1
|
Findlay AR. Dominantly inherited muscle disorders: understanding their complexity and exploring therapeutic approaches. Dis Model Mech 2024; 17:dmm050720. [PMID: 39501809 PMCID: PMC11574355 DOI: 10.1242/dmm.050720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
Treatments for disabling and life-threatening hereditary muscle disorders are finally close to becoming a reality. Research has thus far focused primarily on recessive forms of muscle disease. The gene replacement strategies that are commonly employed for recessive, loss-of-function disorders are not readily translatable to most dominant myopathies owing to the presence of a normal chromosome in each nucleus, hindering the development of novel treatments for these dominant disorders. This is largely due to their complex, heterogeneous disease mechanisms that require unique therapeutic approaches. However, as viral and RNA interference-based therapies enter clinical use, key tools are now in place to develop treatments for dominantly inherited disorders of muscle. This article will review what is known about dominantly inherited disorders of muscle, specifically their genetic basis, how mutations lead to disease, and the pathomechanistic implications for therapeutic approaches.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University Saint Louis, Neuromuscular Disease Center, 660 S. Euclid Ave., St Louis, MO 63110, USA
| |
Collapse
|
2
|
McKaige EA, Lee C, Calcinotto V, Giri S, Crawford S, McGrath MJ, Ramm G, Bryson-Richardson RJ. Mitochondrial abnormalities contribute to muscle weakness in a Dnajb6 deficient zebrafish model. Hum Mol Genet 2024; 33:1195-1206. [PMID: 38621658 PMCID: PMC11227618 DOI: 10.1093/hmg/ddae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/28/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Mutations in DNAJB6 are a well-established cause of limb girdle muscular dystrophy type D1 (LGMD D1). Patients with LGMD D1 develop progressive muscle weakness with histology showing fibre damage, autophagic vacuoles, and aggregates. Whilst there are many reports of LGMD D1 patients, the role of DNAJB6 in the muscle is still unclear. In this study, we developed a loss of function zebrafish model in order to investigate the role of Dnajb6. Using a double dnajb6a and dnajb6b mutant model, we show that loss of Dnajb6 leads to a late onset muscle weakness. Interestingly, we find that adult fish lacking Dnajb6 do not have autophagy or myofibril defects, however, they do show mitochondrial changes and damage. This study demonstrates that loss of Dnajb6 causes mitochondrial defects and suggests that this contributes to muscle weakness in LGMD D1. These findings expand our knowledge of the role of Dnajb6 in the muscle and provides a model to screen novel therapies for LGMD D1.
Collapse
Affiliation(s)
- Emily A McKaige
- School of Biological Sciences Monash University, 25 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Clara Lee
- School of Biological Sciences Monash University, 25 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Vanessa Calcinotto
- School of Biological Sciences Monash University, 25 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Saveen Giri
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, Clayton, VIC 3800, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, 15 Innovation Walk, Clayton, VIC 3800, Australia
| | - Meagan J McGrath
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, Clayton, VIC 3800, Australia
| | - Georg Ramm
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, Clayton, VIC 3800, Australia
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, 15 Innovation Walk, Clayton, VIC 3800, Australia
| | | |
Collapse
|
3
|
Desai M, Hemant, Deo A, Naik J, Dhamale P, Kshirsagar A, Bose T, Majumdar A. Mrj is a chaperone of the Hsp40 family that regulates Orb2 oligomerization and long-term memory in Drosophila. PLoS Biol 2024; 22:e3002585. [PMID: 38648719 PMCID: PMC11034981 DOI: 10.1371/journal.pbio.3002585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Orb2 the Drosophila homolog of cytoplasmic polyadenylation element binding (CPEB) protein forms prion-like oligomers. These oligomers consist of Orb2A and Orb2B isoforms and their formation is dependent on the oligomerization of the Orb2A isoform. Drosophila with a mutation diminishing Orb2A's prion-like oligomerization forms long-term memory but fails to maintain it over time. Since this prion-like oligomerization of Orb2A plays a crucial role in the maintenance of memory, here, we aim to find what regulates this oligomerization. In an immunoprecipitation-based screen, we identify interactors of Orb2A in the Hsp40 and Hsp70 families of proteins. Among these, we find an Hsp40 family protein Mrj as a regulator of the conversion of Orb2A to its prion-like form. Mrj interacts with Hsp70 proteins and acts as a chaperone by interfering with the aggregation of pathogenic Huntingtin. Unlike its mammalian homolog, we find Drosophila Mrj is neither an essential gene nor causes any gross neurodevelopmental defect. We observe a loss of Mrj results in a reduction in Orb2 oligomers. Further, Mrj knockout exhibits a deficit in long-term memory and our observations suggest Mrj is needed in mushroom body neurons for the regulation of long-term memory. Our work implicates a chaperone Mrj in mechanisms of memory regulation through controlling the oligomerization of Orb2A and its association with the translating ribosomes.
Collapse
Affiliation(s)
- Meghal Desai
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Hemant
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Ankita Deo
- Institute of Bioinformatics and Biotechnology (IBB), Savitribai Phule Pune University, Pune, India
| | - Jagyanseni Naik
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Prathamesh Dhamale
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Avinash Kshirsagar
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Tania Bose
- Institute of Bioinformatics and Biotechnology (IBB), Savitribai Phule Pune University, Pune, India
| | - Amitabha Majumdar
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| |
Collapse
|
4
|
Findlay AR, Paing MM, Daw JA, Haller M, Bengoechea R, Pittman SK, Li S, Wang F, Miller TM, True HL, Chou TF, Weihl CC. DNAJB6 isoform specific knockdown: Therapeutic potential for limb girdle muscular dystrophy D1. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:937-948. [PMID: 37346979 PMCID: PMC10280091 DOI: 10.1016/j.omtn.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/10/2023] [Indexed: 06/23/2023]
Abstract
Dominant missense mutations in DNAJB6, a co-chaperone of HSP70, cause limb girdle muscular dystrophy (LGMD) D1. No treatments are currently available. Two isoforms exist, DNAJB6a and DNAJB6b, each with distinct localizations in muscle. Mutations reside in both isoforms, yet evidence suggests that DNAJB6b is primarily responsible for disease pathogenesis. Knockdown treatment strategies involving both isoforms carry risk, as DNAJB6 knockout is embryonic lethal. We therefore developed an isoform-specific knockdown approach using morpholinos. Selective reduction of each isoform was achieved in vitro in primary mouse myotubes and human LGMDD1 myoblasts, as well as in vivo in mouse skeletal muscle. To assess isoform specific knockdown in LGMDD1, we created primary myotube cultures from a knockin LGMDD1 mouse model. Using mass spectrometry, we identified an LGMDD1 protein signature related to protein homeostasis and myofibril structure. Selective reduction of DNAJB6b levels in LGMDD1 myotubes corrected much of the proteomic disease signature toward wild type levels. Additional in vivo functional data is required to determine if selective reduction of DNAJB6b is a viable therapeutic target for LGMDD1.
Collapse
Affiliation(s)
- Andrew R. Findlay
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - May M. Paing
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jil A. Daw
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Meade Haller
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Rocio Bengoechea
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Sara K. Pittman
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shan Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Feng Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Timothy M. Miller
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Heather L. True
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8228, St. Louis, MO 63110, USA
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Conrad C. Weihl
- Department of Neurology, Neuromuscular Division, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
5
|
Javid H, Hashemian P, Yazdani S, Sharbaf Mashhad A, Karimi-Shahri M. The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem 2022; 123:1704-1735. [PMID: 36063530 DOI: 10.1002/jcb.30326] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023]
Abstract
Heat shock proteins (HSPs) are a large molecular chaperone family classified by their molecular weights, including HSP27, HSP40, HSP60, HSP70, HSP90, and HSP110. HSPs are likely to have antiapoptotic properties and participate actively in various processes such as tumor cell proliferation, invasion, metastases, and death. In this review, we discuss comprehensively the functions of HSPs associated with the progression of colorectal cancer (CRC) and metastasis and resistance to cancer therapy. Taken together, HSPs have numerous clinical applications as biomarkers for cancer diagnosis and prognosis and potential therapeutic targets for CRC and its related metastases.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Pedram Hashemian
- Jahad Daneshgahi Research Committee, Jahad Daneshgahi Institute, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
6
|
Capatina N, Burton GJ, Yung HW. Elevated homocysteine activates unfolded protein responses and causes aberrant trophoblast differentiation and mouse blastocyst development. Physiol Rep 2022; 10:e15467. [PMID: 36117391 PMCID: PMC9483615 DOI: 10.14814/phy2.15467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023] Open
Abstract
Hyperhomocysteinemia may arise from folate/vitamin B12 deficiency, genetic polymorphisms, kidney disease, or hypothyroidism. It is associated with an increased risk of early pregnancy loss and placenta-related complications of pregnancy, including pre-eclampsia and fetal growth restriction. While the majority of studies of hyperhomocysteinemia focus on epigenetic changes secondary to metabolic disruption, the effects of homocysteine toxicity on placental development remain unexplored. Here, we investigated the influence of hyperhomocysteinemia on early blastocyst development and trophoblast differentiation. Exposure of cultured blastocysts to high homocysteine levels reduces cell number in the trophectoderm layer, most likely through increased apoptosis. Homocysteine also promotes differentiation of a trophoblast stem cell line. Both effects diminish the stem cell pool, and are mediated in an endoplasmic reticulum (ER) unfolded protein response (UPRER )-dependent manner. Targeted alleviation of UPRER may therefore provide a new therapeutic intervention to improve pregnancy outcome in women with hyperhomocysteinemia.
