In fact, perturbing the apoptosis machinery in any way—through mutations that prevent apoptosis altogether, or through mutations that either slow or accelerate the clearance of dying cells—causes the animal to become more stress resistant. Together our findings raise the possibility that the animal may have a way of detecting defects in the normal programmed cell death pathway, and that in response it induces a new program that protects itself from a harsh environment.
Collapse
Affiliation(s)
- Meredith E. Judy
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Ayumi Nakamura
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Anne Huang
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Harli Grant
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Helen McCurdy
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Kurt F. Weiberth
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Fuying Gao
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Cynthia Kenyon
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Aimee W. Kao
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
12
|
Zhao J, Wei J, Liu M, Xiao L, Wu N, Liu G, Huang H, Zhang Y, Zheng L, Lin X. Cloning, characterization and expression of a cDNA encoding a granulin-like polypeptide in Ciona savignyi. Biochimie 2013; 95:1611-9. [DOI: 10.1016/j.biochi.2013.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 05/03/2013] [Indexed: 11/26/2022]
|
13
|
Kleinberger G, Capell A, Haass C, Van Broeckhoven C. Mechanisms of granulin deficiency: lessons from cellular and animal models. Mol Neurobiol 2012; 47:337-60. [PMID: 23239020 PMCID: PMC3538123 DOI: 10.1007/s12035-012-8380-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/14/2012] [Indexed: 12/12/2022]
Abstract
The identification of causative mutations in the (pro)granulin gene (GRN) has been a major breakthrough in the research on frontotemporal dementia (FTD). So far, all FTD-associated GRN mutations are leading to neurodegeneration through a “loss-of-function” mechanism, encouraging researchers to develop a growing number of cellular and animal models for GRN deficiency. GRN is a multifunctional secreted growth factor, and loss of its function can affect different cellular processes. Besides loss-of-function (i.e., mostly premature termination codons) mutations, which cause GRN haploinsufficiency through reduction of GRN expression, FTD-associated GRN missense mutations have also been identified. Several of these missense mutations are predicted to increase the risk of developing neurodegenerative diseases through altering various key biological properties of GRN-like protein secretion, proteolytic processing, and neurite outgrowth. With the use of cellular and animal models for GRN deficiency, the portfolio of GRN functions has recently been extended to include functions in important biological processes like energy and protein homeostasis, inflammation as well as neuronal survival, neurite outgrowth, and branching. Furthermore, GRN-deficient animal models have been established and they are believed to be promising disease models as they show accelerated aging and recapitulate at least some neuropathological features of FTD. In this review, we summarize the current knowledge on the molecular mechanisms leading to GRN deficiency and the lessons we learned from the established cellular and animal models. Furthermore, we discuss how these insights might help in developing therapeutic strategies for GRN-associated FTD.
Collapse
Affiliation(s)
- Gernot Kleinberger
- Neurodegenerative Brain Diseases Group, VIB Department of Molecular Genetics, University of Antwerp-CDE, Universiteitsplein 1, Antwerp, 2610, Belgium
| | | | | | | |
Collapse
|
14
|
Hu SY, Tai CC, Li YH, Wu JL. Progranulin compensates for blocked IGF-1 signaling to promote myotube hypertrophy in C2C12 myoblasts via the PI3K/Akt/mTOR pathway. FEBS Lett 2012; 586:3485-92. [PMID: 22967900 DOI: 10.1016/j.febslet.2012.07.077] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 12/19/2022]
Abstract
It is well known that growth hormone (GH)-induced IGF-1 signaling plays a dominant role in postnatal muscle growth. Our previous studies have identified a growth factor, progranulin (PGRN), that is co-induced with IGF-1 upon GH administration. This result prompted us to explore the function of PGRN and its association with IGF-1. In the present study, we demonstrated that, similar to IGF-1, PGRN can promote C2C12 myotube hypertrophy via the PI(3)K/Akt/mTOR pathway. Moreover, PGRN can rescue the muscle atrophy phenotypes in C2C12 myotube when IGF-1 signaling is blocked. This result shows that PGRN can substitute for IGF-1 signaling in the regulation of muscle growth. Our findings provide new insights into IGF-1-modulated complicated networks that regulate muscle growth.
Collapse
Affiliation(s)
- Shao-Yang Hu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | | | | | | |
Collapse
|
15
|
Wils H, Kleinberger G, Pereson S, Janssens J, Capell A, Van Dam D, Cuijt I, Joris G, De Deyn PP, Haass C, Van Broeckhoven C, Kumar-Singh S. Cellular ageing, increased mortality and FTLD-TDP-associated neuropathology in progranulin knockout mice. J Pathol 2012; 228:67-76. [PMID: 22733568 DOI: 10.1002/path.4043] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 03/31/2012] [Accepted: 04/14/2012] [Indexed: 12/13/2022]
Abstract
Loss-of-function mutations in progranulin (GRN) are associated with frontotemporal lobar degeneration with intraneuronal ubiquitinated protein accumulations composed primarily of hyperphosphorylated TDP-43 (FTLD-TDP). The mechanism by which GRN deficiency causes TDP-43 pathology or neurodegeneration remains elusive. To explore the role of GRN in vivo, we established Grn knockout mice using a targeted genomic recombination approach and Cre-LoxP technology. Constitutive Grn homozygous knockout (Grn(-/-) ) mice were born in an expected Mendelian pattern of inheritance and showed no phenotypic alterations compared to heterozygous (Grn(+/-) ) or wild-type (Wt) littermates until 10 months of age. From then, Grn(-/-) mice showed reduced survival accompanied by significantly increased gliosis and ubiquitin-positive accumulations in the cortex, hippocampus, and subcortical regions. Although phosphorylated TDP-43 could not be detected in the ubiquitinated inclusions, elevated levels of hyperphosphorylated full-length TDP-43 were recovered from detergent-insoluble brain fractions of Grn(-/-) mice. Phosphorylated TDP-43 increased with age and was primarily extracted from the nuclear fraction. Grn(-/-) mice also showed degenerative liver changes and cathepsin D-positive foamy histiocytes within sinusoids, suggesting widespread defects in lysosomal turnover. An increase in insulin-like growth factor (IGF)-1 was observed in Grn(-/-) brains, and increased IGF-1 signalling has been associated with decreased longevity. Our data suggest that progranulin deficiency in mice leads to reduced survival in adulthood and increased cellular ageing accompanied by hyperphosphorylation of TDP-43, and recapitulates key aspects of FTLD-TDP neuropathology.
