1
|
Sha S, Liu Y, Xie W, Li H, Jing Z, Yin Q, Liu S, Zhao H, Liu W. CREG1 alleviates bone loss in osteoporosis by enhancing the osteogenic differentiation of BMSCs through mitophagy. Int Immunopharmacol 2025; 158:114867. [PMID: 40378431 DOI: 10.1016/j.intimp.2025.114867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/23/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
The pathological process of osteoporosis involves accelerated bone resorption and a decline in bone formation, among which the disruption of the balance between adipogenic and osteogenic differentiation in bone marrow mesenchymal stem cells (BMSCs) is a crucial part. Cellular repressor of E1A-stimulated genes 1 (CREG1), a small glycoprotein, is mainly localized to the endosomal-lysosomal compartment and is associated with the regulation of mitophagy and cell differentiation. However, its roles in BMSCs osteogenic differentiation and skeletal degenerative disorders, including osteoporosis, are poorly understood. We previously identified CREG1 as being highly expressed in the bone marrow through database analysis and found that its expression increased in the process of BMSCs osteogenic differentiation. In the present study, we demonstrated that the expression of CREG1 was reduced in osteoporosis patients and animal models, and the overexpression of CREG1 contributed to higher bone mass compared with ovariectomy (OVX)-induced bone loss models. Further research revealed that the knockdown of CREG1 inhibited the osteogenic differentiation of BMSCs, while CREG1 overexpression promoted this process. Additionally, we found that CREG1 overexpression was accompanied by an increase in mitophagy levels, and the osteogenic differentiation induced by this overexpression was blocked when mitophagy was inhibited, indicating that CREG1 promoted osteogenic differentiation through inducing mitophagy. Therefore, our findings demonstrated that CREG1 is involved in regulating the osteogenic differentiation of BMSCs, thereby providing new therapeutic targets and pathways for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Shiyu Sha
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Yi Liu
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Weiping Xie
- Department of Rehabilitation Medicine, People's Hospital of Dingtao District, Heze, Shandong 274100, People's Republic of China
| | - Haoling Li
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Zhaocheng Jing
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Qingfeng Yin
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Shenghou Liu
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Heng Zhao
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Wenguang Liu
- Department of Joint Surgery/Sports Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China.
| |
Collapse
|
2
|
Grosjean N, Zhang L, Kumaran D, Xie M, Fahey A, Santiago K, Hu F, Regulski M, Blaby IK, Ware D, Blaby-Haas CE. Functional diversification within the heme-binding split-barrel family. J Biol Chem 2024; 300:107888. [PMID: 39395795 PMCID: PMC11602992 DOI: 10.1016/j.jbc.2024.107888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/29/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Due to neofunctionalization, a single fold can be identified in multiple proteins that have distinct molecular functions. Depending on the time that has passed since gene duplication and the number of mutations, the sequence similarity between functionally divergent proteins can be relatively high, eroding the value of sequence similarity as the sole tool for accurately annotating the function of uncharacterized homologs. Here, we combine bioinformatic approaches with targeted experimentation to reveal a large multifunctional family of putative enzymatic and nonenzymatic proteins involved in heme metabolism. This family (homolog of HugZ (HOZ)) is embedded in the "FMN-binding split barrel" superfamily and contains separate groups of proteins from prokaryotes, plants, and algae, which bind heme and either catalyze its degradation or function as nonenzymatic heme sensors. In prokaryotes these proteins are often involved in iron assimilation, whereas several plant and algal homologs are predicted to degrade heme in the plastid or regulate heme biosynthesis. In the plant Arabidopsis thaliana, which contains two HOZ subfamilies that can degrade heme in vitro (HOZ1 and HOZ2), disruption of AtHOZ1 (AT3G03890) or AtHOZ2A (AT1G51560) causes developmental delays, pointing to important biological roles in the plastid. In the tree Populus trichocarpa, a recent duplication event of a HOZ1 ancestor has resulted in localization of a paralog to the cytosol. Structural characterization of this cytosolic paralog and comparison to published homologous structures suggests conservation of heme-binding sites. This study unifies our understanding of the sequence-structure-function relationships within this multilineage family of heme-binding proteins and presents new molecular players in plant and bacterial heme metabolism.
Collapse
Affiliation(s)
- Nicolas Grosjean
- Biology Department, Brookhaven National Laboratory, Upton, New York, USA
| | - Lifang Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Desigan Kumaran
- Biology Department, Brookhaven National Laboratory, Upton, New York, USA
| | - Meng Xie
- Biology Department, Brookhaven National Laboratory, Upton, New York, USA
| | - Audrey Fahey
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Kassandra Santiago
- Biology Department, Brookhaven National Laboratory, Upton, New York, USA
| | - Fangle Hu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Michael Regulski
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Ian K Blaby
- Lawrence Berkeley National Laboratory, US Department of Energy Joint Genome Institute, Berkeley, California, USA
| | - Doreen Ware
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA; USDA ARS NEA Plant, Soil & Nutrition Laboratory Research Unit, Ithaca, New York, USA.
| | - Crysten E Blaby-Haas
- Lawrence Berkeley National Laboratory, US Department of Energy Joint Genome Institute, Berkeley, California, USA; The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
| |
Collapse
|
3
|
Han X, He W, Liang D, Liu X, Zhou J, de Thé H, Zhu J, Yuan H. Creg1 Regulates Erythroid Development via TGF-β/Smad2-Klf1 Axis in Zebrafish. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402804. [PMID: 38953462 PMCID: PMC11434009 DOI: 10.1002/advs.202402804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/04/2024] [Indexed: 07/04/2024]
Abstract
Understanding the regulation of normal erythroid development will help to develop new potential therapeutic strategies for disorders of the erythroid lineage. Cellular repressor of E1A-stimulated genes 1 (CREG1) is a glycoprotein that has been implicated in the regulation of tissue homeostasis. However, its role in erythropoiesis remains largely undefined. In this study, it is found that CREG1 expression increases progressively during erythroid differentiation. In zebrafish, creg1 mRNA is preferentially expressed within the intermediate cell mass (ICM)/peripheral blood island (PBI) region where primitive erythropoiesis occurs. Loss of creg1 leads to anemia caused by defective erythroid differentiation and excessive apoptosis of erythroid progenitors. Mechanistically, creg1 deficiency results in reduced activation of TGF-β/Smad2 signaling pathway. Treatment with an agonist of the Smad2 pathway (IDE2) could significantly restore the defective erythroid development in creg1-/- mutants. Further, Klf1, identified as a key target gene downstream of the TGF-β/Smad2 signaling pathway, is involved in creg1 deficiency-induced aberrant erythropoiesis. Thus, this study reveals a previously unrecognized role for Creg1 as a critical regulator of erythropoiesis, mediated at least in part by the TGF-β/Smad2-Klf1 axis. This finding may contribute to the understanding of normal erythropoiesis and the pathogenesis of erythroid disorders.
Collapse
Affiliation(s)
- Xiao Han
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Wenxin He
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Dongguo Liang
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Xiaohui Liu
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jun Zhou
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Hugues de Thé
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Université de Paris 7/INSERM/CNRS UMR 944/7212Equipe Labellisée Ligue Nationale Contre le CancerHôpital St. LouisParis75010France
| | - Jun Zhu
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Université de Paris 7/INSERM/CNRS UMR 944/7212Equipe Labellisée Ligue Nationale Contre le CancerHôpital St. LouisParis75010France
| | - Hao Yuan
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
4
|
Yuan N, Xiao L, Chen J, Liu B, Ren S, Sheng X, Qi X, Wang Y, Chen C, Guo K, Yang X, Yang L, Wang X. CREG1 promotes bovine placental trophoblast cells exosome release by targeting IGF2R and participates in regulating organoid differentiation via exosomes transport. Int J Biol Macromol 2024; 274:133298. [PMID: 38917918 DOI: 10.1016/j.ijbiomac.2024.133298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Placental exosomes are a kind of intercellular communication media secreted by placental cells during pregnancy, exosomogenesis and release are regulated by many secretory glycoproteins. CREG1 is a kind of secreted glycoprotein widely expressed in various organs and tissues of the body, which inhibits cell proliferation and enhances cell differentiation. The aim of this study was to explore the role of CREG1 in regulating exosomogenesis during the proliferation and differentiation of placental trophoblast cells in early pregnant dairy cows by targeting IGF2R and participating in regulating organoid differentiation via exosomes transport. METHODS Molecular biological methods were firstly used to investigate the expression patterns of CREG1, IGF2R and exosomal marker proteins in early placental development of pregnant dairy cows. Subsequently, the effects of CREG1 on the formation and release of bovine placental trophoblast (BTCs) derived exosomes by targeting IGF2R were investigated. Further, the effects of CREG1 on the change of gene expression patterns along with the transport of exosomes to recipient cells and participate in regulating the differentiation of organoids were explored. RESULTS The expression of CREG1, IGF2R and exosomal marker proteins increased with the increase of pregnancy months during the early evolution of placental trophoblast cells in dairy cows. Overexpression of Creg1 enhanced the genesis and release of exosomes derived from BTCs, while knocking down the expression of Igf2r gene not only inhibited the genesis of exosomes, but also inhibited the genesis and release of exosomes induced by overexpression of CREG1 protein. Interestingly, IGF2R can regulate the expression of CREG1 through reverse secretion. What's more, the occurrence and release of trophoblast-derived exosomes are regulated by CREG1 binding to IGF2R, which subsequently binds to Rab11. CREG1 can not only promote the formation and release of exosomes in donor cells, but also regulate the change of gene expression patterns along with the transport of exosomes to recipient cells and participate in regulating the early development of placenta. CONCLUSIONS Our study confirmed that CREG1 is involved in the exosomogenesis and release of exosomes during the proliferation and differentiation of placental trophoblast cells in early pregnant dairy cows by targeting IGF2R, and is involved in the regulation of organoid differentiation through exosome transport.