Collapse
Affiliation(s)
- Nadejda Capatina
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Graham J. Burton
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
- Department of Clinical NeuroscienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
7
|
Bhadra AK, Rau MJ, Daw JA, Fitzpatrick JAJ, Weihl CC, True HL. Disease-associated mutations within the yeast DNAJB6 homolog Sis1 slow conformer-specific substrate processing and can be corrected by the modulation of nucleotide exchange factors. Nat Commun 2022; 13:4570. [PMID: 35931773 PMCID: PMC9355953 DOI: 10.1038/s41467-022-32318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 07/26/2022] [Indexed: 11/09/2022] Open
Abstract
Molecular chaperones, or heat shock proteins (HSPs), protect against the toxic misfolding and aggregation of proteins. As such, mutations or deficiencies within the chaperone network can lead to disease. Dominant mutations within DNAJB6 (Hsp40)-an Hsp70 co-chaperone-lead to a protein aggregation-linked myopathy termed Limb-Girdle Muscular Dystrophy Type D1 (LGMDD1). Here, we used the yeast prion model client in conjunction with in vitro chaperone activity assays to gain mechanistic insights into the molecular basis of LGMDD1. Here, we show how mutations analogous to those found in LGMDD1 affect Sis1 (a functional homolog of human DNAJB6) function by altering the structure of client protein aggregates, interfering with the Hsp70 ATPase cycle, dimerization and substrate processing; poisoning the function of wild-type protein. These results uncover the mechanisms through which LGMDD1-associated mutations alter chaperone activity, and provide insights relevant to potential therapeutic interventions.
Collapse
Affiliation(s)
- Ankan K Bhadra
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8228, St. Louis, MO, 63110, USA
| | - Michael J Rau
- Washington University Center for Cellular Imaging (WUCCI), Washington University School of Medicine, St. Louis, MO, USA
| | - Jil A Daw
- Department of Neurology, Hope Center for Neurological Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - James A J Fitzpatrick
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8228, St. Louis, MO, 63110, USA
- Washington University Center for Cellular Imaging (WUCCI), Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Conrad C Weihl
- Department of Neurology, Hope Center for Neurological Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Heather L True
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8228, St. Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Asgharzadeh F, Moradi-Marjaneh R, Marjaneh MM. The role of heat shock protein 40 in carcinogenesis and biology of colorectal cancer. Curr Pharm Des 2022; 28:1457-1465. [PMID: 35570564 DOI: 10.2174/1381612828666220513124603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Despite the enormous amount of effort in the diagnosis and treatment of CRC, the overall survival rate of patients remains low. The precise molecular and cellular basis underlying CRC has not been completely understood yet. Over time, new genes and molecular pathways involved in the pathogenesis of the disease are being identified. Accurate discovery of these genes and signaling pathways are important and urgent missions for the next generation of anticancer therapy research. Chaperone DnaJ, also known as Hsp40 (heat shock protein 40), has been of particular interest in CRC pathogenesis, as it is involved in the fundamental cell activities for maintaining cellular homeostasis. Evidence show that protein family members of DnaJ/Hsp40 play both roles; enhancing and reducing the growth of CRC cells. In the present review, we focus on the current knowledge on the molecular mechanisms responsible for the role of DnaJ/Hsp40 in CRC carcinogenesis and biology.
Collapse
Affiliation(s)
- Fereshteh Asgharzadeh
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Moradi-Marjaneh
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mahdi Moradi Marjaneh
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
Guernsey MW, van Kruistum H, Reznick DN, Pollux BJA, Baker JC. Molecular Signatures of Placentation and Secretion Uncovered in Poeciliopsis Maternal Follicles. Mol Biol Evol 2021; 37:2679-2690. [PMID: 32421768 PMCID: PMC7475030 DOI: 10.1093/molbev/msaa121] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Placentation evolved many times independently in vertebrates. Although the core functions of all placentas are similar, we know less about how this similarity extends to the molecular level. Here, we study Poeciliopsis, a unique genus of live-bearing fish that have independently evolved complex placental structures at least three times. The maternal follicle is a key component of these structures. It envelops yolk-rich eggs and is morphologically simple in lecithotrophic species but has elaborate villous structures in matrotrophic species. Through sequencing, the follicle transcriptome of a matrotrophic, Poeciliopsis retropinna, and lecithotrophic, P. turrubarensis, species we found genes known to be critical for placenta function expressed in both species despite their difference in complexity. Additionally, when we compare the transcriptome of different river populations of P. retropinna, known to vary in maternal provisioning, we find differential expression of secretory genes expressed specifically in the top layer of villi cells in the maternal follicle. This provides some of the first evidence that the placental structures of Poeciliopsis function using a secretory mechanism rather than direct contact with maternal circulation. Finally, when we look at the expression of placenta proteins at the maternal–fetal interface of a larger sampling of Poeciliopsis species, we find expression of key maternal and fetal placenta proteins in their cognate tissue types of all species, but follicle expression of prolactin is restricted to only matrotrophic species. Taken together, we suggest that all Poeciliopsis follicles are poised for placenta function but require expression of key genes to form secretory villi.
Collapse
Affiliation(s)
- Michael W Guernsey
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Henri van Kruistum
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - David N Reznick
- Department of Biology, University of California Riverside, Riverside, CA
| | - Bart J A Pollux
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
10
|
Liu X, Wu Z, Li J, Bao H, Wu C. Genome-Wide Association Study and Transcriptome Differential Expression Analysis of the Feather Rate in Shouguang Chickens. Front Genet 2021; 11:613078. [PMID: 33414812 PMCID: PMC7783405 DOI: 10.3389/fgene.2020.613078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/02/2020] [Indexed: 12/01/2022] Open
Abstract
The feather rate phenotype in chicks, including early-feathering and late-feathering phenotypes, are widely used as a sexing system in the poultry industry. The objective of this study was to obtain candidate genes associated with the feather rate in Shouguang chickens. In the present study, we collected 56 blood samples and 12 hair follicle samples of flight feathers from female Shouguang chickens. Then we identified the chromosome region associated with the feather rate by genome-wide association analysis (GWAS). We also performed RNA sequencing and analyzed differentially expressed genes between the early-feathering and late-feathering phenotypes using HISAT2, StringTie, and DESeq2. We identified a genomic region of 10.0–13.0 Mb of chromosome Z, which is statistically associated with the feather rate of Shouguang chickens at one-day old. After RNA sequencing analysis, 342 differentially expressed known genes between the early-feathering (EF) and late-feathering (LF) phenotypes were screened out, which were involved in epithelial cell differentiation, intermediate filament organization, protein serine kinase activity, peptidyl-serine phosphorylation, retinoic acid binding, and so on. The sperm flagellar 2 gene (SPEF2) and prolactin receptor (PRLR) gene were the only two overlapping genes between the results of GWAS and differential expression analysis, which implies that SPEF2 and PRLR are possible candidate genes for the formation of the chicken feathering phenotype in the present study. Our findings help to elucidate the molecular mechanism of the feather rate in chicks.
Collapse
Affiliation(s)
- Xiayi Liu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhou Wu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Animal Breeding and Genomics, Wageningen University & Research, Wageningen, Netherlands
| | - Junying Li
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Haigang Bao
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Changxin Wu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Neuromuscular Diseases Due to Chaperone Mutations: A Review and Some New Results. Int J Mol Sci 2020; 21:ijms21041409. [PMID: 32093037 PMCID: PMC7073051 DOI: 10.3390/ijms21041409] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle and the nervous system depend on efficient protein quality control, and they express chaperones and cochaperones at high levels to maintain protein homeostasis. Mutations in many of these proteins cause neuromuscular diseases, myopathies, and hereditary motor and sensorimotor neuropathies. In this review, we cover mutations in DNAJB6, DNAJB2, αB-crystallin (CRYAB, HSPB5), HSPB1, HSPB3, HSPB8, and BAG3, and discuss the molecular mechanisms by which they cause neuromuscular disease. In addition, previously unpublished results are presented, showing downstream effects of BAG3 p.P209L on DNAJB6 turnover and localization.
Collapse
|
12
|
Eaton M, Davies AH, Devine J, Zhao X, Simmons DG, Maríusdóttir E, Natale DRC, Matyas JR, Bering EA, Workentine ML, Hallgrimsson B, Cross JC. Complex patterns of cell growth in the placenta in normal pregnancy and as adaptations to maternal diet restriction. PLoS One 2020; 15:e0226735. [PMID: 31917811 PMCID: PMC6952106 DOI: 10.1371/journal.pone.0226735] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
The major milestones in mouse placental development are well described, but our understanding is limited to how the placenta can adapt to damage or changes in the environment. By using stereology and expression of cell cycle markers, we found that the placenta grows under normal conditions not just by hyperplasia of trophoblast cells but also through extensive polyploidy and cell hypertrophy. In response to feeding a low protein diet to mothers prior to and during pregnancy, to mimic chronic malnutrition, we found that this normal program was altered and that it was influenced by the sex of the conceptus. Male fetuses showed intrauterine growth restriction (IUGR) by embryonic day (E) 18.5, just before term, whereas female fetuses showed IUGR as early as E16.5. This difference was correlated with differences in the size of the labyrinth layer of the placenta, the site of nutrient and gas exchange. Functional changes were implied based on up-regulation of nutrient transporter genes. The junctional zone was also affected, with a reduction in both glycogen trophoblast and spongiotrophoblast cells. These changes were associated with increased expression of Phlda2 and reduced expression of Egfr. Polyploidy, which results from endoreduplication, is a normal feature of trophoblast giant cells (TGC) but also spongiotrophoblast cells. Ploidy was increased in sinusoidal-TGCs and spongiotrophoblast cells, but not parietal-TGCs, in low protein placentas. These results indicate that the placenta undergoes a range of changes in development and function in response to poor maternal diet, many of which we interpret are aimed at mitigating the impacts on fetal and maternal health.
