1
|
Xia L, Li J, Pang Y, Dai C, Xu M, Du Y, Tian Q, Yi L, Wu B, Chen M, Qiu Y, Cheng C, Wang YT, Song W, Dong Z. Disruption of BAG3-mediated BACE1 stabilization alleviates neuropathology and memory deficits in a mouse model of Alzheimer's disease. SCIENCE ADVANCES 2025; 11:eadt7981. [PMID: 40408490 PMCID: PMC12101485 DOI: 10.1126/sciadv.adt7981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 04/18/2025] [Indexed: 05/25/2025]
Abstract
β-Site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) is the rate-limiting enzyme for amyloid-β (Aβ) generation and is considered promising drug target for Alzheimer's disease (AD). The co-chaperone BAG3 (Bcl-2-associated athanogene 3) plays an important role in maintaining intracellular protein homeostasis by regulating heat shock protein 70 (HSP70). Here, we reported that BAG3 expression was significantly elevated in AD. It interacted with and stabilized BACE1 by delaying its degradation through ubiquitin-proteasome and autophagy-lysosomal pathways. BAG3E455K and BAG3R480A mutations reduced their interaction with BACE1. SPOT peptide arrays revealed that BACE1 carboxyl-terminal peptide fragments bound to the RQ domain of BAG3. This interaction can be disrupted by BACE1-derived peptide (Tat-BACE1480-494), leading to decreased BACE1 stability. In APP23/PS45 double transgenic mice, Tat-BACE1480-494 reduced BACE1 levels, decreased Aβ production, and improved synaptic and cognitive deficits. These findings indicate that BAG3 forms complex with HSP70 and BACE1 to stabilize BACE1, suggesting that Tat-BACE1480-494, may represent an ideal class of neuroprotective therapeutics against AD.
Collapse
Affiliation(s)
- Lei Xia
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Junjie Li
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yayan Pang
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Chunfang Dai
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Children Health Care, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Mingliang Xu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yehong Du
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiuyun Tian
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Lilin Yi
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Bin Wu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Mulan Chen
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yiqiong Qiu
- Clinical Laboratory of Changshou District Hospital of Traditional Chinese Medicine, Chongqing 401220, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yu Tian Wang
- Department of Medicine, Brain Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Weihong Song
- Center for Geriatric Medicine, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, The First Affiliated Hospital and Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
2
|
Khan I, Baig MH, Mahfooz S, Rahim M, Karacam B, Elbasan EB, Ulasov I, Dong JJ, Hatiboglu MA. Deciphering the Role of Autophagy in Treatment of Resistance Mechanisms in Glioblastoma. Int J Mol Sci 2021; 22:ijms22031318. [PMID: 33525678 PMCID: PMC7865981 DOI: 10.3390/ijms22031318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a process essential for cellular energy consumption, survival, and defense mechanisms. The role of autophagy in several types of human cancers has been explicitly explained; however, the underlying molecular mechanism of autophagy in glioblastoma remains ambiguous. Autophagy is thought to be a “double-edged sword”, and its effect on tumorigenesis varies with cell type. On the other hand, autophagy may play a significant role in the resistance mechanisms against various therapies. Therefore, it is of the utmost importance to gain insight into the molecular mechanisms deriving the autophagy-mediated therapeutic resistance and designing improved treatment strategies for glioblastoma. In this review, we discuss autophagy mechanisms, specifically its pro-survival and growth-suppressing mechanisms in glioblastomas. In addition, we try to shed some light on the autophagy-mediated activation of the cellular mechanisms supporting radioresistance and chemoresistance in glioblastoma. This review also highlights autophagy’s involvement in glioma stem cell behavior, underlining its role as a potential molecular target for therapeutic interventions.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea;
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Moniba Rahim
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 226026, India;
| | - Busra Karacam
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Elif Burce Elbasan
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey;
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea;
- Correspondence: (J.-J.D.); (M.A.H.)
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey;
- Correspondence: (J.-J.D.); (M.A.H.)
| |
Collapse
|
3
|
Riew TR, Kim S, Jin X, Kim HL, Yoo K, Seo SB, Lee JH, Lee MY. Induction of BIS Protein During Astroglial and Fibrotic Scar Formation After Mitochondrial Toxin-Mediated Neuronal Injury in Rats. Mol Neurobiol 2020; 57:3846-3859. [PMID: 32607834 DOI: 10.1007/s12035-020-02000-6] [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: 03/19/2020] [Accepted: 06/22/2020] [Indexed: 10/24/2022]
Abstract
B cell leukemia/lymphoma-2 (Bcl-2)-interacting death suppressor (BIS), also identified as Bcl-2-associated athanogene 3 (BAG3), has been reported to be upregulated in reactive astrocytes after brain insults. The present study was designed to further substantiate the involvement of BIS protein in the astroglial reaction in the striatum of rats treated with the mitochondrial toxin, 3-nitropropionic acid. Weak constitutive immunoreactivity for BIS was observed in astrocytes in the control striatum, whereas its expression was upregulated, along with that of nestin, in the lesioned striatum. In the lesion core, where astrocytes are virtually absent, BIS/nestin double-labeled cells were associated with the vasculature and were identified as perivascular adventitial fibroblasts. By contrast, BIS/nestin double-labeled cells in the perilesional area were reactive astrocytes, which were confined to the border zone contributing to the formation of the astroglial scar; this was evident 3 days post-lesion and increased thereafter progressively throughout the 28-day experimental period. At the ultrastructural level, BIS protein was diffusely localized throughout the cytoplasm within the stained cells. Collectively, our results demonstrate the phenotypic and functional heterogeneity of BIS-positive cells in the lesioned striatum, suggesting the involvement of BIS in the formation of astroglial scar and its potential role in the development of fibrotic scar after brain insults.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Soojin Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kyunghyun Yoo
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sung Bin Seo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
4
|
Wang JM, Liu BQ, Du ZX, Li C, Sun J, Yan J, Jiang JY, Wang HQ. p53-dependent transcriptional suppression of BAG3 protects cells against metabolic stress via facilitation of p53 accumulation. J Cell Mol Med 2019; 24:562-572. [PMID: 31657880 PMCID: PMC6933324 DOI: 10.1111/jcmm.14764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/22/2019] [Accepted: 09/13/2019] [Indexed: 01/15/2023] Open
Abstract
Solid tumour frequently undergoes metabolic stress during tumour development because of inadequate blood supply and the high nutrient expenditure. p53 is activated by glucose limitation and maintains cell survival via triggering metabolic checkpoint. However, the exact downstream contributors are not completely identified. BAG3 is a cochaperone with multiple cellular functions and is implicated in metabolic reprogramming of pancreatic cancer cells. The current study demonstrated that glucose limitation transcriptionally suppressed BAG3 expression in a p53‐dependent manner. Importantly, hinderance of its down‐regulation compromised cellular adaptation to metabolic stress triggered by glucose insufficiency, supporting that BAG3 might be one of p53 downstream contributors for cellular adaptation to metabolic stress. Our data showed that ectopic BAG3 expression suppressed p53 accumulation via direct interaction under metabolic stress. Thereby, the current study highlights the significance of p53‐mediated BAG3 suppression in cellular adaptation to metabolic stress via facilitating p53 accumulation.