Collapse
Affiliation(s)
- Naihan Yuan
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Jiaxi Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Bingying Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Siqi Ren
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Yingqiu Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Chang Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Kaijun Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiaowen Yang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lin Yang
- Animal Epidemic prevention and Quarantine center, Huimin District, Hohhot, Inner Mongolia Autonomous Region 010030, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China.
| |
Collapse
|
5
|
Tan YW, Teo FMS, Ler SG, Alli-Shaik A, Nyo M, Chong CY, Tan NWH, Wang RYL, Gunaratne J, Chu JJH. Potential relevance of salivary legumain for the clinical diagnostic of hand, foot, and mouth disease. J Med Virol 2023; 95:e29243. [PMID: 38009231 DOI: 10.1002/jmv.29243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/28/2023]
Abstract
The fight against hand, foot, and mouth disease (HFMD) remains an arduous challenge without existing point-of-care (POC) diagnostic platforms for accurate diagnosis and prompt case quarantine. Hence, the purpose of this salivary biomarker discovery study is to set the fundamentals for the realization of POC diagnostics for HFMD. Whole salivary proteome profiling was performed on the saliva obtained from children with HFMD and healthy children, using a reductive dimethylation chemical labeling method coupled with high-resolution mass spectrometry-based quantitative proteomics technology. We identified 19 upregulated (fold change = 1.5-5.8) and 51 downregulated proteins (fold change = 0.1-0.6) in the saliva samples of HFMD patients in comparison to that of healthy volunteers. Four upregulated protein candidates were selected for dot blot-based validation assay, based on novelty as biomarkers and exclusions in oral diseases and cancers. Salivary legumain was validated in the Singapore (n = 43 healthy, 28 HFMD cases) and Taiwan (n = 60 healthy, 47 HFMD cases) cohorts with an area under the receiver operating characteristic curve of 0.7583 and 0.8028, respectively. This study demonstrates the feasibility of a broad-spectrum HFMD POC diagnostic test based on legumain, a virus-specific host systemic signature, in saliva.
Collapse
Affiliation(s)
- Yong Wah Tan
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Fiona Mei Shan Teo
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siok Ghee Ler
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Min Nyo
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chia Yin Chong
- Infectious Disease Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Natalie Woon Hui Tan
- Infectious Disease Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Robert Y L Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial and Children's Hospital, Linkou, Taiwan
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Xiong M, Liu C, Li W, Jiang H, Long W, Zhou M, Yang C, Kazobinka G, Sun Y, Zhao J, Hou T. PABPN1 promotes clear cell renal cell carcinoma progression by suppressing the alternative polyadenylation of SGPL1 and CREG1. Carcinogenesis 2023; 44:576-586. [PMID: 37452741 DOI: 10.1093/carcin/bgad049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/15/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023] Open
Abstract
Alternative polyadenylation (APA) is an important post-transcriptional regulatory mechanism in cancer development and progression. Poly(A) binding protein nuclear 1 (PABPN1) is a gene that encodes abundant nuclear protein, binds with high affinity to nascent poly(A) tails, and is crucial for 3'-UTR (3'-untranslated region) APA. Although PABPN1 has been recently reported as a dominant master APA regulator in clear cell renal cell carcinoma (ccRCC), the underlying functional mechanism remain unclear and the genes subject to PABPN1 regulation that contribute to ccRCC progression have not been identified. Here, we found that PABPN1 is upregulated in ccRCC, and its expression is highly associated with the clinical prognosis of ccRCC patients. PABPN1 promotes ccRCC cell proliferation, migration, invasion, and exerts an influence on sphingolipid metabolism and cell cycle. Moreover, PABPN1 depletion significantly suppressed cancer cell growth via induction of cell cycle arrest and apoptosis. In particular, we characterized PABPN1-regulated 3'-UTR APA of sphingosine-1-phosphate lyase 1 (SGPL1) and cellular repressor of E1A stimulated genes 1 (CREG1), which contribute to ccRCC progression. Collectively, our data revealed that PABPN1 promotes ccRCC progression at least in part, by suppressing SGPL1 and CREG1. Thus, PABPN1 may be a potential therapeutic target in ccRCC.
Collapse
Affiliation(s)
- Ming Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyu Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wencheng Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Jiang
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, China
| | - Wulin Long
- Department of Urology, Wuhan Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Menghao Zhou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Yang
- Department of Gynecology and Obstetrics, Women and Children Hospital of Guangdong Province, Guangzhou 510080, China
| | - Gallina Kazobinka
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Urology Unit, La Nouvelle Polyclinique Centrale de Bujumbura, Bujumbura 378, Burundi
| | - Yi Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teng Hou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, China
| |
Collapse
|
7
|
Goto A, Endo Y, Yamashita H. CREG1 stimulates AMPK phosphorylation and glucose uptake in skeletal muscle cells. Biochem Biophys Res Commun 2023; 641:162-167. [PMID: 36528955 DOI: 10.1016/j.bbrc.2022.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
The cellular repressor of adenovirus early region 1A-stimulated gene 1 (CREG1) is a secreted glycoprotein involved in cell differentiation and energy metabolism. It also binds to insulin-like growth factor 2 receptor (IGF2R), a protein implicated in muscle regeneration. However, whether CREG1 regulates the regeneration and metabolism of skeletal muscles via IGF2R remains unclear. This study investigates the role of CREG1 in skeletal muscle regeneration and glucose uptake in C2C12 myotubes and a cardiotoxin (CTX)-induced mouse skeletal muscle regeneration model. CTX-treated skeletal muscle showed significantly higher levels of IGF2R, CREG1, phospho-AMPKα Thr172, and GLUT4 proteins. Similarly, treatment of myotubes with CREG1 also stimulated AMPKα phosphorylation and GLUT4 expression. CREG1-induced AMPKα phosphorylation and 2DG uptake in myotubes were suppressed by IGF2R knockdown and Compound C, an AMPK inhibitor. These results suggest that CREG1 stimulates glucose uptake in skeletal muscles partially through AMPK activation. Hence, CREG1 plays an essential role in muscle regeneration by affecting glucose metabolism in skeletal muscles.
Collapse
Affiliation(s)
- Ayumi Goto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.
| | - Yuki Endo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.
| |
Collapse
|
8
|
Wan HT, Ng AH, Lee WK, Shi F, Wong CKC. Identification and characterization of a membrane receptor that binds to human STC1. Life Sci Alliance 2022; 5:5/11/e202201497. [PMID: 35798563 PMCID: PMC9263378 DOI: 10.26508/lsa.202201497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
A study using TriCEPS-based ligand–receptor methodology and surface plasmon resonance assays identified that human stanniocalcin-1 binds to insulin-like growth factor-2 receptors in human leukemia monocytic cells with high affinity. Stanniocalcin-1 (STC1) is a hypocalcemic hormone originally identified in bony fishes. The mammalian homolog is found to be involved in inflammation and carcinogenesis, among other physiological functions. In this study, we used the TriCEPS-based ligand–receptor methodology to identify the putative binding proteins of human STC1 (hSTC1) in the human leukemia monocytic cell line, ThP-1. LC–MS/MS analysis of peptides from shortlisted hSTC1-binding proteins detected 32 peptides that belong to IGF2/MPRI. Surface plasmon resonance assay demonstrated that hSTC1 binds to immobilized IGF2R/MPRI with high affinity (10–20 nM) and capacity (Rmax 70–100%). The receptor binding data are comparable with those of (CREG) cellular repressor of E1A-stimulated gene a known ligand of IGF2R/MPRI, with Rmax of 75–80% and affinity values of 1–2 nM. The surface plasmon resonance competitive assays showed CREG competed with hSTC1 in binding to IGF2R/MPRI. The biological effects of hSTC1 on ThP-1 cells were demonstrated via IGF2R/MPRI to significantly reduce secreted levels of IL-1β. This is the first study to reveal the high-affinity binding of hSTC1 to the membrane receptor IGF2R/MPRI.
Collapse
Affiliation(s)
- Hin Ting Wan
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Alice Hm Ng
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wang Ka Lee
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Feng Shi
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chris Kong-Chu Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
9
|
Berg AL, Rowson-Hodel A, Wheeler MR, Hu M, Free SR, Carraway KL. Engaging the Lysosome and Lysosome-Dependent Cell Death in Cancer. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-lysosome] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Kusudo T, Okada T, Hashimoto M, Takeuchi T, Endo Y, Niwa A, Yamashita H. CREG1 administration stimulates BAT thermogenesis and improves diet-induced obesity in mice. J Biochem 2021; 171:63-73. [PMID: 34647124 DOI: 10.1093/jb/mvab106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/10/2021] [Indexed: 01/11/2023] Open
Abstract
Brown and beige adipocytes, which express thermogenic uncoupling protein-1 (UCP1), stimulate glucose and lipid metabolism, improving obesity and metabolic diseases such as type 2 diabetes and hyperlipidemia. Overexpression of cellular repressor of E1A-stimulated genes 1 (CREG1) promotes adipose tissue browning and inhibits diet-induced obesity (DIO) in mice. In this study, we investigated the effects of CREG1 administration on DIO inhibition and adipose browning. Subcutaneous administration of recombinant CREG1 protein to C57BL/6 mice stimulated UCP1 expression in interscapular brown adipose tissue (IBAT) and improved DIO, glucose tolerance, and fatty liver compared with those in PBS-treated mice. Injection of Creg1-expressing adenovirus into inguinal white adipose tissue (IWAT) significantly increased browning and mRNA expression of beige adipocyte marker genes compared with that in mice injected with control virus. The effect of Creg1 induction on beige adipocyte differentiation was supported in primary culture using preadipocytes isolated from IWAT of Creg1-transgenic mice compared with that of wild-type mice. Our results indicate a therapeutic effect of CREG1 on obesity and its associated pathology and a potential of CREG1 to stimulate brown/beige adipocyte formation.
Collapse
Affiliation(s)
| | | | | | - Tamaki Takeuchi
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Yuki Endo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| | | | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai 487-8501, Japan
| |
Collapse
|
11
|
Li Y, Tian X, Zhang Q, Yan C, Han Y. A novel function of CREG in metabolic disorders. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:18-22. [PMID: 37724076 PMCID: PMC10388757 DOI: 10.1515/mr-2021-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/13/2021] [Indexed: 09/20/2023]
Abstract
Metabolic disorders are public health problems that require prevention and new efficient drugs for treatment. Cellular repressor of E1A-stimulated genes (CREG) is ubiquitously expressed in mature tissues and cells in mammals and plays a critical role in keeping cells or tissues in a mature, homeostatic state. Recently, CREG turns to be an important mediator in the development of metabolic disorders. Here in this review, we briefly discuss the structure and molecular regulation of CREG along with the therapeutic strategy to combat the metabolic disorders.
Collapse
Affiliation(s)
- Yang Li
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Quanyu Zhang
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| |
Collapse
|
12
|
Huang Z, Yang P, Wang F. Shrimp Plasma CREG Is a Hemocyte Activation Factor. Front Immunol 2021; 12:707770. [PMID: 34484206 PMCID: PMC8415475 DOI: 10.3389/fimmu.2021.707770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
Cytokines are a class of immunoregulatory proteins that are secreted by cells. Although vertebrate cytokine, especially mammalian cytokine has been well studied for the past decades. Much less attention has been paid to invertebrate so that only some cytokines have been identified in invertebrates. We have chosen Peaneus vannamei as a model to explore novel invertebrate cytokines. To achieve this, we previously purified shrimp plasma low abundance proteins and identified more than 400 proteins with proteomics analyses. In this study, a cellular repressor of E1A-stimulated gene (CREG)-like protein, which is highly conserved from Drosophila melanogaster to Homo sapiens, was further characterized in shrimp plasma. We found that shrimp plasma CREG was a glycoprotein which was strongly induced in hemolymph at 8 h post-LPS injection. Further function experiment unveiled that recombinant shrimp CREG protein injection significantly increased phagocytic hemocyte and lysosome-high hemocyte proportion in hemolymph. After that, hemocytes from rEGFP- and rCREG-protein injected shrimps were subjected to transcriptome analyses, which revealed that shrimp CREG protein could comprehensively promote hemocyte maturation and activation. Taken together, our data clearly indicated that shrimp plasma CREG protein is a novel hemocyte activation factor, which is probably a conserved myeloid cell lineage activation factor from invertebrate to vertebrate.