Collapse
Affiliation(s)
- Malcolm Eaton
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - Alastair H. Davies
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Jay Devine
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - Xiang Zhao
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - David G. Simmons
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Elín Maríusdóttir
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - David R. C. Natale
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - John R. Matyas
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
| | - Elizabeth A. Bering
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | | | - Benedikt Hallgrimsson
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
| | - James C. Cross
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary Alberta
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary Alberta
- * E-mail:
| |
Collapse
|
13
|
Findlay AR, Bengoechea R, Pittman SK, Chou TF, True HL, Weihl CC. Lithium chloride corrects weakness and myopathology in a preclinical model of LGMD1D. NEUROLOGY-GENETICS 2019; 5:e318. [PMID: 31123706 PMCID: PMC6510529 DOI: 10.1212/nxg.0000000000000318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/04/2019] [Indexed: 12/12/2022]
Abstract
Objective To understand DNAJB6's function in skeletal muscle and identify therapeutic targets for limb-girdle muscular dystrophy 1D (LGMD1D). Methods DNAJB6 knockout (KO) myoblasts were generated with Crispr/cas9 technology, and differentially accumulated proteins were identified using stable isotope labeling, followed by quantitative mass spectrometry. Cultured KO myotubes and mouse muscle from DNAJB6b-WT or DNAJB6b-F93L mice were analyzed using histochemistry, immunohistochemistry, and immunoblot. Mouse functional strength measures included forelimb grip strength and inverted wire hang. Results DNAJB6 inactivation leads to the accumulation of sarcomeric proteins and hypertrophic myotubes with an enhanced fusion index. The increased fusion in DNAJB6 KO myotubes correlates with diminished glycogen synthase kinase-β (GSK3β) activity. In contrast, LGMD1D mutations in DNAJB6 enhance GSK3β activation and suppress β-catenin and NFAT3c signaling. GSK3β inhibition with lithium chloride improves muscle size and strength in an LGMD1D preclinical mouse model. Conclusions Our results suggest that DNAJB6 facilitates protein quality control and negatively regulates myogenic signaling. In addition, LGMD1D-associated DNAJB6 mutations inhibit myogenic signaling through augmented GSK3β activity. GSK3β inhibition with lithium chloride may be a therapeutic option in LGMD1D.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| | - Rocio Bengoechea
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| | - Sara K Pittman
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| | - Tsui-Fen Chou
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| | - Heather L True
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| | - Conrad C Weihl
- Washington University School of Medicine (A.R.F., R.B., S.K.P., H.L.T., C.C.W); Department of Neurology (A.R.F., R.B., S.K.P., C.C.W), Hope Center for Neurological Diseases, St. Louis, MO; Harbor-UCLA Medical Center (T.-F.C.), Department of Pediatrics, Division of Medical Genetics, Torrance, CA; Department of Cell Biology and Physiology (H.L.T.), Saint Louis, MO
| |
Collapse
|
14
|
Söderberg CAG, Månsson C, Bernfur K, Rutsdottir G, Härmark J, Rajan S, Al-Karadaghi S, Rasmussen M, Höjrup P, Hebert H, Emanuelsson C. Structural modelling of the DNAJB6 oligomeric chaperone shows a peptide-binding cleft lined with conserved S/T-residues at the dimer interface. Sci Rep 2018; 8:5199. [PMID: 29581438 PMCID: PMC5979959 DOI: 10.1038/s41598-018-23035-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/05/2018] [Indexed: 12/28/2022] Open
Abstract
The remarkably efficient suppression of amyloid fibril formation by the DNAJB6 chaperone is dependent on a set of conserved S/T-residues and an oligomeric structure, features unusual among DNAJ chaperones. We explored the structure of DNAJB6 using a combination of structural methods. Lysine-specific crosslinking mass spectrometry provided distance constraints to select a homology model of the DNAJB6 monomer, which was subsequently used in crosslink-assisted docking to generate a dimer model. A peptide-binding cleft lined with S/T-residues is formed at the monomer-monomer interface. Mixed isotope crosslinking showed that the oligomers are dynamic entities that exchange subunits. The purified protein is well folded, soluble and composed of oligomers with a varying number of subunits according to small-angle X-ray scattering (SAXS). Elongated particles (160 × 120 Å) were detected by electron microscopy and single particle reconstruction resulted in a density map of 20 Å resolution into which the DNAJB6 dimers fit. The structure of the oligomer and the S/T-rich region is of great importance for the understanding of the function of DNAJB6 and how it can bind aggregation-prone peptides and prevent amyloid diseases.
Collapse
Affiliation(s)
| | - Cecilia Månsson
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Katja Bernfur
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Gudrun Rutsdottir
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Johan Härmark
- School of Technology and Health, KTH Royal Institute of Technology and Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Sreekanth Rajan
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Salam Al-Karadaghi
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Morten Rasmussen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Peter Höjrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Hans Hebert
- School of Technology and Health, KTH Royal Institute of Technology and Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Cecilia Emanuelsson
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, SE-221 00, Lund, Sweden.
| |
Collapse
|
15
|
Meng E, Shevde LA, Samant RS. Emerging roles and underlying molecular mechanisms of DNAJB6 in cancer. Oncotarget 2018; 7:53984-53996. [PMID: 27276715 PMCID: PMC5288237 DOI: 10.18632/oncotarget.9803] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/26/2016] [Indexed: 12/29/2022] Open
Abstract
DNAJB6 also known as mammalian relative of DnaJ (MRJ) encodes a highly conserved member of the DnaJ/Hsp40 family of co-chaperone proteins that function with Hsp70 chaperones. DNAJB6 is widely expressed in all tissues, with higher expression levels detected in the brain. DNAJB6 is involved in diverse cellular functions ranging from murine placental development, reducing the formation and toxicity of mis-folded protein aggregates, to self-renewal of neural stem cells. Involvement of DNAJB6 is implicated in multiple pathologies such as Huntington's disease, Parkinson's diseases, limb-girdle muscular dystrophy, cardiomyocyte hypertrophy and cancer. This review summarizes the important involvement of the spliced isoforms of DNAJB6 in various pathologies with a specific focus on the emerging roles of human DNAJB6 in cancer and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Erhong Meng
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Beijing DOING Biomedical Technology Co. Ltd., Beijing,China
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
16
|
Ruggieri A, Saredi S, Zanotti S, Pasanisi MB, Maggi L, Mora M. DNAJB6 Myopathies: Focused Review on an Emerging and Expanding Group of Myopathies. Front Mol Biosci 2016; 3:63. [PMID: 27747217 PMCID: PMC5043021 DOI: 10.3389/fmolb.2016.00063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022] Open
Abstract
Mutations in the DNAJB6 gene have been associated with the autosomal dominant limb girdle muscular dystrophy type 1D (LGMD1D), a disorder characterized by abnormal protein aggregates and rimmed vacuoles in muscle fibers. DNAJB6 is a ubiquitously expressed Hsp40 co-chaperone characterized by a J domain that specifies Hsp70 functions in the cellular environment. DNAJB6 is also a potent inhibitor of expanded polyglutamine (polyQ) aggregation preventing aggregate toxicity in cells. In DNAJB6-mutated patients this anti-aggregation property is significantly reduced, albeit not completely lost. To elucidate the pathogenetic mechanisms underlying the DNAJB6-related myopathy, animal models have been created showing that, indeed, conditional muscular expression of a DNAJB6 mutant in the mouse causes a LGMD1D myofibrillary muscle tissue phenotype. Both mutations and phenotypes reported until recently were rather homogeneous, being exclusively missense mutations of a few amino acids of the protein G/F domain, and with a phenotype characterized by adult-onset slowly progressive muscular dystrophy predominantly affecting proximal muscles. Lately, several novel mutations and new phenotypes of DNAJB6 have been described. These mutations once more affect the G/F domain of DNAJB6 with missense changes and a splice site mutation; and the phenotypes include childhood onset and distal involvement of muscles, or childhood-onset LGMD1D with loss of ambulation in early adulthood and respiratory involvement. Thus, the spectrum of DNAJB6-related phenotypes is widening. Although our knowledge about the role of DNAJB6 in the pathogenesis of muscle diseases has made great progression, several questions remain unsolved, including why a ubiquitous protein affects only, or predominantly, skeletal muscle; why only the G/F domain is involved; and what is the possible role of the DNAJB6a isoform. Clarification of these issues will provide clues to implement possible therapeutic strategies for DNAJB6-related myopathies.