Collapse
Affiliation(s)
- Hans Wils
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang D, Bai X, Tian Q, Lai Y, Lin EA, Shi Y, Mu X, Feng JQ, Carlson CS, Liu CJ. GEP constitutes a negative feedback loop with MyoD and acts as a novel mediator in controlling skeletal muscle differentiation. Cell Mol Life Sci 2012; 69:1855-73. [PMID: 22179841 PMCID: PMC3319484 DOI: 10.1007/s00018-011-0901-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 11/22/2011] [Accepted: 11/28/2011] [Indexed: 01/16/2023]
Abstract
Granulin-epithelin precursor (GEP) is an autocrine growth factor that has been implicated in embryonic development, tissue repair, tumorigenesis, and inflammation. Here we report that GEP was expressed in skeletal muscle tissue and its level was differentially altered in the course of C2C12 myoblast fusion. The GEP expression during myoblast fusion was a consequence of MyoD transcription factor binding to several E-box (CANNTG) sequences in the 5'-flanking regulatory region of GEP gene, followed by transcription. Recombinant GEP potently inhibited myotube formation from C2C12 myoblasts whereas the knockdown of endogenous of GEP via a siRNA approach accelerated the fusion of myoblasts to myotubes. Interestingly, the muscle fibers of GEP knockdown mice were larger in number but noticeably smaller in size when compared to the wild-type. Mechanistic studies revealed that during myoblast fusion, the addition of GEP led to remarkable reductions in the expressions of muscle-specific transcription factors, including MyoD. In addition, the regulation of myotube formation by GEP is mediated by the anti-myogenic factor JunB, which is upregulated following GEP stimulation. Thus, GEP growth factor, JunB, and MyoD transcription factor form a regulatory loop and act in concert in the course of myogenesis.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Orthopedics, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Xiaohui Bai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Otorhinolaryngology Head and Neck Surgery, Provincial Hospital affiliated to Shandong University, Jinan 250021, China
| | - Qingyun Tian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Yongjie Lai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Edward A. Lin
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Yongxiang Shi
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Xiaodong Mu
- Stem Cell Research Center, Children’s Hospital of Pittsburgh and Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219
| | - Jian Q. Feng
- Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX 75246
| | - Cathy S. Carlson
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
17
|
Serrero G, Hawkins DM, Yue B, Ioffe O, Bejarano P, Phillips JT, Head JF, Elliott RL, Tkaczuk KR, Godwin AK, Weaver J, Kim WE. Progranulin (GP88) tumor tissue expression is associated with increased risk of recurrence in breast cancer patients diagnosed with estrogen receptor positive invasive ductal carcinoma. Breast Cancer Res 2012; 14:R26. [PMID: 22316048 PMCID: PMC3496144 DOI: 10.1186/bcr3111] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 01/16/2012] [Accepted: 02/08/2012] [Indexed: 01/10/2023] Open
Abstract
Introduction GP88 (progranulin) has been implicated in tumorigenesis and resistance to anti-estrogen therapies for estrogen receptor positive (ER+) breast cancer. Previous pathological studies showed that GP88 is expressed in invasive ductal carcinoma (IDC), but not in normal mammary epithelial tissue, benign lesions or lobular carcinoma. Based on these results, the present study examines GP88 prognostic significance in association with recurrence and death risks for ER+ IDC patients. Methods Two retrospective multi-site clinical studies examined GP88 expression by immunohistochemistry (IHC) analysis of paraffin-embedded breast tumor tissue sections from ER+ IDC patients (lymph node positive and negative, stage 1 to 3) in correlation with patients' survival outcomes. The training study established a GP88 cut-off value associated with decreased disease-free (DFS) and overall (OS) survivals. The validation study verified the GP88 cut-off value and compared GP88 prognostic information with other prognostic factors, particularly tumor size, grade, disease stage and lymph node status in multivariate analysis. Results GP88 expression is associated with a statistically significant increase in recurrence risk for ER+ IDC patients. The training study established that GP88 3+ score was associated with decreased DFS (P = 0.0004) and OS (P = 0.0036). The independent validation study verified that GP88 3+ score was associated with a 5.9-fold higher hazard of disease recurrence and a 2.5-fold higher mortality hazard compared to patients with tumor GP88 < 3+. GP88 remained an independent risk predictor after considering age, ethnicity, nodal status, tumor size, tumor grade, disease stage, progesterone receptor expression and treatments. Conclusions The survival factor GP88 is a novel prognostic biomarker, predictive of recurrence risk and increased mortality for non-metastatic ER+ IDC patients. Of importance, our data show that GP88 continues to be a prognostic factor even after five years. These results also provide evidence that GP88 provides prognostic information independent of tumor and clinical characteristics and would support prospective study to examine whether GP88 expression could help stratify patients with ER+ tumors for adjuvant therapy.
Collapse
Affiliation(s)
- Ginette Serrero
- A&G Pharmaceutical Inc,, 9130 Red Branch Rd,, Columbia, MD 21045, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gass J, Prudencio M, Stetler C, Petrucelli L. Progranulin: an emerging target for FTLD therapies. Brain Res 2012; 1462:118-28. [PMID: 22338605 DOI: 10.1016/j.brainres.2012.01.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/13/2012] [Accepted: 01/18/2012] [Indexed: 12/12/2022]
Abstract
Frontotemporal lobar degeneration (FTLD), a neurodegenerative disease primarily affecting the frontal and temporal lobes, is one of the most common types of dementia. While the majority of FTLD cases are sporadic, approximately 10-40% of patients have an inherited form of FTLD. Mutations in the progranulin gene (GRN) have recently been identified as a major cause of FTLD with ubiquitin positive inclusions (FTLD-U). Because over 70 disease-linked GRN mutations cause abnormal deficiencies in the production of PGRN, a protein that plays a crucial role in embryogenesis, cell growth and survival, wound repair and inflammation, researchers now aim to design therapies that would increase PGRN levels in affected individuals, thereby alleviating the symptoms associated with disease. Several compounds and genetic factors, as well as PGRN receptors, have recently been identified because of their ability to regulate PGRN levels. Strict quality control measures are needed given that extreme PGRN levels at either end of the spectrum - too low or too high - can lead to neurodegeneration or cancer, respectively. The aim of this review is to highlight what is known regarding PGRN biology; to improve understanding of the mechanisms involved in regulating PGRN levels and highlight studies that are laying the groundwork for the development of effective therapeutic modulators of PGRN. This article is part of a Special Issue entitled RNA-Binding Proteins.
Collapse
Affiliation(s)
- Jennifer Gass
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | | | |
Collapse
|
19
|
Nicholson AM, Gass J, Petrucelli L, Rademakers R. Progranulin axis and recent developments in frontotemporal lobar degeneration. ALZHEIMERS RESEARCH & THERAPY 2012; 4:4. [PMID: 22277331 PMCID: PMC3372369 DOI: 10.1186/alzrt102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is a devastating neurodegenerative disease that is the second most common form of dementia affecting individuals under age 65. The most common pathological subtype, FTLD with transactive response DNA-binding protein with a molecular weight of 43 kDa inclusions (FTLD-TDP), is often caused by autosomal dominant mutations in the progranulin gene (GRN) encoding the progranulin protein (PGRN). GRN pathogenic mutations result in haploinsufficiency, usually by nonsense-mediated decay of the mRNA. Since the discovery of these mutations in 2006, several groups have published data and animal models that provide further insight into the genetic and functional relevance of PGRN in the context of FTLD-TDP. These studies were critical in initiating our understanding of the role of PGRN in neural development, degeneration, synaptic transmission, cell signaling, and behavior. Furthermore, recent publications have now identified the receptors for PGRN, which will hopefully lead to additional therapeutic targets. Additionally, drug screens have been conducted to identify pharmacological regulators of PGRN levels to be used as potential treatments for PGRN haploinsufficiency. Here we review recent literature describing relevant data on GRN genetics, cell culture experiments describing the potential role and regulators of PGRN in the central nervous system, animal models of PGRN deficiency, and potential PGRN-related FTLD therapies that are currently underway. The present review aims to underscore the necessity of further elucidation of PGRN biology in FTLD-related neurodegeneration.
Collapse
Affiliation(s)
- Alexandra M Nicholson
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | | | | | | |
Collapse
|
20
|
Dennissen FJA, Kholod N, van Leeuwen FW. The ubiquitin proteasome system in neurodegenerative diseases: culprit, accomplice or victim? Prog Neurobiol 2012; 96:190-207. [PMID: 22270043 DOI: 10.1016/j.pneurobio.2012.01.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/18/2011] [Accepted: 01/05/2012] [Indexed: 12/14/2022]
Abstract
A shared hallmark for many neurodegenerative disorders is the accumulation of toxic protein species which is assumed to be the cause for these diseases. Since the ubiquitin proteasome system (UPS) is the most important pathway for selective protein degradation it is likely that it is involved in the aetiology neurodegenerative disorders. Indeed, impairment of the UPS has been reported to occur during neurodegeneration. Although accumulation of toxic protein species (amyloid β) are in turn known to impair the UPS the relationship is not necessarily causal. We provide an overview of the most recent insights in the roles the UPS plays in protein degradation and other processes. Additionally, we discuss the role of the UPS in clearance of the toxic proteins known to accumulate in the hallmarks of neurodegenerative diseases. The present paper will focus on critically reviewing the involvement of the UPS in specific neurodegenerative diseases and will discuss if UPS impairment is a cause, a consequence or both of the disease.