Collapse
Affiliation(s)
- Zhiqi Huang
- Department of Biology, College of Science, Shantou University, Shantou, China
| | - Peng Yang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Fan Wang
- Department of Biology, College of Science, Shantou University, Shantou, China.,Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China.,STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, China
| |
Collapse
|
13
|
Liu J, Qi Y, Chao J, Sathuvalli P, Y Lee L, Li S. CREG1 promotes lysosomal biogenesis and function. Autophagy 2021; 17:4249-4265. [PMID: 33966596 PMCID: PMC8726608 DOI: 10.1080/15548627.2021.1909997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
CREG1 is a small glycoprotein which has been proposed as a transcription repressor, a secretory ligand, a lysosomal, or a mitochondrial protein. This is largely because of lack of antibodies for immunolocalization validated through gain- and loss-of-function studies. In the present study, we demonstrate, using antibodies validated for immunofluorescence microscopy, that CREG1 is mainly localized to the endosomal-lysosomal compartment. Gain- and loss-of-function analyses reveal an important role for CREG1 in both macropinocytosis and clathrin-dependent endocytosis. CREG1 also promotes acidification of the endosomal-lysosomal compartment and increases lysosomal biogenesis. Functionally, overexpression of CREG1 enhances macroautophagy/autophagy and lysosome-mediated degradation, whereas knockdown or knockout of CREG1 has opposite effects. The function of CREG1 in lysosomal biogenesis is likely attributable to enhanced endocytic trafficking. Our results demonstrate that CREG1 is an endosomal-lysosomal protein implicated in endocytic trafficking and lysosomal biogenesis.Abbreviations: AIFM1/AIF: apoptosis inducing factor mitochondria associated 1; AO: acridine orange; ATP6V1H: ATPase H+ transporting V1 subunit H; CALR: calreticulin; CREG: cellular repressor of E1A stimulated genes; CTSC: cathepsin C; CTSD: cathepsin D; EBAG9/RCAS1: estrogen receptor binding site associated antigen 9; EIPA: 5-(N-ethyl-N-isopropyl)amiloride; ER: endoplasmic reticulum; GFP: green fluorescent protein; HEXA: hexosaminidase subunit alpha; IGF2R: insulin like growth factor 2 receptor; LAMP1: lysosomal associated membrane protein 1; M6PR: mannose-6-phosphate receptor, cation dependent; MAPK1/ERK2: mitogen-activated protein kinase 1; MTORC1: mechanistic target of rapamycin kinase complex 1; PDIA2: protein disulfide isomerase family A member 2; SQSTM1/p62: sequestosome 1; TF: transferrin; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Jie Liu
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Yanmei Qi
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Joshua Chao
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Pranav Sathuvalli
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Leonard Y Lee
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Shaohua Li
- Department of Surgery, Rutgers University-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
14
|
Turnšek J, Brunson JK, Viedma MDPM, Deerinck TJ, Horák A, Oborník M, Bielinski VA, Allen AE. Proximity proteomics in a marine diatom reveals a putative cell surface-to-chloroplast iron trafficking pathway. eLife 2021; 10:e52770. [PMID: 33591270 PMCID: PMC7972479 DOI: 10.7554/elife.52770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Iron is a biochemically critical metal cofactor in enzymes involved in photosynthesis, cellular respiration, nitrate assimilation, nitrogen fixation, and reactive oxygen species defense. Marine microeukaryotes have evolved a phytotransferrin-based iron uptake system to cope with iron scarcity, a major factor limiting primary productivity in the global ocean. Diatom phytotransferrin is endocytosed; however, proteins downstream of this environmentally ubiquitous iron receptor are unknown. We applied engineered ascorbate peroxidase APEX2-based subcellular proteomics to catalog proximal proteins of phytotransferrin in the model marine diatom Phaeodactylum tricornutum. Proteins encoded by poorly characterized iron-sensitive genes were identified including three that are expressed from a chromosomal gene cluster. Two of them showed unambiguous colocalization with phytotransferrin adjacent to the chloroplast. Further phylogenetic, domain, and biochemical analyses suggest their involvement in intracellular iron processing. Proximity proteomics holds enormous potential to glean new insights into iron acquisition pathways and beyond in these evolutionarily, ecologically, and biotechnologically important microalgae.
Collapse
Affiliation(s)
- Jernej Turnšek
- Biological and Biomedical Sciences, The Graduate School of Arts and Sciences, Harvard UniversityCambridgeUnited States
- Department of Systems Biology, Harvard Medical SchoolBostonUnited States
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonUnited States
- Integrative Oceanography Division, Scripps Institution of Oceanography, University of California San DiegoLa JollaUnited States
- Center for Research in Biological Systems, University of California San DiegoLa JollaUnited States
- Microbial and Environmental Genomics, J. Craig Venter InstituteLa JollaUnited States
| | - John K Brunson
- Microbial and Environmental Genomics, J. Craig Venter InstituteLa JollaUnited States
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San DiegoLa JollaUnited States
| | | | - Thomas J Deerinck
- National Center for Microscopy and Imaging Research, University of California San DiegoLa JollaUnited States
| | - Aleš Horák
- Biology Centre CAS, Institute of ParasitologyČeské BudějoviceCzech Republic
- University of South Bohemia, Faculty of ScienceČeské BudějoviceCzech Republic
| | - Miroslav Oborník
- Biology Centre CAS, Institute of ParasitologyČeské BudějoviceCzech Republic
- University of South Bohemia, Faculty of ScienceČeské BudějoviceCzech Republic
| | - Vincent A Bielinski
- Synthetic Biology and Bioenergy, J. Craig Venter InstituteLa JollaUnited States
| | - Andrew Ellis Allen
- Integrative Oceanography Division, Scripps Institution of Oceanography, University of California San DiegoLa JollaUnited States
- Microbial and Environmental Genomics, J. Craig Venter InstituteLa JollaUnited States
| |
Collapse
|
15
|
Gomez-Auli A, Hillebrand LE, Christen D, Günther SC, Biniossek ML, Peters C, Schilling O, Reinheckel T. The secreted inhibitor of invasive cell growth CREG1 is negatively regulated by cathepsin proteases. Cell Mol Life Sci 2020; 78:733-755. [PMID: 32385587 PMCID: PMC7873128 DOI: 10.1007/s00018-020-03528-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/31/2020] [Accepted: 04/13/2020] [Indexed: 01/15/2023]
Abstract
Previous clinical and experimental evidence strongly supports a breast cancer-promoting function of the lysosomal protease cathepsin B. However, the cathepsin B-dependent molecular pathways are not completely understood. Here, we studied the cathepsin-mediated secretome changes in the context of the MMTV-PyMT breast cancer mouse model. Employing the cell-conditioned media from tumor-macrophage co-cultures, as well as tumor interstitial fluid obtained by a novel strategy from PyMT mice with differential cathepsin B expression, we identified an important proteolytic and lysosomal signature, highlighting the importance of this organelle and these enzymes in the tumor micro-environment. The Cellular Repressor of E1A Stimulated Genes 1 (CREG1), a secreted endolysosomal glycoprotein, displayed reduced abundance upon over-expression of cathepsin B as well as increased abundance upon cathepsin B deletion or inhibition. Moreover, it was cleaved by cathepsin B in vitro. CREG1 reportedly could act as tumor suppressor. We show that treatment of PyMT tumor cells with recombinant CREG1 reduced proliferation, migration, and invasion; whereas, the opposite was observed with reduced CREG1 expression. This was further validated in vivo by orthotopic transplantation. Our study highlights CREG1 as a key player in tumor–stroma interaction and suggests that cathepsin B sustains malignant cell behavior by reducing the levels of the growth suppressor CREG1 in the tumor microenvironment.
Collapse
Affiliation(s)
- Alejandro Gomez-Auli
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Larissa Elisabeth Hillebrand
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Daniel Christen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Sira Carolin Günther
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Martin Lothar Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Christoph Peters
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany. .,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
16
|
Robledo D, Hamilton A, Gutiérrez AP, Bron JE, Houston RD. Characterising the mechanisms underlying genetic resistance to amoebic gill disease in Atlantic salmon using RNA sequencing. BMC Genomics 2020; 21:271. [PMID: 32228433 PMCID: PMC7106639 DOI: 10.1186/s12864-020-6694-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
Background Gill health is one of the main concerns for Atlantic salmon aquaculture, and Amoebic Gill Disease (AGD), attributable to infection by the amoeba Neoparamoeba perurans, is a frequent cause of morbidity. In the absence of preventive measures, increasing genetic resistance of salmon to AGD via selective breeding can reduce the incidence of the disease and mitigate gill damage. Understanding the mechanisms leading to AGD resistance and the underlying causative genomic features can aid in this effort, while also providing critical information for the development of other control strategies. AGD resistance is considered to be moderately heritable, and several putative QTL have been identified. The aim of the current study was to improve understanding of the mechanisms underlying AGD resistance, and to identify putative causative genomic factors underlying the QTL. To achieve this, RNA was extracted from the gill and head kidney of AGD resistant and susceptible animals following a challenge with N. perurans, and sequenced. Results Comparison between resistant and susceptible animals primarily highlighted differences mainly in the local immune response in the gill, involving red blood cell genes and genes related to immune function and cell adhesion. Differentially expressed immune genes pointed to a contrast in Th2 and Th17 responses, which is consistent with the increased heritability observed after successive challenges with the amoeba. Five QTL-region candidate genes showed differential expression, including a gene connected to interferon responses (GVINP1), a gene involved in systemic inflammation (MAP4K4), and a positive regulator of apoptosis (TRIM39). Analyses of allele-specific expression highlighted a gene in the QTL region on chromosome 17, cellular repressor of E1A-stimulated genes 1 (CREG1), showing allelic differential expression suggestive of a cis-acting regulatory variant. Conclusions In summary, this study provides new insights into the mechanisms of resistance to AGD in Atlantic salmon, and highlights candidate genes for further functional studies that can further elucidate the genomic mechanisms leading to resistance and contribute to enhancing salmon health via improved genomic selection.