Collapse
Affiliation(s)
- Jia-Mei Wang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Department of Laboratory Medicine, The 1st affiliated Hospital, China Medical University, Shenyang, China
| | - Bao-Qin Liu
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Zhen-Xian Du
- Department of Endocrinology and Metabolism, The 1st affiliated Hospital, China Medical University, Shenyang, China
| | - Chao Li
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Jia Sun
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Jing Yan
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Jing-Yi Jiang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | - Hua-Qin Wang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Myers VD, McClung JM, Wang J, Tahrir FG, Gupta MK, Gordon J, Kontos CH, Khalili K, Cheung JY, Feldman AM. The Multifunctional Protein BAG3: A Novel Therapeutic Target in Cardiovascular Disease. JACC Basic Transl Sci 2018; 3:122-131. [PMID: 29938246 PMCID: PMC6013050 DOI: 10.1016/j.jacbts.2017.09.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The B-cell lymphoma 2–associated anthanogene (BAG3) protein is expressed most prominently in the heart, the skeletal muscle, and in many forms of cancer. In the heart, it serves as a co-chaperone with heat shock proteins in facilitating autophagy; binds to B-cell lymphoma 2, resulting in inhibition of apoptosis; attaches actin to the Z disk, providing structural support for the sarcomere; and links the α-adrenergic receptor with the L-type Ca2+ channel. When BAG3 is overexpressed in cancer cells, it facilitates prosurvival pathways that lead to insensitivity to chemotherapy, metastasis, cell migration, and invasiveness. In contrast, in the heart, mutations in BAG3 have been associated with a variety of phenotypes, including both hypertrophic/restrictive and dilated cardiomyopathy. In murine skeletal muscle and vasculature, a mutation in BAG3 leads to critical limb ischemia after femoral artery ligation. An understanding of the biology of BAG3 is relevant because it may provide a therapeutic target in patients with both cardiac and skeletal muscle disease.
Collapse
Affiliation(s)
- Valerie D Myers
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - JuFang Wang
- Center for Translational Medicine, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Farzaneh G Tahrir
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Manish K Gupta
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Jennifer Gordon
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Christopher H Kontos
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, North Carolina
| | - Kamel Khalili
- Department of Neuroscience, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Joseph Y Cheung
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania.,Center for Translational Medicine, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Arthur M Feldman
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Muhammad K, Rudolf R, Pham DAT, Klein-Hessling S, Takata K, Matsushita N, Ellenrieder V, Kondo E, Serfling E. Induction of Short NFATc1/αA Isoform Interferes with Peripheral B Cell Differentiation. Front Immunol 2018; 9:32. [PMID: 29416540 PMCID: PMC5787671 DOI: 10.3389/fimmu.2018.00032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/04/2018] [Indexed: 11/15/2022] Open
Abstract
In lymphocytes, immune receptor signals induce the rapid nuclear translocation of preformed cytosolic NFAT proteins. Along with co-stimulatory signals, persistent immune receptor signals lead to high levels of NFATc1/αA, a short NFATc1 isoform, in effector lymphocytes. Whereas NFATc1 is not expressed in plasma cells, in germinal centers numerous centrocytic B cells express nuclear NFATc1/αA. When overexpressed in chicken DT40 B cells or murine WEHI 231 B cells, NFATc1/αA suppressed their cell death induced by B cell receptor signals and affected the expression of genes controlling the germinal center reaction and plasma cell formation. Among those is the Prdm1 gene encoding Blimp-1, a key factor of plasma cell formation. By binding to a regulatory DNA element within exon 1 of the Prdm1 gene, NFATc1/αA suppresses Blimp-1 expression. Since expression of a constitutive active version of NFATc1/αA interfered with Prdm1 RNA expression, LPS-mediated differentiation of splenic B cells to plasmablasts in vitro and reduced immunoglobulin production in vivo, one may conclude that NFATc1/αA plays an important role in controlling plasmablast/plasma cell formation.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center (CCC) Mainfranken, University of Würzburg, Würzburg, Germany
| | - Ronald Rudolf
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center (CCC) Mainfranken, University of Würzburg, Würzburg, Germany
| | - Duong Anh Thuy Pham
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center (CCC) Mainfranken, University of Würzburg, Würzburg, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center (CCC) Mainfranken, University of Würzburg, Würzburg, Germany
| | - Katsuyoshi Takata
- Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama University, Okayama, Japan
| | - Nobuko Matsushita
- Laboratory of Molecular Biochemistry, School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Eisaku Kondo
- Division of Oncological Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center (CCC) Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Jennewein L, Ronellenfitsch MW, Antonietti P, Ilina EI, Jung J, Stadel D, Flohr LM, Zinke J, von Renesse J, Drott U, Baumgarten P, Braczynski AK, Penski C, Burger MC, Theurillat JP, Steinbach JP, Plate KH, Dikic I, Fulda S, Brandts C, Kögel D, Behrends C, Harter PN, Mittelbronn M. Diagnostic and clinical relevance of the autophago-lysosomal network in human gliomas. Oncotarget 2018; 7:20016-32. [PMID: 26956048 PMCID: PMC4991435 DOI: 10.18632/oncotarget.7910] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Recently, the conserved intracellular digestion mechanism ‘autophagy’ has been considered to be involved in early tumorigenesis and its blockade proposed as an alternative treatment approach. However, there is an ongoing debate about whether blocking autophagy has positive or negative effects in tumor cells. Since there is only poor data about the clinico-pathological relevance of autophagy in gliomas in vivo, we first established a cell culture based platform for the in vivo detection of the autophago-lysosomal components. We then investigated key autophagosomal (LC3B, p62, BAG3, Beclin1) and lysosomal (CTSB, LAMP2) molecules in 350 gliomas using immunohistochemistry, immunofluorescence, immunoblotting and qPCR. Autophagy was induced pharmacologically or by altering oxygen and nutrient levels. Our results show that autophagy is enhanced in astrocytomas as compared to normal CNS tissue, but largely independent from the WHO grade and patient survival. A strong upregulation of LC3B, p62, LAMP2 and CTSB was detected in perinecrotic areas in glioblastomas suggesting micro-environmental changes as a driver of autophagy induction in gliomas. Furthermore, glucose restriction induced autophagy in a concentration-dependent manner while hypoxia or amino acid starvation had considerably lesser effects. Apoptosis and autophagy were separately induced in glioma cells both in vitro and in vivo. In conclusion, our findings indicate that autophagy in gliomas is rather driven by micro-environmental changes than by primary glioma-intrinsic features thus challenging the concept of exploitation of the autophago-lysosomal network (ALN) as a treatment approach in gliomas.
Collapse
Affiliation(s)
- Lukas Jennewein
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Michael W Ronellenfitsch
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Antonietti
- Experimental Neurosurgery, Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Elena I Ilina
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Jennifer Jung
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Daniela Stadel
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Lisa-Marie Flohr
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Jenny Zinke
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Janusz von Renesse
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Ulrich Drott
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Peter Baumgarten
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Anne K Braczynski
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany
| | - Cornelia Penski
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael C Burger
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany
| | | | - Joachim P Steinbach
- Senckenberg Institute of Neurooncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl-Heinz Plate
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivan Dikic
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Simone Fulda
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Experimental Cancer Research in Pediatrics, Goethe University, Frankfurt am Main, Germany
| | - Christian Brandts
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Donat Kögel
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Experimental Neurosurgery, Department of Neurosurgery, Goethe University, Frankfurt am Main, Germany
| | - Christian Behrends
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Patrick N Harter
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michel Mittelbronn
- Neurological Institute (Edinger Institute), Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
Role of BAG3 in cancer progression: A therapeutic opportunity. Semin Cell Dev Biol 2017; 78:85-92. [PMID: 28864347 DOI: 10.1016/j.semcdb.2017.08.049] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
BAG3 is a multifunctional protein that can bind to heat shock proteins (Hsp) 70 through its BAG domain and to other partners through its WW domain, proline-rich (PXXP) repeat and IPV (Ile-Pro-Val) motifs. Its intracellular expression can be induced by stressful stimuli, while is constitutive in skeletal muscle, cardiac myocytes and several tumour types. BAG3 can modulate the levels, localisation or activity of its partner proteins, thereby regulating major cell pathways and functions, including apoptosis, autophagy, mechanotransduction, cytoskeleton organisation, motility. A secreted form of BAG3 has been identified in studies on pancreatic ductal adenocarcinoma (PDAC). Secreted BAG3 can bind to a specific receptor, IFITM2, expressed on macrophages, and induce the release of factors that sustain tumour growth and the metastatic process. BAG3 neutralisation therefore appears to constitute a novel potential strategy in the therapy of PDAC and, possibly, other tumours.