Collapse
Affiliation(s)
- F J A Dennissen
- Department of Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | | | | |
Collapse
|
21
|
Kumar-Singh S. Progranulin and TDP-43: mechanistic links and future directions. J Mol Neurosci 2011; 45:561-73. [PMID: 21863317 PMCID: PMC3207122 DOI: 10.1007/s12031-011-9625-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022]
Abstract
Loss-of-function mutations in the multifunctional growth factor progranulin (GRN) cause frontotemporal lobar degeneration (FTLD) with TDP-43 protein accumulation. Nuclear TDP-43 protein with key roles in RNA metabolism is also aggregated in amyotrophic lateral sclerosis (ALS), suggesting that ALS and FTLD constitute a broad disease continuum. However, the fact that mutations in GRN are associated with FTLD, while mutations in TDP-43 cause a preferential loss of motor neurons resulting in ALS-end of the disease spectrum, suggests involvement of both cell-autonomous and non-autonomous mechanisms. Studies on animal models and in vitro studies have been instrumental in understanding the link between GRN and TDP-43 and also their role in neurodegeneration. For instance, in mouse models, allelic deficiencies of Grn do not recapitulate human pathology of TDP-43 brain accumulations, but embryonic neurons derived from these mice do show abnormal TDP-43 accumulation after additional cellular challenges, suggesting that TDP-43 changes observed in GRN mutation carriers might also relate to stress. Recent results have shown that the dual action of GRN in growth modulation and inflammation could be due to its negative regulation of TNF-α signaling. In addition, GRN also interacts with sortilin and is endocytosed, thereby regulating its own levels and possibly also modulating the turnover of other proteins including that of TDP-43. Accumulating evidence suggests that TDP-43 abnormal cellular aggregation causes a possible gain of function, also suggested by recently constructed mouse models of TDP-43 proteinopathy; however, it would be inconvincible that sequestration of physiological TDP-43 within cellular aggregates observed in patients would be innocuous for disease pathogenesis. This review discusses some of these data on the possible link between GRN and TDP-43 as well as mechanisms involved in TDP-43-led neurodegeneration. Continued multitiered efforts on genetic, cell biological, and animal modeling approaches would prove crucial in finding a cure for GRN-related diseases.
Collapse
Affiliation(s)
- Samir Kumar-Singh
- Laboratory of Molecular and Cellular Neuropathology, University of Antwerp, Universiteitsplein 1, 2610, Antwerpen, Belgium.
| |
Collapse
|
22
|
Liu CJ, Bosch X. Progranulin: a growth factor, a novel TNFR ligand and a drug target. Pharmacol Ther 2011; 133:124-32. [PMID: 22008260 DOI: 10.1016/j.pharmthera.2011.10.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 09/21/2011] [Indexed: 01/05/2023]
Abstract
Progranulin (PGRN) is abundantly expressed in epithelial cells, immune cells, neurons, and chondrocytes, and reportedly contributes to tumorigenesis. PGRN is a crucial mediator of wound healing and tissue repair. PGRN also functions as a neurotrophic factor and mutations in the PGRN gene resulting in partial loss of the PGRN protein cause frontotemporal dementia. PGRN has been found to be a novel chondrogenic growth factor and to play an important role in cartilage development and inflammatory arthritis. Although research has shown that PGRN exhibits anti-inflammatory properties, the details about the exact molecular pathway of such effects, and, in particular, the PGRN binding receptor, have not been identified so far. Recently, researchers have shown that PGRN binds to tumor necrosis factor (TNF)-receptors (TNFR), interfering with the interaction between TNFα and TNFR. They further demonstrated that mice deficient in PGRN are susceptible to collagen-induced arthritis, an experimental model of rheumatoid arthritis, and that administration of PGRN reversed the arthritic process. An engineered protein made of three PGRN fragments (Atsttrin), displayed selective TNFR binding and was more active than natural PGRN. Both PGRN and Atsttrin prevented inflammation in various arthritis mouse models and inhibited TNFα-induced intracellular signaling pathways. Thus, PGRN is a key regulator of inflammation and it may mediate its anti-inflammatory effects, at least in part, by blocking TNF binding to its receptors. As we discuss here, TNFR-based interventions may both stimulate and suppress the growth of cancer cells, and the same may be true in analogy for Atsttrin as a new player.
Collapse
Affiliation(s)
- Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003, United States
| | | |
Collapse
|
23
|
Tkaczuk KR, Yue B, Zhan M, Tait N, Yarlagadda L, Dai H, Serrero G. Increased Circulating Level of the Survival Factor GP88 (Progranulin) in the Serum of Breast Cancer Patients When Compared to Healthy Subjects. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2011; 5:155-62. [PMID: 21792312 PMCID: PMC3140268 DOI: 10.4137/bcbcr.s7224] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Introduction: GP88 (PC-Cell Derived Growth Factor, progranulin) is a glycoprotein overexpressed in breast tumors and involved in their proliferation and survival. Since GP88 is secreted, an exploratory study was established to compare serum GP88 level between breast cancer patients (BC) and healthy volunteers (HV). Methods: An IRB approved prospective study enrolled 189 stage 1–4 BC patients and 18 HV. GP88 serum concentration was determined by immunoassay. Results: Serum GP88 level was 28.7 + 5.8 ng/ml in HV and increased to 40.7 + 16.0 ng/ml (P = 0.007) for stage 1–3 and 45.3 + 23.3 ng/ml (P = 0.0007) for stage 4 BC patients. There was no correlation between the GP88 level and BC characteristics such as age, race, tumor grade, ER, PR and HER-2 expression. Conclusion: These data suggest that serial testing of serum GP88 levels may have value as a circulating biomarker for detection, monitoring and follow up of BC.
Collapse
Affiliation(s)
- Katherine Rak Tkaczuk
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Cellular effects of progranulin in health and disease. J Mol Neurosci 2011; 45:549-60. [PMID: 21611805 DOI: 10.1007/s12031-011-9553-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/10/2011] [Indexed: 12/12/2022]
Abstract
Progranulin is a fascinating multifunctional protein, which has been implicated in cell growth, wound repair, tumorigenesis, inflammation, neurodevelopment, and more recently in neurodegeneration. The mechanism of action of this protein is still largely unknown, but the knowledge about the cellular effects on various cell types is expanding. In the current review, we will summarize what is known about the cell biology of progranulin. A better understanding of the biology of progranulin will impact diverse areas of research.
Collapse
|
25
|
Nedachi T, Kawai T, Matsuwaki T, Yamanouchi K, Nishihara M. Progranulin enhances neural progenitor cell proliferation through glycogen synthase kinase 3β phosphorylation. Neuroscience 2011; 185:106-15. [PMID: 21540081 DOI: 10.1016/j.neuroscience.2011.04.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 03/28/2011] [Accepted: 04/13/2011] [Indexed: 02/06/2023]
Abstract
Progranulin (PGRN) is an estrogen-inducible growth factor thought to affect multiple processes in the CNS, including brain sexual differentiation, adult neurogenesis in the hippocampus, and development of neurodegenerative diseases. However, the precise physiological functions of PGRN in individual nerve cells are not fully understood. The aim of the present study was to enhance the understanding of PGRN function in the CNS by investigating the effects of PGRN on neural progenitor cells (NPCs). We found that significant amounts of endogenous PGRN were secreted from isolated NPCs in cultures. To assess the bioactivities of endogenous and exogenous PGRN, we studied NPCs derived from wild-type mice (WT-NPCs) and PGRN-deficient mice (KO-NPCs). We found that proliferation of KO-NPCs was significantly enhanced by PGRN treatment; however, PGRN treatment apparently did not affect proliferation of WT-NPCs perhaps because of the high levels of endogenous PGRN expression. NPC death and asymmetric cellular division of KO-NPCs and WT-NPCs, which results in production of neural stem cells, astrocytes, or oligodendrocytes, were not affected by PGRN treatment. We also investigated the signaling mechanism(s) that mediate PGRN-induced NPC proliferation and found that phosphorylation of serine 9 (S9) of glycogen synthase kinase 3-beta (GSK3β), which was dependent on phosphatidylinositol 3-kinase (PI3K) activity, was induced by PGRN treatment. In addition, a GSK3β-specific inhibitor enhanced NPC proliferation. Taken together, our observations indicate that PGRN enhanced NPC proliferation, at least in part, via inducing GSK3β phosphorylation.
