401
|
Hattori N, Arano T, Hatano T, Mori A, Imai Y. Mitochondrial-Associated Membranes in Parkinson's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:157-169. [PMID: 28815529 DOI: 10.1007/978-981-10-4567-7_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder, with ageing being a major risk factor. Accordingly, estimates predict an increasing number of PD patients due to our expanding life span. Consequently, developing a true disease-modifying therapy is necessary. In this regard, monogenic PD offers a suitable means for determining pathogenesis. Among monogenic forms of PD, mitochondrial dysfunction may be a major cause and is also likely to be involved in sporadic PD. Thus, mitochondrial impairment may be a common pathway. Recently, mitochondria-associated membranes (MAM) were identified as dynamic sites between mitochondria and endoplasmic reticulum. Indeed, the gene product of α-synuclein is a major component of MAM, with other gene products also involved. This review focuses on the possibility of using MAM as novel therapeutic targets.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
| | - Taku Arano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Akio Mori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
402
|
Bonneau B, Ando H, Kawaai K, Hirose M, Takahashi-Iwanaga H, Mikoshiba K. IRBIT controls apoptosis by interacting with the Bcl-2 homolog, Bcl2l10, and by promoting ER-mitochondria contact. eLife 2016; 5. [PMID: 27995898 PMCID: PMC5173324 DOI: 10.7554/elife.19896] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/24/2016] [Indexed: 12/15/2022] Open
Abstract
IRBIT is a molecule that interacts with the inositol 1,4,5-trisphosphate (IP3)-binding pocket of the IP3 receptor (IP3R), whereas the antiapoptotic protein, Bcl2l10, binds to another part of the IP3-binding domain. Here we show that Bcl2l10 and IRBIT interact and exert an additive inhibition of IP3R in the physiological state. Moreover, we found that these proteins associate in a complex in mitochondria-associated membranes (MAMs) and that their interplay is involved in apoptosis regulation. MAMs are a hotspot for Ca2+ transfer between endoplasmic reticulum (ER) and mitochondria, and massive Ca2+ release through IP3R in mitochondria induces cell death. We found that upon apoptotic stress, IRBIT is dephosphorylated, becoming an inhibitor of Bcl2l10. Moreover, IRBIT promotes ER mitochondria contact. Our results suggest that by inhibiting Bcl2l10 activity and promoting contact between ER and mitochondria, IRBIT facilitates massive Ca2+ transfer to mitochondria and promotes apoptosis. This work then describes IRBIT as a new regulator of cell death.
Collapse
Affiliation(s)
- Benjamin Bonneau
- Laboratory for Developmental Neurobiology, RIKEN Brain Science institute, Wako-shi, Japan
| | - Hideaki Ando
- Laboratory for Developmental Neurobiology, RIKEN Brain Science institute, Wako-shi, Japan
| | - Katsuhiro Kawaai
- Laboratory for Developmental Neurobiology, RIKEN Brain Science institute, Wako-shi, Japan
| | - Matsumi Hirose
- Laboratory for Developmental Neurobiology, RIKEN Brain Science institute, Wako-shi, Japan
| | | | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science institute, Wako-shi, Japan
| |
Collapse
|
403
|
Watanabe S, Ilieva H, Tamada H, Nomura H, Komine O, Endo F, Jin S, Mancias P, Kiyama H, Yamanaka K. Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR1- and SOD1-linked ALS. EMBO Mol Med 2016; 8:1421-1437. [PMID: 27821430 PMCID: PMC5167132 DOI: 10.15252/emmm.201606403] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 09/21/2016] [Accepted: 09/27/2016] [Indexed: 01/11/2023] Open
Abstract
A homozygous mutation in the gene for sigma 1 receptor (Sig1R) is a cause of inherited juvenile amyotrophic lateral sclerosis (ALS16). Sig1R localizes to the mitochondria-associated membrane (MAM), which is an interface of mitochondria and endoplasmic reticulum. However, the role of the MAM in ALS is not fully elucidated. Here, we identified a homozygous p.L95fs mutation of Sig1R as a novel cause of ALS16. ALS-linked Sig1R variants were unstable and incapable of binding to inositol 1,4,5-triphosphate receptor type 3 (IP3R3). The onset of mutant Cu/Zn superoxide dismutase (SOD1)-mediated ALS disease in mice was accelerated when Sig1R was deficient. Moreover, either deficiency of Sig1R or accumulation of mutant SOD1 induced MAM disruption, resulting in mislocalization of IP3R3 from the MAM, calpain activation, and mitochondrial dysfunction. Our findings indicate that a loss of Sig1R function is causative for ALS16, and collapse of the MAM is a common pathomechanism in both Sig1R- and SOD1-linked ALS Furthermore, our discovery of the selective enrichment of IP3R3 in motor neurons suggests that integrity of the MAM is crucial for the selective vulnerability in ALS.
Collapse
Affiliation(s)
- Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Hristelina Ilieva
- Houston Methodist Hospital, Houston, TX, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Hiromi Tamada
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hanae Nomura
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Shijie Jin
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Pedro Mancias
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, USA
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
404
|
Sariki SK, Sahu PK, Golla U, Singh V, Azad GK, Tomar RS. Sen1, the homolog of human Senataxin, is critical for cell survival through regulation of redox homeostasis, mitochondrial function, and the TOR pathway inSaccharomyces cerevisiae. FEBS J 2016; 283:4056-4083. [DOI: 10.1111/febs.13917] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/30/2016] [Accepted: 10/05/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Santhosh Kumar Sariki
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| | - Pushpendra Kumar Sahu
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| | - Upendarrao Golla
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| | - Vikash Singh
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| | - Gajendra Kumar Azad
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| | - Raghuvir S. Tomar
- Laboratory of Chromatin Biology; Department of Biological Sciences; Indian Institute of Science Education and Research; Bhopal India
| |
Collapse
|
405
|
Brobeil A, Dietel E, Gattenlöhner S, Wimmer M. Orchestrating cellular signaling pathways-the cellular "conductor" protein tyrosine phosphatase interacting protein 51 (PTPIP51). Cell Tissue Res 2016; 368:411-423. [PMID: 27734150 DOI: 10.1007/s00441-016-2508-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/07/2016] [Indexed: 12/28/2022]
Abstract
The protein tyrosine phosphatase interacting protein 51 (PTPIP51) is thought to regulate crucial cellular functions such as mitosis, apoptosis, migration, differentiation and communication between organelles as a scaffold protein. These diverse functions are modulated by the tyrosine/serine phosphorylation status of PTPIP51. This review interconnects the insights obtained about the action of PTPIP51 in mitogen-activated protein kinase signaling, nuclear factor kB signaling, calcium homeostasis and chromosomal segregation and identifies important signaling hubs. The interference of PTPIP51 in such multiprotein complexes and their PTPIP51-modulated cross-talk makes PTPIP51 an ideal target for novel drugs such as the small molecule LDC-3. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Alexander Brobeil
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, 35392, Giessen, Germany. .,Institute of Pathology, Justus-Liebig-University, 35392, Giessen, Germany.
| | - Eric Dietel
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, 35392, Giessen, Germany
| | | | - Monika Wimmer
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, 35392, Giessen, Germany
| |
Collapse
|
406
|
Rodríguez-Arribas M, Yakhine-Diop SMS, Pedro JMBS, Gómez-Suaga P, Gómez-Sánchez R, Martínez-Chacón G, Fuentes JM, González-Polo RA, Niso-Santano M. Mitochondria-Associated Membranes (MAMs): Overview and Its Role in Parkinson's Disease. Mol Neurobiol 2016; 54:6287-6303. [PMID: 27714635 DOI: 10.1007/s12035-016-0140-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/19/2016] [Indexed: 12/21/2022]
Abstract
Mitochondria-associated membranes (MAMs) are structures that regulate physiological functions between endoplasmic reticulum (ER) and mitochondria in order to maintain calcium signaling and mitochondrial biogenesis. Several proteins located in MAMs, including those encoded by PARK genes and some of neurodegeneration-related proteins (huntingtin, presenilin, etc.), ensure this regulation. In this regard, MAM alteration is associated with neurodegenerative diseases such as Parkinson's (PD), Alzheimer's (AD), and Huntington's diseases (HD) and contributes to the appearance of the pathogenesis features, i.e., autophagy dysregulation, mitochondrial dysfunction, oxidative stress, and lately, neuronal death. Moreover,, ER stress and/or damaged mitochondria can be the cause of these disruptions. Therefore, ER-mitochondria contact structure and function are crucial to multiple cellular processes. This review is focused on the molecular interaction between ER and mitochondria indispensable to MAM formation and on MAM alteration-induced etiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- M Rodríguez-Arribas
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain.,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain
| | - S M S Yakhine-Diop
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain.,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain
| | - J M Bravo-San Pedro
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006, Paris, France.,INSERM U1138, 75006, Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006, Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006, Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805, Villejuif, France
| | - P Gómez-Suaga
- Department Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute Kings College London, London, SE5 9RX, UK
| | - R Gómez-Sánchez
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - G Martínez-Chacón
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain.,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain
| | - J M Fuentes
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain.,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain
| | - R A González-Polo
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain. .,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain.
| | - M Niso-Santano
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Extremadura, Avda. De la Universidad S/N, C.P, 10003, Cáceres, Cáceres, Spain. .,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Avda. de la Universidad s/n, C.P, 10003, Cáceres, Cáceres, Spain.
| |
Collapse
|
407
|
Piecing Together the Patchwork of Contact Sites. Trends Cell Biol 2016; 27:214-229. [PMID: 27717534 DOI: 10.1016/j.tcb.2016.08.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/10/2016] [Accepted: 08/25/2016] [Indexed: 11/23/2022]
Abstract
Contact sites are places where two organelles join together to carry out a shared activity requiring nonvesicular communication. A large number of contact sites have been discovered, and almost any two organelles can contact each other. General rules about contacts include constraints on bridging proteins, with only a minority of bridges physically creating contacts by acting as 'tethers'. The downstream effects of contacts include changing the physical behaviour of organelles, and also forming biochemically heterogeneous subdomains. However, some functions typically localized to contact sites, such as lipid transfer, have no absolute requirement to be situated there. Therefore, the key aspect of contacts is the directness of communication, which allows metabolic channelling and collective regulation.