Collapse
Affiliation(s)
- Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.
| | - Alastair Hamilton
- Landcatch Natural Selection Ltd., Roslin Innovation Centre, University of Edinburgh, Midlothian, EH25 9RG, UK.,Hendrix Genetics Aquaculture BV/ Netherlands, Villa 'de Körver', Spoorstraat 69, 5831 CK, Boxmeer, Netherlands
| | - Alejandro P Gutiérrez
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - James E Bron
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Ross D Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.
| |
Collapse
|
17
|
Azimi A, Yang P, Ali M, Howard V, Mann GJ, Kaufman KL, Fernandez-Penas P. Data Independent Acquisition Proteomic Analysis Can Discriminate between Actinic Keratosis, Bowen’s Disease, and Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2020; 140:212-222.e11. [DOI: 10.1016/j.jid.2019.06.128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
|
18
|
Hashimoto M, Kusudo T, Takeuchi T, Kataoka N, Mukai T, Yamashita H. CREG1 stimulates brown adipocyte formation and ameliorates diet-induced obesity in mice. FASEB J 2019; 33:8069-8082. [PMID: 30917000 DOI: 10.1096/fj.201802147rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased formation of brown and beige adipocytes is critical for adaptive thermogenesis to maintain homeothermy in cold or to circumvent diet-induced obesity (DIO). Cellular repressor of adenovirus early region 1A-stimulated genes 1 (CREG1) exhibits the ability to stimulate brown adipogenesis, including the induction of uncoupling protein 1 (UCP1), in vitro. Thus, we aimed to clarify whether CREG1 promotes brown adipocyte formation and inhibits DIO at the whole-animal level. In mouse brown adipose tissue (BAT), CREG1 expression was markedly increased in cold but was decreased under thermoneutrality, suggesting CREG1 involvement in BAT thermogenesis. Moreover, in BAT and white adipose tissue, expression of UCP1 and fibroblast growth factor-21 and browning were both significantly higher in adipocyte P2-Creg1-transgenic (Tg) mice than in wild-type (WT) littermates. Following stimulation with a β3-adrenergic agonist, energy consumption was elevated in the Tg mice, which showed increased resistance to DIO and improvement of obesity-associated complications including fatty liver relative to WT mice. The CREG1 stimulatory effect on brown adipogenesis was confirmed in Tg-BAT primary cultures. It was also found that CREG1 binds to retinoid X receptor α, which interacts with thyroid hormone receptor for brown adipogenesis. Our findings demonstrate that CREG1 stimulates brown adipocyte formation and browning, ameliorating obesity and its related pathology in vivo.-Hashimoto, M., Kusudo, T., Takeuchi, T., Kataoka, N., Mukai, T., Yamashita, H. CREG1 stimulates brown adipocyte formation and ameliorates diet-induced obesity in mice.
Collapse
Affiliation(s)
- Michihiro Hashimoto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Tatsuya Kusudo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.,Department of Nutrition and Food Sciences, Faculty of Human Sciences, Tezukayama Gakuin University, Sakai, Japan
| | - Tamaki Takeuchi
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Naoya Kataoka
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.,Department of Integrative Physiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Takako Mukai
- Department of Nutrition and Food Sciences, Faculty of Human Sciences, Tezukayama Gakuin University, Sakai, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
19
|
Kusudo T, Hashimoto M, Kataoka N, Li Y, Nozaki A, Yamashita H. CREG1 promotes uncoupling protein 1 expression and brown adipogenesis in vitro. J Biochem 2019; 165:47-55. [PMID: 30295852 DOI: 10.1093/jb/mvy083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/05/2018] [Indexed: 12/31/2022] Open
Abstract
Brown adipocytes play a critical role for adaptive thermogenesis to regulate body temperature in cold or to circumvent diet-induced obesity. In this study, we investigated the role of cellular repressor of E1A-stimulated genes 1 (CREG1) on brown adipogenesis and uncoupling protein 1 (UCP1) expression by using in vitro culture models. In murine mesenchymal stem cell line C3H10T1/2, Creg1 mRNA expression significantly increased in a time-dependent manner along with Ucp1 mRNA induction in brown adipogenesis. Creg1 gene overexpression upregulated the expression of brown fat-related genes including Ucp1 but its suppression downregulated these gene expression in C3H10T1/2 cells. Unlike the brown adipogenesis, Creg1 mRNA expression decreased significantly after differentiation stimulation in white adipogenesis of 3T3-L1 cells. Either Creg1 gene overexpression or suppression hardly affected white adipogenesis. In addition, CREG1 protein stimulated brown adipogenesis and rescued the adipogenesis in the absence of thyroid hormone in C3H10T1/2 cells. In reporter assay, CREG1 induction stimulated Ucp1 promoter activity, which was enhanced by co-expression with thyroid hormone receptors. The effect of CREG1 on Ucp1 promoter activity was also stimulated by retinoic acid. These results strongly suggest that CREG1 plays an important role on the regulation of UCP1 expression and brown adipogenesis.
Collapse
Affiliation(s)
- Tatsuya Kusudo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.,Department of Nutrition & Food Sciences, Faculty of Human Sciences, Tezukayama Gakuin University, Sakai, Japan
| | - Michihiro Hashimoto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Naoya Kataoka
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan.,Department of Integrative Physiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Yongxue Li
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Aya Nozaki
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
20
|
Ghobrial G, Araujo L, Jinwala F, Li S, Lee LY. The Structure and Biological Function of CREG. Front Cell Dev Biol 2018; 6:136. [PMID: 30416997 PMCID: PMC6212480 DOI: 10.3389/fcell.2018.00136] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/25/2018] [Indexed: 11/18/2022] Open
Abstract
The cellular repressor of E1A-stimulated genes (CREG) is a 220 amino acid glycoprotein structurally similar to oxidoreductases. However, CREG does not have enzymatic activities because it cannot bind to the cofactor flavin mononucleotide. Although CREG can be secreted, it is mainly an intracellular protein localized in the endocytic-lysosomal compartment. It undergoes proteolytic maturation mediated by lysosomal cysteine proteases. Biochemical studies have demonstrated that CREG interacts with mannose-6-phosphate/insulin-like growth factor-2 receptor (M6P/IGF2R) and exocyst Sec8. CREG inhibits proliferation and induces differentiation and senescence when overexpressed in cultured cells. In Drosophila, RNAi-mediated knockdown of CREG causes developmental lethality at the pupal stage. In mice, global deletion of the CREG1 gene leads to early embryonic death. These findings establish an essential role for CREG in development. CREG1 haploinsufficient and liver-specific knockout mice are susceptible to high fat diet-induced obesity, hepatic steatosis and insulin resistance. The purpose of this review is to provide an overview of what we know about the biochemistry and biology of CREG and to discuss the important questions that remain to be addressed in the future.
Collapse
Affiliation(s)
- Gaby Ghobrial
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Luiz Araujo
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Felecia Jinwala
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Shaohua Li
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Leonard Y Lee
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
21
|
Inhibition of insulin-like growth factor II (IGF-II)-dependent cell growth by multidentate pentamannosyl 6-phosphate-based ligands targeting the mannose 6-phosphate/IGF-II receptor. Oncotarget 2018; 7:62386-62410. [PMID: 27694692 PMCID: PMC5308735 DOI: 10.18632/oncotarget.11493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/28/2016] [Indexed: 01/24/2023] Open
Abstract
The mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R) binds M6P-capped ligands and IGF-II at different binding sites within the ectodomain and mediates ligand internalization and trafficking to the lysosome. Multivalent M6P-based ligands can cross-bridge the M6P/IGF2R, which increases the rate of receptor internalization, permitting IGF-II binding as a passenger ligand and subsequent trafficking to the lysosome, where the IGF-II is degraded. This unique feature of the receptor may be exploited to design novel therapeutic agents against IGF-II-dependent cancers that will lead to decreased bioavailable IGF-II within the tumor microenvironment. We have designed a panel of M6P-based ligands that bind to the M6P/IGF2R with high affinity in a bivalent manner and cause decreased cell viability. We present evidence that our ligands bind through the M6P-binding sites of the receptor and facilitate internalization and degradation of IGF-II from conditioned medium to mediate this cellular response. To our knowledge, this is the first panel of synthetic bivalent ligands for the M6P/IGF2R that can take advantage of the ligand-receptor interactions of the M6P/IGF2R to provide proof-of-principle evidence for the feasibility of novel chemotherapeutic agents that decrease IGF-II-dependent growth of cancer cells.
Collapse
|
22
|
Tian X, Yan C, Liu M, Zhang Q, Liu D, Liu Y, Li S, Han Y. CREG1 heterozygous mice are susceptible to high fat diet-induced obesity and insulin resistance. PLoS One 2017; 12:e0176873. [PMID: 28459882 PMCID: PMC5411056 DOI: 10.1371/journal.pone.0176873] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022] Open
Abstract
Cellular repressor of E1A-stimulated genes 1 (CREG1) is a small glycoprotein whose physiological function is unknown. In cell culture studies, CREG1 promotes cellular differentiation and maturation. To elucidate its physiological functions, we deleted the Creg1 gene in mice and found that loss of CREG1 leads to early embryonic death, suggesting that it is essential for early development. In the analysis of Creg1 heterozygous mice, we unexpectedly observed that they developed obesity as they get older. In this study, we further studied this phenotype by feeding wild type (WT) and Creg1 heterozygote (Creg1+/-) mice a high fat diet (HFD) for 16 weeks. Our data showed that Creg1+/- mice exhibited a more prominent obesity phenotype with no change in food intake compared with WT controls when challenged with HFD. Creg1 haploinsufficiency also exacerbated HFD-induced liver steatosis, dyslipidemia and insulin resistance. In addition, HFD markedly increased pro-inflammatory cytokines in plasma and epididymal adipose tissue in Creg1+/- mice as compared with WT controls. The activation level of NF-κB, a major regulator of inflammatory response, in epididymal adipose tissue was also elevated in parallel with the cytokines in Creg1+/- mice. These pro-inflammatory responses elicited by CREG1 reduction were confirmed in 3T3-L1-derived adipocytes with CREG1 depletion by siRNA transfection. Given that adipose tissue inflammation has been shown to play a key role in obesity-induced insulin resistance and metabolic syndrome, our results suggest that Creg1 haploinsufficiency confers increased susceptibility of adipose tissue to inflammation, leading to aggravated obesity and insulin resistance when challenged with HFD. This study uncovered a novel function of CREG1 in metabolic disorders.