Collapse
Affiliation(s)
- T Nedachi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
26
|
Kleinberger G, Wils H, Ponsaerts P, Joris G, Timmermans JP, Van Broeckhoven C, Kumar-Singh S. Increased caspase activation and decreased TDP-43 solubility in progranulin knockout cortical cultures. J Neurochem 2010; 115:735-47. [PMID: 20731760 DOI: 10.1111/j.1471-4159.2010.06961.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Null mutations in progranulin (GRN) are associated with frontotemporal lobar degeneration characterized by intraneuronal accumulation of TAR DNA-binding protein-43 (TDP-43). However, the mechanism by which GRN deficiency leads to neurodegeneration remains largely unknown. In primary cortical neurons derived from Grn knockout (Grn(-/-) ) mice, we found that Grn-deficiency causes significantly reduced neuronal survival and increased caspase-mediated apoptosis, which was not observed in primary mouse embryonic fibroblasts derived from Grn(-/-) mice. Also, neurons derived from Grn(-/-) mice showed an increased amount of pTDP-43 accumulations. Furthermore, proteasomal inhibition with MG132 caused increased caspase-mediated TDP-43 fragmentation and accumulation of detergent-insoluble 35- and 25-kDa C-terminal fragments in Grn(-/-) neurons and mouse embryonic fibroblasts. Interestingly, full-length TDP-43 also accumulated in the detergent-insoluble fraction, and caspase-inhibition prevented MG132-induced generation of TDP-43 C-terminal fragments but did not block the pathological conversion of full-length TDP-43 from soluble to insoluble species. These data suggest that GRN functions as a survival factor for cortical neurons and GRN-deficiency causes increased susceptibility to cellular stress. This leads to increased aggregation and accumulation of full-length TDP-43 along with its C-terminal derivatives by both caspase-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Gernot Kleinberger
- Department of Molecular Genetics, VIB, Universiteitsplein 1, Antwerpen, Belgium
| | | | | | | | | | | | | |
Collapse
|
27
|
Guo F, Lai Y, Tian Q, Lin EA, Kong L, Liu C. Granulin-epithelin precursor binds directly to ADAMTS-7 and ADAMTS-12 and inhibits their degradation of cartilage oligomeric matrix protein. ACTA ACUST UNITED AC 2010; 62:2023-36. [PMID: 20506400 DOI: 10.1002/art.27491] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine 1) whether a protein interaction network exists between granulin-epithelin precursor (GEP), ADAMTS-7/ADAMTS-12, and cartilage oligomeric matrix protein (COMP); 2) whether GEP interferes with the interactions between ADAMTS-7/ADAMTS-12 metalloproteinases and COMP substrate, including the cleavage of COMP; 3) whether GEP affects tumor necrosis factor alpha (TNFalpha)-mediated induction of ADAMTS-7/ADAMTS-12 expression and COMP degradation; and 4) whether GEP levels are altered during the progression of arthritis. METHODS Yeast two-hybrid, in vitro glutathione S-transferase pull-down, and coimmunoprecipitation assays were used to 1) examine the interactions between GEP, ADAMTS-7/ADAMTS-12, and COMP, and 2) map the binding sites required for the interactions between GEP and ADAMTS-7/ADAMTS-12. Immunofluorescence cell staining was performed to visualize the subcellular localization of GEP and ADAMTS-7/ADAMTS-12. An in vitro digestion assay was employed to determine whether GEP inhibits ADAMTS-7/ADAMTS-12-mediated digestion of COMP. The role of GEP in inhibiting TNFalpha-induced ADAMTS-7/ADAMTS-12 expression and COMP degradation in cartilage explants was also analyzed. RESULTS GEP bound directly to ADAMTS-7 and ADAMTS-12 in vitro and in chondrocytes, and the 4 C-terminal thrombospondin motifs of ADAMTS-7/ADAMTS-12 and each granulin unit of GEP mediated their interactions. Additionally, GEP colocalized with ADAMTS-7 and ADAMTS-12 on the cell surface of chondrocytes. More importantly, GEP inhibited COMP degradation by ADAMTS-7/ADAMTS-12 in a dose-dependent manner through 1) competitive inhibition through direct protein-protein interactions with ADAMTS-7/ADAMTS-12 and COMP, and 2) inhibition of TNFalpha-induced ADAMTS-7/ADAMTS-12 expression. Furthermore, GEP levels were significantly elevated in patients with either osteoarthritis or rheumatoid arthritis. CONCLUSION Our observations demonstrate a novel protein-protein interaction network between GEP, ADAMTS-7/ADAMTS-12, and COMP. Furthermore, GEP is a novel specific inhibitor of ADAMTS-7/ADAMTS-12-mediated COMP degradation and may play a significant role in preventing the destruction of joint cartilage in arthritis.
Collapse
Affiliation(s)
- Fengjin Guo
- New York University School of Medicine, New York, New York 10003, USA
| | | | | | | | | | | |
Collapse
|
28
|
Wang WX, Wilfred BR, Madathil SK, Tang G, Hu Y, Dimayuga J, Stromberg AJ, Huang Q, Saatman KE, Nelson PT. miR-107 regulates granulin/progranulin with implications for traumatic brain injury and neurodegenerative disease. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:334-45. [PMID: 20489155 PMCID: PMC2893676 DOI: 10.2353/ajpath.2010.091202] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/12/2010] [Indexed: 11/20/2022]
Abstract
Granulin (GRN, or progranulin) is a protein involved in wound repair, inflammation, and neoplasia. GRN has also been directly implicated in frontotemporal dementia and may contribute to Alzheimer's disease pathogenesis. However, GRN regulation expression is poorly understood. A high-throughput experimental microRNA assay showed that GRN is the strongest target for miR-107 in human H4 neuroglioma cells. miR-107 has been implicated in Alzheimer's disease pathogenesis, and sequence elements in the open reading frame-rather than the 3' untranslated region-of GRN mRNA are recognized by miR-107 and are highly conserved among vertebrate species. To better understand the mechanism of this interaction, FLAG-tagged Argonaute constructs were used following miR-107 transfection. GRN mRNA interacts preferentially with Argonaute 2. In vitro and in vivo studies indicate that regulation of GRN by miR-107 may be functionally important. Glucose supplementation in cultured cells that leads to increased miR-107 levels also results in decreased GRN expression, including changes in cell compartmentation and decreased secretion of GRN protein. This effect was eliminated following miR-107 transfection. We also tested a mouse model where miR-107 has been shown to be down-regulated. In brain tissue subjacent to 1.0 mm depth controlled cortical impact, surviving hippocampal neurons show decreased miR-107 with augmentation of neuronal GRN expression. These findings indicate that miR-107 contributes to GRN expression regulation with implications for brain disorders.
Collapse
Affiliation(s)
- Wang-Xia Wang
- Department of Pathology, Division of Neuropathology, and the Sanders-Brown Center on Aging, Rm 311, Sanders-Brown Center, 800 S. Limestone, University of Kentucky, Lexington, KY 40536-0230, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sleegers K, Brouwers N, Van Broeckhoven C. Role of progranulin as a biomarker for Alzheimer's disease. Biomark Med 2010; 4:37-50. [PMID: 20387302 DOI: 10.2217/bmm.09.82] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Serum or plasma progranulin (GRN) is a highly accurate of GRN-related frontotemporal lobar degeneration, which is caused by loss-of-function mutations in the GRN gene. Both null mutations and missense mutations in GRN have also been observed in patients with Alzheimer's disease. Here, the evidence for a role of circulating GRN as a biochemical biomarker in neurodegeneration is reviewed, with a specific focus on its relevance in Alzheimer's disease. We conclude that circulating GRN is a promising, nonintrusive biomarker that warrants screening in both patients with dementia of the Alzheimer type and people with mild cognitive impairment; specifically for, but not limited to, those that have a positive family history of neurodegenerative disease. Once a cure for GRN-related neurodegeneration becomes available, this biomarker will be an important tool in the effort to personalize treatment of dementia.
Collapse
|
30
|
Hoque M, Mathews MB, Pe'ery T. Progranulin (granulin/epithelin precursor) and its constituent granulin repeats repress transcription from cellular promoters. J Cell Physiol 2010; 223:224-33. [PMID: 20054825 DOI: 10.1002/jcp.22031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Progranulin (also known as granulin/epithelin precursor, GEP) is composed of seven granulin/epithelin repeats (granulins) and functions both as a full-length protein and as individual granulins. It is a secretory protein but a substantial amount of GEP is found inside cells, some in complexes with positive transcription elongation factor b (P-TEFb). GEP and certain granulins interact with the cyclin T1 subunit of P-TEFb, and with its HIV-1 Tat co-factor, leading to repression of transcription from the HIV promoter. We show that GEP lacking the signal peptide (GEPspm) remains inside cells and, like wild-type GEP, interacts with cyclin T1 and Tat. GEPspm represses transcription from the HIV-1 promoter at the RNA level. Granulins that bind cyclin T1 are phosphorylated by P-TEFb in vivo and in vitro on serine residues. GEPspm and those granulins that interact with cyclin T1 also inhibit transcription from cellular cad and c-myc promoters, which are highly dependent on P-TEFb, but not from the PCNA promoter. In addition, GEPspm and granulins repress transcriptional activation by VP16 or c-Myc, proteins that bind and recruit P-TEFb to responsive promoters. These data suggest that intracellular GEP is a promoter-specific transcriptional repressor that modulates the function of cellular and viral transcription factors.