Collapse
|
408
|
East DA, Campanella M. Mitophagy and the therapeutic clearance of damaged mitochondria for neuroprotection. Int J Biochem Cell Biol 2016; 79:382-387. [DOI: 10.1016/j.biocel.2016.08.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/20/2016] [Indexed: 12/23/2022]
|
409
|
Giedt RJ, Fumene Feruglio P, Pathania D, Yang KS, Kilcoyne A, Vinegoni C, Mitchison TJ, Weissleder R. Computational imaging reveals mitochondrial morphology as a biomarker of cancer phenotype and drug response. Sci Rep 2016; 6:32985. [PMID: 27609668 PMCID: PMC5017129 DOI: 10.1038/srep32985] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/16/2016] [Indexed: 12/27/2022] Open
Abstract
Mitochondria, which are essential organelles in resting and replicating cells, can vary in number, mass and shape. Past research has primarily focused on short-term molecular mechanisms underlying fission/fusion. Less is known about longer-term mitochondrial behavior such as the overall makeup of cell populations’ morphological patterns and whether these patterns can be used as biomarkers of drug response in human cells. We developed an image-based analytical technique to phenotype mitochondrial morphology in different cancers, including cancer cell lines and patient-derived cancer cells. We demonstrate that (i) cancer cells of different origins, including patient-derived xenografts, express highly diverse mitochondrial phenotypes; (ii) a given phenotype is characteristic of a cell population and fairly constant over time; (iii) mitochondrial patterns correlate with cell metabolic measurements and (iv) therapeutic interventions can alter mitochondrial phenotypes in drug-sensitive cancers as measured in pre- versus post-treatment fine needle aspirates in mice. These observations shed light on the role of mitochondrial dynamics in the biology and drug response of cancer cells. On the basis of these findings, we propose that image-based mitochondrial phenotyping can provide biomarkers for assessing cancer phenotype and drug response.
Collapse
Affiliation(s)
- Randy J Giedt
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA
| | - Paolo Fumene Feruglio
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA.,Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Divya Pathania
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA
| | - Katherine S Yang
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA
| | - Aoife Kilcoyne
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge St., CPZN 5206, Boston, MA 02114, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| |
Collapse
|
410
|
Stoica R, Paillusson S, Gomez-Suaga P, Mitchell JC, Lau DH, Gray EH, Sancho RM, Vizcay-Barrena G, De Vos KJ, Shaw CE, Hanger DP, Noble W, Miller CC. ALS/FTD-associated FUS activates GSK-3β to disrupt the VAPB-PTPIP51 interaction and ER-mitochondria associations. EMBO Rep 2016; 17:1326-42. [PMID: 27418313 PMCID: PMC5007559 DOI: 10.15252/embr.201541726] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/06/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
Defective FUS metabolism is strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), but the mechanisms linking FUS to disease are not properly understood. However, many of the functions disrupted in ALS/FTD are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling is facilitated by close physical associations between the two organelles that are mediated by binding of the integral ER protein VAPB to the outer mitochondrial membrane protein PTPIP51, which act as molecular scaffolds to tether the two organelles. Here, we show that FUS disrupts the VAPB-PTPIP51 interaction and ER-mitochondria associations. These disruptions are accompanied by perturbation of Ca(2+) uptake by mitochondria following its release from ER stores, which is a physiological read-out of ER-mitochondria contacts. We also demonstrate that mitochondrial ATP production is impaired in FUS-expressing cells; mitochondrial ATP production is linked to Ca(2+) levels. Finally, we demonstrate that the FUS-induced reductions to ER-mitochondria associations and are linked to activation of glycogen synthase kinase-3β (GSK-3β), a kinase already strongly associated with ALS/FTD.
Collapse
Affiliation(s)
- Radu Stoica
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Sébastien Paillusson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Jacqueline C Mitchell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Dawn Hw Lau
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Emma H Gray
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Rosa M Sancho
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | | | - Kurt J De Vos
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher Cj Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| |
Collapse
|
411
|
Eden ER. The formation and function of ER-endosome membrane contact sites. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:874-879. [PMID: 26898183 PMCID: PMC4917889 DOI: 10.1016/j.bbalip.2016.01.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 02/02/2023]
Abstract
Recent advances in membrane contact site (MCS) biology have revealed key roles for MCSs in inter-organellar exchange, the importance of which is becoming increasingly apparent. Roles for MCSs in many essential physiological processes including lipid transfer, calcium exchange, receptor tyrosine kinase signalling, lipid droplet formation, autophagosome formation, organelle dynamics and neurite outgrowth have been reported. The ER forms an extensive and dynamic network of MCSs with a diverse range of functionally distinct organelles. MCSs between the ER and endocytic pathway are particularly abundant, suggesting important physiological roles. Here, our current knowledge of the formation and function of ER contact sites with endocytic organelles from studies in mammalian systems is reviewed. Their relatively poorly defined molecular composition and recently identified functions are discussed. In addition, likely, but yet to be established, roles for these contacts in lipid transfer and calcium signalling are considered. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
412
|
Dimmer KS, Rapaport D. Mitochondrial contact sites as platforms for phospholipid exchange. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:69-80. [PMID: 27477677 DOI: 10.1016/j.bbalip.2016.07.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/21/2016] [Accepted: 07/22/2016] [Indexed: 12/15/2022]
Abstract
Mitochondria are unique organelles that contain their own - although strongly reduced - genome, and are surrounded by two membranes. While most cellular phospholipid biosynthesis takes place in the ER, mitochondria harbor the whole spectrum of glycerophospholipids common to biological membranes. Mitochondria also contribute to overall phospholipid biosynthesis in cells by producing phosphatidylethanolamine, phosphatidylglycerol, and cardiolipin. Considering these features, it is not surprising that mitochondria maintain highly active exchange of phospholipids with other cellular compartments. In this contribution we describe the transport of phospholipids between mitochondria and other organelles, and discuss recent developments in our understanding of the molecular functions of the protein complexes that mediate these processes. This article is part of a Special Issue entitled: Lipids of Mitochondria edited by Guenther Daum.
Collapse
Affiliation(s)
- Kai Stefan Dimmer
- Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany.
| | - Doron Rapaport
- Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| |
Collapse
|
413
|
Carrì MT, D'Ambrosi N, Cozzolino M. Pathways to mitochondrial dysfunction in ALS pathogenesis. Biochem Biophys Res Commun 2016; 483:1187-1193. [PMID: 27416757 DOI: 10.1016/j.bbrc.2016.07.055] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/10/2016] [Indexed: 12/20/2022]
Abstract
Alterations in the structure and functions of mitochondria are a typical trait of Amyotrophic Lateral Sclerosis, a neurodegenerative disease characterized by a prominent degeneration of upper and lower motor neurons. The known gene mutations that are responsible for a small fraction of ALS cases point to a complex interplay between different mechanisms in the disease pathogenesis. Here we will briefly overview the genetic and mechanistic evidence that make dysfunction of mitochondria a candidate major player in this process.
Collapse
Affiliation(s)
- Maria Teresa Carrì
- Department of Biology, Università di Roma "Tor Vergata", Via della Ricerca Scientifica, 00133, Rome, Italy; Fondazione Santa Lucia, IRCCS, Via del Fosso di Fiorano 64, 00143, Rome, Italy.
| | - Nadia D'Ambrosi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Mauro Cozzolino
- Institute of Translational Pharmacology, CNR, Via del Fosso del Cavaliere 100, 00133, Rome, Italy.
| |
Collapse
|
414
|
Ansoleaga B, Garcia-Esparcia P, Llorens F, Hernández-Ortega K, Carmona Tech M, Antonio Del Rio J, Zerr I, Ferrer I. Altered Mitochondria, Protein Synthesis Machinery, and Purine Metabolism Are Molecular Contributors to the Pathogenesis of Creutzfeldt-Jakob Disease. J Neuropathol Exp Neurol 2016; 75:755-769. [PMID: 27297670 DOI: 10.1093/jnen/nlw048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuron loss, synaptic decline, and spongiform change are the hallmarks of sporadic Creutzfeldt-Jakob disease (sCJD), and may be related to deficiencies in mitochondria, energy metabolism, and protein synthesis. To investigate these relationships, we determined the expression levels of genes encoding subunits of the 5 protein complexes of the electron transport chain, proteins involved in energy metabolism, nucleolar and ribosomal proteins, and enzymes of purine metabolism in frontal cortex samples from 15 cases of sCJD MM1 and age-matched controls. We also assessed the protein expression levels of subunits of the respiratory chain, initiation and elongation translation factors of protein synthesis, and localization of selected mitochondrial components. We identified marked, generalized alterations of mRNA and protein expression of most subunits of all 5 mitochondrial respiratory chain complexes in sCJD cases. Expression of molecules involved in protein synthesis and purine metabolism were also altered in sCJD. These findings point to altered mRNA and protein expression of components of mitochondria, protein synthesis machinery, and purine metabolism as components of the pathogenesis of CJD.