Collapse
Affiliation(s)
- Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Meili Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Quanyu Zhang
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Yanxia Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
| | - Shaohua Li
- Department of Surgery, Robert Wood Johnson Medical School, Rutgers-the State University of New Jersey, New Brunswick, United States of America
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
- Cardiovascular Center for Translational Medicine of Liaoning Province, Shenyang, China
- Cardiovascular Core Lab for Translational Medicine of Liaoning Province, Shenyang, China
- * E-mail:
| |
Collapse
|
23
|
LeBoeuf AC, Waridel P, Brent CS, Gonçalves AN, Menin L, Ortiz D, Riba-Grognuz O, Koto A, Soares ZG, Privman E, Miska EA, Benton R, Keller L. Oral transfer of chemical cues, growth proteins and hormones in social insects. eLife 2016; 5:e20375. [PMID: 27894417 PMCID: PMC5153251 DOI: 10.7554/elife.20375] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/14/2016] [Indexed: 01/02/2023] Open
Abstract
Social insects frequently engage in oral fluid exchange - trophallaxis - between adults, and between adults and larvae. Although trophallaxis is widely considered a food-sharing mechanism, we hypothesized that endogenous components of this fluid might underlie a novel means of chemical communication between colony members. Through protein and small-molecule mass spectrometry and RNA sequencing, we found that trophallactic fluid in the ant Camponotus floridanus contains a set of specific digestion- and non-digestion related proteins, as well as hydrocarbons, microRNAs, and a key developmental regulator, juvenile hormone. When C. floridanus workers' food was supplemented with this hormone, the larvae they reared via trophallaxis were twice as likely to complete metamorphosis and became larger workers. Comparison of trophallactic fluid proteins across social insect species revealed that many are regulators of growth, development and behavioral maturation. These results suggest that trophallaxis plays previously unsuspected roles in communication and enables communal control of colony phenotypes.
Collapse
Affiliation(s)
- Adria C LeBoeuf
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Patrice Waridel
- Protein Analysis FacilityUniversity of LausanneLausanneSwitzerland
| | - Colin S Brent
- Arid Land Agricultural Research Center, USDA-ARSMaricopaUnited States
| | - Andre N Gonçalves
- Department of Biochemistry and ImmunologyInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisMinas GeraisBrazil
- Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Laure Menin
- Institute of Chemical Sciences and EngineeringEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Daniel Ortiz
- Institute of Chemical Sciences and EngineeringEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Oksana Riba-Grognuz
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Akiko Koto
- The Department of Genetics, Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Zamira G Soares
- Department of Biochemistry and ImmunologyInstituto de Ciências Biológicas, Universidade Federal de Minas GeraisMinas GeraisBrazil
- Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Eyal Privman
- Department of Evolutionary and Environmental Biology, Institute of EvolutionUniversity of HaifaHaifaIsrael
| | - Eric A Miska
- Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
- Department of GeneticsUniversity of CambridgeCambridgeUnited Kingdom
- Wellcome Trust Sanger InstituteWellcome Trust Genome CampusCambridgeUnited Kingdom
| | - Richard Benton
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Laurent Keller
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
24
|
Liu J, Qi Y, Li S, Hsu SC, Saadat S, Hsu J, Rahimi SA, Lee LY, Yan C, Tian X, Han Y. CREG1 Interacts with Sec8 to Promote Cardiomyogenic Differentiation and Cell-Cell Adhesion. Stem Cells 2016; 34:2648-2660. [PMID: 27334848 DOI: 10.1002/stem.2434] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 04/29/2016] [Accepted: 05/28/2016] [Indexed: 02/06/2023]
Abstract
Understanding the regulation of cell-cell interactions during the formation of compact myocardial structures is important for achieving true cardiac regeneration through enhancing the integration of stem cell-derived cardiomyocytes into the recipient myocardium. In this study, we found that cellular repressor of E1A-stimulated genes 1 (CREG1) is highly expressed in both embryonic and adult hearts. Gain- and loss-of-function analyses demonstrated that CREG1 is required for differentiation of mouse embryonic stem (ES) cell into cardiomyocytes and the formation of cohesive myocardium-like structures in a cell-autonomous fashion. Furthermore, CREG1 directly interacts with Sec8 of the exocyst complex, which tethers vesicles to the plasma membrane. Site-directed mutagenesis and rescue of CREG1 knockout ES cells showed that CREG1 binding to Sec8 is required for cardiomyocyte differentiation and cohesion. Mechanistically, CREG1, Sec8, and N-cadherin colocalize at intercalated discs in vivo and are enriched at cell-cell junctions in cultured cardiomyocytes. CREG1 overexpression enhances the assembly of adherens and gap junctions. By contrast, its knockout inhibits the Sec8-N-cadherin interaction and induces their degradation. These results suggest that the CREG1 binding to Sec8 enhances the assembly of intercellular junctions and promotes cardiomyogenesis. Stem Cells 2016;34:2648-2660.
Collapse
Affiliation(s)
- Jie Liu
- Department of Surgery, Robert Wood Johnson Medical School
| | - Yanmei Qi
- Department of Surgery, Robert Wood Johnson Medical School
| | - Shaohua Li
- Department of Surgery, Robert Wood Johnson Medical School
| | - Shu-Chan Hsu
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers-the State University of New Jersey, USA
| | - Siavash Saadat
- Department of Surgery, Robert Wood Johnson Medical School
| | - June Hsu
- Department of Surgery, Robert Wood Johnson Medical School
| | - Saum A Rahimi
- Department of Surgery, Robert Wood Johnson Medical School
| | - Leonard Y Lee
- Department of Surgery, Robert Wood Johnson Medical School
| | - Chenghui Yan
- Department of Cardiology, The General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Xiaoxiang Tian
- Department of Cardiology, The General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Yaling Han
- Department of Cardiology, The General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| |
Collapse
|
25
|
Clark DJ, Mei Y, Sun S, Zhang H, Yang AJ, Mao L. Glycoproteomic Approach Identifies KRAS as a Positive Regulator of CREG1 in Non-small Cell Lung Cancer Cells. Am J Cancer Res 2016; 6:65-77. [PMID: 26722374 PMCID: PMC4679355 DOI: 10.7150/thno.12350] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/09/2015] [Indexed: 01/05/2023] Open
Abstract
Protein glycosylation plays a fundamental role in a multitude of biological processes, and the associated aberrant expression of glycoproteins in cancer has made them attractive biomarkers and therapeutic targets. In this study, we examined differentially expressed glycoproteins in cell lines derived from three different states of lung tumorigenesis: an immortalized bronchial epithelial cell (HBE) line, a non-small cell lung cancer (NSCLC) cell line harboring a Kirsten rat sarcoma viral oncogene homolog (KRAS) activation mutation and a NSCLC cell line harboring an epidermal growth factor receptor (EGFR) activation deletion. Using a Triple SILAC proteomic quantification strategy paired with hydrazide chemistry N-linked glycopeptide enrichment, we quantified 118 glycopeptides in the three cell lines derived from 82 glycoproteins. Proteomic profiling revealed 27 glycopeptides overexpressed in both NSCLC cell lines, 6 glycopeptides overexpressed only in the EGFR mutant cells and 19 glycopeptides overexpressed only in the KRAS mutant cells. Further investigation of a panel of NSCLC cell lines found that Cellular repressor of E1A-stimulated genes (CREG1) overexpression was closely correlated with KRAS mutation status in NSCLC cells and could be down-regulated by inhibition of KRAS expression. Our results indicate that CREG1 is a down-stream effector of KRAS in a sub-type of NSCLC cells and a novel candidate biomarker or therapeutic target for KRAS mutant NSCLC.
Collapse
|
26
|
Barzelay A, Levy R, Kohn E, Sella M, Shani N, Meilik B, Entin-Meer M, Gur E, Loewenstein A, Barak A. Power-Assisted Liposuction Versus Tissue Resection for the Isolation of Adipose Tissue-Derived Mesenchymal Stem Cells: Phenotype, Senescence, and Multipotency at Advanced Passages. Aesthet Surg J 2015; 35:NP230-40. [PMID: 26319084 DOI: 10.1093/asj/sjv055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Adipose tissue-derived mesenchymal stem cells (ASCs) can be isolated from subcutaneous fat harvested by tissue resection or liposuction. OBJECTIVES The authors compared ASCs isolated by tissue resection or power-assisted liposuction (PAL) to determine whether either surgical procedure yielded ASCs with improved purity and competence that was preserved for several passages. METHODS For this experimental study, ASCs were isolated from fat harvested by tissue resection or PAL from six patients who underwent abdominoplasty. ASCs were counted to determine cell yields, and viabilities were assessed with an amine-reactive dye and by fluorescence-activated cell sorting (FACS). Cell phenotypes were determined by immunostaining and FACS, and doubling times were calculated. Senescence ratios of the cells were detected by gene profiling and by assaying β-galactosidase activity. Multipotency was evaluated by induced differentiation analyses. RESULTS No significant differences were observed in cell numbers or viabilities of ASCs isolated following either surgical method of fat harvesting. Both populations of cultured ASCs expressed markers of mesenchymal stem cells and preserved this expression pattern through the third passage. PAL and tissue resection yielded ASCs with similar division rates, similar senescence ratios into the fourth passage, and similar capacities to differentiate into osteocytes or adipocytes. CONCLUSIONS Fat harvested by PAL or tissue resection yielded uniform cultures of ASCs with high division rates, low senescence ratios, and multipotency preserved into passages 3 and 4. Because PAL is less invasive, it may be preferable for the isolation of ASCs.
Collapse
Affiliation(s)
- Aya Barzelay
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Ran Levy
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Emmanulle Kohn
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Meirav Sella
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Nir Shani
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Benjamin Meilik
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Michal Entin-Meer
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Eyal Gur
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Anat Loewenstein
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| | - Adiel Barak
- Dr Barzelay is a resident, Drs Levy and Kohn are Assistant Researchers, Dr Loewenstein is a Professor of Ophthalmology and Chair of the Department of Ophthalmology, and Dr Barak is a Professor of Ophthalmology and Director of the Vitreoretinal Surgery Unit in the Ophthalmology Laboratory and Department of Ophthalmology at the Tel-Aviv Sourasky Medical Center in Tel-Aviv, Israel. Dr Sella is an Assistant Researcher, Dr Meilik is an Associate Professor and Assistant Researcher, and Dr Gur is a Professor and Chair of the Department of Plastic and Reconstructive Surgery at the Tel-Aviv Sourasky Medical Center. Dr Entin-Meer is an Assistant Researcher in the Department of Cardiology at the Tel-Aviv Sourasky Medical Center
| |
Collapse
|
27
|
Overexpression of the Insulin-Like Growth Factor II Receptor Increases β-Amyloid Production and Affects Cell Viability. Mol Cell Biol 2015; 35:2368-84. [PMID: 25939386 DOI: 10.1128/mcb.01338-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 04/20/2015] [Indexed: 12/26/2022] Open
Abstract
Amyloid β (Aβ) peptides originating from amyloid precursor protein (APP) in the endosomal-lysosomal compartments play a critical role in the development of Alzheimer's disease (AD), the most common type of senile dementia affecting the elderly. Since insulin-like growth factor II (IGF-II) receptors facilitate the delivery of nascent lysosomal enzymes from the trans-Golgi network to endosomes, we evaluated their role in APP metabolism and cell viability using mouse fibroblast MS cells deficient in the murine IGF-II receptor and corresponding MS9II cells overexpressing the human IGF-II receptors. Our results show that IGF-II receptor overexpression increases the protein levels of APP. This is accompanied by an increase of β-site APP-cleaving enzyme 1 levels and an increase of β- and γ-secretase enzyme activities, leading to enhanced Aβ production. At the cellular level, IGF-II receptor overexpression causes localization of APP in perinuclear tubular structures, an increase of lipid raft components, and increased lipid raft partitioning of APP. Finally, MS9II cells are more susceptible to staurosporine-induced cytotoxicity, which can be attenuated by β-secretase inhibitor. Together, these results highlight the potential contribution of IGF-II receptor to AD pathology not only by regulating expression/processing of APP but also by its role in cellular vulnerability.