Collapse
Affiliation(s)
- Alessandra Ruggieri
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| | - Simona Saredi
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| | - Simona Zanotti
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| | - Maria Barbara Pasanisi
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| | - Lorenzo Maggi
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta Milan, Italy
| |
Collapse
|
17
|
Salas PJ, Forteza R, Mashukova A. Multiple roles for keratin intermediate filaments in the regulation of epithelial barrier function and apico-basal polarity. Tissue Barriers 2016; 4:e1178368. [PMID: 27583190 PMCID: PMC4993576 DOI: 10.1080/21688370.2016.1178368] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 12/27/2022] Open
Abstract
As multicellular organisms evolved a family of cytoskeletal proteins, the keratins (types I and II) expressed in epithelial cells diversified in more than 20 genes in vertebrates. There is no question that keratin filaments confer mechanical stiffness to cells. However, such a number of genes can hardly be explained by evolutionary advantages in mechanical features. The use of transgenic mouse models has revealed unexpected functional relationships between keratin intermediate filaments and intracellular signaling. Accordingly, loss of keratins or mutations in keratins that cause or predispose to human diseases, result in increased sensitivity to apoptosis, regulation of innate immunity, permeabilization of tight junctions, and mistargeting of apical proteins in different epithelia. Precise mechanistic explanations for these phenomena are still lacking. However, immobilization of membrane or cytoplasmic proteins, including chaperones, on intermediate filaments (“scaffolding”) appear as common molecular mechanisms and may explain the need for so many different keratin genes in vertebrates.
Collapse
Affiliation(s)
- Pedro J Salas
- Department of Cell Biology, Miller School of Medicine, University of Miami , Miami, FL, USA
| | - Radia Forteza
- Department of Cell Biology, Miller School of Medicine, University of Miami , Miami, FL, USA
| | - Anastasia Mashukova
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Physiology, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
18
|
Kakkar V, Månsson C, de Mattos EP, Bergink S, van der Zwaag M, van Waarde MAWH, Kloosterhuis NJ, Melki R, van Cruchten RTP, Al-Karadaghi S, Arosio P, Dobson CM, Knowles TPJ, Bates GP, van Deursen JM, Linse S, van de Sluis B, Emanuelsson C, Kampinga HH. The S/T-Rich Motif in the DNAJB6 Chaperone Delays Polyglutamine Aggregation and the Onset of Disease in a Mouse Model. Mol Cell 2016; 62:272-283. [PMID: 27151442 DOI: 10.1016/j.molcel.2016.03.017] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 01/14/2016] [Accepted: 03/10/2016] [Indexed: 01/30/2023]
Abstract
Expanded CAG repeats lead to debilitating neurodegenerative disorders characterized by aggregation of proteins with expanded polyglutamine (polyQ) tracts. The mechanism of aggregation involves primary and secondary nucleation steps. We show how a noncanonical member of the DNAJ-chaperone family, DNAJB6, inhibits the conversion of soluble polyQ peptides into amyloid fibrils, in particular by suppressing primary nucleation. This inhibition is mediated by a serine/threonine-rich region that provides an array of surface-exposed hydroxyl groups that bind to polyQ peptides and may disrupt the formation of the H bonds essential for the stability of amyloid fibrils. Early prevention of polyQ aggregation by DNAJB6 occurs also in cells and leads to delayed neurite retraction even before aggregates are visible. In a mouse model, brain-specific coexpression of DNAJB6 delays polyQ aggregation, relieves symptoms, and prolongs lifespan, pointing to DNAJB6 as a potential target for disease therapy and tool for unraveling early events in the onset of polyQ diseases.
Collapse
Affiliation(s)
- Vaishali Kakkar
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Cecilia Månsson
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Eduardo P de Mattos
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands; Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Steven Bergink
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Marianne van der Zwaag
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Maria A W H van Waarde
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, Molecular Genetics Section, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Ronald Melki
- Neuroscience Paris-Saclay Institute of Neuroscience, Centre National de la Recherche Scientifique, 91190 Gif-Sur-Yvette, France
| | - Remco T P van Cruchten
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Salam Al-Karadaghi
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Paolo Arosio
- Department of Chemistry, University of Cambridge, Cambridge CB2 1TN, UK
| | | | | | - Gillian P Bates
- Department of Medical and Molecular Genetics, King's College London, London WC2R 2LS, UK
| | | | - Sara Linse
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Cecilia Emanuelsson
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Harm H Kampinga
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| |
Collapse
|
19
|
Walentin K, Hinze C, Schmidt-Ott KM. The basal chorionic trophoblast cell layer: An emerging coordinator of placenta development. Bioessays 2016; 38:254-65. [PMID: 26778584 DOI: 10.1002/bies.201500087] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
During gestation, fetomaternal exchange occurs in the villous tree (labyrinth) of the placenta. Development of this structure depends on tightly coordinated cellular processes of branching morphogenesis and differentiation of specialized trophoblast cells. The basal chorionic trophoblast (BCT) cell layer that localizes next to the chorioallantoic interface is of critical importance for labyrinth morphogenesis in rodents. Gcm1-positive cell clusters within this layer initiate branching morphogenesis thereby guiding allantoic fetal blood vessels towards maternal blood sinuses. Later these cells differentiate and contribute to the syncytiotrophoblast of the fetomaternal barrier. Additional cells within the BCT layer sustain continued morphogenesis, possibly through a repopulating progenitor population. Several mouse mutants highlight the importance of a structurally intact BCT epithelium, and a growing number of studies addresses its patterning and epithelial architecture. Here, we review and discuss emerging concepts in labyrinth development focussing on the biology of the BCT cell layer.
Collapse
Affiliation(s)
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Bengoechea R, Pittman SK, Tuck EP, True HL, Weihl CC. Myofibrillar disruption and RNA-binding protein aggregation in a mouse model of limb-girdle muscular dystrophy 1D. Hum Mol Genet 2015; 24:6588-602. [PMID: 26362252 PMCID: PMC4634370 DOI: 10.1093/hmg/ddv363] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022] Open
Abstract
Limb-girdle muscular dystrophy type 1D (LGMD1D) is caused by dominantly inherited missense mutations in DNAJB6, an Hsp40 co-chaperone. LGMD1D muscle has rimmed vacuoles and inclusion bodies containing DNAJB6, Z-disc proteins and TDP-43. DNAJB6 is expressed as two isoforms; DNAJB6a and DNAJB6b. Both isoforms contain LGMD1D mutant residues and are expressed in human muscle. To identify which mutant isoform confers disease pathogenesis and generate a mouse model of LGMD1D, we evaluated DNAJB6 expression and localization in skeletal muscle as well as generating DNAJB6 isoform specific expressing transgenic mice. DNAJB6a localized to myonuclei while DNAJB6b was sarcoplasmic. LGMD1D mutations in DNAJB6a or DNAJB6b did not alter this localization in mouse muscle. Transgenic mice expressing the LGMD1D mutant, F93L, in DNAJB6b under a muscle-specific promoter became weak, had early lethality and developed muscle pathology consistent with myopathy after 2 months; whereas mice expressing the same F93L mutation in DNAJB6a or overexpressing DNAJB6a or DNAJB6b wild-type transgenes remained unaffected after 1 year. DNAJB6b localized to the Z-disc and DNAJB6b-F93L expressing mouse muscle had myofibrillar disorganization and desmin inclusions. Consistent with DNAJB6 dysfunction, keratin 8/18, a DNAJB6 client also accumulated in DNAJB6b-F93L expressing mouse muscle. The RNA-binding proteins hnRNPA1 and hnRNPA2/B1 accumulated and co-localized with DNAJB6 at sarcoplasmic stress granules suggesting that these proteins maybe novel DNAJB6b clients. Similarly, hnRNPA1 and hnRNPA2/B1 formed sarcoplasmic aggregates in patients with LGMD1D. Our data support that LGMD1D mutations in DNAJB6 disrupt its sarcoplasmic function suggesting a role for DNAJB6b in Z-disc organization and stress granule kinetics.
Collapse
Affiliation(s)
| | | | | | - Heather L True
- Department of Cell Biology and Physiology and and The Hope Center for Neurological Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Conrad C Weihl
- Department of Neurology, The Hope Center for Neurological Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
21
|
Functional Analysis of Keratin-Associated Proteins in Intestinal Epithelia: Heat-Shock Protein Chaperoning and Kinase Rescue. Methods Enzymol 2015. [PMID: 26778557 DOI: 10.1016/bs.mie.2015.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
A growing body of evidence from several laboratories points at nonmechanical functions of keratin intermediate filaments (IF), such as control of apoptosis, modulation of signaling, or regulation of innate immunity, among others. While these functions are generally assigned to the ability of IF to scaffold other proteins, direct mechanistic causal relationships between filamentous keratins and the observed effects of keratin knockout or mutations are still missing. We have proposed that the scaffolding of chaperones such as Hsp70/40 may be key to understand some IF nonmechanical functions if unique features or specificity of the chaperoning activity in the IF scaffold can be demonstrated. The same criteria of uniqueness could be applied to other biochemical functions of the IF scaffold. Here, we describe a subcellular fractionation technique based on established methods of keratin purification. The resulting keratin-enriched fraction contains several proteins tightly associated with the IF scaffold, including Hsp70/40 chaperones. Being nondenaturing, this fractionation method enables direct testing of chaperoning and other enzymatic activities associated with IF, as well as supplementation experiments to determine the need for soluble (cytosolic) proteins. This method also permits to analyze inhibitory activity of cytosolic proteins at independently characterized physiological concentrations. When used as complementary approaches to knockout, knockdown, or site-directed mutagenesis, these techniques are expected to shed light on molecular mechanisms involved in the effects of IF loss of function.