Collapse
Affiliation(s)
- Belén Ansoleaga
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Paula Garcia-Esparcia
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Franc Llorens
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Karina Hernández-Ortega
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Margarita Carmona Tech
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - José Antonio Del Rio
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Inga Zerr
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF)
| | - Isidro Ferrer
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain (BA, PG-E, KH-O, MC, IF); CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain (PG-E, KH-O, MC, JAR, IF); Department of Neurology, Clinical Dementia Center, University Medical School, Georg-August University and German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany (FL, IZ); Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain (JAR); and Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain (IF).
| |
Collapse
|
415
|
Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P. Presenilin 2 Modulates Endoplasmic Reticulum-Mitochondria Coupling by Tuning the Antagonistic Effect of Mitofusin 2. Cell Rep 2016; 15:2226-2238. [PMID: 27239030 DOI: 10.1016/j.celrep.2016.05.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/23/2016] [Accepted: 04/28/2016] [Indexed: 01/31/2023] Open
Abstract
Communication between organelles plays key roles in cell biology. In particular, physical and functional coupling of the endoplasmic reticulum (ER) and mitochondria is crucial for regulation of various physiological and pathophysiological processes. Here, we demonstrate that Presenilin 2 (PS2), mutations in which underlie familial Alzheimer's disease (FAD), promotes ER-mitochondria coupling only in the presence of mitofusin 2 (Mfn2). PS2 is not necessary for the antagonistic effect of Mfn2 on organelle coupling, although its abundance can tune it. The two proteins physically interact, whereas their homologues Mfn1 and PS1 are dispensable for this interplay. Moreover, PS2 mutants associated with FAD are more effective than the wild-type form in modulating ER-mitochondria tethering because their binding to Mfn2 in mitochondria-associated membranes is favored. We propose a revised model for ER-mitochondria interaction to account for these findings and discuss possible implications for FAD pathogenesis.
Collapse
Affiliation(s)
- Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, Padua 35131, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, Padua 35131, Italy; Department of Biomedical Sciences, Institute of Neuroscience, Italian National Research Council (CNR), via U. Bassi 58/B, Padua 35131, Italy
| | - Gabriele Turacchio
- Department of Biomedical Sciences, Institute of Protein Biochemistry, Italian National Research Council (CNR), via P. Castellino 111, Naples 80131, Italy
| | - Alberto Luini
- Department of Biomedical Sciences, Institute of Protein Biochemistry, Italian National Research Council (CNR), via P. Castellino 111, Naples 80131, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, Padua 35131, Italy; Venetian Institute of Molecular Medicine, via Orus 2, Padua 35131, Italy; Department of Biomedical Sciences, Institute of Neuroscience, Italian National Research Council (CNR), via U. Bassi 58/B, Padua 35131, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, Padua 35131, Italy.
| |
Collapse
|
416
|
Gautier CA, Erpapazoglou Z, Mouton-Liger F, Muriel MP, Cormier F, Bigou S, Duffaure S, Girard M, Foret B, Iannielli A, Broccoli V, Dalle C, Bohl D, Michel PP, Corvol JC, Brice A, Corti O. The endoplasmic reticulum-mitochondria interface is perturbed in PARK2 knockout mice and patients with PARK2 mutations. Hum Mol Genet 2016; 25:2972-2984. [PMID: 27206984 DOI: 10.1093/hmg/ddw148] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/10/2016] [Accepted: 05/10/2016] [Indexed: 12/13/2022] Open
Abstract
Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.
Collapse
Affiliation(s)
- Clément A Gautier
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Zoi Erpapazoglou
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - François Mouton-Liger
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Marie Paule Muriel
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Florence Cormier
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
| | - Stéphanie Bigou
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Sophie Duffaure
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Mathilde Girard
- CECS, I-Stem, AFM, Institute of Stem Cell Therapy and Exploration of Monogenic Diseases, 91030 Evry cedex, France
| | - Benjamin Foret
- CECS, I-Stem, AFM, Institute of Stem Cell Therapy and Exploration of Monogenic Diseases, 91030 Evry cedex, France
| | - Angelo Iannielli
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vania Broccoli
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Carine Dalle
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Delphine Bohl
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Patrick P Michel
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Jean-Christophe Corvol
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
- Assistance-publique Hôpitaux de Paris, Inserm, CIC-1422, Department of Neurology, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Alexis Brice
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Olga Corti
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| |
Collapse
|
417
|
Alsultan AA, Waller R, Heath PR, Kirby J. The genetics of amyotrophic lateral sclerosis: current insights. Degener Neurol Neuromuscul Dis 2016; 6:49-64. [PMID: 30050368 PMCID: PMC6053097 DOI: 10.2147/dnnd.s84956] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that results in loss of the upper and lower motor neurons from motor cortex, brainstem, and spinal cord. While the majority of cases are sporadic, approximately 10% show familial inheritance. ALS is usually inherited in an autosomal dominant manner, although autosomal recessive and X-linked inheritance do occur. To date, 24 of the genes at 26 loci have been identified; these include loci linked to ALS and to frontotemporal dementia-ALS, where family pedigrees contain individuals with frontotemporal dementia with/without ALS. The most commonly established genetic causes of familial ALS (FALS) to date are the presence of a hexanucleotide repeat expansion in the C9ORF72 gene (39.3% FALS) and mutation of SOD1, TARDBP, and FUS, with frequencies of 12%-23.5%, 5%, and 4.1%, respectively. However, with the increasing use of next-generation sequencing of small family pedigrees, this has led to an increasing number of genes being associated with ALS. This review provides a comprehensive review on the genetics of ALS and an update of the pathogenic mechanisms associated with these genes. Commonly implicated pathways have been established, including RNA processing, the protein degradation pathways of autophagy and ubiquitin-proteasome system, as well as protein trafficking and cytoskeletal function. Elucidating the role genetics plays in both FALS and sporadic ALS is essential for understanding the subsequent cellular dysregulation that leads to motor neuron loss, in order to develop future effective therapeutic strategies.
Collapse
Affiliation(s)
- Afnan A Alsultan
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Rachel Waller
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| |
Collapse
|
418
|
Joshi AU, Kornfeld OS, Mochly-Rosen D. The entangled ER-mitochondrial axis as a potential therapeutic strategy in neurodegeneration: A tangled duo unchained. Cell Calcium 2016; 60:218-34. [PMID: 27212603 DOI: 10.1016/j.ceca.2016.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) and mitochondrial function have both been shown to be critical events in neurodegenerative diseases. The ER mediates protein folding, maturation, sorting as well acts as calcium storage. The unfolded protein response (UPR) is a stress response of the ER that is activated by the accumulation of misfolded proteins within the ER lumen. Although the molecular mechanisms underlying ER stress-induced apoptosis are not completely understood, increasing evidence suggests that ER and mitochondria cooperate to signal cell death. Similarly, calcium-mediated mitochondrial function and dynamics not only contribute to ATP generation and calcium buffering but are also a linchpin in mediating cell fate. Mitochondria and ER form structural and functional networks (mitochondria-associated ER membranes [MAMs]) essential to maintaining cellular homeostasis and determining cell fate under various pathophysiological conditions. Regulated Ca(2+) transfer from the ER to the mitochondria is important in maintaining control of pro-survival/pro-death pathways. In this review, we summarize the latest therapeutic strategies that target these essential organelles in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Opher S Kornfeld
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA.
| |
Collapse
|
419
|
Galmes R, Houcine A, van Vliet AR, Agostinis P, Jackson CL, Giordano F. ORP5/ORP8 localize to endoplasmic reticulum-mitochondria contacts and are involved in mitochondrial function. EMBO Rep 2016; 17:800-10. [PMID: 27113756 DOI: 10.15252/embr.201541108] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 03/09/2016] [Indexed: 12/20/2022] Open
Abstract
The oxysterol-binding protein (OSBP)-related proteins ORP5 and ORP8 have been shown recently to transport phosphatidylserine (PS) from the endoplasmic reticulum (ER) to the plasma membrane (PM) at ER-PM contact sites. PS is also transferred from the ER to mitochondria where it acts as precursor for mitochondrial PE synthesis. Here, we show that, in addition to ER-PM contact sites, ORP5 and ORP8 are also localized to ER-mitochondria contacts and interact with the outer mitochondrial membrane protein PTPIP51. A functional lipid transfer (ORD) domain was required for this localization. Interestingly, ORP5 and ORP8 depletion leads to defects in mitochondria morphology and respiratory function.
Collapse
Affiliation(s)
- Romain Galmes
- Institut Jacques Monod, CNRS, UMR7592, Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| | - Audrey Houcine
- Institut Jacques Monod, CNRS, UMR7592, Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| | - Alexander R van Vliet
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Catherine L Jackson
- Institut Jacques Monod, CNRS, UMR7592, Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| | - Francesca Giordano
- Institut Jacques Monod, CNRS, UMR7592, Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| |
Collapse
|
420
|
Krols M, van Isterdael G, Asselbergh B, Kremer A, Lippens S, Timmerman V, Janssens S. Mitochondria-associated membranes as hubs for neurodegeneration. Acta Neuropathol 2016; 131:505-23. [PMID: 26744348 PMCID: PMC4789254 DOI: 10.1007/s00401-015-1528-7] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
There is a growing appreciation that membrane-bound organelles in eukaryotic cells communicate directly with one another through direct membrane contact sites. Mitochondria-associated membranes are specialized subdomains of the endoplasmic reticulum that function as membrane contact sites between the endoplasmic reticulum and mitochondria. These sites have emerged as major players in lipid metabolism and calcium signaling. More recently also autophagy and mitochondrial dynamics have been found to be regulated at ER-mitochondria contact sites. Neurons critically depend on mitochondria-associated membranes as a means to exchange metabolites and signaling molecules between these organelles. This is underscored by the fact that genes affecting mitochondrial and endoplasmic reticulum homeostasis are clearly overrepresented in several hereditary neurodegenerative disorders. Conversely, the processes affected by the contact sites between the endoplasmic reticulum and mitochondria are widely implicated in neurodegeneration. This review will focus on the most recent data addressing the structural composition and function of the mitochondria-associated membranes. In addition, the 3D morphology of the contact sites as observed using volume electron microscopy is discussed. Finally, it will highlight the role of several key proteins associated with these contact sites that are involved not only in dementias, amyotrophic lateral sclerosis and Parkinson's disease, but also in axonopathies such as hereditary spastic paraplegia and Charcot-Marie-Tooth disease.