Collapse
|
28
|
Peng C, Pei H, Wei F, Tian X, Deng J, Yan C, Li Y, Sun M, Zhang J, Liu D, Rong J, Wang J, Gao E, Li S, Han Y. Cellular repressor of E1A-stimulated gene overexpression in bone mesenchymal stem cells protects against rat myocardial infarction. Int J Cardiol 2015; 183:232-41. [DOI: 10.1016/j.ijcard.2015.01.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/14/2014] [Accepted: 01/25/2015] [Indexed: 12/13/2022]
|
29
|
Kowalewski-Nimmerfall E, Schähs P, Maresch D, Rendic D, Krämer H, Mach L. Drosophila melanogaster cellular repressor of E1A-stimulated genes is a lysosomal protein essential for fly development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2900-12. [PMID: 25173815 PMCID: PMC4331662 DOI: 10.1016/j.bbamcr.2014.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 01/01/2023]
Abstract
Mammalian cellular repressor of E1A-stimulated genes is a lysosomal glycoprotein implicated in cellular growth and differentiation. The genome of the fruit fly Drosophila melanogaster encodes a putative orthologue (dCREG), suggesting evolutionarily conserved physiological functions of this protein. In D. melanogaster S2 cells, dCREG was found to localize in lysosomes. Further studies revealed that intracellular dCREG is subject of proteolytic maturation. Processing and turnover could be substantially reduced by RNAi-mediated silencing of cathepsin L. In contrast to mammalian cells, lysosomal delivery of dCREG does not depend on its carbohydrate moiety. Furthermore, depletion of the putative D. melanogaster lysosomal sorting receptor lysosomal enzyme receptor protein did not compromise cellular retention of dCREG. We also investigated the developmental consequences of dCREG ablation in whole D. melanogaster flies. Ubiquitous depletion of dCREG proved lethal at the late pupal stage once a knock-down efficiency of > 95% was achieved. These results demonstrate that dCREG is essential for proper completion of fly development. The lysosomal localization of CREG is evolutionarily conserved. Lysosomal delivery of CREG is mediated by different mechanisms in mammals and flies. Cathepsin L is the main protease responsible for CREG processing and turnover. CREG deficiency causes developmental lethality in D. melanogaster.
Collapse
Affiliation(s)
- Elisabeth Kowalewski-Nimmerfall
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Philipp Schähs
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Daniel Maresch
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Dubravko Rendic
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Helmut Krämer
- Department of Neuroscience, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9111, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9111, USA
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria.
| |
Collapse
|
30
|
Wang Y, Thinakaran G, Kar S. Overexpression of the IGF-II/M6P receptor in mouse fibroblast cell lines differentially alters expression profiles of genes involved in Alzheimer's disease-related pathology. PLoS One 2014; 9:e98057. [PMID: 24846272 PMCID: PMC4028253 DOI: 10.1371/journal.pone.0098057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/25/2014] [Indexed: 11/29/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of senile dementia affecting elderly people. The processing of amyloid precursor protein (APP) leading to the generation of β-amyloid (Aβ) peptide contributes to neurodegeneration and development of AD pathology. The endocytic trafficking pathway, which comprises of the endosomes and lysosomes, acts as an important site for Aβ generation, and endocytic dysfunction has been linked to increased Aβ production and loss of neurons in AD brains. Since insulin-like growth factor-II (IGF-II) receptor plays a critical role in the transport of lysosomal enzymes from the trans-Golgi network to endosomes, it is likely that the receptor may have a role in regulating Aβ metabolism in AD pathology. However, very little is known on how altered levels of the IGF-II receptor can influence the expression/function of various molecules involved in AD pathology. To address this issue, we evaluated the expression profiles of 87 selected genes related to AD pathology in mouse fibroblast MS cells that are deficient in murine IGF-II receptor and corresponding MS9II cells overexpressing ∼500 times the human IGF-II receptors. Our results reveal that an elevation in IGF-II receptor levels alters the expression profiles of a number of genes including APP as well as enzymes regulating Aβ production, degradation and clearance mechanisms. Additionally, it influences the expression of various lysosomal enzymes and protein kinases that are involved in Aβ toxicity. IGF-II receptor overexpression also alters expression of several genes involved in intracellular signalling as well as cholesterol metabolism, which play a critical role in AD pathology. The altered gene profiles observed in this study closely match with the corresponding protein levels, with a few exceptions. These results, taken together, suggest that an elevation in IGF-II receptor levels can influence the expression profiles of transcripts as well as proteins that are involved in AD pathogenesis.
Collapse
Affiliation(s)
- Yanlin Wang
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Gopal Thinakaran
- Departments of Neurobiology, Neurology and Pathology, The University of Chicago, Chicago, Illinois, United States of America
| | - Satyabrata Kar
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
31
|
Tao J, Yan C, Tian X, Liu S, Li Y, Zhang J, Sun M, Ma X, Han Y. CREG promotes the proliferation of human umbilical vein endothelial cells through the ERK/cyclin E signaling pathway. Int J Mol Sci 2013; 14:18437-56. [PMID: 24018888 PMCID: PMC3794788 DOI: 10.3390/ijms140918437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/15/2013] [Accepted: 08/28/2013] [Indexed: 11/26/2022] Open
Abstract
Cellular repressor of E1A-stimulated genes (CREG) is a recently discovered secreted glycoprotein involved in homeostatic modulation. We previously reported that CREG is abundantly expressed in the adult vascular endothelium and dramatically downregulated in atherosclerotic lesions. In addition, CREG participates in the regulation of apoptosis, inflammation and wound healing of vascular endothelial cells. In the present study, we attempted to investigate the effect of CREG on the proliferation of vascular endothelial cells and to decipher the underlying molecular mechanisms. Overexpression of CREG in human umbilical vein endothelial cells (HUVEC) was obtained by infection with adenovirus carrying CREG. HUVEC proliferation was investigated by flow cytometry and 5-bromo-2′-deoxy-uridine (BrdU) incorporation assays. The expressions of cyclins, cyclin-dependent kinases and signaling molecules were also examined. In CREG-overexpressing cells, we observed a marked increase in the proportion of the S and G2 population and a decrease in the G0/G1 phase population. The number of BrdU positively-stained cells also increased, obviously. Furthermore, silencing of CREG expression by specific short hairpin RNA effectively inhibited the proliferation of human umbilical vein endothelial cells (HUVEC). CREG overexpression induced the expression of cyclin E in both protein and mRNA levels to regulate cell cycle progression. Further investigation using inhibitor blocking analysis identified that ERK activation mediated the CREG modulation of the proliferation and cyclin E expression in HUVEC. In addition, blocking vascular endothelial growth factor (VEGF) in CREG-overexpressed HUVEC and supplementation of VEGF in CREG knocked-down HUVEC identified that the pro-proliferative effect of CREG was partially mediated by VEGF-induced ERK/cyclin E activation. These results suggest a novel role of CREG to promote HUVEC proliferation through the ERK/cyclin E signaling pathway.
Collapse
Affiliation(s)
- Jie Tao
- Graduate School of Third Military Medical University, Chongqing 400038, China; E-Mail:
| | - Chenghui Yan
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Shaowei Liu
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Yang Li
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Jian Zhang
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Mingyu Sun
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; E-Mail:
| | - Yaling Han
- Cardiovascular Research Institute and Key laboratory of Cardiology, Shenyang Northern Hospital, Shenyang 110840, China; E-Mails: (C.Y.); (X.T.); (S.L.); (Y.L.); (J.Z.); (M.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-24-2305-6123; Fax: +86-24-2392-2184
| |
Collapse
|
32
|
Pellagatti A, Fidler C, Wainscoat JS, Boultwood J. Gene expression profiling in the myelodysplastic syndromes. Hematology 2013; 10:281-7. [PMID: 16085540 DOI: 10.1080/10245330500065680] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The myelodysplastic syndromes (MDS) are a heterogeneous group of haematopoietic malignancies, characterized by blood cytopenias, ineffective hematopoiesis and hypercellular bone marrow. Several genetic alterations have been reported in MDS but these are not MDS-specific and the underlying molecular causes of the disease remain poorly understood. Gene expression microarray technology allows the simultaneous parallel analysis of many thousands of genes and has already provided novel insights into cancer pathogenesis. In this review we discuss the results of several recent studies which utilize the enormous power of microarray technology for the study of MDS. Several exciting findings have emerged from these early studies that highlight the potential of this technology to further our understanding of the molecular pathogenesis of this disorder. It is clear, however, that these findings should be confirmed in larger sets of MDS patients.