Collapse
|
9
|
BAG3 promotes stem cell-like phenotype in breast cancer by upregulation of CXCR4 via interaction with its transcript. Cell Death Dis 2017; 8:e2933. [PMID: 28703799 PMCID: PMC5550869 DOI: 10.1038/cddis.2017.324] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/14/2017] [Accepted: 06/08/2017] [Indexed: 01/05/2023]
Abstract
BAG3 is an evolutionarily conserved co-chaperone expressed at high levels and has a prosurvival role in many tumor types. The current study reported that BAG3 was induced under specific floating culture conditions that enrich breast cancer stem cell (BCSC)-like cells in spheres. Ectopic BAG3 overexpression increased CD44+/CD24− CSC subpopulations, first-generation and second-generation mammosphere formation, indicating that BAG3 promotes CSC self-renewal and maintenance in breast cancer. We further demonstrated that mechanically, BAG3 upregulated CXCR4 expression at the post-transcriptional level. Further studies showed that BAG3 interacted with CXCR4 mRNA and promoted its expression via its coding and 3′-untranslational regions. BAG3 was also found to be positively correlated with CXCR4 expression and unfavorable prognosis in patients with breast cancer. Taken together, our data demonstrate that BAG3 promotes BCSC-like phenotype through CXCR4 via interaction with its transcript. Therefore, this study establishes BAG3 as a potential adverse prognostic factor and a therapeutic target of breast cancer.
Collapse
|
10
|
Yan J, Liu C, Jiang JY, Liu H, Li C, Li XY, Yuan Y, Zong ZH, Wang HQ. BAG3 promotes proliferation of ovarian cancer cells via post-transcriptional regulation of Skp2 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28624440 DOI: 10.1016/j.bbamcr.2017.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bcl-2 associated athanogene 3 (BAG3) contains a modular structure, through which BAG3 interacts with a wide range of proteins, thereby affording its capacity to regulate multifaceted biological processes. BAG3 is often highly expressed and functions as a pro-survival factor in many cancers. However, the oncogenic potential of BAG3 remains not fully understood. The cell cycle regulator, S-phase kinase associated protein 2 (Skp2) is increased in various cancers and plays an important role in tumorigenesis. The current study demonstrated that BAG3 promoted proliferation of ovarian cancer cells via upregulation of Skp2. BAG3 stabilized Skp2 mRNA via its 3'-untranslated region (UTR). The current study demonstrated that BAG3 interacted with Skp2 mRNA. In addition, miR-21-5p suppressed Skp2 expression, which was compromised by forced BAG3 expression. These results indicated that at least some oncogenic functions of BAG3 were mediated through posttranscriptional regulation of Skp2 via antagonizing suppressive action of miR-21-5p in ovarian cancer cells.
Collapse
Affiliation(s)
- Jing Yan
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China; Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang 110026, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110026, China
| | - Chuan Liu
- Department of Gynecology & Obstetrics, Sheng Jing Hospital, China Medical University, Shenyang 110005, China
| | - Jing-Yi Jiang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China
| | - Hans Liu
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Chao Li
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China
| | - Xin-Yu Li
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China
| | - Ye Yuan
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China
| | - Zhi-Hong Zong
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China
| | - Hua-Qin Wang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110026, China; Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang 110026, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110026, China.
| |
Collapse
|
11
|
The prosurvival protein BAG3: a new participant in vascular homeostasis. Cell Death Dis 2016; 7:e2431. [PMID: 27763645 PMCID: PMC5133988 DOI: 10.1038/cddis.2016.321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 02/03/2023]
Abstract
Bcl2-associated athanogene 3 (BAG3), is constitutively expressed in a few normal cell types, including myocytes, peripheral nerves and in the brain, and is also expressed in certain tumors. To date, the main studies about the role of BAG3 are focused on its pro-survival effect in tumors through various mechanisms that vary according to cellular type. Recently, elevated concentrations of a soluble form of BAG3 were described in patients affected by advanced stage of heart failure (HF), identifying BAG3 as a potentially useful biomarker in monitoring HF progression. Despite the finding of high levels of BAG3 in the sera of HF patients, there are no data on its possible role on the modulation of vascular tone and blood pressure levels. The aim of this study was to investigate the possible hemodynamic effects of BAG3 performing both in vitro and in vivo experiments. Through vascular reactivity studies, we demonstrate that BAG3 is capable of evoking dose-dependent vasorelaxation. Of note, BAG3 exerts its vasorelaxant effect on resistance vessels, typically involved in the blood pressure regulation. Our data further show that the molecular mechanism through which BAG3 exerts this effect is the activation of the PI3K/Akt signalling pathway leading to nitric oxide release by endothelial cells. Finally, we show that in vivo BAG3 administration is capable of regulating blood pressure and that this is dependent on eNOS regulation since this ability is lost in eNOS KO animals.
Collapse
|
12
|
Kim HY, Kim YS, Yun HH, Im CN, Ko JH, Lee JH. ERK-mediated phosphorylation of BIS regulates nuclear translocation of HSF1 under oxidative stress. Exp Mol Med 2016; 48:e260. [PMID: 27659916 PMCID: PMC5050300 DOI: 10.1038/emm.2016.84] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 12/28/2022] Open
Abstract
B-cell lymphoma (BCL)-2-interacting cell death suppressor (BIS) has diverse cellular functions depending on its binding partners. However, little is known about the effects of biochemical modification of BIS on its various activities under oxidative stress conditions. In this study, we showed that H2O2 reduced BIS mobility on SDS–polyacrylamide gels in a time-dependent manner via the activation of extracellular signaling-regulated kinase (ERK). The combined results of mass spectroscopy and computational prediction identified Thr285 and Ser289 in BIS as candidate residues for phosphorylation by ERK under oxidative stress conditions. Deletion of these sites resulted in a partial reduction in the H2O2-induced mobility shift relative to that of the wild-type BIS protein; overexpression of the deletion mutant sensitized A172 cells to H2O2-induced cell death without increasing the level of intracellular reactive oxygen species. Expression of the BIS deletion mutant decreased the level of heat shock protein (HSP) 70 mRNA following H2O2 treatment, which was accompanied by impaired nuclear translocation of heat shock transcription factor (HSF) 1. Co-immunoprecipitation assays revealed that the binding of wild-type BIS to HSF1 was decreased by oxidative stress, while the binding of the BIS deletion mutant to HSF1 was not affected. These results indicate that ERK-dependent phosphorylation of BIS has a role in the regulation of nuclear translocation of HSF1 likely through modulation of its interaction affinity with HSF1, which affects HSP70 expression and sensitivity to oxidative stress.
Collapse
Affiliation(s)
- Hye Yun Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-Sam Kim
- Aging Intervention Research Center, Aging Research Institute, KRIBB, Daejeon, Republic of Korea.,Korea University of Science and Technology. Daejeon, Republic of Korea
| | - Hye Hyeon Yun
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang-Nim Im
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Heon Ko
- Aging Intervention Research Center, Aging Research Institute, KRIBB, Daejeon, Republic of Korea.,Korea University of Science and Technology. Daejeon, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
13
|
Yoo HJ, Im CN, Youn DY, Yun HH, Lee JH. Bis is Induced by Oxidative Stress via Activation of HSF1. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:403-9. [PMID: 25352760 PMCID: PMC4211124 DOI: 10.4196/kjpp.2014.18.5.403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/24/2014] [Accepted: 07/26/2014] [Indexed: 12/15/2022]
Abstract
The Bis protein is known to be involved in a variety of cellular processes including apoptosis, migration, autophagy as well as protein quality control. Bis expression is induced in response to a number of types of stress, such as heat shock or a proteasome inhibitor via the activation of heat shock factor (HSF)1. We report herein that Bis expression is increased at the transcriptional level in HK-2 kidney tubular cells and A172 glioma cells by exposure to oxidative stress such as H2O2 treatment and oxygen-glucose deprivation, respectively. The pretreatment of HK-2 cells with N-acetyl cysteine, suppressed Bis induction. Furthermore, HSF1 silencing attenuated Bis expression that was induced by H2O2, accompaniedby increase in reactive oxygen species (ROS) accumulation. Using a series of deletion constructs of the bis gene promoter, two putative heat shock elements located in the proximal region of the bis gene promoter were found to be essential for the constitutive expression is as well as the inducible expression of Bis. Taken together, our results indicate that oxidative stress induces Bis expression at the transcriptional levels via activation of HSF1, which might confer an expansion of antioxidant capacity against pro-oxidant milieu. However, the possible role of the other cis-element in the induction of Bis remains to be determined.