Collapse
|
421
|
VAP, a Versatile Access Point for the Endoplasmic Reticulum: Review and analysis of FFAT-like motifs in the VAPome. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:952-961. [PMID: 26898182 DOI: 10.1016/j.bbalip.2016.02.009] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 11/24/2022]
Abstract
Dysfunction of VAMP-associated protein (VAP) is associated with neurodegeneration, both Amyotrophic Lateral Sclerosis and Parkinson's disease. Here we summarize what is known about the intracellular interactions of VAP in humans and model organisms. VAP is a simple, small and highly conserved protein on the cytoplasmic face of the endoplasmic reticulum (ER). It is the sole protein on that large organelle that acts as a receptor for cytoplasmic proteins. This may explain the extremely wide range of interacting partners of VAP, with components of many cellular pathways binding it to access the ER. Many proteins that bind VAP also target other intracellular membranes, so VAP is a component of multiple molecular bridges at membrane contact sites between the ER and other organelles. So far approximately 100 proteins have been identified in the VAP interactome (VAPome), of which a small minority have a "two phenylalanines in an acidic tract" (FFAT) motif as it was originally defined. We have analyzed the entire VAPome in humans and yeast using a simple algorithm that identifies many more FFAT-like motifs. We show that approximately 50% of the VAPome binds directly or indirectly via the VAP-FFAT interaction. We also review evidence on pathogenesis in genetic disorders of VAP, which appear to arise from reduced overall VAP levels, leading to ER stress. It is not possible to identify one single interaction that underlies disease. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
422
|
There's Something Wrong with my MAM; the ER-Mitochondria Axis and Neurodegenerative Diseases. Trends Neurosci 2016; 39:146-157. [PMID: 26899735 PMCID: PMC4780428 DOI: 10.1016/j.tins.2016.01.008] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis with associated frontotemporal dementia (ALS/FTD) are major neurodegenerative diseases for which there are no cures. All are characterised by damage to several seemingly disparate cellular processes. The broad nature of this damage makes understanding pathogenic mechanisms and devising new treatments difficult. Can the different damaged functions be linked together in a common disease pathway and which damaged function should be targeted for therapy? Many functions damaged in neurodegenerative diseases are regulated by communications that mitochondria make with a specialised region of the endoplasmic reticulum (ER; mitochondria-associated ER membranes or 'MAM'). Moreover, several recent studies have shown that disturbances to ER-mitochondria contacts occur in neurodegenerative diseases. Here, we review these findings.
Collapse
|
423
|
Lipid transfer and metabolism across the endolysosomal-mitochondrial boundary. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:880-894. [PMID: 26852832 DOI: 10.1016/j.bbalip.2016.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/30/2016] [Accepted: 02/03/2016] [Indexed: 01/10/2023]
Abstract
Lysosomes and mitochondria occupy a central stage in the maintenance of cellular homeostasis, by playing complementary roles in nutrient sensing and energy metabolism. Specifically, these organelles function as signaling hubs that integrate environmental and endogenous stimuli with specific metabolic responses. In particular, they control various lipid biosynthetic and degradative pipelines, either directly or indirectly, by regulating major cellular metabolic pathways, and by physical and functional connections established with each other and with other organelles. Membrane contact sites allow the exchange of ions and molecules between organelles, even without membrane fusion, and are privileged routes for lipid transfer among different membrane compartments. These inter-organellar connections typically involve the endoplasmic reticulum. Direct membrane contacts have now been described also between lysosomes, autophagosomes, lipid droplets, and mitochondria. This review focuses on these recently identified membrane contact sites, and on their role in lipid biosynthesis, exchange, turnover and catabolism. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
424
|
Garofalo T, Manganelli V, Grasso M, Mattei V, Ferri A, Misasi R, Sorice M. Role of mitochondrial raft-like microdomains in the regulation of cell apoptosis. Apoptosis 2015; 20:621-34. [PMID: 25652700 DOI: 10.1007/s10495-015-1100-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipid rafts are envisaged as lateral assemblies of specific lipids and proteins that dissociate and associate rapidly and form functional clusters in cell membranes. These structural platforms are not confined to the plasma membrane; indeed lipid microdomains are similarly formed at subcellular organelles, which include endoplasmic reticulum, Golgi and mitochondria, named raft-like microdomains. In addition, some components of raft-like microdomains are present within ER-mitochondria associated membranes. This review is focused on the role of mitochondrial raft-like microdomains in the regulation of cell apoptosis, since these microdomains may represent preferential sites where key reactions take place, regulating mitochondria hyperpolarization, fission-associated changes, megapore formation and release of apoptogenic factors. These structural platforms appear to modulate cytoplasmic pathways switching cell fate towards cell survival or death. Main insights on this issue derive from some pathological conditions in which alterations of microdomains structure or function can lead to severe alterations of cell activity and life span. In the light of the role played by raft-like microdomains to integrate apoptotic signals and in regulating mitochondrial dynamics, it is conceivable that these membrane structures may play a role in the mitochondrial alterations observed in some of the most common human neurodegenerative diseases, such as Amyotrophic lateral sclerosis, Huntington's chorea and prion-related diseases. These findings introduce an additional task for identifying new molecular target(s) of pharmacological agents in these pathologies.
Collapse
Affiliation(s)
- Tina Garofalo
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
425
|
Haase G, Rabouille C. Golgi Fragmentation in ALS Motor Neurons. New Mechanisms Targeting Microtubules, Tethers, and Transport Vesicles. Front Neurosci 2015; 9:448. [PMID: 26696811 PMCID: PMC4672084 DOI: 10.3389/fnins.2015.00448] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022] Open
Abstract
Pathological alterations of the Golgi apparatus, such as its fragmentation represent an early pre-clinical feature of many neurodegenerative diseases and have been widely studied in the motor neuron disease amyotrophic lateral sclerosis (ALS). Yet, the underlying molecular mechanisms have remained cryptic. In principle, Golgi fragmentation may result from defects in three major classes of proteins: structural Golgi proteins, cytoskeletal proteins and molecular motors, as well as proteins mediating transport to and through the Golgi. Here, we present the different mechanisms that may underlie Golgi fragmentation in animal and cellular models of ALS linked to mutations in SOD1, TARDBP (TDP-43), VAPB, and C9Orf72 and we propose a novel one based on findings in progressive motor neuronopathy (pmn) mice. These mice are mutated in the TBCE gene encoding the cis-Golgi localized tubulin-binding cofactor E, one of five chaperones that assist in tubulin folding and microtubule polymerization. Loss of TBCE leads to alterations in Golgi microtubules, which in turn impedes on the maintenance of the Golgi architecture. This is due to down-regulation of COPI coat components, dispersion of Golgi tethers and strong accumulation of ER-Golgi SNAREs. These effects are partially rescued by the GTPase ARF1 through recruitment of TBCE to the Golgi. We hypothesize that defects in COPI vesicles, microtubules and their interaction may also underlie Golgi fragmentation in human ALS linked to other mutations, spinal muscular atrophy (SMA), and related motor neuron diseases. We also discuss the functional relevance of pathological Golgi alterations, in particular their potential causative, contributory, or compensatory role in the degeneration of motor neuron cell bodies, axons and synapses.