Collapse
Affiliation(s)
- Andrea Pellagatti
- Leukaemia Research Fund Molecular Haematology Unit, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | |
Collapse
|
33
|
Li Y, Tao J, Zhang J, Tian X, Liu S, Sun M, Zhang X, Yan C, Han Y. Cellular repressor E1A-stimulated genes controls phenotypic switching of adventitial fibroblasts by blocking p38MAPK activation. Atherosclerosis 2012; 225:304-14. [PMID: 23040447 DOI: 10.1016/j.atherosclerosis.2012.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 08/02/2012] [Accepted: 08/13/2012] [Indexed: 11/16/2022]
Abstract
AIMS Phenotypic modulation of adventitial fibroblasts (AFs) plays an important role in the pathogenesis of proliferative vascular diseases. The current study aimed to identify the role of cellular repressor E1A-stimulated genes (CREG), a critical mediator in the maintenance of vascular homeostasis, in AF phenotypic modulation and adventitial remodeling. METHOD AND RESULTS Using in situ double-immunofluorescence staining, we ascertained that CREG expression was significantly down-regulated in the adventitia after vascular injury, and its expression pattern was conversely correlated with the expression of smooth muscle α-actin (α-SMA), a marker for differentiation of AFs into myofibroblasts. In vitro data confirmed the association of CREG in angiotensin II (Ang II)-induced AF differentiation. Additionally, overexpression of CREG attenuated Ang II-induced α-SMA expression in AFs. CREGoverexpressing AFs showed decreased levels of proliferation on days 2-5 following stimulation by Ang II compared with controls, with changes in the cell cycle profile as shown by BrdU incorporation assay and fluorescence activated cell sorting analysis. Moreover, wound healing assay and transwell migration model demonstrated that upregulation of CREG expression inhibited Ang II-induced AF migration. We found that CREG-mediated its counterbalancing effects in Ang II-induced phenotypic modulation, proliferation and migration by inhibition of the p38MAPK signaling pathway, validated by pharmacological blockade of p38MAPK with SB 203580 and by overexpression of p38MAPK with transfectants expressing constitutively active p38αMAPK. CONCLUSION Our findings suggest that CREG is a novel AF phenotypic modulator in a p38MAPK-dependent manner. Modulating CREG on the local vascular wall may become a new therapeutic target against proliferative vascular diseases.
Collapse
Affiliation(s)
- Yang Li
- Graduate School of Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kreiling JL, Montgomery MA, Wheeler JR, Kopanic JL, Connelly CM, Zavorka ME, Allison JL, Macdonald RG. Dominant-negative effect of truncated mannose 6-phosphate/insulin-like growth factor II receptor species in cancer. FEBS J 2012; 279:2695-713. [PMID: 22681933 DOI: 10.1111/j.1742-4658.2012.08652.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oligomerization of the mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R) is important for optimal ligand binding and internalization. M6P/IGF2R is a tumor suppressor gene that exhibits loss of heterozygosity and is mutated in several cancers. We tested the potential dominant-negative effects of two cancer-associated mutations that truncate M6P/IGF2R in ectodomain repeats 9 and 14. Our hypothesis was that co-expression of the truncated receptors with the wild-type/endogenous full-length M6P/IGF2R would interfere with M6P/IGF2R function by heterodimer interference. Immunoprecipitation confirmed formation of heterodimeric complexes between full-length M6P/IGF2Rs and the truncated receptors, termed Rep9F and Rep14F. Remarkably, increasing expression of either Rep9F or Rep14F provoked decreased levels of full-length M6P/IGF2Rs in both cell lysates and plasma membranes, indicating a dominant-negative effect on receptor availability. Loss of full-length M6P/IGF2R was not due to increased proteasomal or lysosomal degradation, but instead arose from increased proteolytic cleavage of cell-surface M6P/IGF2Rs, resulting in ectodomain release, by a mechanism that was inhibited by metal ion chelators. These data suggest that M6P/IGF2R truncation mutants may contribute to the cancer phenotype by decreasing the availability of full-length M6P/IGF2Rs to perform tumor-suppressive functions such as binding/internalization of receptor ligands such as insulin-like growth factor II.
Collapse
Affiliation(s)
- Jodi L Kreiling
- Department of Chemistry, University of Nebraska at Omaha, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Li Y, Yan CH, Han YL. CREG mediated adventitial fibroblast phenotype modulation: A possible therapeutic target for proliferative vascular disease. Med Hypotheses 2012; 79:95-7. [DOI: 10.1016/j.mehy.2012.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/03/2012] [Indexed: 11/16/2022]
|
36
|
Zhang H, Han Y, Tao J, Liu S, Yan C, Li S. Cellular repressor of E1A-stimulated genes regulates vascular endothelial cell migration by the ILK/AKT/mTOR/VEGF(165) signaling pathway. Exp Cell Res 2011; 317:2904-13. [PMID: 21939655 DOI: 10.1016/j.yexcr.2011.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 08/19/2011] [Accepted: 08/20/2011] [Indexed: 02/07/2023]
Abstract
The migration of vascular endothelial cells plays a critical role in a variety of vascular physiological and pathological processes, such as embryonic development, angiogenesis, wound healing, re-endothelialization, and vascular remodeling. This study clarified the role and mechanism of a new vascular homeostasis regulator, Cellular repressor of E1A-stimulated genes (CREG), in the migration of primary human umbilical vein endothelial cells (HUVECs). A wound healing assay and transwell migration model showed that upregulation of CREG expression induced HUVEC migration and it was positively correlated with the expression of vascular endothelial growth factor. Furthermore, wild type integrin-linked kinase reversed the poor mobility of CREG knock-down HUVECs; in contrast, kinase-dead integrin-linked kinase weakened the migration of HUVECs. We also studied the effect of CREG on HUVEC migration by the addition of an mTOR inhibitor, recombinant vascular endothelial growth factor(165), neutralizing antibody of vascular endothelial growth factor(165) and AKT siRNA, and we concluded that CREG induces endothelial cell migration by activating the integrin-linked kinase/AKT/mTOR/VEGF(165) signaling pathway.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Cardiology, Cardiovascular Research Institute, Shenyang Northern Hospital, Shenyang, China
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Cellular senescence is an irreversible growth arrest that is activated in normal cells upon shortening of telomere and other cellular stresses. Bypassing cellular senescence is a necessary step for cells to become immortal during oncogenic transformation. During the spontaneous immortalization of Li-Fraumeni Syndrome (LFS) fibroblasts, we found that CREG1 (Cellular Repressor of E1A-stimulated Genes 1) expression was decreased during immortalization and increased in senescence. Moreover, we found that repression of CREG1 expression occurs via an epigenetic mechanism, promoter DNA methylation. Ectopic expression of CREG1 in the immortal LFS cell lines decreases cell proliferation but does not directly induce senescence. We confirmed this in osteosarcoma and fibrosarcoma cancer cell lines, cancers commonly seen in Li-Fraumeni Syndrome. In addition, we found that p16 (INK4a) is also downregulated in immortal cells and that coexpression of CREG1 and p16 (INK4a) , an inhibitor of CDK4/6 and Rb phosphorylation, has a greater effect than either CREG1 and p16 (INK4a) alone to reduce cell growth, induce cell cycle arrest and cellular senescence in immortal LFS fibroblasts, osteosarcoma and fibrosarcoma cell lines. Moreover, cooperation of CREG1 and p16 (INK4a) inhibits the expression of cyclin A and cyclin B by inhibiting promoter activity thereby decreasing mRNA and protein levels; these proteins are required for S-phase entry and G2/M transition. In conclusion, this is the first evidence to demonstrate that CREG1 enhances p16 (INK4a) -induced senescence by transcriptional repression of cell cycle-regulated genes.
Collapse
Affiliation(s)
- Benchamart Moolmuang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
38
|
Wang T, Zhang M, Han Y, Zhang X, Yan C, Liang Z, Sun Y, Kang J. Lack of association between cellular repressor of E1A-stimulated genes (GREG) polymorphisms and coronary artery disease in the Han population of North China. Clin Chim Acta 2011; 412:249-52. [DOI: 10.1016/j.cca.2010.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 10/08/2010] [Accepted: 10/09/2010] [Indexed: 12/01/2022]
|
39
|
Pattern of expression of the CREG gene and CREG protein in the mouse embryo. Mol Biol Rep 2010; 38:2133-40. [PMID: 20857207 DOI: 10.1007/s11033-010-0340-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 09/04/2010] [Indexed: 10/19/2022]
Abstract
The cellular repressor of E1A-stimulated genes (CREG) is a secreted glycoprotein that inhibits cell proliferation and/or enhances differentiation. CREG is widely expressed in adult tissues such as the brain, heart, lungs, liver, intestines and kidneys in mice. We investigated the level of CREG expression during mouse embryogenesis and its distribution at 18.5 days post coitus (dpc) using immunohistochemical staining with diaminobenzidine, western blotting and reverse transcription-polymerase chain reaction. CREG expression was first detected in mouse embryos at 4.5 dpc. It was expressed at almost all stages up to 18.5 dpc. The level of CREG was found to increase gradually and was highest at 18.5 dpc. Western blotting showed that the CREG protein was expressed at higher levels in the brain, heart, intestines and kidneys than in the lungs and liver at 18.5 dpc. In 9.5 dpc embryos, CREG was expressed only in the endothelial cells of blood vessels, after the vascular lumen had formed. With advanced differentiation, vascular smooth muscle cells developed in the embryonic vascular structures; the expression of smooth muscle α-actin protein and CREG were positive and increased gradually in 10.5 dpc embryonic vessels. CREG expression in the embryonic blood vessels peaked at 15.5 dpc and was reduced slightly at 18.5 dpc. These results indicate that CREG is expressed during mouse embryogenesis and might participate in the differentiation of these organs during embryogenesis.
Collapse
|
40
|
Han Y, Wu G, Deng J, Tao J, Guo L, Tian X, Kang J, Zhang X, Yan C. Cellular repressor of E1A-stimulated genes inhibits human vascular smooth muscle cell apoptosis via blocking P38/JNK MAP kinase activation. J Mol Cell Cardiol 2010; 48:1225-35. [DOI: 10.1016/j.yjmcc.2009.12.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 12/21/2009] [Accepted: 12/21/2009] [Indexed: 02/06/2023]
|
41
|
WU GZ, YAN CH, HAN YL, TAO J, DENG J, TIAN XX, ZHANG BH, WANG T, KANG J, ZHANG XL. Overexpression of The Cellular Repressor of E1A-stimulated Genes Inhibits The Apoptosis of Human Vascular Smooth Muscle Cells <I>via</I> Blocking p38/JNK MAP Kinase Activation*. PROG BIOCHEM BIOPHYS 2010. [DOI: 10.3724/sp.j.1206.2009.00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
CREG inhibits migration of human vascular smooth muscle cells by mediating IGF-II endocytosis. Exp Cell Res 2009; 315:3301-11. [PMID: 19769965 DOI: 10.1016/j.yexcr.2009.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 11/23/2022]
Abstract
We previously determined that the cellular repressor of E1A-stimulated genes, (CREG) plays a role in the maintenance of the mature phenotype of vascular smooth muscle cells (SMCs). This study aimed to identify the role of CREG in modulating the migration of SMCs. Recombinant virus-mediated CREG expression inhibited the cellular migration of cultured SMCs associated with down-regulated activity of matrix metalloproteinase-9 (MMP-9). In contrast, CREG knockdown via the retroviral transfer of short hairpin RNAs promoted cellular migration. Enzyme-linked immunosorbent assay and endocytosis analysis revealed that CREG knockdown attenuated the internalization and increased secretion of insulin-like growth factor (IGF)-II. Western blot analysis demonstrated that both phosphoinositide 3-kinase (PI3K) and phosphatase Akt were enhanced in CREG knockdown SMCs. Furthermore, the effect of CREG knockdown on SMC migration was abrogated in a dose-dependent manner by the addition of either IGF-II neutralizing antibody or the PI3K inhibitor, LY294002. These results indicate that the CREG knockdown-mediated increase in IGF-II secretion promoted cellular migration in SMCs via the PI3K/Akt signal pathway. Additionally, blockage of IGF-II binding to the mannose-6-phosphate/IGF-II receptor (M6P/IGF2R) by IGF2R antibody or recombinant IGF2R fragment attenuated the endocytosis of IGF-II in cells overexpressing CREG. This indicates that M6P/IGF2R is involved in the regulation of CREG-mediated IGF-II endocytosis. In summary, these data demonstrate for the first time that CREG plays a critical role in the inhibition of SMC migration, as well as maintaining SMCs in a mature phenotype. These results may provide a new therapeutic target for vascular disease associated with neointimal hyperplasia.