Collapse
Affiliation(s)
- Hyung Jae Yoo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Chang-Nim Im
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea. ; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea. ; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Dong-Ye Youn
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Hye Hyeon Yun
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea. ; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea. ; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea. ; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
14
|
Lim JH, Youn DY, Yoo HJ, Yoon HH, Kim MY, Chung S, Kim YS, Chang YS, Park CW, Lee JH. Aggravation of diabetic nephropathy in BCL-2 interacting cell death suppressor (BIS)-haploinsufficient mice together with impaired induction of superoxide dismutase (SOD) activity. Diabetologia 2014; 57:214-23. [PMID: 24078136 DOI: 10.1007/s00125-013-3064-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/05/2013] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS B cell CLL/lymphoma 2 (BCL-2)-interacting cell death suppressor (BIS), known as an anti-stress and anti-apoptotic protein, has been reported to modulate susceptibility to oxidative stress. This study investigated the potential role of BIS as an antioxidant protein in diabetic nephropathy. METHODS Diabetes was induced in BIS-heterozygote (BIS-HT) mice via streptozotocin injections and the resulting phenotypes were compared with those of BIS-wild-type (BIS-WT) mice over the 20 weeks following diabetes induction. RESULTS Renal injuries, represented by increased plasma creatinine levels and increased albuminuria, were greater in diabetic BIS-HT mice than in diabetic BIS-WT mice, and were accompanied by a significant increase in reactive oxygen species (ROS) and oxidative stress markers. Moreover, renal pathological changes and the apoptotic process were accelerated in diabetic BIS-HT mice compared with diabetic BIS-WT mice with the same degree of hyperglycaemia; all were restored by 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (tempol) treatment. The levels of NADPH oxidase and related proteins were not significantly higher in diabetic BIS-HT mice compared with diabetic BIS-WT mice. However, levels of superoxide dismutase (SOD)1 and SOD2 increased on the induction of diabetes in BIS-WT mice but not in BIS-HT mice, correlating with the total SOD activity. An in vitro study showed that knockdown of BIS production also resulted in impaired induction of SOD activity as well as SOD levels in HK-2 and NMS cells, concomitant with significant ROS accumulation. CONCLUSION/INTERPRETATION Our results suggest that the decreased antioxidant capacity of BIS aggravates diabetic nephropathy in diabetic BIS-HT mice, possibly as a result of the disruption in the regulation of SOD protein quality under oxidative stress.
Collapse
|
15
|
Lee YD, Yoon JS, Yoon HH, Youn HJ, Kim J, Lee JH. Expression of Bis in the mouse gastrointestinal system. Anat Cell Biol 2012; 45:160-9. [PMID: 23094204 PMCID: PMC3472142 DOI: 10.5115/acb.2012.45.3.160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/08/2012] [Accepted: 09/19/2012] [Indexed: 12/13/2022] Open
Abstract
The Bcl-2 interacting death suppressor (Bis) protein is known to be involved in a variety of pathophysiological conditions. We recently generated bis-deficient mice, which exhibited early lethality with typical nutritional deprivation status. To further investigate the molecular basis for the malnutrition phenotype of bis deficient mice, we explored Bis expression in the digestive system of normal mice. Western blot analysis and quantitative real time reverse transcription polymerase chain reaction analysis indicated that Bis expression is highest in the esophagus, followed by the stomach, colon, jejunum and ileum. Immunohistochemical data indicated that Bis expression is restricted to the stratified squamous epitheliums in the esophagus and forestomach, and was not notable in the columnar epitheliums in the stomach, small intestine and colon. In addition, strong Bis immunoreactivity was detected in the striated muscles surrounding the esophagus and smooth muscles at a lesser intensity throughout the gastrointestinal (GI) tract. Ganglionated plexuses, located in submucous layers, as well as intermuscular layers, were specifically immunoreactive for Bis. Immunofluorescence studies revealed that Bis is co-localized in glial fibrillary acidic protein-expressing enteric glial cells. Immunostaining with neuron specific esterase antibodies indicate that Bis is also present in the cell bodies of ganglions in the enteric nervous system (ENS). Our findings indicate that Bis plays a role in regulating GI functions, such as motility and absorption, through modulating signal transmission between the ENS and smooth muscles or the intestinal epitheliums.
Collapse
Affiliation(s)
- Young Dae Lee
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
16
|
Cho KO, Lee KE, Youn DY, Jeong KH, Kim JY, Yoon HH, Lee JH, Kim SY. Decreased vulnerability of hippocampal neurons after neonatal hypoxia-ischemia in bis-deficient mice. Glia 2012; 60:1915-29. [PMID: 22907804 DOI: 10.1002/glia.22407] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 07/24/2012] [Indexed: 11/07/2022]
Abstract
The Bcl-2-interacting death suppressor (Bis) protein is involved in antiapoptosis and antistress pathways. However, its roles after neonatal hypoxia-ischemia remain obscure. Therefore, we investigated the effects of Bis deletion on hippocampal cell death following neonatal hypoxia-ischemia. We transected the right common carotid artery of bis(+/+) and bis(-/-) mice at postnatal Day 7 and subjected them to hypoxia for 35 min. Cresyl violet staining showed that hypoxia-ischemia induced progressive cell death in the hippocampi of bis(+/+) mice. Moreover, Bis was expressed in astrocytes, not microglia, in sham-manipulated hippocampi of bis(+/+) mice, and was markedly enhanced after hypoxia-ischemia. Immunoblotting showed that Bis expression significantly increased 3 and 7 days following hypoxia-ischemia. Unexpectedly, 7 days after hypoxia-ischemia, the number of hippocampal NeuN-positive cells was higher in the bis(-/-) mice than in the bis(+/+) mice. We subsequently performed transcriptomic analysis and quantitative real time polymerase chain reaction to search for the underlying genes responsible for resistance to hypoxia-ischemia in the bis(-/-) hippocampus. These studies showed that 6 h after hypoxia-ischemia, galectin 3 and filamin C levels increased to a lesser extent in the bis(-/-) hippocampi compared with the bis(+/+) hippocampi. Finally, our in vitro hypoxia-ischemia model, using A172 glioma cells and primary astrocytes, showed that downregulation of Bis blocked the enhanced expression of galectin 3 after oxygen-glucose deprivation. This study demonstrated that Bis was upregulated in the astrocytes after hypoxia-ischemia. In addition, we showed that hippocampal neurons are less vulnerable to hypoxia-ischemia in mice lacking Bis, possibly because of the modulation of galectin 3 induction.
Collapse
Affiliation(s)
- Kyung-Ok Cho
- Department of Pharmacology, Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Role of BAG3 protein in leukemia cell survival and response to therapy. Biochim Biophys Acta Rev Cancer 2012; 1826:365-9. [PMID: 22710027 DOI: 10.1016/j.bbcan.2012.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/05/2012] [Accepted: 06/08/2012] [Indexed: 01/19/2023]
Abstract
The ability of BAG3, a member of the BAG family of heat shock protein (Hsp) 70 - cochaperones, to sustain the survival of human primary B-CLL and ALL cells was recognized about nine years ago. Since then, the anti-apoptotic activity of BAG3 has been confirmed in other tumor types, where it has been shown to regulate the intracellular concentration and localization of apoptosis-regulating factors, including NF-κB-activating (IKKγ) and Bcl2-family (Bax) proteins. Furthermore, growing evidences support its role in lymphoid and myeloid leukemia response to therapy. Moreover in the last years, the contribution of BAG3 to autophagy, a process known to be involved in the pathogenesis and response to therapy of leukemia cells, has been disclosed, opening a new avenue for the interpretation of the role of this protein in leukemias' biology.