Collapse
Affiliation(s)
- Georg Haase
- Centre National de la Recherche Scientifique and Aix-Marseille Université UMR 7289, Institut de Neurosciences de la Timone Marseille, France
| | - Catherine Rabouille
- The Department of Cell Biology, Hubrecht Institute of the Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
426
|
Esmaeili MA, Yadav S, Gupta RK, Waggoner GR, Deloach A, Calingasan NY, Beal MF, Kiaei M. Preferential PPAR-α activation reduces neuroinflammation, and blocks neurodegeneration in vivo. Hum Mol Genet 2015; 25:317-27. [PMID: 26604138 DOI: 10.1093/hmg/ddv477] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation, immune reactivity and mitochondrial abnormalities are considered as causes and/or contributors to neuronal degeneration. Peroxisome proliferator-activated receptors (PPARs) regulate both inflammatory and multiple other pathways that are implicated in neurodegeneration. In the present study, we investigated the efficacy of fenofibrate (Tricor), a pan-PPAR agonist that activates PPAR-α as well as other PPARs. We administered fenofibrate to superoxide dismutase 1 (SOD1(G93A)) mice daily prior to any detectable phenotypes and then animal behavior, pathology and longevity were assessed. Treated animals showed a significant slowing of the progression of disease with weight loss attenuation, enhanced motor performance, delayed onset and survival extension. Histopathological analysis of the spinal cords showed that neuronal loss was significantly attenuated in fenofibrate-treated mice. Mitochondria were preserved as indicated by Cytochrome c immunostaining in the spinal cord, which maybe partly due to increased expression of the PPAR-γ co-activator 1-α. The total mRNA analysis revealed that neuroprotective and anti-inflammatory genes were elevated, while neuroinflammatory genes were down-regulated. This study demonstrates that the activation of PPAR-α action via fenofibrate leads to neuroprotection by both reducing neuroinflammation and protecting mitochondria, which leads to a significant increase in survival in SOD1(G93A) mice. Therefore, the development of therapeutic strategies to activate PPAR-α as well as other PPARs may lead to new therapeutic agents to slow or halt the progression of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Mohammad A Esmaeili
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience and
| | - Shilpi Yadav
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience and
| | - Ravi Kr Gupta
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, 72205 AR, USA and
| | - Garrett R Waggoner
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience and
| | - Abigail Deloach
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience and
| | - Noel Y Calingasan
- Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - M Flint Beal
- Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Mahmoud Kiaei
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience and,
| |
Collapse
|
427
|
Norkett R, Modi S, Birsa N, Atkin TA, Ivankovic D, Pathania M, Trossbach SV, Korth C, Hirst WD, Kittler JT. DISC1-dependent Regulation of Mitochondrial Dynamics Controls the Morphogenesis of Complex Neuronal Dendrites. J Biol Chem 2015; 291:613-29. [PMID: 26553875 PMCID: PMC4705382 DOI: 10.1074/jbc.m115.699447] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Indexed: 01/09/2023] Open
Abstract
The DISC1 protein is implicated in major mental illnesses including schizophrenia, depression, bipolar disorder, and autism. Aberrant mitochondrial dynamics are also associated with major mental illness. DISC1 plays a role in mitochondrial transport in neuronal axons, but its effects in dendrites have yet to be studied. Further, the mechanisms of this regulation and its role in neuronal development and brain function are poorly understood. Here we have demonstrated that DISC1 couples to the mitochondrial transport and fusion machinery via interaction with the outer mitochondrial membrane GTPase proteins Miro1 and Miro2, the TRAK1 and TRAK2 mitochondrial trafficking adaptors, and the mitochondrial fusion proteins (mitofusins). Using live cell imaging, we show that disruption of the DISC1-Miro-TRAK complex inhibits mitochondrial transport in neurons. We also show that the fusion protein generated from the originally described DISC1 translocation (DISC1-Boymaw) localizes to the mitochondria, where it similarly disrupts mitochondrial dynamics. We also show by super resolution microscopy that DISC1 is localized to endoplasmic reticulum contact sites and that the DISC1-Boymaw fusion protein decreases the endoplasmic reticulum-mitochondria contact area. Moreover, disruption of mitochondrial dynamics by targeting the DISC1-Miro-TRAK complex or upon expression of the DISC1-Boymaw fusion protein impairs the correct development of neuronal dendrites. Thus, DISC1 acts as an important regulator of mitochondrial dynamics in both axons and dendrites to mediate the transport, fusion, and cross-talk of these organelles, and pathological DISC1 isoforms disrupt this critical function leading to abnormal neuronal development.
Collapse
Affiliation(s)
- Rosalind Norkett
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Souvik Modi
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Nicol Birsa
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Talia A Atkin
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Davor Ivankovic
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Manav Pathania
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Svenja V Trossbach
- the Department of Neuropathology, Heinrich Heine University, Moorenstrasse 5, 40225 Dusseldorf, Germany
| | - Carsten Korth
- the Department of Neuropathology, Heinrich Heine University, Moorenstrasse 5, 40225 Dusseldorf, Germany
| | - Warren D Hirst
- the Neuroscience Research Unit, Pfizer, Cambridge, Massachusetts 02139, and
| | - Josef T Kittler
- From the Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom,
| |
Collapse
|
428
|
van Bergeijk P, Hoogenraad CC, Kapitein LC. Right Time, Right Place: Probing the Functions of Organelle Positioning. Trends Cell Biol 2015; 26:121-134. [PMID: 26541125 DOI: 10.1016/j.tcb.2015.10.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 10/22/2022]
Abstract
The proper spatial arrangement of organelles underlies many cellular processes including signaling, polarization, and growth. Despite the importance of local positioning, the precise connection between subcellular localization and organelle function is often not fully understood. To address this, recent studies have developed and employed different strategies to directly manipulate organelle distributions, such as the use of (light-sensitive) heterodimerization to control the interaction between selected organelles and specific motor proteins, adaptor molecules, or anchoring factors. We review here the importance of subcellular localization as well as tools to control local organelle positioning. Because these approaches allow spatiotemporal control of organelle distribution, they will be invaluable tools to unravel local functioning and the mechanisms that control positioning.
Collapse
Affiliation(s)
- Petra van Bergeijk
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
429
|
Huttlin EL, Ting L, Bruckner RJ, Gebreab F, Gygi MP, Szpyt J, Tam S, Zarraga G, Colby G, Baltier K, Dong R, Guarani V, Vaites LP, Ordureau A, Rad R, Erickson BK, Wühr M, Chick J, Zhai B, Kolippakkam D, Mintseris J, Obar RA, Harris T, Artavanis-Tsakonas S, Sowa ME, De Camilli P, Paulo JA, Harper JW, Gygi SP. The BioPlex Network: A Systematic Exploration of the Human Interactome. Cell 2015; 162:425-440. [PMID: 26186194 DOI: 10.1016/j.cell.2015.06.043] [Citation(s) in RCA: 1011] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/04/2015] [Accepted: 06/12/2015] [Indexed: 01/05/2023]
Abstract
Protein interactions form a network whose structure drives cellular function and whose organization informs biological inquiry. Using high-throughput affinity-purification mass spectrometry, we identify interacting partners for 2,594 human proteins in HEK293T cells. The resulting network (BioPlex) contains 23,744 interactions among 7,668 proteins with 86% previously undocumented. BioPlex accurately depicts known complexes, attaining 80%-100% coverage for most CORUM complexes. The network readily subdivides into communities that correspond to complexes or clusters of functionally related proteins. More generally, network architecture reflects cellular localization, biological process, and molecular function, enabling functional characterization of thousands of proteins. Network structure also reveals associations among thousands of protein domains, suggesting a basis for examining structurally related proteins. Finally, BioPlex, in combination with other approaches, can be used to reveal interactions of biological or clinical significance. For example, mutations in the membrane protein VAPB implicated in familial amyotrophic lateral sclerosis perturb a defined community of interactors.
Collapse
Affiliation(s)
- Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lily Ting
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Raphael J Bruckner
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Fana Gebreab
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Melanie P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Stanley Tam
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela Zarraga
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Greg Colby
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kurt Baltier
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Dong
- Department of Cell Biology and Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT 06519, USA
| | - Virginia Guarani
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ramin Rad
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian K Erickson
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Wühr
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joel Chick
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bo Zhai
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Deepak Kolippakkam
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Julian Mintseris
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert A Obar
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Biogen, Cambridge, MA 02142, USA
| | | | - Spyros Artavanis-Tsakonas
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Biogen, Cambridge, MA 02142, USA
| | - Mathew E Sowa
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Pietro De Camilli
- Department of Cell Biology and Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT 06519, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
430
|
Schrader M, Godinho LF, Costello JL, Islinger M. The different facets of organelle interplay-an overview of organelle interactions. Front Cell Dev Biol 2015; 3:56. [PMID: 26442263 PMCID: PMC4585249 DOI: 10.3389/fcell.2015.00056] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/08/2015] [Indexed: 12/28/2022] Open
Abstract
Membrane-bound organelles such as mitochondria, peroxisomes, or the endoplasmic reticulum (ER) create distinct environments to promote specific cellular tasks such as ATP production, lipid breakdown, or protein export. During recent years, it has become evident that organelles are integrated into cellular networks regulating metabolism, intracellular signaling, cellular maintenance, cell fate decision, and pathogen defence. In order to facilitate such signaling events, specialized membrane regions between apposing organelles bear distinct sets of proteins to enable tethering and exchange of metabolites and signaling molecules. Such membrane associations between the mitochondria and a specialized site of the ER, the mitochondria associated-membrane (MAM), as well as between the ER and the plasma membrane (PAM) have been partially characterized at the molecular level. However, historical and recent observations imply that other organelles like peroxisomes, lysosomes, and lipid droplets might also be involved in the formation of such apposing membrane contact sites. Alternatively, reports on so-called mitochondria derived-vesicles (MDV) suggest alternative mechanisms of organelle interaction. Moreover, maintenance of cellular homeostasis requires the precise removal of aged organelles by autophagy—a process which involves the detection of ubiquitinated organelle proteins by the autophagosome membrane, representing another site of membrane associated-signaling. This review will summarize the available data on the existence and composition of organelle contact sites and the molecular specializations each site uses in order to provide a timely overview on the potential functions of organelle interaction.
Collapse
Affiliation(s)
- Michael Schrader
- Department of Biosciences, College of Life and Environmental Sciences, University of Exeter Exeter, UK
| | - Luis F Godinho
- Centre for Cell Biology and Department of Biology, University of Aveiro Aveiro, Portugal
| | - Joseph L Costello
- Department of Biosciences, College of Life and Environmental Sciences, University of Exeter Exeter, UK
| | - Markus Islinger
- Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, University of Heidelberg Mannheim, Germany
| |
Collapse
|
431
|
Lim Y, Cho IT, Schoel LJ, Cho G, Golden JA. Hereditary spastic paraplegia-linked REEP1 modulates endoplasmic reticulum/mitochondria contacts. Ann Neurol 2015. [PMID: 26201691 DOI: 10.1002/ana.24488] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Mutations in receptor expression enhancing protein 1 (REEP1) are associated with hereditary spastic paraplegias (HSPs). Although axonal degeneration is thought to be a predominant feature in HSP, the role of REEP1 mutations in degeneration is largely unknown. Previous studies have implicated a role for REEP1 in the endoplasmic reticulum (ER), whereas others localized REEP1 with mitochondria. We sought to resolve the cellular localization of REEP1 and further elucidate the pathobiology underlying REEP1 mutations in patients. METHODS A combination of cellular imaging and biochemical approaches was used to refine the cellular localization of REEP1. Next, Reep1 mutations associated with HSP were functionally tested in neuritic growth and degeneration assays using mouse cortical culture. Finally, a novel assay was developed and used with wild-type and mutant Reep1s to measure the interactions between the ER and mitochondria. RESULTS We found that REEP1 is present at the ER-mitochondria interface, and it contains subdomains for mitochondrial as well as ER localization. Knockdown of Reep1 and expression of pathological Reep1 mutations resulted in neuritic growth defects and degeneration. Finally, using our novel split-RLuc8 assay, we show that REEP1 facilitates ER-mitochondria interactions, a function diminished by disease-associated mutations. INTERPRETATION Our data potentially reconcile the current conflicting reports regarding REEP1 being either an ER or a mitochondrial protein. Furthermore, our results connect, for the first time, the disrupted ER-mitochondria interactions to a failure in maintaining health of long axons in HSPs. Finally, the split-RLuc8 assay offers a new tool to identify potential drugs for multiple neurodegenerative diseases with ER-mitochondria interaction defects.