Collapse
|
22
|
Szabo S, Wögenstein KL, Österreicher CH, Guldiken N, Chen Y, Doler C, Wiche G, Boor P, Haybaeck J, Strnad P, Fuchs P. Epiplakin attenuates experimental mouse liver injury by chaperoning keratin reorganization. J Hepatol 2015; 62:1357-66. [PMID: 25617501 PMCID: PMC4451473 DOI: 10.1016/j.jhep.2015.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/08/2014] [Accepted: 01/05/2015] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Epiplakin is a member of the plakin protein family and exclusively expressed in epithelial tissues where it binds to keratins. Epiplakin-deficient (Eppk1(-/-)) mice displayed no obvious spontaneous phenotype, but their keratinocytes showed a faster keratin network breakdown in response to stress. The role of epiplakin in the stressed liver remained to be elucidated. METHODS Wild-type (WT) and Eppk1(-/-) mice were subjected to common bile duct ligation (CBDL) or fed with a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing diet. The importance of epiplakin during keratin reorganization was assessed in primary hepatocytes. RESULTS Our experiments revealed that epiplakin is expressed in hepatocytes and cholangiocytes, and binds to keratin 8 (K8) and K18 via multiple domains. In several liver stress models epiplakin and K8 genes displayed identical expression patterns and transgenic K8 overexpression resulted in elevated hepatic epiplakin levels. After CBDL and DDC treatment, Eppk1(-/-) mice developed a more pronounced liver injury and their livers contained larger amounts of hepatocellular keratin granules, indicating impaired disease-induced keratin network reorganization. In line with these findings, primary Eppk1(-/-) hepatocytes showed increased formation of keratin aggregates after treatment with the phosphatase inhibitor okadaic acid, a phenotype which was rescued by the chemical chaperone trimethylamine N-oxide (TMAO). Finally, transfection experiments revealed that Eppk1(-/-) primary hepatocytes were less able to tolerate forced K8 overexpression and that TMAO treatment rescued this phenotype. CONCLUSION Our data indicate that epiplakin plays a protective role during experimental liver injuries by chaperoning disease-induced keratin reorganization.
Collapse
Affiliation(s)
- Sandra Szabo
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Karl L Wögenstein
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Christoph H Österreicher
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nurdan Guldiken
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Yu Chen
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Carina Doler
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gerhard Wiche
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Peter Boor
- Division of Nephrology and Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | | | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Peter Fuchs
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria.
| |
Collapse
|
23
|
Stein KC, Bengoechea R, Harms MB, Weihl CC, True HL. Myopathy-causing mutations in an HSP40 chaperone disrupt processing of specific client conformers. J Biol Chem 2015; 289:21120-30. [PMID: 24920671 DOI: 10.1074/jbc.m114.572461] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The molecular chaperone network protects against the toxic misfolding and aggregation of proteins. Disruption of this network leads to a variety of protein conformational disorders. One such example recently discovered is limb-girdle muscular dystrophy type 1D (LGMD1D), which is caused by mutation of the HSP40 chaperone DNAJB6. All LGMD1D-associated mutations localize to the conserved G/F domain of DNAJB6, but the function of this domain is largely unknown. Here, we exploit the yeast HSP40 Sis1, which has known aggregation-prone client proteins, to gain insight into the role of the G/F domain and its significance in LGMD1D pathogenesis. Strikingly, we demonstrate that LGMD1D mutations in a Sis1-DNAJB6 chimera differentially impair the processing of specific conformers of two yeast prions, [RNQ+] and [PSI+]. Importantly, these differences do not simply correlate to the sensitivity of these prion strains to changes in chaperone levels. Additionally, we analyzed the effect of LGMD1D-associated DNAJB6 mutations on TDP-43, a protein known to form inclusions in LGMD1D. We show that the DNAJB6 G/F domain mutants disrupt the processing of nuclear TDP-43 stress granules in mammalian cells. These data suggest that the G/F domain mediates chaperone-substrate interactions in a manner that extends beyond recognition of a particular client and to a subset of client conformers. We propose that such selective chaperone disruption may lead to the accumulation of toxic aggregate conformers and result in the development of LGMD1D and perhaps other protein conformational disorders.
Collapse
|
24
|
Wögenstein KL, Szabo S, Lunova M, Wiche G, Haybaeck J, Strnad P, Boor P, Wagner M, Fuchs P. Epiplakin deficiency aggravates murine caerulein-induced acute pancreatitis and favors the formation of acinar keratin granules. PLoS One 2014; 9:e108323. [PMID: 25232867 PMCID: PMC4169488 DOI: 10.1371/journal.pone.0108323] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/19/2014] [Indexed: 11/23/2022] Open
Abstract
Epiplakin, a member of the plakin protein family, is exclusively expressed in epithelial tissues and was shown to bind to keratins. Epiplakin-deficient (EPPK−/−) mice showed no obvious spontaneous phenotype, however, EPPK−/− keratinocytes displayed faster keratin network breakdown in response to stress. The role of epiplakin in pancreas, a tissue with abundant keratin expression, was not yet known. We analyzed epiplakin’s expression in healthy and inflamed pancreatic tissue and compared wild-type and EPPK−/− mice during caerulein-induced acute pancreatitis. We found that epiplakin was expressed primarily in ductal cells of the pancreas and colocalized with apicolateral keratin bundles in murine pancreatic acinar cells. Epiplakin’s diffuse subcellular localization in keratin filament-free acini of K8-deficient mice indicated that its filament-associated localization in acinar cells completely depends on its binding partner keratin. During acute pancreatitis, epiplakin was upregulated in acinar cells and its redistribution closely paralleled keratin reorganization. EPPK−/− mice suffered from aggravated pancreatitis but showed no obvious regeneration phenotype. At the most severe stage of the disease, EPPK−/− acinar cells displayed more keratin aggregates than those of wild-type mice. Our data propose epiplakin to be a protective protein during acute pancreatitis, and that its loss causes impaired disease-associated keratin reorganization.
Collapse
Affiliation(s)
- Karl L. Wögenstein
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Sandra Szabo
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Mariia Lunova
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Gerhard Wiche
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | | | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Peter Boor
- Division of Nephrology and Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Martin Wagner
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Peter Fuchs
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
25
|
Koutras C, Braun JEA. J protein mutations and resulting proteostasis collapse. Front Cell Neurosci 2014; 8:191. [PMID: 25071450 PMCID: PMC4086201 DOI: 10.3389/fncel.2014.00191] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/21/2014] [Indexed: 01/20/2023] Open
Abstract
Despite a century of intensive investigation the effective treatment of protein aggregation diseases remains elusive. Ordinarily, molecular chaperones ensure that proteins maintain their functional conformation. The appearance of misfolded proteins that aggregate implies the collapse of the cellular chaperone quality control network. That said, the cellular chaperone network is extensive and functional information regarding the detailed action of specific chaperones is not yet available. J proteins (DnaJ/Hsp40) are a family of chaperone cofactors that harness Hsc70 (heat shock cognate protein of 70 kDa) for diverse conformational cellular tasks and, as such, represent novel clinically relevant targets for diseases resulting from the disruption of proteostasis. Here we review incisive reports identifying mutations in individual J protein chaperones and the proteostasis collapse that ensues.
Collapse
Affiliation(s)
- Carolina Koutras
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - Janice E. A. Braun
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| |
Collapse
|
26
|
Crish J, Conti MA, Sakai T, Adelstein RS, Egelhoff TT. Keratin 5-Cre-driven excision of nonmuscle myosin IIA in early embryo trophectoderm leads to placenta defects and embryonic lethality. Dev Biol 2013; 382:136-148. [PMID: 23911870 PMCID: PMC4186751 DOI: 10.1016/j.ydbio.2013.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 11/21/2022]
Abstract
In studies initially focused on roles of nonmuscle myosin IIA (NMIIA) in the developing mouse epidermis, we have discovered that a previously described cytokeratin 5 (K5)-Cre gene construct is expressed in early embryo development. Mice carrying floxed alleles of the nonmuscle myosin II heavy chain gene (NMHC IIA(flox/flox)) were crossed with the K5-Cre line. The progeny of newborn pups did not show a Mendelian genotype distribution, suggesting embryonic lethality. Analysis of post-implantation conceptuses from embryonic day (E)9.5 to E13.5 revealed poorly developed embryos and defective placentas, with significantly reduced labyrinth surface area and blood vessel vascularization. These results suggested the novel possibility that the bovine K5 promoter-driven Cre-recombinase was active early in trophoblast-lineage cells that give rise to the placenta. To test this possibility, K5-Cre transgenic mice were crossed with the mT/mG reporter mouse in which activation of GFP expression indicates Cre transgene expression. We observed activation of K5-Cre-driven GFP expression in the ectoplacental cone, in the extraembryonic ectoderm, and in trophoblast giant cells in the E6.5 embryo. In addition, we observed GFP expression at E11.5 to E13.5 in both the labyrinth of the placenta and the yolk sac. NMIIA expression was detected in these same cell types in normal embryos, as well as in E13.5 yolk sac and labyrinth. These findings taken together suggest that NMHC IIA may play critical roles in the early trophoblast-derived ectoplacental cone and extraembryonic ectoderm, as well as in the yolk sac and labyrinth tissues that form later. Our findings are consistent with phenotypes of constitutive NMIIA knockout mice made earlier, that displayed labyrinth and yolk sac-specific defects, but our findings extend those observations by suggesting possible NMIIA roles in trophoblast lineages as well. These results furthermore demonstrate that K5-Cre gene constructs, previously reported to be activated starting at approximately E12.5 in the forming epidermis, may be widely useful as drivers for activation of cre/lox based gene excision in early embryo extraembronic trophoblast tissues as well.