Collapse
|
18
|
Youn DY, Yoon JS, Kim YK, Yeum CE, Lee SB, Youn HJ, Tsujimoto Y, Lee JH. Deletion of the bis gene results in a marked increase in the production of corticosterone that is associated with thymic atrophy in mice. Am J Physiol Endocrinol Metab 2011; 301:E223-31. [PMID: 21540452 DOI: 10.1152/ajpendo.00604.2010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Bis (Bag3) is known to be involved in cell survival, migration, the regulating of chaperones, and protein quality control. We reported recently on the production of bis gene-deleted mice, which show early lethality within 3 wk after birth with a phenotype showing severe malnutrition and shrinkage of the thymus. In this report, we provide evidence to show that an intrinsic problem of adrenal gland is the the primary cause for the severe atrophy of the thymus in bis(-/-) mice. The bis(-/-) mice show significantly higher levels of corticosterone, but CRH and ACTH levels were considerably lower than those of wild littermates. The transcription of steroidogenic enzymes was increased in the adrenal glands of bis(-/-) mice, accompanied by an increase in the thickness of the zona reticularis. An analysis of thymus tissue from bis(-/-) mice revealed that the severe atrophy of the thymus is due to the specific loss of immature double-positive (CD4(+)CD8(+)) cortical thymocytes by apoptosis, as evidenced by immunohistochemical examination and flow cytometric analysis, which were restored by injection of an inhibitor of glucocorticoid synthesis. In vitro cultures of thymocytes with increasing doses of dexamethasone exhibited a similar degree of apoptosis between wild and bis(-/-) thymocytes. The corticosterone levels from fasted wild littermates were one-half those of bis(-/-) mice, although serum glucose levels were similar. Thus, the deletion of the bis gene resulted in the intrinsic defect in the adrenal gland, leading to a marked increase in glucocorticoid levels, probably upon starvation stress, which accounts for the massive apoptosis of the thymus.
Collapse
Affiliation(s)
- Dong-Ye Youn
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Gout E, Gutkowska M, Takayama S, Reed JC, Chroboczek J. Co-chaperone BAG3 and adenovirus penton base protein partnership. J Cell Biochem 2011; 111:699-708. [PMID: 20607728 PMCID: PMC7166384 DOI: 10.1002/jcb.22756] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The BAG family of Hsp70/Hsc70 co‐chaperones is characterised by the presence of a conserved BAG domain at the carboxyl‐terminus. BAG3 protein is the only member of this family containing also the N‐terminally located WW domain. We describe here the identification of adenovirus (Ad) penton base protein as the first BAG3 partner recognising BAG3 WW domain. Ad penton base is the viral capsid constituent responsible for virus internalisation. It contains in the N‐terminal part two conserved PPxY motifs, known ligands of WW domains. In cells producing Ad penton base protein, cytoplasmic endogenous BAG3 interacts with it and co‐migrates to the nucleus. Preincubation of BAG3 with Ad base protein results in only slight modulation of BAG3 co‐chaperone activity, suggesting that this interaction is not related to the classical BAG3 co‐chaperone function. However, depletion of BAG3 impairs the cell entry of the virus and viral progeny production in Ad‐infected cells, suggesting that the interaction between virus penton base protein and cellular co‐chaperone BAG3 positively influences virus life cycle. These results thus demonstrate a novel host–pathogen interaction, which contributes to the successful infectious life cycle of adenoviruses. In addition, these data enrich our knowledge about the multifunctionality of the BAG3 co‐chaperone. J. Cell. Biochem. 111: 699–708, 2010. © 2010 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
- E Gout
- Institut de Biologie Structurale, 41 rue Jules Horowitz, CEA, CNRS, Université Joseph Fourier, 38027 Grenoble, France
| | | | | | | | | |
Collapse
|
20
|
Gentilella A, Khalili K. BAG3 expression is sustained by FGF2 in neural progenitor cells and impacts cell proliferation. Cell Cycle 2010; 9:4245-7. [PMID: 20962586 DOI: 10.4161/cc.9.20.13517] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
21
|
Jung SE, Kim YK, Youn DY, Lim MH, Ko JH, Ahn YS, Lee JH. Down-modulation of Bis sensitizes cell death in C6 glioma cells induced by oxygen–glucose deprivation. Brain Res 2010; 1349:1-10. [DOI: 10.1016/j.brainres.2010.06.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 06/15/2010] [Accepted: 06/18/2010] [Indexed: 12/28/2022]
|
22
|
Chalothorn D, Faber JE. Strain-dependent variation in collateral circulatory function in mouse hindlimb. Physiol Genomics 2010; 42:469-79. [PMID: 20551146 DOI: 10.1152/physiolgenomics.00070.2010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The extent (density and diameter) of the native (preexisting) collateral circulation in healthy tissues and the capacity of collaterals to enlarge/remodel in obstructive arterial disease are important determinants of ischemic injury. Evidence suggests that these parameters vary widely from yet-to-be-identified genetic and environmental factors. Recently, a locus on chromosome 7 was linked to less recovery of perfusion after femoral artery ligation in BALB/c and A/J versus C57BL/6 mouse strains. Moreover, evidence suggested that BALB/c and A/J share an allele(s) at this locus that is different from C57BL/6 mice. Here we tested the hypothesis that differences in collateral extent and/or remodeling underlie these findings. Compared with C57BL/6, BALB/c and A/J strains have fewer native collaterals in hindlimb (also confirmed in brain)-associated with greater reduction in perfusion immediately after femoral ligation, slower recovery of perfusion, greater hindlimb use impairment, and worse ischemia. However, A/J also differed from BALB/c in a number of these parameters, including having more robust collateral remodeling. Analysis of A/J --> C57BL/6 chromosome substitution strains confirmed that a difference in an allele(s) on chromosome 7 conferred most, but not all, of the magnitude of the differences in collateral function. Additional studies of C57BL/6 x BALB/c F1 mice demonstrated that alleles of the C57BL/6 strain exert dominance for collateral traits. Finally, negative results were obtained from studies examining a previously identified candidate gene potentially responsible for these differences-Bcl2-associated athanogene-3. These findings emphasize the major contribution of genetic background to variation in the collateral circulation and its capacity to lessen ischemia in obstructive disease.
Collapse
Affiliation(s)
- Dan Chalothorn
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, North Carolina 27599-7545, USA
| | | |
Collapse
|
23
|
Yoon JS, Lee MY, Lee JS, Park CS, Youn HJ, Lee JH. Bis is involved in glial differentiation of p19 cells induced by retinoic Acid. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2009; 13:251-6. [PMID: 19885044 DOI: 10.4196/kjpp.2009.13.3.251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 06/09/2009] [Accepted: 06/12/2009] [Indexed: 11/15/2022]
Abstract
Previous observations suggest that Bis, a Bcl-2-binding protein, may play a role the neuronal and glial differentiation in vivo. To examine this further, we investigated Bis expression during the in vitro differentiation of P19 embryonic carcinoma cells induced by retinoic acid (RA). Western blotting and RT-PCR assays showed that Bis expression was temporarily decreased during the free floating stage and then began to increase on day 6 after the induction of differentiation. Double immunostaining indicated that Bis-expressing cells do not express several markers of differentiation, including NeuN, MAP-2 and Tuj-1. However, some of the Bis-expressing cells also were stained with GFAP-antibodies, indicating that Bis is involved glial differentiation. Using an shRNA strategy, we developed bis-knock down P19 cells and compared them with control P19 cells for the expression of NeuroD, Mash-1 and GFAP during RA-induced differentiation. Among these, only GFAP induction was significantly attenuated in P19-dnbis cells and the population showing GFAP immunoreactivity was also decreased. It is noteworthy that distribution of mature neurons and migrating neurons was disorganized, and the close association of migrating neuroblasts with astrocytes was not observed in P19-dnbis cells. These results suggest that Bis is involved in the migration-inducing activity of glial cells.