Collapse
Affiliation(s)
- Youngshin Lim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Il-Taeg Cho
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leah J Schoel
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ginam Cho
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
432
|
Manfredi G, Kawamata H. Mitochondria and endoplasmic reticulum crosstalk in amyotrophic lateral sclerosis. Neurobiol Dis 2015; 90:35-42. [PMID: 26282323 DOI: 10.1016/j.nbd.2015.08.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/31/2015] [Accepted: 08/12/2015] [Indexed: 12/13/2022] Open
Abstract
Physical and functional interactions between mitochondria and the endoplasmic reticulum (ER) are crucial for cell life. These two organelles are intimately connected and collaborate to essential processes, such as calcium homeostasis and phospholipid biosynthesis. The connections between mitochondria and endoplasmic reticulum occur through structures named mitochondria associated membranes (MAMs), which contain lipid rafts and a large number of proteins, many of which serve multiple functions at different cellular sites. Growing evidence strongly suggests that alterations of ER-mitochondria interactions are involved in neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), a devastating and rapidly fatal motor neuron disease. Mutations in proteins that participate in ER-mitochondria interactions and MAM functions are increasingly being associated with genetic forms of ALS and other neurodegenerative diseases. This evidence strongly suggests that, rather than considering the two organelles separately, a better understanding of the disease process can derive from studying the alterations in their crosstalk. In this review we discuss normal and pathological ER-mitochondria interactions and the evidence that link them to ALS.
Collapse
Affiliation(s)
- Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 401 East 61st Street, New York, NY 10065, United States.
| | - Hibiki Kawamata
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 401 East 61st Street, New York, NY 10065, United States.
| |
Collapse
|
433
|
Orieux G, Slembrouck A, Bensaïd M, Sahel JA, Goureau O. The protein tyrosine phosphatase interacting protein 51 (PTPIP51) is required for the differentiation of photoreceptors. Neuroscience 2015; 300:276-85. [DOI: 10.1016/j.neuroscience.2015.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/24/2015] [Accepted: 05/12/2015] [Indexed: 02/05/2023]
|
434
|
Sreedharan J, Neukomm LJ, Brown RH, Freeman MR. Age-Dependent TDP-43-Mediated Motor Neuron Degeneration Requires GSK3, hat-trick, and xmas-2. Curr Biol 2015; 25:2130-6. [PMID: 26234214 DOI: 10.1016/j.cub.2015.06.045] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/24/2015] [Accepted: 06/18/2015] [Indexed: 12/12/2022]
Abstract
The RNA-processing protein TDP-43 is central to the pathogenesis of amyotrophic lateral sclerosis (ALS), the most common adult-onset motor neuron (MN) disease. TDP-43 is conserved in Drosophila, where it has been the topic of considerable study, but how TDP-43 mutations lead to age-dependent neurodegeneration is unclear and most approaches have not directly examined changes in MN morphology with age. We used a mosaic approach to study age-dependent MN loss in the adult fly leg where it is possible to resolve single motor axons, NMJs and active zones, and perform rapid forward genetic screens. We show that expression of TDP-43(Q331K) caused dying-back of NMJs and axons, which could not be suppressed by mutations that block Wallerian degeneration. We report the identification of three genes that suppress TDP-43 toxicity, including shaggy/GSK3, a known modifier of neurodegeneration. The two additional novel suppressors, hat-trick and xmas-2, function in chromatin modeling and RNA export, two processes recently implicated in human ALS. Loss of shaggy/GSK3, hat-trick, or xmas-2 does not suppress Wallerian degeneration, arguing TDP-43(Q331K)-induced and Wallerian degeneration are genetically distinct processes. In addition to delineating genetic factors that modify TDP-43 toxicity, these results establish the Drosophila adult leg as a valuable new tool for the in vivo study of adult MN phenotypes.
Collapse
Affiliation(s)
- Jemeen Sreedharan
- Howard Hughes Medical Institute and Department of Neurobiology, LRB-740A1, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01655, USA; Department of Neurology, S5-755, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA; Signalling ISP, The Babraham Institute, Cambridge CB22 3AT, UK.
| | - Lukas J Neukomm
- Howard Hughes Medical Institute and Department of Neurobiology, LRB-740A1, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01655, USA
| | - Robert H Brown
- Department of Neurology, S5-755, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - Marc R Freeman
- Howard Hughes Medical Institute and Department of Neurobiology, LRB-740A1, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01655, USA.
| |
Collapse
|
435
|
Abstract
The transactive response DNA binding protein (TDP-43) has long been characterized as a main hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U, also known as FTLD-TDP). Several studies have indicated TDP-43 deposits in Alzheimer's disease (AD) brains and have robust connection with AD clinical phenotype. FTLD-U, which was symptomatically connected with AD, may be predictable for the comprehension of the role TDP-43 in AD. TDP-43 may contribute to AD through both β-amyloid (Aβ)-dependent and Aβ-independent pathways. In this article, we summarize the latest studies concerning the role of TDP-43 in AD and explore TDP-43 modulation as a potential therapeutic strategy for AD. However, to date, little of pieces of the research on TDP-43 have been performed to investigate the role in AD; more investigations need to be confirmed in the future.
Collapse
|
436
|
Leal SS, Gomes CM. Calcium dysregulation links ALS defective proteins and motor neuron selective vulnerability. Front Cell Neurosci 2015; 9:225. [PMID: 26136661 PMCID: PMC4468822 DOI: 10.3389/fncel.2015.00225] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/28/2015] [Indexed: 12/12/2022] Open
Abstract
More than 20 distinct gene loci have so far been implicated in amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder characterized by progressive neurodegeneration of motor neurons (MN) and death. Most of this distinct set of ALS-related proteins undergoes toxic deposition specifically in MN for reasons which remain unclear. Here we overview a recent body of evidence indicative that mutations in ALS-related proteins can disrupt fundamental Ca2+ signalling pathways in MN, and that Ca2+ itself impacts both directly or indirectly in many ALS critical proteins and cellular processes that result in MN neurodegeneration. We argue that the inherent vulnerability of MN to dysregulation of intracellular Ca2+ is deeply associated with discriminating pathogenicity and aberrant crosstalk of most of the critical proteins involved in ALS. Overall, Ca2+ deregulation in MN is at the cornerstone of different ALS processes and is likely one of the factors contributing to the selective susceptibility of these cells to this particular neurodegenerative disease.
Collapse
Affiliation(s)
- Sónia S Leal
- Faculdade de Ciências, Biosystems and Integrative Sciences Institute and Department of Chemistry and Biochemistry, Universidade de Lisboa Campo Grande, Lisboa, Portugal ; Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa Oeiras, Portugal
| | - Cláudio M Gomes
- Faculdade de Ciências, Biosystems and Integrative Sciences Institute and Department of Chemistry and Biochemistry, Universidade de Lisboa Campo Grande, Lisboa, Portugal ; Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa Oeiras, Portugal
| |
Collapse
|
437
|
De Rosa P, Marini ES, Gelmetti V, Valente EM. Candidate genes for Parkinson disease: Lessons from pathogenesis. Clin Chim Acta 2015; 449:68-76. [PMID: 26048192 DOI: 10.1016/j.cca.2015.04.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 01/06/2023]
Abstract
Parkinson disease (PD) is a multifactorial neurodegenerative disease characterized by the progressive loss of specific neuronal populations and accumulation of Lewy bodies in the brain, leading to motor and non-motor symptoms. In a small subset of patients, PD is dominantly or recessively inherited, while a number of susceptibility genetic loci have been identified through genome wide association studies. The discovery of genes mutated in PD and functional studies on their protein products have provided new insights into the molecular events leading to neurodegeneration, suggesting that few interconnected molecular pathways may be deranged in all forms of PD, triggering neuronal loss. Here, we summarize the most relevant findings implicating the main PD-related proteins in biological processes such as mitochondrial dysfunction, misfolded protein damage, alteration of cellular clearance systems, abnormal calcium handling and altered inflammatory response, which represent key targets for neuroprotection.