Collapse
Affiliation(s)
- Mainul Hoque
- Department of Biochemistry and Molecular Biology, UMDNJ-New Jersey Medical School, Newark, New Jersey, USA
| | | | | |
Collapse
|
31
|
Feng JQ, Guo FJ, Jiang BC, Zhang Y, Frenkel S, Wang DW, Tang W, Xie Y, Liu CJ. Granulin epithelin precursor: a bone morphogenic protein 2-inducible growth factor that activates Erk1/2 signaling and JunB transcription factor in chondrogenesis. FASEB J 2010; 24:1879-92. [PMID: 20124436 DOI: 10.1096/fj.09-144659] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Granulin epithelin precursor (GEP) has been implicated in development, tissue regeneration, tumorigenesis, and inflammation. Herein we report that GEP stimulates chondrocyte differentiation from mesenchymal stem cells in vitro and endochondral ossification ex vivo, and GEP-knockdown mice display skeleton defects. Similar to bone morphogenic protein (BMP) 2, application of the recombinant GEP accelerates rabbit cartilage repair in vivo. GEP is a key downstream molecule of BMP2, and it is required for BMP2-mediated chondrocyte differentiation. We also show that GEP activates chondrocyte differentiation through Erk1/2 signaling and that JunB transcription factor is one of key downstream molecules of GEP in chondrocyte differentiation. Collectively, these findings reveal a novel critical role of GEP growth factor in chondrocyte differentiation and the molecular events both in vivo and in vitro.
Collapse
Affiliation(s)
- Jian Q Feng
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M University System Health Science Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The growth factor progranulin (PGRN) regulates cell division, survival, and migration. PGRN is an extracellular glycoprotein bearing multiple copies of the cysteine-rich granulin motif. With PGRN family members in plants and slime mold, it represents one of the most ancient of the extracellular regulatory proteins still extant in modern animals. PRGN has multiple biological roles. It contributes to the regulation of early embryogenesis, to adult tissue repair and inflammation. Elevated PGRN levels often occur in cancers, and PGRN immunotherapy inhibits the growth of hepatic cancer xenografts in mice. Recent studies have demonstrated roles for PGRN in neurobiology. An autosomal dominant mutation in GRN, the gene for PGRN, leads to neuronal atrophy in the frontal and temporal lobes, resulting in the disease frontotemporal lobar dementia. In this review we will discuss current knowledge of the multifaceted biology of PGRN.
Collapse
Affiliation(s)
- Andrew Bateman
- Endocrine Research Laboratory, McGill University Health Centre, Royal Victoria Hospital, Montreal, Canada.
| | | |
Collapse
|
33
|
Lovat F, Bitto A, Xu SQ, Fassan M, Goldoni S, Metalli D, Wubah V, McCue P, Serrero G, Gomella LG, Baffa R, Iozzo RV, Morrione A. Proepithelin is an autocrine growth factor for bladder cancer. Carcinogenesis 2009; 30:861-8. [PMID: 19237611 DOI: 10.1093/carcin/bgp050] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The growth factor proepithelin functions as an important regulator of proliferation and motility. Proepithelin is overexpressed in a great variety of cancer cell lines and clinical specimens of breast, ovarian and renal cancer, as well as glioblastomas. Using recombinant proepithelin on 5637 transitional cell carcinoma-derived cells, we have shown previously that proepithelin plays a critical role in bladder cancer by promoting motility of bladder cancer cells. In this study, we used the ONCOMINE database and gene microarray analysis tool to analyze proepithelin expression in several bladder cancer microarray studies. We found a statistically significant increase in proepithelin messenger RNA expression in bladder cancers vis-à-vis non-neoplastic tissues, and this was associated with pathologic and prognostic parameters. Targeted downregulation of proepithelin in T24 transitional carcinoma cells with small hairpin RNA inhibited both Akt and mitogen-activated protein kinase pathways, severely reduced the ability of T24 cells to proliferate in the absence of serum and inhibited migration, invasion and wound healing. In support of these in vitro results, we discovered that proepithelin expression was significantly upregulated in invasive bladder cancer tissues compared with normal urothelium. In addition, proepithelin was secreted in the urine, where it was detectable by immunoblotting and enzyme-linked immunosorbent assay. Collectively, these results support the hypothesis that proepithelin may play a critical role as an autocrine growth factor in the establishment and progression of bladder cancer and suggest that proepithelin may prove a novel biomarker for the diagnosis and prognosis of bladder neoplasms.
Collapse
Affiliation(s)
- Francesca Lovat
- Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Monami G, Emiliozzi V, Bitto A, Lovat F, Xu SQ, Goldoni S, Fassan M, Serrero G, Gomella LG, Baffa R, Iozzo RV, Morrione A. Proepithelin regulates prostate cancer cell biology by promoting cell growth, migration, and anchorage-independent growth. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1037-47. [PMID: 19179604 DOI: 10.2353/ajpath.2009.080735] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The growth factor proepithelin has recently emerged as an important regulator of transformation in several physiological and pathological systems. In this study, we determined the biological roles of proepithelin in prostate cancer cells using purified human recombinant proepithelin as well as proepithelin-depletion strategies. Proepithelin promoted the migration of androgen-dependent and -independent human prostate cancer cells; androgen-independent DU145 cells were the more responsive. In these cells, proepithelin additionally stimulated wound closure, invasion, and promotion of cell growth in vitro. These effects required the activation of both the Akt and mitogen-activated protein kinase pathways. We have analyzed proepithelin expression levels in different available prostate cancer microarray studies using the Oncomine database and found a statistically significant increase in proepithelin mRNA expression levels in prostate cancers compared with nonneoplastic controls. Notably, depletion of endogenous proepithelin by siRNA and antisense strategies impaired the ability of DU145 cells to grow and migrate after serum withdrawal and inhibited anchorage-independent growth. Our results provide the first evidence for a role of proepithelin in stimulating the migration, invasion, proliferation, and anchorage-independent growth of prostate cancer cells. This study supports the hypothesis that proepithelin may play a critical role as an autocrine growth factor in the establishment and initial progression of prostate cancer. Furthermore, proepithelin may prove to be a useful clinical marker for the diagnosis of prostate tumors.
Collapse
Affiliation(s)
- Giada Monami
- Department of Urology, Thomas Jefferson University, 233 South 10th St., BLSB Room 620, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gijselinck I, Van Broeckhoven C, Cruts M. Granulin mutations associated with frontotemporal lobar degeneration and related disorders: an update. Hum Mutat 2009; 29:1373-86. [PMID: 18543312 DOI: 10.1002/humu.20785] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the gene encoding granulin (HUGO gene symbol GRN, also referred to as progranulin, PGRN), located at chromosome 17q21, were recently linked to tau-negative ubiquitin-positive frontotemporal lobar degeneration (FTLDU). Since then, 63 heterozygous mutations were identified in 163 families worldwide, all leading to loss of functional GRN, implicating a haploinsufficiency mechanism. Together, these mutations explained 5 to 10% of FTLD. The high mutation frequency, however, might still be an underestimation because not all patient samples were examined for all types of loss-of-function mutations and because several variants, including missense mutations, have a yet uncertain pathogenic significance. Although the complete phenotypic spectrum associated with GRN mutations is not yet fully characterized, it was shown that it is highly heterogeneous, suggesting the influence of modifying factors. A role of GRN in neuronal survival was suggested but the exact mechanism by which neurodegeneration and deposition of pathologic brain inclusions occur still has to be clarified.