Collapse
|
43
|
Tsalavouta M, Astudillo O, Byrnes L, Nolan CM. Regulation of expression of zebrafish(Danio rerio) insulin-like growth factor 2 receptor: implications for evolution at theIGF2Rlocus. Evol Dev 2009; 11:546-58. [DOI: 10.1111/j.1525-142x.2009.00361.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Martin-Kleiner I, Gall Troselj K. Mannose-6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R) in carcinogenesis. Cancer Lett 2009; 289:11-22. [PMID: 19646808 DOI: 10.1016/j.canlet.2009.06.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 06/29/2009] [Accepted: 06/30/2009] [Indexed: 01/18/2023]
Abstract
The cation-independent mannose-6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R) is a multifunctional receptor. It is involved in a variety of cellular processes which become dysregulated in cancer. Its tumor suppressor role was recognized a long time ago. However, due to its multifunctionality, it is not easy to understand the extent of its relevance to normal cellular physiology. Accordingly, it is even more difficult understanding its role in carcinogenesis. This review presents critical and focused highlights of data relating to M6P/IGF2R, obtained during more than 25 years of cancer research.
Collapse
|
45
|
Lübke T, Lobel P, Sleat D. Proteomics of the lysosome. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1793:625-35. [PMID: 18977398 PMCID: PMC2684028 DOI: 10.1016/j.bbamcr.2008.09.018] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 09/24/2008] [Accepted: 09/30/2008] [Indexed: 01/05/2023]
Abstract
Defects in lysosomal function have been associated with numerous monogenic human diseases typically classified as lysosomal storage diseases. However, there is increasing evidence that lysosomal proteins are also involved in more widespread human diseases including cancer and Alzheimer disease. Thus, there is a continuing interest in understanding the cellular functions of the lysosome and an emerging approach to this is the identification of its constituent proteins by proteomic analyses. To date, the mammalian lysosome has been shown to contain approximately 60 soluble luminal proteins and approximately 25 transmembrane proteins. However, recent proteomic studies based upon affinity purification of soluble components or subcellular fractionation to obtain both soluble and membrane components suggest that there may be many more of both classes of protein resident within this organelle than previously appreciated. Discovery of such proteins has important implications for understanding the function and the dynamics of the lysosome but can also lead the way towards the discovery of the genetic basis for human diseases of hitherto unknown etiology. Here, we describe current approaches to lysosomal proteomics and data interpretation and review the new lysosomal proteins that have recently emerged from such studies.
Collapse
Affiliation(s)
- Torben Lübke
- Zentrum Biochemie und Molekulare Zellbiologie, Abteilung Biochemie II, Georg-August Universität Göttingen, 37073 Göttingen, Germany
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine and Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854, USA. Piscataway, NJ 08854, USA
| | - David Sleat
- Center for Advanced Biotechnology and Medicine and Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854, USA. Piscataway, NJ 08854, USA
| |
Collapse
|
46
|
Schähs P, Weidinger P, Probst OC, Svoboda B, Stadlmann J, Beug H, Waerner T, Mach L. Cellular repressor of E1A-stimulated genes is a bona fide lysosomal protein which undergoes proteolytic maturation during its biosynthesis. Exp Cell Res 2008; 314:3036-47. [PMID: 18621046 DOI: 10.1016/j.yexcr.2008.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 06/18/2008] [Accepted: 06/21/2008] [Indexed: 11/20/2022]
Abstract
Cellular repressor of E1A-stimulated genes (CREG) has been reported to be a secretory glycoprotein implicated in cellular growth and differentiation. We now show that CREG is predominantly localized within intracellular compartments. Intracellular CREG was found to lack an N-terminal peptide present in the secreted form of the protein. In contrast to normal cells, CREG is largely secreted by fibroblasts missing both mannose 6-phosphate receptors. This is not observed in cells lacking only one of them. Mass spectrometric analysis of recombinant CREG revealed that the protein contains phosphorylated oligosaccharides at either of its two N-glycosylation sites. Cellular CREG was found to cosediment with lysosomal markers upon subcellular fractionation by density-gradient centrifugation. In fibroblasts expressing a CREG-GFP fusion construct, the heterologous protein was detected in compartments containing lysosomal proteins. Immunolocalization of endogenous CREG confirmed that intracellular CREG is localized in lysosomes. Proteolytic processing of intracellular CREG involves the action of lysosomal cysteine proteinases. These results establish that CREG is a lysosomal protein that undergoes proteolytic maturation in the course of its biosynthesis, carries the mannose 6-phosphate recognition marker and depends on the interaction with mannose 6-phosphate receptors for efficient delivery to lysosomes.
Collapse
Affiliation(s)
- Philipp Schähs
- Institut für Angewandte Genetik und Zellbiologie, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lv K, Guo Y, Zhang Y, Wang K, Jia Y, Sun S. Allele-specific targeting of hsa-miR-657 to human IGF2R creates a potential mechanism underlying the association of ACAA-insertion/deletion polymorphism with type 2 diabetes. Biochem Biophys Res Commun 2008; 374:101-5. [DOI: 10.1016/j.bbrc.2008.06.102] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Accepted: 06/26/2008] [Indexed: 12/21/2022]
|
48
|
Fridman AL, Tainsky MA. Critical pathways in cellular senescence and immortalization revealed by gene expression profiling. Oncogene 2008; 27:5975-87. [PMID: 18711403 DOI: 10.1038/onc.2008.213] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Bypassing cellular senescence and becoming immortal is a prerequisite step in the tumorigenic transformation of a cell. It has long been known that loss of a key tumor suppressor gene, such as p53, is necessary, but not sufficient, for spontaneous cellular immortalization. Therefore, there must be additional mutations and/or epigenetic alterations required for immortalization to occur. Early work on these processes included somatic cell genetic studies to estimate the number of senescence genes, and microcell-mediated transfer of chromosomes into immortalized cells to identify putative senescence-inducing genetic loci. These principal studies laid the foundation for the field of senescence/immortalization, but were labor intensive and the results were somewhat limited. The advent of gene expression profiling and bioinformatics analysis greatly facilitated the identification of genes and pathways that regulate cellular senescence/immortalization. In this review, we present the findings of several gene expression profiling studies and supporting functional data, where available. We identified universal genes regulating senescence/immortalization and found that the key regulator genes represented six pathways: the cell cycle pRB/p53, cytoskeletal, interferon-related, insulin growth factor-related, MAP kinase and oxidative stress pathway. The identification of the genes and pathways regulating senescence/immortalization could provide novel molecular targets for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- A L Fridman
- Department of Pathology, Program in Molecular Biology and Genetics, Barbara Ann Karmanos Cancer Institute, Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | |
Collapse
|
49
|
Han YL, Guo P, Sun MY, Guo L, Luan B, Kang J, Yan CH, Li SH. Secreted CREG inhibits cell proliferation mediated by mannose 6-phosphate/insulin-like growth factor II receptor in NIH3T3 fibroblasts. Genes Cells 2008; 13:977-86. [PMID: 18691225 DOI: 10.1111/j.1365-2443.2008.01221.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cellular repressor of E1A-stimulated genes (CREG) is a recently described glycoprotein that plays a critical role in keeping cells or tissues in mature, homeostatic states. To understand the relationship between CREG and its membrane receptor, mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R), we first generated stable NIH3T3 fibroblasts by transfection of pDS_shCREGs vectors, which produced an approximately 80% decrease in CREG levels both in the lysate and in the media. We used fluorescence activated cell sorting and a bromide deoxyuridine incorporation assay to identify whether CREG knockdown promoted the cell proliferation associated with the increase of IGF-II in NIH3T3 fibroblasts. Proliferation was markedly inhibited in a concentration-dependent manner by re-addition of recombinant CREG protein into the media, and this was mediated by the membrane receptor M6P/IGF2R. We subsequently confirmed the direct interaction of CREG and M6P/IGF2R by both immunoprecipitation-Western blotting and immunofluorescence staining. We found that expression of CREG correlated with localization of the receptor in NIH3T3 fibroblasts but did not affect its expression. Our findings indicated that CREG might act as a functional regulator of M6P/IGF2R to facilitate binding and trafficking of IGF-II endocytosis, leading to growth inhibition.
Collapse
Affiliation(s)
- Ya-Ling Han
- Department of Cardiology, Cardiovascular Research Institute, Shenyang Northern Hospital, Shenyang, China.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Dahms NM, Olson LJ, Kim JJP. Strategies for carbohydrate recognition by the mannose 6-phosphate receptors. Glycobiology 2008; 18:664-78. [PMID: 18621992 DOI: 10.1093/glycob/cwn061] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The two members of the P-type lectin family, the 46 kDa cation-dependent mannose 6-phosphate receptor (CD-MPR) and the 300 kDa cation-independent mannose 6-phosphate receptor (CI-MPR), are ubiquitously expressed throughout the animal kingdom and are distinguished from all other lectins by their ability to recognize phosphorylated mannose residues. The best-characterized function of the MPRs is their ability to direct the delivery of approximately 60 different newly synthesized soluble lysosomal enzymes bearing mannose 6-phosphate (Man-6-P) on their N-linked oligosaccharides to the lysosome. In addition to its intracellular role in lysosome biogenesis, the CI-MPR, but not the CD-MPR, participates in a number of other biological processes by interacting with various molecules at the cell surface. The list of extracellular ligands recognized by this multifunctional receptor has grown to include a diverse spectrum of Man-6-P-containing proteins as well as several non-Man-6-P-containing ligands. Recent structural studies have given us a clearer view of how these two receptors use related, but yet distinct, approaches in the recognition of phosphomannosyl residues.
Collapse
Affiliation(s)
- Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | |
Collapse
|