Collapse
Affiliation(s)
- James Crish
- Department of Cellular and Molecular Medicine NC10, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Mary Anne Conti
- Laboratory of Molecular Cardiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Takao Sakai
- Department of Biomedical Engineering, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Robert S. Adelstein
- Laboratory of Molecular Cardiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas T. Egelhoff
- Department of Cellular and Molecular Medicine NC10, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| |
Collapse
|
27
|
DNAJB6 governs a novel regulatory loop determining Wnt/β-catenin signalling activity. Biochem J 2012; 444:573-80. [PMID: 22455953 DOI: 10.1042/bj20120205] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
DKK1 (dickkopf 1 homologue) is a secreted inhibitor of the Wnt signalling pathway and a critical modulator of tumour promotion and the tumour microenvironment. However, mechanisms regulating DKK1 expression are understudied. DNAJB6 {DnaJ [HSP40 (heat-shock protein 40 kDa)] homologue, subfamily B, member 6} is an HSP40 family member whose expression is compromised during progression of breast cancer and melanoma. Inhibition of the Wnt/β-catenin signalling pathway by up-regulation of DKK1 is one of the key mechanisms by which DNAJB6 suppresses tumour metastasis and EMT (epithelial-mesenchymal transition). Analysis of the DKK1 promoter to define the cis-site responsible for its up-regulation by DNAJB6 revealed the presence of two binding sites for a transcriptional repressor, MSX1 (muscle segment homeobox 1). Our investigations showed that MSX1 binds the DKK1 promoter and inhibits DKK1 transcription. Interestingly, silencing DNAJB6 resulted in up-regulation of MSX1 concomitant with increased stabilization of β-catenin. ChIP (chromatin immunoprecipitation) studies revealed that β-catenin binds the MSX1 promoter and stabilization of β-catenin elevates MSX1 transcription, indicating that β-catenin works as a transcription co-activator for MSX1. Functionally, exogenous expression of MSX1 in DNAJB6-expressing cells promotes the mesenchymal phenotype by suppression of DKK1. Thus we have identified a novel regulatory mechanism of DNAJB6-mediated DKK1 transcriptional up-regulation that can influence EMT. DKK1 is a feedback regulator of β-catenin levels and thus our studies also define an additional negative control of this β-catenin/DKK1 feedback loop by MSX1, which may potentially contribute to excessive stabilization of β-catenin.
Collapse
|
28
|
Hemberger M. Health during pregnancy and beyond: Fetal trophoblast cells as chief co-ordinators of intrauterine growth and reproductive success. Ann Med 2012; 44:325-37. [PMID: 22409432 DOI: 10.3109/07853890.2012.663930] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Abstract Differentiation of extra-embryonic tissues and organs, notably the placenta, is vital for embryonic development and growth throughout gestation, starting from a few days after fertilization when the trophoblast cell lineage arises until parturition. In utero metabolic programming events may even extend the impact of placental function well into adulthood as they may predispose the offspring to common pathologies such as diabetes and cardiovascular disease. This review summarizes key steps that lead up to formation of a functional placenta. It highlights recent insights that have advanced our view of how early trophoblast expansion is achieved and how sufficient maternal blood supply to the developing fetus is secured. Exciting cumulative data have revealed the importance of a close cross-talk between the embryo proper and extra-embryonic trophoblast cells that involves extracellular matrix components in the establishment of a stem cell-like niche and proliferation compartment. Remarkably, placental function also relies on beneficial interactions between trophoblast cells and maternal immune cells at the implantation site. Our growing knowledge of the molecular mechanisms involved in trophoblast differentiation and function will help to devise informed approaches aimed at deciphering how placentation is controlled in humans as an essential process for reproductive success and long-term health.
Collapse
|
29
|
Carnini A, Scott LOM, Ahrendt E, Proft J, Winkfein RJ, Kim SW, Colicos MA, Braun JEA. Cell line specific modulation of extracellular aβ42 by Hsp40. PLoS One 2012; 7:e37755. [PMID: 22666389 PMCID: PMC3362613 DOI: 10.1371/journal.pone.0037755] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 04/24/2012] [Indexed: 01/17/2023] Open
Abstract
Heat shock proteins (Hsps) are a set of molecular chaperones involved in cellular repair. They provide protective mechanisms that allow cells to survive potentially lethal insults, In response to a conditioning stress their expression is increased. Here we examined the connection between Hsps and Aβ(42), the amyloid peptide involved in the pathological sequence of Alzheimer's disease (AD). Extracellular Aβ(42) associates with neuronal cells and is a major constituent of senile plaques, one of the hallmarks of AD. Although Hsps are generally thought to prevent accumulation of misfolded proteins, there is a lack of mechanistic evidence that heat shock chaperones directly modulate Aβ(42) toxicity. In this study we show that neither extracellular Aβ(42) nor Aβ(42/)PrP(C) trigger the heat shock response in neurons. To address the influence of the neuroprotective heat shock response on cellular Aβ(42), Western analysis of Aβ(42) was performed following external Aβ(42) application. Five hours after a conditioning heat shock, Aβ(42) association with CAD cells was increased compared to control neurons. However, at forty-eight hours following heat shock Aβ(42) levels were reduced compared to that found for control cells. Moreover, transient transfection of the stress induced Hsp40, decreased CAD levels of Aβ(42). In contrast to CAD cells, hippocampal neurons transfected with Hsp40 retained Aβ(42) indicating that Hsp40 modulation of Aβ(42) proteostasis is cell specific. Mutation of the conserved HPD motif within Hsp40 significantly reduced the Hsp40-mediated Aβ(42) increase in hippocampal cultures indicating the importance of this motif in regulating cellular Aβ(42). Our data reveal a biochemical link between Hsp40 expression and Aβ(42) proteostasis that is cell specific. Therefore, increasing Hsp40 therapeutically with the intention of interfering with the pathogenic cascade leading to neurodegeneration in AD should be pursued with caution.
Collapse
Affiliation(s)
- Anna Carnini
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Lucas O. M. Scott
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Eva Ahrendt
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Juliane Proft
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Robert J. Winkfein
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sung-Woo Kim
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Michael A. Colicos
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Janice E. A. Braun
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
30
|
Andrews JF, Sykora LJ, Letostak TB, Menezes ME, Mitra A, Barik S, Shevde LA, Samant RS. Cellular stress stimulates nuclear localization signal (NLS) independent nuclear transport of MRJ. Exp Cell Res 2012; 318:1086-93. [PMID: 22504047 DOI: 10.1016/j.yexcr.2012.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 03/12/2012] [Accepted: 03/23/2012] [Indexed: 12/14/2022]
Abstract
HSP40 family member MRJ (DNAJB6) has been in the spot light for its relevance to Huntington's, Parkinson's diseases, limb-girdle muscular dystrophy, placental development, neural stem cells, cell cycle and malignancies such as breast cancer and melanoma. This gene has two spliced variants coding for 2 distinct proteins with significant homology. However, MRJ(L) (large variant) is predominantly localized to the nucleus whereas MRJ(S) (small variant) is predominantly cytoplasmic. Interestingly MRJ(S) translocates to the nucleus in response to heat shock. The classical heat shock proteins respond to crises (stress) by increasing the number of molecules, usually by transcriptional up-regulation. Our studies imply that a quick increase in the molar concentration of MRJ in the nuclear compartment is a novel method by which MRJ responds to stress. We found that MRJ(S) shows NLS (nuclear localization signal) independent nuclear localization in response to heat shock and hypoxia. The specificity of this response is realized due to lack of such response by MRJ(S) when challenged by other stressors, such as some cytokines or UV light. Deletion analysis has allowed us to narrow down on a 20 amino acid stretch at the C-terminal region of MRJ(S) as a potential stress sensing region. Functional studies indicated that constitutive nuclear localization of MRJ(S) promoted attributes of malignancy such as proliferation and invasiveness overall indicating distinct phenotypic characteristics of nuclear MRJ(S).
Collapse
Affiliation(s)
- Joel F Andrews
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Sarparanta J, Jonson PH, Golzio C, Sandell S, Luque H, Screen M, McDonald K, Stajich JM, Mahjneh I, Vihola A, Raheem O, Penttilä S, Lehtinen S, Huovinen S, Palmio J, Tasca G, Ricci E, Hackman P, Hauser M, Katsanis N, Udd B. Mutations affecting the cytoplasmic functions of the co-chaperone DNAJB6 cause limb-girdle muscular dystrophy. Nat Genet 2012; 44:450-5, S1-2. [PMID: 22366786 PMCID: PMC3315599 DOI: 10.1038/ng.1103] [Citation(s) in RCA: 190] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 01/11/2012] [Indexed: 12/13/2022]
Abstract
Limb-girdle muscular dystrophy type 1D (LGMD1D) was linked to chromosome 7q36 over a decade ago, but its genetic cause has remained elusive. Here we studied nine LGMD-affected families from Finland, the United States and Italy and identified four dominant missense mutations leading to p.Phe93Leu or p.Phe89Ile changes in the ubiquitously expressed co-chaperone DNAJB6. Functional testing in vivo showed that the mutations have a dominant toxic effect mediated specifically by the cytoplasmic isoform of DNAJB6. In vitro studies demonstrated that the mutations increase the half-life of DNAJB6, extending this effect to the wild-type protein, and reduce its protective anti-aggregation effect. Further, we show that DNAJB6 interacts with members of the CASA complex, including the myofibrillar myopathy-causing protein BAG3. Our data identify the genetic cause of LGMD1D, suggest that its pathogenesis is mediated by defective chaperone function and highlight how mutations in a ubiquitously expressed gene can exert effects in a tissue-, isoform- and cellular compartment-specific manner.