Collapse
Affiliation(s)
- Jung-Sook Yoon
- Department of Biomedical Science, Graduate School, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | |
Collapse
|
24
|
Choi JS, Lee JH, Shin YJ, Lee JY, Yun H, Chun MH, Lee MY. Transient expression of Bis protein in midline radial glia in developing rat brainstem and spinal cord. Cell Tissue Res 2009; 337:27-36. [DOI: 10.1007/s00441-009-0794-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 03/17/2009] [Indexed: 10/20/2022]
|
25
|
Rosati A, Khalili K, Deshmane SL, Radhakrishnan S, Pascale M, Turco MC, Marzullo L. BAG3 protein regulates caspase-3 activation in HIV-1-infected human primary microglial cells. J Cell Physiol 2009; 218:264-7. [PMID: 18821563 PMCID: PMC4503248 DOI: 10.1002/jcp.21604] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BAG3, a member of the BAG co-chaperones family, is expressed in several cell types subjected to stressful conditions, such as exposure to high temperature, heavy metals, drugs. Furthermore, it is constitutively expressed in some tumors. Among the biological activities of the protein, there is apoptosis downmodulation; this appears to be exerted through BAG3 interaction with the heat shock protein (Hsp) 70, that influences cell apoptosis at several levels. We recently reported that BAG3 protein was detectable in the cytoplasm of reactive astrocytes in HIV-1-associated encephalopathy biopsies. Here we report that downmodulation of BAG3 protein levels allows caspase-3 activation by HIV-1 infection in human primary microglial cells. This is the first reported evidence of a role for BAG3 in the balance of death versus survival during viral infection.
Collapse
Affiliation(s)
- Alessandra Rosati
- Department of Pharmaceutical Sciences (DiFarma), University of Salerno, Salerno, Italy
| | - Kamel Khalili
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Satish L. Deshmane
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Sujatha Radhakrishnan
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Maria Pascale
- Department of Pharmaceutical Sciences (DiFarma), University of Salerno, Salerno, Italy
| | - M. Caterina Turco
- Department of Pharmaceutical Sciences (DiFarma), University of Salerno, Salerno, Italy
| | | |
Collapse
|
26
|
Youn DY, Lee DH, Lim MH, Yoon JS, Lim JH, Jung SE, Yeum CE, Park CW, Youn HJ, Lee JS, Lee SB, Ikawa M, Okabe M, Tsujimoto Y, Lee JH. Bis deficiency results in early lethality with metabolic deterioration and involution of spleen and thymus. Am J Physiol Endocrinol Metab 2008; 295:E1349-57. [PMID: 18840758 DOI: 10.1152/ajpendo.90704.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bcl-2 interacting cell death suppressor (Bis), also known as Bag3 or CAIR-1, is involved in antistress and antiapoptotic pathways. In addition to Bcl-2, Bis binds to several proteins, suggesting it has diverse functions in normal and pathological conditions. To better define the physiological function of Bis in vivo, we developed bis-deficient mice with a cre-loxP system. Targeted disruption of exon 4 of the bis gene was demonstrated by Southern blotting and PCR, and Western blotting showed that no intact or truncated Bis protein was synthesized in bis(-/-) mice. While heterozygotes were fertile and appeared normal, Bis-deficient mice showed growth retardation and died by 3 wk after birth. The relative weight of the thymus and spleen was reduced and the total numbers of white blood cells, splenocytes, and thymocytes were significantly reduced compared with wild-type littermates. Serum profiles indicated significant hypoglycemia as well as decrease in triglyceride and cholesterol levels. Expression profiles of metabolic genes indicated that gluconeogenesis and beta-oxidation are activated in the liver of bis(-/-) mice. This activation, as well as a decrease in peripheral fat and an induction of fatty liver, appears to be an adaptive response to hypoglycemia. Our study reveals that the absence of Bis has considerable influences on postnatal growth and survival, possibly due to a nutritional impairment.
Collapse
Affiliation(s)
- Dong-Ye Youn
- Department of Biochemistry, Catholic University of Korea, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang HQ, Zhang HY, Hao FJ, Meng X, Guan Y, Du ZX. Induction of BAG2 protein during proteasome inhibitor-induced apoptosis in thyroid carcinoma cells. Br J Pharmacol 2008; 155:655-60. [PMID: 18660828 DOI: 10.1038/bjp.2008.302] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Proteasome inhibitors exhibit cytotoxic against tumours of different histology. However, the mechanism of apoptosis induction by these compounds remains unclear and is likely to be a complex cascade of events. Bcl-2-associated athanogene (BAG) family proteins are characterized by their property of interaction with a variety of partners involved in modulating the proliferation/death balance, including heat shock proteins (HSP), Bcl-2, Raf-1. The role of BAG family proteins in proteasome inhibition has not been elucidated. EXPERIMENTAL APPROACH Effects of proteasome inhibitors on BAG2 expression were evaluated using real-time reverse transcription-polymerase chain reaction (RT-PCR). BAG2 expression was knocked down by small interfering RNAs (siRNA). Cell death was evaluated using Annexin V/propidium iodide staining and subsequent FACS. KEY RESULTS The proteasome inhibitors, MG132, PSI, lactacystin and epoxomicin, induced BAG2 at the transcriptional level. MG132-induced apoptosis was significantly suppressed by BAG2 knockdown using RNA interference. CONCLUSIONS AND IMPLICATIONS Our results suggest that BAG2 is a novel molecule induced by proteasome inhibition, which exhibits a pro-apoptotic property in death of thyroid cancer cells induced by proteasome inhibition.
Collapse
Affiliation(s)
- H-Q Wang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
The co-chaperone protein, BAG3, which belongs to the BAG protein family, has an established antiapoptotic function in different tumor cell lines. Here we demonstrated that treatment of the human neuroblastoma cell line, SK-N-MC, with fibroblast growth factor-2 (FGF-2) results in induction of BAG3 expression. Induction of BAG3 protein by FGF-2 occurs at the transcriptional level; it requires the extracellular regulated kinase1/2 pathway and is dependent on the activity of Egr-1 upon the BAG3 promoter. Targeted suppression of BAG3 by small-interfering RNA results in dysregulation of cell-cycle progression most notably at S and G(2) phases, which corroborates the decreased level of cyclin B1 expression. These observations suggest a new role for BAG3 in regulation of the cell cycle.
Collapse
|
29
|
Kobayashi MS, Asai S, Ishikawa K, Nishida Y, Nagata T, Takahashi Y. Global profiling of influence of intra-ischemic brain temperature on gene expression in rat brain. ACTA ACUST UNITED AC 2008; 58:171-91. [PMID: 18440647 DOI: 10.1016/j.brainresrev.2008.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2007] [Revised: 02/08/2008] [Accepted: 03/08/2008] [Indexed: 12/20/2022]
Abstract
Mild to moderate differences in brain temperature are known to greatly affect the outcome of cerebral ischemia. The impact of brain temperature on ischemic disorders has been mainly evaluated through pathological analysis. However, no comprehensive analyses have been conducted at the gene expression level. Using a high-density oligonucleotide microarray, we screened 24000 genes in the hippocampus under hypothermic (32 degrees C), normothermic (37 degrees C), and hyperthermic (39 degrees C) conditions in a rat ischemia-reperfusion model. When the ischemic group at each intra-ischemic brain temperature was compared to a sham-operated control group, genes whose expression levels changed more than three-fold with statistical significance could be detected. In our screening condition, thirty-three genes (some of them novel) were obtained after screening, and extensive functional surveys and literature reviews were subsequently performed. In the hypothermic condition, many neuroprotective factor genes were obtained, whereas cell death- and cell damage-associated genes were detected as the brain temperature increased. At all intra-ischemic brain temperatures, multiple molecular chaperone genes were obtained. The finding that intra-ischemic brain temperature affects the expression level of many genes related to neuroprotection or neurotoxicity coincides with the different pathological outcomes at different brain temperatures, demonstrating the utility of the genetic approach.