Collapse
Affiliation(s)
- I Gijselinck
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, Flanders Institute for Biotechnology (VIB), Antwerpen, Belgium
| | | | | |
Collapse
|
36
|
Tolkatchev D, Malik S, Vinogradova A, Wang P, Chen Z, Xu P, Bennett HPJ, Bateman A, Ni F. Structure dissection of human progranulin identifies well-folded granulin/epithelin modules with unique functional activities. Protein Sci 2008; 17:711-24. [PMID: 18359860 DOI: 10.1110/ps.073295308] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Progranulin is a secreted protein with important functions in several physiological and pathological processes, such as embryonic development, host defense, and wound repair. Autosomal dominant mutations in the progranulin gene cause frontotemporal dementia, while overexpression of progranulin promotes the invasive progression of a range of tumors, including those of the breast and the brain. Structurally, progranulin consists of seven-and-a-half tandem repeats of the granulin/epithelin module (GEM), several of which have been isolated as discrete 6-kDa GEM peptides. We have expressed all seven human GEMs using recombinant DNA in Escherichia coli. High-resolution NMR showed that only the three GEMs, hGrnA, hGrnC, and hGrnF, contain relatively well-defined three-dimensional structures in solution, while others are mainly mixtures of poorly structured disulfide isomers. The three-dimensional structures of hGrnA, hGrnC, and hGrnF contain a stable stack of two beta-hairpins in their N-terminal subdomains, but showed a more flexible C-terminal subdomain. Interestingly, of the well-structured GEMs, hGrnA demonstrated potent growth inhibition of a breast cancer cell line, while hGrnF was stimulatory. Poorly folded peptides were either weakly inhibitory or without activity. The functionally active and structurally well-characterized human hGrnA offers a unique opportunity for detailed structure-function studies of these important GEM proteins as novel members of mammalian growth factors.
Collapse
Affiliation(s)
- Dmitri Tolkatchev
- Bio-NMR and Protein Research Laboratory, Biotechnology Research Institute, National Research Council of Canada, Montreal, Quebec H4P 2R2, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Expression of PC cell-derived growth factor and vascular endothelial growth factor in esophageal squamous cell carcinoma and their clinicopathologic significance. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200805020-00004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
38
|
Guerra RR, Kriazhev L, Hernandez-Blazquez FJ, Bateman A. Progranulin is a stress-response factor in fibroblasts subjected to hypoxia and acidosis. Growth Factors 2007; 25:280-5. [PMID: 18092235 DOI: 10.1080/08977190701781222] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The growth factor progranulin (granulin-epithelin precursor, PC-derived growth factor or acrogranin) regulates proliferation and migration and is implicated in cancer, development, wound repair and neurodegenerative diseases. Under most conditions fibroblasts do not express progranulin in vivo, however its expression is activated following wounding. We hypothesised that progranulin is part of a fibroblast stress response. Fibroblasts in culture were exposed to two physiologically and clinically relevant microenvironmental stresses; hypoxia (1% oxygen) and acidosis, both of which increase progranulin expression. The greatest increases occurred when hypoxia and acidosis were combined. Increased progranulin expression is not a direct response to apoptosis since it occurred under conditions of pH and hypoxia under which cell viability remained high. Low concentrations of progranulin (2 nM) protected fibroblasts from apoptosis induced by extreme acidosis (pH 5.0 and 4.0). We propose that progranulin is part of a fibroblast stress response and is cytoprotective to acidotic stress.
Collapse
Affiliation(s)
- Ricardo R Guerra
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, Sao Paulo University, São Paulo, Brazil
| | | | | | | |
Collapse
|
39
|
Kumar-Singh S, Van Broeckhoven C. Frontotemporal lobar degeneration: current concepts in the light of recent advances. Brain Pathol 2007; 17:104-14. [PMID: 17493044 PMCID: PMC8095552 DOI: 10.1111/j.1750-3639.2007.00055.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Work done over the past decade has led to a molecular understanding of frontotemporal lobar degeneration (FTLD), a deadly disease that afflicts patients in mid-life. It is a common cause of dementia, second only to Alzheimer's disease in the population below 65 years of age. Neuroanatomical and neurobiological substrates have been identified for the three major subtypes of FTLD and these discoveries have broadened the FTLD spectrum to include amyotrophic lateral sclerosis (ALS). Mutations in MAPT were found to cause frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), a familial disorder with filamentous tau inclusions in nerve cells and glial cells. FTDP-17 can result in clinical syndromes that closely resemble progressive supranuclear palsy, corticobasal degeneration and Pick's disease. More recently, mutations in three genes (VCP, CHMP2B and PGRN) have been found to cause FTLD with ubiquitin-positive, tau-negative neuronal inclusions (FTLD-U). They explain a large proportion of inherited FTLD-U. It remains to be seen whether dementia lacking distinctive histopathology (DLDH) constitutes a third disease category, as many of these cases are now being reclassified as FTLD-U. Recently, TAR DNA-binding protein-43 (TDP-43) has been identified as a key protein of the ubiquitin inclusions of FTLD-U and ALS. Thus, for familial forms of FTLD and related disorders, we now know the primary etiologies and accumulating proteins. These findings are pivotal for dissecting the pathways by which different etiologies lead to the varied clinicopathological presentations of FTLD.
Collapse
Affiliation(s)
- Samir Kumar-Singh
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, Laboratory of Neurogenetics, VIB, Institute Born-Bunge and University of Antwerp, BE-2610 Antwerpen, Belgium.
| | | |
Collapse
|
40
|
Xu K, Zhang Y, Ilalov K, Carlson CS, Feng JQ, Di Cesare PE, Liu CJ. Cartilage oligomeric matrix protein associates with granulin-epithelin precursor (GEP) and potentiates GEP-stimulated chondrocyte proliferation. J Biol Chem 2007; 282:11347-55. [PMID: 17307734 DOI: 10.1074/jbc.m608744200] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mutations in human cartilage oligomeric matrix protein (COMP) have been linked to the development of pseudoachondroplasia and multiple epiphyseal dysplasia; however, the functions of both wild-type and mutant COMP in the skeletogenesis remain unknown. In an effort to define the biological functions of COMP, a functional genetic screen based on the yeast two-hybrid system was performed. This led to the identification of granulin-epithelin precursor (GEP), an autocrine growth factor, as a COMP-associated partner. COMP directly binds to GEP both in vitro and in vivo, as revealed by in vitro pull down and co-immunoprecipitation assays. GEP selectively interacts with the epidermal growth factor repeat domain of COMP but not with the other three functional domains of COMP. The granulin A repeat unit of GEP is required and sufficient for association with COMP. COMP co-localizes with GEP predominantly in the pericellular matrix of transfected rat chondrosarcoma cell and primary human chondrocytes. Staining of musculoskeletal tissues of day 19 mouse embryo with antibodies to GEP is restricted to chondrocytes in the lower proliferative and upper hypertrophic zones. Overexpression of GEP stimulates the proliferation of chondrocytes, and this stimulation is enhanced by COMP. In addition, COMP appears to be required for GEP-mediated chondrocyte proliferation, since chondrocyte proliferation induced by GEP is dramatically inhibited by an anti-COMP antibody. These findings provide the first evidence linking the association of COMP and GEP and identifying a previously unrecognized growth factor (i.e. GEP) in cartilage.
Collapse
Affiliation(s)
- Ke Xu
- Department of Orthopaedic Surgery, New York University Hospital for Joint Diseases, New York, New York 10003, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Inhibition of PC cell-derived growth factor (PCDGF)/granulin-epithelin precursor (GEP) decreased cell proliferation and invasion through downregulation of cyclin D and CDK4 and inactivation of MMP-2. BMC Cancer 2007; 7:22. [PMID: 17261172 PMCID: PMC1794415 DOI: 10.1186/1471-2407-7-22] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 01/29/2007] [Indexed: 11/10/2022] Open
Abstract
Background PC cell-derived growth factor (PCDGF), also called epithelin/granulin precursor (GEP), is an 88-kDa secreted glycoprotein with the ability to stimulate cell proliferation in an autocrine fashion. In addition, some studies indicated that PCDGF participated in invasion, metastasis and survival of cancer cells by regulating cell migration, adhesion and proliferation. Yet the effects of PCDGF on proliferation and invasion of ovarian cancer cells in vitro and the mechanisms by which PCDGF mediates biological behaviors of ovarian cancer have rarely been reported. In the present study we investigated whether and how PCDGF/GEP mediated cell proliferation and invasion in ovarian cancer. Methods PCDGF/GEP expression level in three human ovarian cancer cell lines of different invasion potential were detected by RT-PCR and western blot. Effects of inhibition of PCDGF expression on cell proliferation and invasion capability were determined by MTT assay and Boyden chamber assay. Expression levels of cyclin D1 and CDK4 and MMP-2 activity were evaluated in a pilot study. Results PCDGF mRNA and protein were expressed at a high level in SW626 and A2780 and at a low level in SKOV3. PCDGF expression level correlated well with malignant phenotype including proliferation and invasion in ovarian cancer cell lines. In addition, the proliferation rate and invasion index decreased after inhibition of PCDGF expression by antisense PCDGF cDNA transfection in SW626 and A2780. Furthermore expression of CyclinD1 and CDK4 were downregulated and MMP-2 was inactivated after PCDGF inhibition in the pilot study. Conclusion PCDGF played an important role in stimulating proliferation and promoting invasion in ovarian cancer. Inhibition of PCDGF decreased proliferation and invasion capability through downregulation of cyclin D1 and CDK4 and inactivation of MMP-2. PCDGF could serve as a potential therapeutic target in ovarian cancer.