Collapse
Affiliation(s)
- Jaakko Sarparanta
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Harms MB, Sommerville RB, Allred P, Bell S, Ma D, Cooper P, Lopate G, Pestronk A, Weihl CC, Baloh RH. Exome sequencing reveals DNAJB6 mutations in dominantly-inherited myopathy. Ann Neurol 2012; 71:407-16. [PMID: 22334415 DOI: 10.1002/ana.22683] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/23/2011] [Accepted: 11/18/2011] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To identify the causative gene in an autosomal dominant limb-girdle muscular dystrophy (LGMD) with skeletal muscle vacuoles. METHODS Exome sequencing was used to identify candidate mutations in the studied pedigree. Genome-wide linkage was then used to narrow the list of candidates to a single disease-associated mutation. Additional pedigrees with dominant or sporadic myopathy were screened for mutations in the same gene (DNAJB6) using exome sequencing. Skeletal muscle from affected patients was evaluated with histochemistry and immunohistochemical stains for dystrophy-related proteins, SMI-31, TDP43, and DNAJB6. RESULTS Exome analysis in 3 affected individuals from a family with dominant LGMD and vacuolar pathology identified novel candidate mutations in 22 genes. Linkage analysis excluded all variants except a Phe93Leu mutation in the G/F domain of the DNAJB6 gene, which resides within the LGMD locus at 7q36. Analysis of exome sequencing data from other pedigrees with dominant myopathy identified a second G/F domain mutation (Pro96Arg) in DNAJB6. Affected muscle showed mild dystrophic changes, vacuoles, and abnormal aggregation of proteins, including TDP-43 and DNAJB6 itself. INTERPRETATION Mutations within the G/F domain of DNAJB6 are a novel cause of dominantly-inherited myopathy. DNAJB6 is a member of the HSP40/DNAJ family of molecular co-chaperones tasked with protecting client proteins from irreversible aggregation during protein synthesis or during times of cellular stress. The abnormal accumulation of several proteins in patient muscle, including DNAJB6 itself, suggest that DNAJB6 function is compromised by the identified G/F domain mutations.
Collapse
Affiliation(s)
- Matthew B Harms
- Department of Neurology, Hope Center for Neurological Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
DNAJB6 chaperones PP2A mediated dephosphorylation of GSK3β to downregulate β-catenin transcription target, osteopontin. Oncogene 2012; 31:4472-83. [PMID: 22266849 DOI: 10.1038/onc.2011.623] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated levels of the oncoprotein, osteopontin (OPN), are associated with poor outcome of several types of cancers including melanoma. We have previously reported an important involvement of DNAJB6, a member of heat-shock protein 40 (HSP40) family, in negatively impacting tumor growth. The current study was prompted by our observations reported here which revealed a reciprocal relationship between DNAJB6 and OPN in melanoma specimens. The 'J domain' is the most conserved domain of HSP40 family of proteins. Hence, we assessed the functional role of the J domain in activities of DNAJB6. We report that the J domain of DNAJB6 is involved in mediating OPN suppression. Deletion of the J domain renders DNAJB6 incapable of impeding malignancy and suppressing OPN. Our mechanistic investigations reveal that DNAJB6 binds HSPA8 (heat-shock cognate protein, HSC70) and causes dephosphorylation of glycogen synthase kinase 3β (GSK3β) at Ser 9 by recruiting protein phosphatase, PP2A. This dephosphorylation activates GSK3β, leading to degradation of β-catenin and subsequent loss of TCF/LEF (T cell factor1/lymphoid enhancer factor1) activity. Deletion of the J domain abrogates assembly of this multiprotein complex and renders GSK3β inactive, thus, stabilizing β-catenin, a transcription co-activator for OPN expression. Our in-vitro and in-vivo functional analyses show that silencing OPN expression in the background of deletion of the J domain renders the resultant tumor cells less malignant despite the presence of stabilized β-catenin. Thus, we have uncovered a new mechanism for regulation of GSK3β activity leading to inhibition of Wnt/β-catenin signaling.
Collapse
|
34
|
Mitra A, Menezes ME, Pannell LK, Mulekar MS, Honkanen RE, Shevde LA, Samant RS. DNAJB6 chaperones PP2A mediated dephosphorylation of GSK3β to downregulate β-catenin transcription target, osteopontin. Oncogene 2012. [PMID: 22266849 DOI: 10.1038/onc.2011.623.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elevated levels of the oncoprotein, osteopontin (OPN), are associated with poor outcome of several types of cancers including melanoma. We have previously reported an important involvement of DNAJB6, a member of heat-shock protein 40 (HSP40) family, in negatively impacting tumor growth. The current study was prompted by our observations reported here which revealed a reciprocal relationship between DNAJB6 and OPN in melanoma specimens. The 'J domain' is the most conserved domain of HSP40 family of proteins. Hence, we assessed the functional role of the J domain in activities of DNAJB6. We report that the J domain of DNAJB6 is involved in mediating OPN suppression. Deletion of the J domain renders DNAJB6 incapable of impeding malignancy and suppressing OPN. Our mechanistic investigations reveal that DNAJB6 binds HSPA8 (heat-shock cognate protein, HSC70) and causes dephosphorylation of glycogen synthase kinase 3β (GSK3β) at Ser 9 by recruiting protein phosphatase, PP2A. This dephosphorylation activates GSK3β, leading to degradation of β-catenin and subsequent loss of TCF/LEF (T cell factor1/lymphoid enhancer factor1) activity. Deletion of the J domain abrogates assembly of this multiprotein complex and renders GSK3β inactive, thus, stabilizing β-catenin, a transcription co-activator for OPN expression. Our in-vitro and in-vivo functional analyses show that silencing OPN expression in the background of deletion of the J domain renders the resultant tumor cells less malignant despite the presence of stabilized β-catenin. Thus, we have uncovered a new mechanism for regulation of GSK3β activity leading to inhibition of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- A Mitra
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Watson ED, Hughes M, Simmons DG, Natale DR, Sutherland AE, Cross JC. Cell-cell adhesion defects in Mrj mutant trophoblast cells are associated with failure to pattern the chorion during early placental development. Dev Dyn 2011; 240:2505-19. [DOI: 10.1002/dvdy.22755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2011] [Indexed: 11/12/2022] Open
|
36
|
Stetler RA, Gan Y, Zhang W, Liou AK, Gao Y, Cao G, Chen J. Heat shock proteins: cellular and molecular mechanisms in the central nervous system. Prog Neurobiol 2010; 92:184-211. [PMID: 20685377 DOI: 10.1016/j.pneurobio.2010.05.002] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 05/23/2010] [Accepted: 05/27/2010] [Indexed: 12/30/2022]
Abstract
Emerging evidence indicates that heat shock proteins (HSPs) are critical regulators in normal neural physiological function as well as in cell stress responses. The functions of HSPs represent an enormous and diverse range of cellular activities, far beyond the originally identified roles in protein folding and chaperoning. HSPs are now understood to be involved in processes such as synaptic transmission, autophagy, ER stress response, protein kinase and cell death signaling. In addition, manipulation of HSPs has robust effects on the fate of cells in neurological injury and disease states. The ongoing exploration of multiple HSP superfamilies has underscored the pluripotent nature of HSPs in the cellular context, and has demanded the recent revamping of the nomenclature referring to these families to reflect a re-organization based on structure and function. In keeping with this re-organization, we first discuss the HSP superfamilies in terms of protein structure, regulation, expression and distribution in the brain. We then explore major cellular functions of HSPs that are relevant to neural physiological states, and from there we discuss known and proposed HSP impacts on major neurological disease states. This review article presents a three-part discussion on the array of HSP families relevant to neuronal tissue, their cellular functions, and the exploration of therapeutic targets of these proteins in the context of neurological diseases.
Collapse
Affiliation(s)
- R Anne Stetler
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, United States.
| | | | | | | | | | | | | |
Collapse
|
37
|
Hageman J, Rujano MA, van Waarde MAWH, Kakkar V, Dirks RP, Govorukhina N, Oosterveld-Hut HMJ, Lubsen NH, Kampinga HH. A DNAJB chaperone subfamily with HDAC-dependent activities suppresses toxic protein aggregation. Mol Cell 2010; 37:355-69. [PMID: 20159555 DOI: 10.1016/j.molcel.2010.01.001] [Citation(s) in RCA: 277] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 09/22/2009] [Accepted: 11/24/2009] [Indexed: 11/25/2022]
Abstract
Misfolding and aggregation are associated with cytotoxicity in several protein folding diseases. A large network of molecular chaperones ensures protein quality control. Here, we show that within the Hsp70, Hsp110, and Hsp40 (DNAJ) chaperone families, members of a subclass of the DNAJB family (particularly DNAJB6b and DNAJB8) are superior suppressors of aggregation and toxicity of disease-associated polyglutamine proteins. The antiaggregation activity is largely independent of the N-terminal Hsp70-interacting J-domain. Rather, a C-terminal serine-rich (SSF-SST) region and the C-terminal tail are essential. The SSF-SST region is involved in substrate binding, formation of polydisperse oligomeric complexes, and interaction with histone deacetylases (HDAC4, HDAC6, SIRT2). Inhibiting HDAC4 reduced DNAJB8 function. DNAJB8 is (de)acetylated at two conserved C-terminal lysines that are not involved in substrate binding, but do play a role in suppressing protein aggregation. Combined, our data provide a functional link between HDACs and DNAJs in suppressing cytotoxic protein aggregation.