Collapse
Affiliation(s)
- Megumi Sugahara Kobayashi
- Division of Genomic Epidemiology and Clinical Trials, Advanced Medical Research Center, Nihon University School of Medicine, Oyaguchi-Kami Machi, Itabashi-ku, Tokyo 173-8610, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Du ZX, Meng X, Zhang HY, Guan Y, Wang HQ. Caspase-dependent cleavage of BAG3 in proteasome inhibitors-induced apoptosis in thyroid cancer cells. Biochem Biophys Res Commun 2008; 369:894-8. [PMID: 18325327 DOI: 10.1016/j.bbrc.2008.02.112] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 02/23/2008] [Indexed: 01/10/2023]
Abstract
Proteasome inhibitors are emerging as effective drugs for the treatment of relapsed/refractory multiple myeloma and possibly some solid tumors. Bcl-2-associated athanogene 3 (BAG3) is a survival protein that has been shown to be stimulated during cell response to stressful conditions, such as exposure to high temperature, heavy metals. We have recently demonstrated that BAG3 is also induced by proteasome inhibitors at the transcriptional level and the induction of BAG3 by proteasome inhibition is antiapoptotic. Here, we demonstrated that although proteasome inhibitors triggered similar upregulation of BAG3 transcript in sensitive and insensitive thyroid cancer cells, persistent increase of BAG3 protein was detected in insensitive cells, whereas less increase or even decrease was observed in sensitive cells. Notably, decrease of BAG3 protein was associated with the appearance of a BAG3 fragment of approximately 40kDa, which appeared to be caspase-dependent. Therefore, caspase-dependent cleavage of BAG3 might facilitate apoptosis in sensitive cells.
Collapse
Affiliation(s)
- Zhen-Xian Du
- Department of Endocrinology and Metabolism, The 1st Affiliated Hospital, China Medical University, Shenyang 110001, China
| | | | | | | | | |
Collapse
|
31
|
Yao S, Peng M, Zhu X, Cheng M, Qi X. Heat shock protein72 protects hippocampal neurons from apoptosis induced by chronic psychological stress. Int J Neurosci 2007; 117:1551-64. [PMID: 17917925 DOI: 10.1080/00207450701239285] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
When exposed to nonlethal heat stress (i.e., heat shock preconditioning), HSP72 expression increased in the mammalian brain. HSP72 enhance the viability of neurons and decrease TUNEL-positive neurons under several kinds of stress (e.g., ischemic). Chronic psychological stress is a kind of stress that could cause hippocampal neuron apoptosis. But whether overexpression of HSP72 can decrease TUNEL-positive hippocampal neurons caused by chronic psychological stress is unclear. To investigate the possible protective role of HSP72 in decreasing chronic psychological stress-induced hippocampal neuron apoptosis, this study analyzed HSP72 expression, apoptotic neurons in the hippocampus of mice. Adult mice were divided into four groups unstressed group; chronic psychological stress group; heat shock stress group; heat shock preconditioning plus psychological stress group; receiving no experimental stress, chronic psychological stress, heat shock stress, heat shock preconditioning plus psychological stress separately. Mice were killed after one month, two months, or three months of stress. A three-way ANOVA (psychological stress x heat shock stress x time) revealed a significant effect of heat shock stress in increasing HSP72 expression, decreasing neuronal apoptosis in hippocampus CA3 region caused by chronic psychological stress, and showed that HSP72 protected hippocampus CA3 neurons from chronic psychological stress.
Collapse
Affiliation(s)
- Shuqiao Yao
- The Medical Psychological Research Center 2nd Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.
| | | | | | | | | |
Collapse
|
32
|
Wang HQ, Liu HM, Zhang HY, Guan Y, Du ZX. Transcriptional upregulation of BAG3 upon proteasome inhibition. Biochem Biophys Res Commun 2007; 365:381-5. [PMID: 17996194 DOI: 10.1016/j.bbrc.2007.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 11/01/2007] [Indexed: 10/22/2022]
Abstract
Proteasome inhibitors exhibit antitumoral activity against malignancies of different histology. Emerging evidence indicates that antiapoptotic factors may also accumulate as a consequence of exposure to these drugs, thus it seems plausible that activation of survival signaling cascades might compromise their antitumoral effects. Bcl-2-associated athanogene (BAG) family proteins are characterized by their property of interaction with a variety of partners involved in modulating the proliferation/death balance, including heat shock proteins (HSP), Bcl-2, Raf-1. In this report, we demonstrated that BAG3 is a novel antiapoptotic molecule induced by proteasome inhibitors in various cancer cells at the transcriptional level. Moreover, we demonstrated that BAG3 knockdown by siRNA sensitized cancer cells to MG132-induced apoptosis. Taken together, our results suggest that BAG3 induction might represents as an unwanted molecular consequence of utilizing proteasome inhibitors to combat tumors.
Collapse
Affiliation(s)
- Hua-Qin Wang
- Department of Biochemistry & Molecular Biology, China Medical University, Shenyang 110001, China.
| | | | | | | | | |
Collapse
|
33
|
Rosati A, Ammirante M, Gentilella A, Basile A, Festa M, Pascale M, Marzullo L, Belisario MA, Tosco A, Franceschelli S, Moltedo O, Pagliuca G, Lerose R, Turco MC. Apoptosis inhibition in cancer cells: A novel molecular pathway that involves BAG3 protein. Int J Biochem Cell Biol 2007; 39:1337-42. [PMID: 17493862 DOI: 10.1016/j.biocel.2007.03.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 02/06/2007] [Accepted: 03/07/2007] [Indexed: 12/16/2022]
Abstract
Stress-induced apoptosis regulates neoplasia pathogenesis and response to therapy. Indeed, cell transformation induces a stress response, that is overcome, in neoplastic cells, by alterations in apoptosis modulators; on the other hand, antineoplastic therapies largely trigger the apoptosis stress pathway, whose impairment results in resistance. Therefore, the study of the roles of apoptosis-modulating molecules in neoplasia development and response to therapy is of key relevance for our understanding of these processes. Among molecules that regulate apoptosis, a role is emerging for BAG3, a member of the BAG co-chaperone protein family. Proteins that share the BAG domain are characterized by their interaction with a variety of partners (heat shock proteins, steroid hormone receptors, Raf-1 and others), involved in regulating a number of cellular processes, including proliferation and apoptosis. BAG3, also known as CAIR-1 or Bis, forms a complex with the heat shock protein (Hsp) 70. This assists polypeptide folding, can mediate protein delivery to proteasome and is able to modulate apoptosis by interfering with cytochrome c release, apoptosome assembly and other events in the death process. It has been recently shown that, in human primary lymphoid and myeloblastic leukemias and other neoplastic cell types, BAG3 expression sustains cell survival and underlies resistance to therapy, through downmodulation of apoptosis. This review summarizes findings that assign an apoptotic role to BAG3 in some neoplastic cell types and identify the protein as a candidate target of therapy.
Collapse
Affiliation(s)
- Alessandra Rosati
- Department of Pharmaceutical Sciences, University of Salerno, via ponte don Melillo, 84084 Fisciano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Homma S, Iwasaki M, Shelton GD, Engvall E, Reed JC, Takayama S. BAG3 deficiency results in fulminant myopathy and early lethality. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:761-73. [PMID: 16936253 PMCID: PMC1698816 DOI: 10.2353/ajpath.2006.060250] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bcl-2-associated athanogene 3 (BAG3) is a member of a conserved family of cyto-protective proteins that bind to and regulate Hsp70 family molecular chaperones. Here, we show that BAG3 is prominently expressed in striated muscle and colocalizes with Z-disks. Mice with homozygous disruption of the bag3 gene developed normally but deteriorated postnatally with stunted growth evident by 1 to 2 weeks of age and death by 4 weeks. BAG3-deficient animals developed a fulminant myopathy characterized by noninflammatory myofibrillar degeneration with apoptotic features. Knockdown of bag3 expression in cultured C2C12 myoblasts increased apoptosis on induction of differentiation, suggesting a need for bag3 for maintenance of myotube survival and confirming a cell autonomous role for bag3 in muscle. We conclude that although BAG3 is not required for muscle development, this co-chaperone appears to be critically important for maintenance of mature skeletal muscle.