Collapse
|
42
|
Chiba S, Suzuki M, Yamanouchi K, Nishihara M. Involvement of Granulin in Estrogen-Induced Neurogenesis in the Adult Rat Hippocampus. J Reprod Dev 2007; 53:297-307. [PMID: 17179653 DOI: 10.1262/jrd.18108] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies have demonstrated the presence of neurogenesis in the adult mammalian hippocampus, and it has been suggested that estrogen and various growth factors influence the processes of adult neurogenesis. The present study assessed cell proliferation in the dentate gyrus and the mRNA expression levels of granulin, insulin-like growth factor-I (IGF-I), and brain-derived neurotrophic factor (BDNF) in the hippocampus 4 h after treatment with estradiol benzoate (EB) in 3- and 12-month old ovariectomized rats. At 3 months of age, mRNA expression of granulin precursor and cell proliferation were increased by EB treatment, although the mRNA expressions of IGF-I and BDNF remained unchanged. At 12 months of age, however, neither mRNA expression of the three genes nor cell proliferation in the dentate gyrus were affected by EB treatment. In addition, 17beta-estradiol enhanced the proliferation of neural progenitor cells derived from hippocampal tissue of 3-month-old female rats in vitro; this was inhibited by neutralization of granulin with specific antibody. These results suggest that estrogen induces granulin gene expression in the hippocampus and that the product of this gene is involved in the mitogenic effects of estrogen in the dentate gyrus, although the responses to estrogen decline with age.
Collapse
Affiliation(s)
- Shuichi Chiba
- Department of Veterinary Physiology, Veterinary Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
43
|
Monami G, Gonzalez EM, Hellman M, Gomella LG, Baffa R, Iozzo RV, Morrione A. Proepithelin promotes migration and invasion of 5637 bladder cancer cells through the activation of ERK1/2 and the formation of a paxillin/FAK/ERK complex. Cancer Res 2006; 66:7103-10. [PMID: 16849556 DOI: 10.1158/0008-5472.can-06-0633] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The growth factor proepithelin (also known as progranulin, acrogranin, PC-derived growth factor, or granulin-epithelin precursor) is a secreted glycoprotein that functions as an important regulator of cell growth, migration, and transformation. Proepithelin is overexpressed in a great variety of cancer cell lines and clinical specimens of breast, ovarian, and renal cancer as well as glioblastomas. In this study, we have investigated the effects of proepithelin on bladder cancer cells using human recombinant proepithelin purified to homogeneity from 293-EBNA cells. Although proepithelin did not appreciably affect cell growth, it did promote migration of 5637 bladder cancer cells and stimulate in vitro wound closure and invasion. These effects required the activation of the mitogen-activated protein kinase pathway and paxillin, which upon proepithelin stimulation formed a complex with focal adhesion kinase and active extracellular signal-regulated kinase. Our results provide the first evidence for a role of proepithelin in stimulating migration and invasion of bladder cancer cells, and support the hypothesis that this growth factor may play a critical role in the establishment of the invasive phenotype.
Collapse
Affiliation(s)
- Giada Monami
- Department of Urology, Anatomy and Cell Biology and Cellular Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Jones MB, Houwink AP, Freeman BK, Greenwood TM, Lafky JM, Lingle WL, Berchuck A, Maxwell GL, Podratz KC, Maihle NJ. The granulin-epithelin precursor is a steroid-regulated growth factor in endometrial cancer. ACTA ACUST UNITED AC 2006; 13:304-11. [PMID: 16697948 DOI: 10.1016/j.jsgi.2006.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2005] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The majority of endometrial cancers arise as a result of estrogen stimulation, the molecular targets of which remain incompletely defined. We hypothesize that the granulin-epithelin precursor (GEP) may be one such target. In this study, we examined the frequency of GEP and estrogen receptor (ER) co-expression in human endometrial cancers. Once we established the co-expression of GEP with the estrogen receptor we examined the potential estrogen regulation of GEP expression, as well as the functional significance of GEP expression in vitro. METHODS Double immunofluorescence and confocal microscopy were used to compare GEP and ER expression among 41 endometrial cancers. The effects of estradiol and tamoxifen treatment on GEP expression in two endometrial cancer cell lines, KLE and HEC-1-A, were assessed through reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis. The antiproliferative effect of GEP silencing by short hairpin (sh)RNA, was evaluated in HEC-1-A cells using an MTT assay. RESULTS GEP co-expression with ER was observed in 63% of the cancers examined. A two- to fivefold increase in GEP expression with estradiol and/or tamoxifen treatment was observed in KLE cells. Silencing of GEP in HEC-1-A cells using shRNA resulted in a decrease in proliferation among transfected cells. CONCLUSIONS Co-expression of GEP and ER in endometrial cancer cells, and the regulation of GEP by estrogen, suggests a role for GEP in steroid-mediated endometrial cancer cell growth. Further characterization of GEP as a steroid-mediated growth factor in these cells may broaden our understanding of endometrial cancer biology and also provide guidance in the development of novel therapeutic targets.
Collapse
|
45
|
Hanington PC, Barreda DR, Belosevic M. A Novel Hematopoietic Granulin Induces Proliferation of Goldfish (Carassius auratus L.) Macrophages. J Biol Chem 2006; 281:9963-70. [PMID: 16473876 DOI: 10.1074/jbc.m600631200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Granulins are a group of highly conserved growth factors that have been described from a variety of organisms spanning the metazoa. In this study, goldfish granulin was one of the most commonly identified transcripts in the differential cross-screening of macrophage cDNA libraries and was preferentially expressed in proliferating macrophages. Unlike mammalian granulins, which possess 7.5 repeats of a characteristic signature of 12 cysteine residues, the goldfish granulin encoded a putative peptide possessing only 1.5 cysteine repeats. Northern blot and real-time PCR analyses indicated that goldfish granulin was expressed only in the hematopoietic tissues of the goldfish, specifically the kidney and spleen, and in activated peripheral blood mononuclear cells. We expressed granulin using a prokaryotic expression system and produced an affinity-purified rabbit anti-goldfish granulin IgG. Recombinant goldfish granulin induced a dose-dependent proliferative response of goldfish macrophages that was inversely related to the myeloid differentiation stage of the cells studied. The highest proliferative response was observed in macrophage progenitor cells and monocytes. This proliferative response of macrophages was abrogated by the addition of anti-granulin IgG. These results indicate that goldfish granulin is a growth factor that positively modulates cell proliferation at distinct junctures of macrophage differentiation.
Collapse
Affiliation(s)
- Patrick C Hanington
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta T6G E9, Canada
| | | | | |
Collapse
|
46
|
Matsumura N, Mandai M, Miyanishi M, Fukuhara K, Baba T, Higuchi T, Kariya M, Takakura K, Fujii S. Oncogenic Property of Acrogranin in Human Uterine Leiomyosarcoma: Direct Evidence of Genetic Contribution inIn vivoTumorigenesis. Clin Cancer Res 2006; 12:1402-11. [PMID: 16533762 DOI: 10.1158/1078-0432.ccr-05-2003] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To identify potential oncogenes that contribute to the development of uterine leiomyosarcoma, we conducted a cDNA microarray analysis between normal uterine smooth muscle and uterine leiomyosarcoma. We found that acrogranin (also named PCDGF or progranulin) is overexpressed in uterine leiomyosarcoma. With immunohistochemical staining of 12 leiomyosarcoma cases, we verified acrogranin expression in tumor cells. Furthermore, the intensity of acrogranin expression correlated with high histologic grade and poor prognosis. To directly analyze the oncogenic properties of acrogranin, we established an immortalized uterine smooth muscle cell line by transfection of human telomerase reverse transcriptase into primary culture. This cell line retained the original characteristics of uterine smooth muscle cells, including spindle-shaped extension as well as expression of vimentin, estrogen receptor alpha, progesterone receptor, and alpha smooth muscle actin. Transfection of acrogranin into the immortalized uterine smooth muscle cells resulted in colony formation in soft agar, but the diameter of the colonies did not exceed 100 mum. Transfection of both acrogranin and SV40 early region (SV40ER) into the immortalized uterine smooth muscle cells resulted in an increased number of colonies and increased colony size in soft agar versus transfection of SV40ER alone. We show that only immortalized uterine smooth muscle cells expressing both acrogranin and SV40ER are capable of tumor formation in nude mice. Thus, acrogranin is overexpressed in uterine leiomyosarcoma cells, particularly in high-grade cases, and forced expression of acrogranin in immortalized uterine smooth muscle cells contributes to malignant transformation, which suggest that acrogranin plays an important role in the pathogenesis of uterine leiomyosarcoma.