Collapse
Affiliation(s)
- Jurre Hageman
- Department of Cell Biology, Section of Radiation and Stress Cell Biology, University Medical Center Groningen and University of Groningen, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Löffek S, Wöll S, Höhfeld J, Leube RE, Has C, Bruckner-Tuderman L, Magin TM. The ubiquitin ligase CHIP/STUB1 targets mutant keratins for degradation. Hum Mutat 2010; 31:466-76. [DOI: 10.1002/humu.21222] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Watson ED, Mattar P, Schuurmans C, Cross JC. Neural stem cell self-renewal requires the Mrj co-chaperone. Dev Dyn 2009; 238:2564-74. [DOI: 10.1002/dvdy.22088] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
40
|
Durrenberger PF, Filiou MD, Moran LB, Michael GJ, Novoselov S, Cheetham ME, Clark P, Pearce RKB, Graeber MB. DnaJB6 is present in the core of Lewy bodies and is highly up-regulated in parkinsonian astrocytes. J Neurosci Res 2009; 87:238-45. [PMID: 18711724 DOI: 10.1002/jnr.21819] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
DnaJ/Hsp40 chaperones determine the activity of Hsp70s by stabilizing their interaction with substrate proteins. We have predicted, based on the in silico analysis of a brain-derived whole-genome transcriptome data set, an increased expression of DnaJ/Hsp40 homologue, subfamily B, member 6 (DnaJB6) in Parkinson's disease (PD; Moran et al. [2006] Neurogenetics 7:1-11). We now show that DnaJB6 is a novel component of Lewy bodies (LBs) in both PD substantia nigra and PD cortex and that it is strongly up-regulated in parkinsonian astrocytes. The presence of DnaJB6 in the center of LBs suggests an early and direct involvement of this chaperone in the neuronal disease process associated with PD. The strong concomitant expression of DnaJB6 in astrocytes emphasizes the involvement of glial cells in PD and could indicate a route for therapeutic intervention. Extracellular alpha-synuclein originating from intravesicular alpha-synuclein is prone to aggregation and the potential source of extracellular aggregates (Lee [2008] J. Mol. Neurosci. 34:17-22). The observed strong expression of DnaJB6 by astrocytes could reflect a protective reaction, so reducing the neuronal release of toxic alpha-synuclein and supporting the astrocyte response in PD might limit the progression of the disease process.
Collapse
Affiliation(s)
- P F Durrenberger
- University Department of Neuropathology, Imperial College, University of London, and Hammersmith Hospitals Trust, London, United Kindom
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lens intermediate filaments. Exp Eye Res 2008; 88:165-72. [PMID: 19071112 DOI: 10.1016/j.exer.2008.11.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 11/04/2008] [Accepted: 11/04/2008] [Indexed: 12/12/2022]
Abstract
The ocular lens assembles two separate intermediate filament systems sequentially with differentiation. Canonical 8-11 nm IFs composed of Vimentin are assembled in lens epithelial cells and younger fiber cells, while the fiber cell-specific beaded filaments are switched on as fiber cell elongation initiates. Some of the key features of both filament systems are reviewed.
Collapse
|
42
|
Dey S, Banerjee P, Saha P. Cell cycle specific expression and nucleolar localization of human J-domain containing co-chaperone Mrj. Mol Cell Biochem 2008; 322:137-42. [PMID: 19002655 DOI: 10.1007/s11010-008-9950-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Accepted: 10/22/2008] [Indexed: 01/28/2023]
Abstract
J-domain containing co-chaperone Mrj (mammalian relative to DnaJ) has been implicated in diverse cellular functions including placental development and inhibition of Huntingtin mediated cytotoxicity. It has also been shown to interact with keratin intermediate filaments. Since keratins undergo extensive reorganization during cell division, its interactor Mrj might also play an important role in the regulation of cell cycle. In support of this hypothesis, we report the up-regulation of Mrj protein in M-phase of HeLa cells implicating its role in mitosis related activities. The protein is dispersed throughout the cell during late mitosis and is localized in nucleolus during interphase, confirming that the activity of Mrj is regulated by its cell cycle specific expression together with its differential subcellular localization.
Collapse
Affiliation(s)
- Sanjib Dey
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Sector I, Block AF, Bidhannagar, Kolkata, 700064, India
| | | | | |
Collapse
|
43
|
Maurer J, Nelson B, Ceceña G, Bajpai R, Mercola M, Terskikh A, Oshima RG. Contrasting expression of keratins in mouse and human embryonic stem cells. PLoS One 2008; 3:e3451. [PMID: 18941637 PMCID: PMC2565505 DOI: 10.1371/journal.pone.0003451] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 09/28/2008] [Indexed: 02/04/2023] Open
Abstract
RNA expression data reveals that human embryonic stem (hES) cells differ from mouse ES (mES) cells in the expression of RNAs for keratin intermediate filament proteins. These differences were confirmed at the cellular and protein level and may reflect a fundamental difference in the epithelial nature of embryonic stem cells derived from mouse and human blastocysts. Mouse ES cells express very low levels of the simple epithelial keratins K8, K18 and K19. By contrast hES cells express moderate levels of the RNAs for these intermediate filament proteins as do mouse stem cells derived from the mouse epiblast. Expression of K8 and K18 RNAs are correlated with increased c-Jun RNA expression in both mouse and human ES cell cultures. However, decreasing K8 and K18 expression associated with differentiation to neuronal progenitor cells is correlated with increasing expression of the Snai2 (Slug) transcriptional repression and not decreased Jun expression. Increasing K7 expression is correlated with increased CDX2 and decreased Oct4 RNA expression associated with the formation of trophoblast derivatives by hES cells. Our study supports the view that hES cells are more similar to mouse epiblast cells than mouse ES cells and is consistent with the epithelial nature of hES cells. Keratin intermediate filament expression in hES cells may modulate sensitivity to death receptor mediated apoptosis and stress.
Collapse
Affiliation(s)
- Jochen Maurer
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Brandon Nelson
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Grace Ceceña
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Ruchi Bajpai
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Mark Mercola
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Alexey Terskikh
- Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Robert G. Oshima
- Burnham Institute for Medical Research, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Vos MJ, Hageman J, Carra S, Kampinga HH. Structural and functional diversities between members of the human HSPB, HSPH, HSPA, and DNAJ chaperone families. Biochemistry 2008; 47:7001-11. [PMID: 18557634 DOI: 10.1021/bi800639z] [Citation(s) in RCA: 272] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heat shock proteins (HSPs) were originally identified as stress-responsive proteins required to deal with proteotoxic stresses. Besides being stress-protective and possible targets for delaying progression of protein folding diseases, mutations in chaperones also have been shown to cause disease (chaperonopathies). The mechanism of action of the "classical", stress-inducible HSPs in serving as molecular chaperones preventing the irreversible aggregation of stress-unfolded or disease-related misfolded proteins is beginning to emerge. However, the human genome encodes several members for each of the various HSP families that are not stress-related but contain conserved domains. Here, we have reviewed the existing literature on the various members of the human HSPB (HSP27), HSPH (HSP110), HSPA (HSP70), and DNAJ (HSP40) families. Apart from structural and functional homologies, several diversities between members and families can be found that not only point to differences in client specificity but also seem to serve differential client handling and processing. How substrate specificity and client processing is determined is far from being understood.
Collapse
Affiliation(s)
- Michel J Vos
- Department of Cell Biology, Section of Radiation and Stress Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
45
|
Pan Z, Sikandar S, Witherspoon M, Dizon D, Nguyen T, Benirschke K, Wiley C, Vrana P, Lipkin SM. Impaired placental trophoblast lineage differentiation in Alkbh1(-/-) mice. Dev Dyn 2008; 237:316-27. [PMID: 18163532 DOI: 10.1002/dvdy.21418] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
E. coli AlkB has been intensively studied since 1983, but the in vivo roles of its mammalian homologue Alkbh1 are unknown. We, therefore, created null mice for Alkbh1. Alkbh1 mRNA is expressed at highest levels in the trophoblast lineages of the developing placenta. Alkbh1(-/-) placentas have decreased expression of differentiated trophoblast markers including Tpbp, Gcm1, and Pl-1, and increased expression of the trophoblast stem cell marker Eomes. Alkbh1 localizes to nuclear euchromatin, and interacts strongly with Mrj, an essential placental gene that mediates gene repression by recruitment of class II histone deacetylases (HDACs). Competition experiments show Alkbh1 and HDAC4 binding to Mrj are mutually exclusive, which causes decreased HDAC activity and increased target gene expression. Our study demonstrates Alkbh1 performs important functions in placental trophoblast lineage differentiation and participates in mechanisms of transcriptional regulation.
Collapse
Affiliation(s)
- Zishu Pan
- Department of Medicine, University of California, Irvine, Irvine, California 92697, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kim S, Coulombe PA. Intermediate filament scaffolds fulfill mechanical, organizational, and signaling functions in the cytoplasm. Genes Dev 2007; 21:1581-97. [PMID: 17606637 DOI: 10.1101/gad.1552107] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Intermediate filaments (IFs) are cytoskeletal polymers whose protein constituents are encoded by a large family of differentially expressed genes. Owing in part to their properties and intracellular organization, IFs provide crucial structural support in the cytoplasm and nucleus, the perturbation of which causes cell and tissue fragility and accounts for a large number of genetic diseases in humans. A number of additional roles, nonmechanical in nature, have been recently uncovered for IF proteins. These include the regulation of key signaling pathways that control cell survival, cell growth, and vectorial processes including protein targeting in polarized cellular settings. As this discovery process continues to unfold, a rationale for the large size of this family and the context-dependent regulation of its members is finally emerging.
Collapse
Affiliation(s)
- Seyun Kim
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|