Collapse
Affiliation(s)
- Priscilla De Rosa
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy
| | - Elettra Sara Marini
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy; Dept. of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Vania Gelmetti
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy
| | - Enza Maria Valente
- IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute, San Giovanni Rotondo, Italy; Section of Neurosciences, Dept. of Medicine and Surgery, University of Salerno, Salerno, Italy.
| |
Collapse
|
438
|
Guardia-Laguarta C, Area-Gomez E, Schon EA, Przedborski S. A new role for α-synuclein in Parkinson's disease: Alteration of ER-mitochondrial communication. Mov Disord 2015; 30:1026-33. [DOI: 10.1002/mds.26239] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 03/10/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022] Open
Affiliation(s)
| | - Estela Area-Gomez
- Department of Neurology; Columbia University Medical Center; New York NY USA
| | - Eric A. Schon
- Department of Neurology; Columbia University Medical Center; New York NY USA
- Department of Genetics and Development; Columbia University Medical Center; New York NY USA
| | - Serge Przedborski
- Department of Pathology and Cell Biology; Columbia University Medical Center; New York NY USA
| |
Collapse
|
439
|
Ramage HR, Kumar GR, Verschueren E, Johnson JR, Von Dollen J, Johnson T, Newton B, Shah P, Horner J, Krogan NJ, Ott M. A combined proteomics/genomics approach links hepatitis C virus infection with nonsense-mediated mRNA decay. Mol Cell 2015; 57:329-340. [PMID: 25616068 DOI: 10.1016/j.molcel.2014.12.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/28/2014] [Accepted: 12/16/2014] [Indexed: 12/22/2022]
Abstract
Hepatitis C virus (HCV) is a leading cause of liver disease, but insight into virus-host interactions remains limited. We systematically used affinity purification/mass spectrometry to define the host interactions of all ten HCV proteins in hepatoma cells. We combined these studies with RNAi knockdown of corresponding genes using a two-step scoring approach to generate a map of 139 high-confidence HCV-host protein-protein interactions. We found mitochondrial proteins highly involved in HCV infection and characterized an interaction between the viral core protein and host protein within bgcn homolog (WIBG). Expression of core prevents WIBG from binding its regular interaction partners Y14 and Magoh, two known mediators of the nonsense-mediated mRNA decay pathway. We discovered that this surveillance pathway is disrupted in HCV-infected cells, causing potentially harmful transcripts to accumulate. Our study provides a comprehensive view of HCV-host interactions and uncovers mechanisms for how HCV perturbs host functions during infection.
Collapse
Affiliation(s)
- Holly R Ramage
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
| | - G Renuka Kumar
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
| | - Erik Verschueren
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Jeffrey R Johnson
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
| | - John Von Dollen
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Tasha Johnson
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Billy Newton
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Priya Shah
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Julie Horner
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA 95134, USA
| | - Nevan J Krogan
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
- QB3, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA
| | - Melanie Ott
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
- Liver Center, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
440
|
Gaspar R, Santana I, Mendes C, Fernandes AS, Duro D, Simões M, Luís D, Santos MJ, Grazina M. Genetic Variation of MT-ND Genes in Frontotemporal Lobar Degeneration: Biochemical Phenotype-Genotype Correlation. NEURODEGENER DIS 2015; 15:70-80. [DOI: 10.1159/000380766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/05/2015] [Indexed: 11/19/2022] Open
|
441
|
Sanhueza M, Chai A, Smith C, McCray BA, Simpson TI, Taylor JP, Pennetta G. Network analyses reveal novel aspects of ALS pathogenesis. PLoS Genet 2015; 11:e1005107. [PMID: 25826266 PMCID: PMC4380362 DOI: 10.1371/journal.pgen.1005107] [Citation(s) in RCA: 41] [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: 07/03/2014] [Accepted: 02/27/2015] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease characterized by selective loss of motor neurons, muscle atrophy and paralysis. Mutations in the human VAMP-associated protein B (hVAPB) cause a heterogeneous group of motor neuron diseases including ALS8. Despite extensive research, the molecular mechanisms underlying ALS pathogenesis remain largely unknown. Genetic screens for key interactors of hVAPB activity in the intact nervous system, however, represent a fundamental approach towards understanding the in vivo function of hVAPB and its role in ALS pathogenesis. Targeted expression of the disease-causing allele leads to neurodegeneration and progressive decline in motor performance when expressed in the adult Drosophila, eye or in its entire nervous system, respectively. By using these two phenotypic readouts, we carried out a systematic survey of the Drosophila genome to identify modifiers of hVAPB-induced neurotoxicity. Modifiers cluster in a diverse array of biological functions including processes and genes that have been previously linked to hVAPB function, such as proteolysis and vesicular trafficking. In addition to established mechanisms, the screen identified endocytic trafficking and genes controlling proliferation and apoptosis as potent modifiers of ALS8-mediated defects. Surprisingly, the list of modifiers was mostly enriched for proteins linked to lipid droplet biogenesis and dynamics. Computational analysis reveals that most modifiers can be linked into a complex network of interacting genes, and that the human genes homologous to the Drosophila modifiers can be assembled into an interacting network largely overlapping with that in flies. Identity markers of the endocytic process were also found to abnormally accumulate in ALS patients, further supporting the relevance of the fly data for human biology. Collectively, these results not only lead to a better understanding of hVAPB function but also point to potentially relevant targets for therapeutic intervention. Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease causing loss of motor neurons and consequently a progressive deterioration of motor functions. ALS is uniformly fatal with death occurring 5 years after onset of symptoms. There is currently no effective treatment for ALS. Several mutations in a gene called hVAPB have shown that this gene is causative of a type of ALS known as ALS8. In this study we sought to identify genes and cellular processes that are involved in the toxicity conferred by the defective ALS8 allele. By using the power of Drosophila genetics, we performed a large scale genomic screen and identified a number of genes that can affect hVAPB-mediated toxicity. These modifiers cluster into functional pathways known to be involved in ALS as well as novel ones. The relevance of these modifiers and mechanisms for the human disease was confirmed by showing that the human homologues of the fly modifiers can be organized into a network that closely resembles that of the Drosophila genes. Identifying cellular processes and proteins that modulate hVAPB pathological activity can facilitate the discovery of an effective treatment for ALS.
Collapse
Affiliation(s)
- Mario Sanhueza
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrea Chai
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Houston, Texas, United States of America
| | - Colin Smith
- Academic Department of Neuropathology, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Brett A. McCray
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - T. Ian Simpson
- Biomathematics and Statistics Scotland, University of Edinburgh, United Kingdom
- Institute for Adaptive and Neural Computation, School of Informatics, University of Edinburgh, United Kingdom
| | - J. Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Giuseppa Pennetta
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
442
|
Compromised MAPK signaling in human diseases: an update. Arch Toxicol 2015; 89:867-82. [PMID: 25690731 DOI: 10.1007/s00204-015-1472-2] [Citation(s) in RCA: 735] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/09/2015] [Indexed: 02/08/2023]
Abstract
The mitogen-activated protein kinases (MAPKs) in mammals include c-Jun NH2-terminal kinase (JNK), p38 MAPK, and extracellular signal-regulated kinase (ERK). These enzymes are serine-threonine protein kinases that regulate various cellular activities including proliferation, differentiation, apoptosis or survival, inflammation, and innate immunity. The compromised MAPK signaling pathways contribute to the pathology of diverse human diseases including cancer and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. The JNK and p38 MAPK signaling pathways are activated by various types of cellular stress such as oxidative, genotoxic, and osmotic stress as well as by proinflammatory cytokines such as tumor necrosis factor-α and interleukin 1β. The Ras-Raf-MEK-ERK signaling pathway plays a key role in cancer development through the stimulation of cell proliferation and metastasis. The p38 MAPK pathway contributes to neuroinflammation mediated by glial cells including microglia and astrocytes, and it has also been associated with anticancer drug resistance in colon and liver cancer. We here summarize recent research on the roles of MAPK signaling pathways in human diseases, with a focus on cancer and neurodegenerative conditions.
Collapse
|
443
|
Carrì MT, Valle C, Bozzo F, Cozzolino M. Oxidative stress and mitochondrial damage: importance in non-SOD1 ALS. Front Cell Neurosci 2015; 9:41. [PMID: 25741238 PMCID: PMC4330888 DOI: 10.3389/fncel.2015.00041] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/27/2015] [Indexed: 12/12/2022] Open
Abstract
It is well known that mitochondrial damage (MD) is both the major contributor to oxidative stress (OS) (the condition arising from unbalance between production and removal of reactive oxygen species) and one of the major consequences of OS, because of the high dependance of mitochondrial function on redox-sensitive targets such as intact membranes. Conditions in which neuronal cells are not able to cope with MD and OS seem to lead or contribute to several neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS), at least in the most studied superoxide dismutase 1 (SOD1)-linked genetic variant. As summarized in this review, new evidence indicates that MD and OS play a role also in non-SOD1 ALS and thus they may represent a target for therapy despite previous failures in clinical trials.
Collapse
Affiliation(s)
- Maria Teresa Carrì
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia, IRCCS Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia, IRCCS Rome, Italy ; Institute of Cell Biology and Neurobiology, IBCN, National Research Council, CNR Rome, Italy
| | - Francesca Bozzo
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia, IRCCS Rome, Italy
| | - Mauro Cozzolino
- Institute of Translational Pharmacology, National Research Council, CNR, Molecular Mechanisms of Neurodegenerative Diseases Rome, Italy
| |
Collapse
|
444
|
Cozzolino M, Rossi S, Mirra A, Carrì MT. Mitochondrial dynamism and the pathogenesis of Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2015; 9:31. [PMID: 25713513 PMCID: PMC4322717 DOI: 10.3389/fncel.2015.00031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
Research on mitochondria in the last years has been characterized by the fundamental finding that the morphology of mitochondria is deeply connected to the regulation of a vast number of different processes, including oxidative phosphorylation and ATP production, calcium buffering, and apoptosis. This has immediately focused the attention of the neuroscience community to the possible involvement of mitochondrial dynamism, the process underlying morphological features of mitochondria, in neurodegeneration, where mitochondrial dysfunction is believed to represent an important contributing event, or even a primary causative factor. Amyotrophic Lateral Sclerosis (ALS), a disease of motor neurons and their neighboring cells, has long been considered as a neurodegenerative disease with an important mitochondrial issue. Yet, whether mitochondria have a causative, primary role in the pathogenic process has always been debated, and the specific defects which account for this role are elusive. Here we discuss recent genetic advances suggesting that defective mitochondrial dynamism is primarily involved in the pathogenic mechanisms of ALS, and that foster the longstanding concept that disruption of mitochondrial function is a vulnerable factor for motor neurons.