Collapse
Affiliation(s)
- Noriomi Matsumura
- Department of Gynecology and Obstetrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cadieux B, Chitramuthu BP, Baranowski D, Bennett HPJ. The zebrafish progranulin gene family and antisense transcripts. BMC Genomics 2005; 6:156. [PMID: 16277664 PMCID: PMC1310530 DOI: 10.1186/1471-2164-6-156] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Accepted: 11/08/2005] [Indexed: 11/10/2022] Open
Abstract
Background Progranulin is an epithelial tissue growth factor (also known as proepithelin, acrogranin and PC-cell-derived growth factor) that has been implicated in development, wound healing and in the progression of many cancers. The single mammalian progranulin gene encodes a glycoprotein precursor consisting of seven and one half tandemly repeated non-identical copies of the cystine-rich granulin motif. A genome-wide duplication event hypothesized to have occurred at the base of the teleost radiation predicts that mammalian progranulin may be represented by two co-orthologues in zebrafish. Results The cDNAs encoding two zebrafish granulin precursors, progranulins-A and -B, were characterized and found to contain 10 and 9 copies of the granulin motif respectively. The cDNAs and genes encoding the two forms of granulin, progranulins-1 and -2, were also cloned and sequenced. Both latter peptides were found to be encoded by precursors with a simplified architecture consisting of one and one half copies of the granulin motif. A cDNA encoding a chimeric progranulin which likely arises through the mechanism of trans-splicing between grn1 and grn2 was also characterized. A non-coding RNA gene with antisense complementarity to both grn1 and grn2 was identified which may have functional implications with respect to gene dosage, as well as in restricting the formation of the chimeric form of progranulin. Chromosomal localization of the four progranulin (grn) genes reveals syntenic conservation for grna only, suggesting that it is the true orthologue of mammalian grn. RT-PCR and whole-mount in situ hybridization analysis of zebrafish grns during development reveals that combined expression of grna and grnb, but not grn1 and grn2, recapitulate many of the expression patterns observed for the murine counterpart. This includes maternal deposition, widespread central nervous system distribution and specific localization within the epithelial compartments of various organs. Conclusion In support of the duplication-degeneration-complementation model of duplicate gene retention, partitioning of expression between grna and grnb was observed in the intermediate cell mass and yolk syncytial layer, respectively. Taken together these expression patterns suggest that the function of an ancestral grn gene has been devolved upon four paralogues in zebrafish.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- Blotting, Northern
- Chromatography, High Pressure Liquid
- Chromosome Mapping
- Cloning, Molecular
- DNA, Complementary/metabolism
- Gene Dosage
- Gene Expression Regulation, Developmental
- Gene Library
- Humans
- In Situ Hybridization
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Intercellular Signaling Peptides and Proteins/genetics
- Models, Genetic
- Molecular Sequence Data
- Multigene Family
- Oligonucleotides, Antisense/chemistry
- Oligonucleotides, Antisense/genetics
- Phylogeny
- RNA/metabolism
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Amino Acid
- Tissue Distribution
- Transcription, Genetic
- Zebrafish
- Zebrafish Proteins/biosynthesis
- Zebrafish Proteins/chemistry
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Benoît Cadieux
- Endocrine Laboratory, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Cancer Research Institute, UCSF, 2340 Sutter Street, N-231 San Francisco, CA 94143, USA
| | - Babykumari P Chitramuthu
- Endocrine Laboratory, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - David Baranowski
- Endocrine Laboratory, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Hugh PJ Bennett
- Endocrine Laboratory, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Room L2.05, Royal Victoria Hospital, 687 Pine Avenue West, Montreal, Quebec, H3A 1A1, Canada
| |
Collapse
|
48
|
Hoque M, Tian B, Mathews MB, Pe'ery T. Granulin and granulin repeats interact with the Tat.P-TEFb complex and inhibit Tat transactivation. J Biol Chem 2005; 280:13648-57. [PMID: 15653695 DOI: 10.1074/jbc.m409575200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular positive transcription elongation factor b (P-TEFb), containing cyclin T1 and cyclin-dependent kinase 9 (CDK9), interacts with the human immunodeficiency virus, type 1 (HIV-1) regulatory protein Tat to enable viral transcription and replication. Cyclin T1 is an unusually long cyclin and is engaged by cellular regulatory proteins. Previous studies showed that the granulin/epithelin precursor (GEP) binds the histidine-rich region of cyclin T1 and inhibits P-TEFb function. GEP is composed of repeats that vary in sequence and properties. GEP also binds directly to Tat. Here we show that GEP and some of its constituent granulin repeats can inhibit HIV-1 transcription via Tat without directly binding to cyclin T1. The interactions of granulins with Tat and cyclin T1 differ with respect to their binding sites and divalent cation requirements, and we identified granulin repeats that bind differentially to Tat and cyclin T1. Granulins DE and E bind Tat but do not interact directly with cyclin T1. These granulins are present in complexes with Tat and P-TEFb in which Tat forms a bridge between the cellular proteins. Granulins DE and E repress transcription from the HIV-1 LTR and gene expression from the viral genome, raising the possibility of developing granulin-based inhibitors of viral infection.
Collapse
Affiliation(s)
- Mainul Hoque
- Department of Biochemistry and Molecular Biology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103-2714, USA
| | | | | | | |
Collapse
|
49
|
Pan CX, Kinch MS, Kiener PA, Langermann S, Serrero G, Sun L, Corvera J, Sweeney CJ, Li L, Zhang S, Baldridge LA, Jones TD, Koch MO, Ulbright TM, Eble JN, Cheng L. PC Cell-Derived Growth Factor Expression in Prostatic Intraepithelial Neoplasia and Prostatic Adenocarcinoma. Clin Cancer Res 2004; 10:1333-7. [PMID: 14977833 DOI: 10.1158/1078-0432.ccr-1123-03] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE PCDGF (PC cell-derived growth factor), also called progranulin, is a M(r) 88000 glycoprotein precursor of granulin. It is a novel growth factor that stimulates cell proliferation, confers epithelial tumorigenesis, and promotes tumor invasion. Here we investigate the potential of PCDGF as a therapeutic target for prostate cancer. EXPERIMENTAL DESIGN We studied the expression of PCDGF in invasive prostate cancer, adjacent high-grade prostatic intraepithelial neoplasia (PIN), and benign prostate tissue from 99 human prostate specimens. The level of PCDGF expression was correlated with various clinicopathological characteristics. RESULTS Normal prostate tissue did not express (53/99), or expressed low levels (46/99) of PCDGF. In the 46 normal prostate specimens that expressed PCDGF, most of them had less than 10% of cells expressing PCDGF. PCDGF expression could be detected in more than 50% of cells in all specimens of PIN and invasive prostate cancer. The expression of PCDGF in normal prostate tissue was much less intense and in a smaller fraction of cells than in PIN and invasive adenocarcinoma (P < 0.0001). There was no correlation of PCDGF expression with age, Gleason score, pathological stage, status of lymph node metastasis, extraprostatic extension, perineural invasion, surgical margins, and vascular invasion. CONCLUSIONS Our data suggest that the induction of PCDGF expression occurs during the development of PIN. PCDGF may be a new molecular target for the treatment and prevention of prostate cancer.
Collapse
Affiliation(s)
- Chong-Xian Pan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The type 1 insulin-like growth factor receptor (IGF-1R) plays an important role in the establishment and maintenance of the transformed phenotype. It also has a strong antiapoptotic activity and has a significant influence on the control of cell and body size. Downregulation of the IGF-1R leads to massive apoptosis of cancer cells. These characteristics make it an attractive target for anticancer therapy.
Collapse
Affiliation(s)
- Renato Baserga
- Kimmel Cancer Center, Thomas Jefferson University, 233 South 10th Street, 624 BLSB, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|