Collapse
Affiliation(s)
- Sachiko Homma
- Burnham Institute for Medical Research, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|
35
|
Choi JS, Lee JH, Kim HY, Chun MH, Chung JW, Lee MY. Developmental expression of Bis protein in the cerebral cortex and hippocampus of rats. Brain Res 2006; 1092:69-78. [PMID: 16690035 DOI: 10.1016/j.brainres.2006.02.137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 02/09/2006] [Accepted: 02/09/2006] [Indexed: 11/29/2022]
Abstract
Bis (Bcl-2 interacting death suppressor), identified as a Bcl-2-binding protein, has been suggested to have diverse functions in addition to binding to Bcl-2, thereby regulating cell death. To investigate the potential role of Bis in the developing brain, the spatiotemporal expression of Bis protein was studied in the rat forebrain during prenatal and early postnatal development using immunohistochemistry. Initial expression of Bis was detected in the medial telencephalic wall of the lateral ventricle, the area most likely corresponded to the cortical hem from the earliest age examined (E13). There was an abrupt increase of immunoreactive neurons in the cortex and hippocampus during the first postnatal week, which declined thereafter. Two populations of Bis-immunoreactive neurons can be clearly distinguished in the developing forebrain: a population of differentiating and postmitotic neurons coexpressing Bis and microtubule-associated protein-2 (MAP-2), and a population of neurons with the characteristic morphology of Cajal-Retzius cells located exclusively in the marginal zone/layer I of the cortex and in the hippocampal equivalents of the marginal zone. The latter neurons were colabeled with reelin, a marker for Cajal-Retzius cells. While Bis expression in the cerebral cortex and hippocampus exists only transiently by P14, considerable expression was found to be maintained in the rostral migratory stream and the subventricular zone of the lateral ventricle, where Bis-immunoreactive cells were glutamine synthetase-positive glial cells. Our results suggest that Bis may contribute to the developmental processes, including the differentiation and maturation of specific neuronal populations in relation to Bcl-2 in the developing rat forebrain.
Collapse
Affiliation(s)
- Jeong-Sun Choi
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, 137-701 Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
36
|
Benavides A, Pastor D, Santos P, Tranque P, Calvo S. CHOP plays a pivotal role in the astrocyte death induced by oxygen and glucose deprivation. Glia 2006; 52:261-75. [PMID: 16001425 DOI: 10.1002/glia.20242] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ischemia has different consequences on the survival of astrocytes and neurons. Thus, astrocytes show a remarkable resistance to short periods of ischemia that are well known to cause neuronal death. We have used a cell culture model of stroke, oxygen, and glucose deprivation (OGD), to clarify the mechanisms responsible for the exclusive resistance of astrocytes to ischemia. The expression of genes implicated in both ischemia-induced astrocyte death and post-ischemic survival was analysed by the RNA differential display technique. Our study revealed that the expression of the CEBP homologous protein (CHOP)-coding gene is promptly an intensely upregulated following astrocyte oxygen and glucose deprivation. CHOP mRNA induction was accompanied by the activation of other genes (grp78, grp95) that, alike CHOP, are involved in the endoplasmic reticulum (ER) stress response. In addition, drugs that cause ER calcium depletion or protein N-glycosylation inhibition mimicked the effects of OGD on astrocyte survival, further supporting the involvement of ER in the astrocyte responses to OGD. Our experiments also demonstrated that upregulation of CHOP during the ER stress response is required for ischemia to cause astrocyte death. Not only the levels of CHOP mRNA and protein correlate perfectly with the degree of OGD-triggered cell injury, but also astrocyte death induced by OGD is significantly overcome by CHOP antisense oligonucleotide treatment. Nevertheless, we observed that astrocytes undergo apoptosis only when CHOP is permanently upregulated, and not when CHOP increases are transient. Finally, we found that the extent of CHOP induction is determined by the length of the ischemic stimulus. Taken together, our results indicate that permanent upregulation of CHOP is decisive for the induction of astrocyte death by OGD.
Collapse
Affiliation(s)
- Amparo Benavides
- Pharmacology Unit, Facultad de Medicina, Departamento de Ciencias Médicas and Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain
| | | | | | | | | |
Collapse
|
37
|
Götz R, Wiese S, Takayama S, Camarero GC, Rossoll W, Schweizer U, Troppmair J, Jablonka S, Holtmann B, Reed JC, Rapp UR, Sendtner M. Bag1 is essential for differentiation and survival of hematopoietic and neuronal cells. Nat Neurosci 2005; 8:1169-78. [PMID: 16116448 PMCID: PMC1361767 DOI: 10.1038/nn1524] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Accepted: 07/22/2005] [Indexed: 11/08/2022]
Abstract
Bag1 is a cochaperone for the heat-shock protein Hsp70 that interacts with C-Raf, B-Raf, Akt, Bcl-2, steroid hormone receptors and other proteins. Here we use targeted gene disruption in mice to show that Bag1 has an essential role in the survival of differentiating neurons and hematopoietic cells. Cells of the fetal liver and developing nervous system in Bag1-/- mice underwent massive apoptosis. Lack of Bag1 did not disturb the primary function of Akt or Raf, as phosphorylation of the forkhead transcription factor FKHR and activation of extracellular signal-regulated kinase (Erk)-1/2 were not affected. However, the defect was associated with the disturbance of a tripartite complex formed by Akt, B-Raf and Bag1, in addition to the absence of Bad phosphorylation at Ser136. We also observed reduced expression of members of the inhibitor of apoptosis (IAP) family. Our data show that Bag1 is a physiological mediator of extracellular survival signals linked to the cellular mechanisms that prevent apoptosis in hematopoietic and neuronal progenitor cells.
Collapse
Affiliation(s)
- Rudolf Götz
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | - Stefan Wiese
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | | | - Guadalupe C. Camarero
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Wuerzburg, Versbacher Str. 5, D-97078 Wuerzburg, Germany
| | - Wilfried Rossoll
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | - Ulrich Schweizer
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | - Jakob Troppmair
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Wuerzburg, Versbacher Str. 5, D-97078 Wuerzburg, Germany
| | - Sibylle Jablonka
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | - Bettina Holtmann
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
| | - John C. Reed
- The Burnham Institute, La Jolla, California 92037, USA
| | - Ulf R. Rapp
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Wuerzburg, Versbacher Str. 5, D-97078 Wuerzburg, Germany
| | - Michael Sendtner
- Institut für Klinische Neurobiologie, University of Wuerzburg, Josef Schneider Str. 11, D-97080 Wuerzburg, Germany
- Corresponding author: Dr. Michael Sendtner, University of Wuerzburg, Institute for Clinical Neurobiology, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany, Tel.: 0049-(0)931-201-49771, Fax: 0049-(0)931-201-49788,
| |
Collapse
|
38
|
Abstract
Heat-shock proteins are produced in response to different types of stress conditions making cells resistant to stress-induced cell damage. Under normal conditions, heat-shock proteins play numerous roles in cell function, including modulating protein activity by changing protein conformation, promoting multiprotein complex assembly/disassembly, regulating protein degradation within the proteasome pathway, facilitating protein translocation across organellar membranes, and ensuring proper folding of nascent polypeptide chains during protein translation. When cells are stressed, a common response is to undergo cell death by one of two pathways, either 'necrosis' or 'apoptosis'. Recently, both routes to cell death have been revealed to share similar mechanisms, with heat-shock proteins and their cofactors responsible for inhibiting both apoptotic and necrotic pathways. We therefore briefly summarize recent reports showing molecular evidence of cell death regulation by heat-shock proteins and their cochaperones.
Collapse
|