Collapse
Affiliation(s)
| | - Simona Rossi
- Institute of Translational Pharmacology, CNR Rome, Italy ; Department of Biology, Università di Roma Tor Vergata Rome, Italy
| | - Alessia Mirra
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia IRCCS Rome, Italy
| | - Maria Teresa Carrì
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia IRCCS Rome, Italy
| |
Collapse
|
445
|
Burgoyne T, Patel S, Eden ER. Calcium signaling at ER membrane contact sites. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2012-7. [PMID: 25662816 DOI: 10.1016/j.bbamcr.2015.01.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 11/27/2022]
Abstract
Communication between organelles is a necessary consequence of intracellular compartmentalization. Membrane contact sites (MCSs) are regions where the membranes of two organelles come into close apposition allowing exchange of small molecules and ions including Ca²⁺. The ER, the cell's major Ca²⁺ store, forms an extensive and dynamic network of contacts with multiple organelles. Here we review established and emerging roles of ER contacts as platforms for Ca²⁺ exchange and further consider a potential role for Ca²⁺ in the regulation of MCS formation. We additionally discuss the challenges associated with the study of MCS biology and highlight advances in microscopy-based solutions. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
Collapse
Affiliation(s)
| | - Sandip Patel
- Department of Cell and Developmental Biology, UCL, London, UK
| | | |
Collapse
|
446
|
Fukunaga K, Shinoda Y, Tagashira H. The role of SIGMAR1 gene mutation and mitochondrial dysfunction in amyotrophic lateral sclerosis. J Pharmacol Sci 2015; 127:36-41. [PMID: 25704016 DOI: 10.1016/j.jphs.2014.12.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 12/13/2014] [Accepted: 12/15/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) patients exhibit diverse pathologies such as endoplasmic reticulum (ER) stress and mitochondrial dysfunction in motor neurons. Five to ten percent of patients have familial ALS, a form of the disease caused by mutations in ALS-related genes, while sporadic forms of the disease occur in 90-95% of patients. Recently, it was reported that familial ALS patients exhibit a missense mutation in SIGMAR1 (c.304G > C), which encodes sigma-1 receptor (Sig-1R), substituting glutamine for glutamic acid at amino acid residue 102 (p.E102Q). Expression of that mutant Sig-1R(E102Q) protein reduces mitochondrial ATP production, inhibits proteasome activity and causes mitochondrial injury, aggravating ER stress-induced neuronal death in neuro2A cells. In this issue, we discuss mechanisms underlying mitochondrial impairment seen in ALS motor neurons and propose that therapies that protect mitochondria might improve the quality of life (QOL) of ALS patients and should be considered for clinical trials.
Collapse
Affiliation(s)
- Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hideaki Tagashira
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
447
|
Hepler PK, Winship LJ. The pollen tube clear zone: clues to the mechanism of polarized growth. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2015; 57:79-92. [PMID: 25431342 DOI: 10.1111/jipb.12315] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/24/2014] [Indexed: 05/08/2023]
Abstract
Pollen tubes usually exhibit a prominent region at their apex called the "clear zone" because it lacks light refracting amyloplasts. A robust, long clear zone often associates with fast growing pollen tubes, and thus serves as an indicator of pollen tube health. Nevertheless we do not understand how it arises or how it is maintained. Here we review the structure of the clear zone, and attempt to explain the factors that contribute to its formation. While amyloplasts and vacuolar elements are excluded from the clear zone, virtually all other organelles are present including secretory vesicles, mitochondria, Golgi dictyosomes, and the endoplasmic reticulum (ER). Secretory vesicles aggregate into an inverted cone appressed against the apical plasma membrane. ER elements move nearly to the extreme apex, whereas mitochondria and Golgi dictyosomes move less far forward. The cortical actin fringe assumes a central position in the control of clear zone formation and maintenance, given its role in generating cytoplasmic streaming. Other likely factors include the tip-focused calcium gradient, the apical pH gradient, the influx of water, and a host of signaling factors (small G-proteins). We think that the clear zone is an emergent property that depends on the interaction of several factors crucial for polarized growth.
Collapse
Affiliation(s)
- Peter K Hepler
- Biology Department, University of Massachusetts, Amherst, Massachusetts, 01003, USA
| | | |
Collapse
|
448
|
A fungal sarcolemmal membrane-associated protein (SLMAP) homolog plays a fundamental role in development and localizes to the nuclear envelope, endoplasmic reticulum, and mitochondria. EUKARYOTIC CELL 2014; 14:345-58. [PMID: 25527523 DOI: 10.1128/ec.00241-14] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/13/2014] [Indexed: 11/20/2022]
Abstract
Sarcolemmal membrane-associated protein (SLMAP) is a tail-anchored protein involved in fundamental cellular processes, such as myoblast fusion, cell cycle progression, and chromosomal inheritance. Further, SLMAP misexpression is associated with endothelial dysfunctions in diabetes and cancer. SLMAP is part of the conserved striatin-interacting phosphatase and kinase (STRIPAK) complex required for specific signaling pathways in yeasts, filamentous fungi, insects, and mammals. In filamentous fungi, STRIPAK was initially discovered in Sordaria macrospora, a model system for fungal differentiation. Here, we functionally characterize the STRIPAK subunit PRO45, a homolog of human SLMAP. We show that PRO45 is required for sexual propagation and cell-to-cell fusion and that its forkhead-associated (FHA) domain is essential for these processes. Protein-protein interaction studies revealed that PRO45 binds to STRIPAK subunits PRO11 and SmMOB3, which are also required for sexual propagation. Superresolution structured-illumination microscopy (SIM) further established that PRO45 localizes to the nuclear envelope, endoplasmic reticulum, and mitochondria. SIM also showed that localization to the nuclear envelope requires STRIPAK subunits PRO11 and PRO22, whereas for mitochondria it does not. Taken together, our study provides important insights into fundamental roles of the fungal SLMAP homolog PRO45 and suggests STRIPAK-related and STRIPAK-unrelated functions.
Collapse
|
449
|
Joardar A, Menzl J, Podolsky TC, Manzo E, Estes PS, Ashford S, Zarnescu DC. PPAR gamma activation is neuroprotective in a Drosophila model of ALS based on TDP-43. Hum Mol Genet 2014; 24:1741-54. [PMID: 25432537 DOI: 10.1093/hmg/ddu587] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a progressive neuromuscular disease for which there is no cure. We have previously developed a Drosophila model of ALS based on TDP-43 that recapitulates several aspects of disease pathophysiology. Using this model, we designed a drug screening strategy based on the pupal lethality phenotype induced by TDP-43 when expressed in motor neurons. In screening 1200 FDA-approved compounds, we identified the PPARγ agonist pioglitazone as neuroprotective in Drosophila. Here, we show that pioglitazone can rescue TDP-43-dependent locomotor dysfunction in motor neurons and glia but not in muscles. Testing additional models of ALS, we find that pioglitazone is also neuroprotective when FUS, but not SOD1, is expressed in motor neurons. Interestingly, survival analyses of TDP or FUS models show no increase in lifespan, which is consistent with recent clinical trials. Using a pharmacogenetic approach, we show that the predicted Drosophila PPARγ homologs, E75 and E78, are in vivo targets of pioglitazone. Finally, using a global metabolomic approach, we identify a set of metabolites that pioglitazone can restore in the context of TDP-43 expression in motor neurons. Taken together, our data provide evidence that modulating PPARγ activity, although not effective at improving lifespan, provides a molecular target for mitigating locomotor dysfunction in TDP-43 and FUS but not SOD1 models of ALS in Drosophila. Furthermore, our data also identify several 'biomarkers' of the disease that may be useful in developing therapeutics and in future clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniela C Zarnescu
- Department of Molecular and Cellular Biology, Department of Neuroscience Department of Neurology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
450
|
Palomo GM, Manfredi G. Exploring new pathways of neurodegeneration in ALS: the role of mitochondria quality control. Brain Res 2014; 1607:36-46. [PMID: 25301687 DOI: 10.1016/j.brainres.2014.09.065] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/26/2014] [Accepted: 09/27/2014] [Indexed: 12/11/2022]
Abstract
Neuronal cells are highly dependent on mitochondria, and mitochondrial dysfunction is associated with neurodegenerative diseases. As perturbed mitochondrial function renders neurons extremely sensitive to a wide variety of insults, such as oxidative stress and bioenergetic defects, mitochondrial defects can profoundly affect neuronal fate. Several studies have linked ALS with mitochondrial dysfunction, stemming from observations of mitochondrial abnormalities, both in patients and in cellular and mouse models of familial forms of ALS. Mitochondrial changes have been thoroughly investigated in mutants of superoxide dismutase 1 (SOD1), one of the most common causes of familial ALS, for which excellent cellular and animal models are available, but recently evidence is emerging also in other forms of ALS, both familial and sporadic. Mitochondrial defects in ALS involve many critical physiopathological processes, from defective bioenergetics to abnormal calcium homeostasis, altered morphology and impaired trafficking. In this review, we summarize established evidence of mitochondrial dysfunction in ALS, especially in SOD1 mutant models of familial ALS. The main focus of the review is on defective mitochondrial quality control (MQC) in ALS. MQC operates at multiple levels to clear damaged proteins through proteostasis and to eliminate irreparably damaged organelles through mitophagy. However, since ALS motor neurons progressively accumulate damaged mitochondria, it is plausible that the MQC is ineffective or overwhelmed by excessive workload imposed by the chronic and extensive mitochondrial damage. This article is part of a Special Issue entitled ALS complex pathogenesis.
Collapse
Affiliation(s)
- Gloria M Palomo
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, United States
| | - Giovanni Manfredi
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, United States.
| |
Collapse
|