1
|
Sevcikova Tomaskova Z, Mackova K. From function to structure: how myofibrillogenesis influences the transverse-axial tubular system development and its peculiarities. Front Physiol 2025; 16:1576133. [PMID: 40352140 PMCID: PMC12062141 DOI: 10.3389/fphys.2025.1576133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/21/2025] [Indexed: 05/14/2025] Open
Abstract
The transverse-axial tubular system (TATS) is the extension of sarcolemma growing to the cell interior, providing sufficient calcium signaling to induce calcium release from sarcoplasmic reticulum cisternae and stimulate the contraction of neighboring myofibrils. Interestingly, the development of TATS is delayed and matures during the post-partum period. It starts with small invaginations near the sarcolemma, proceeding to grow an irregular network that is later assembled into the notably transversally oriented tubular network. Accumulating evidence supports the idea that the development of TATS is linked to cell dimensions, calcium signaling, and increasing myofibrillar content orchestrated by electromechanical stimulation. However, the overall mechanism has not yet been described. The topic of this review is the development of TATS with an emphasis on the irregular phase of tubule growth. The traditional models of BIN1-related tubulation are also discussed. We summarized the recently described protein interactions during TATS development, mainly mediated by costameric and sarcomeric proteins, supporting the idea of the coupling sites between TATS and the myofibrils. We hypothesize that the formation and final organization of the tubular system is driven by the simultaneous development of the contractile apparatus under cycling electromechanical stimulus.
Collapse
Affiliation(s)
| | - Katarina Mackova
- Department of Biophysics and Electrophysiology, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
2
|
Ashwini P, Subhash B, Amol M, Kumar D, Atmaram P, Ravindra K. Comprehensive investigation of multiple targets in the development of newer drugs for the Alzheimer's disease. Acta Pharm Sin B 2025; 15:1281-1310. [PMID: 40370532 PMCID: PMC12069117 DOI: 10.1016/j.apsb.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/20/2024] [Accepted: 09/06/2024] [Indexed: 05/16/2025] Open
Abstract
Alzheimer's disease, a significant contributor to dementia, is rapidly becoming a serious healthcare concern in the 21st century. The alarming number of patients with Alzheimer's disease is steadily increasing, which is contributed by the dearth of treatment options. The current treatment for Alzheimer's disease is heavily dependent on symptomatic treatment that has failed to cure the disease despite huge investments in the development of drugs. The clinical treatment of Alzheimer's disease with limited drugs is generally targeted towards the inhibition of N-methyl-d-aspartate receptor and acetylcholine esterase, which only elevate cognition levels for a limited period. Beyond the aforementioned molecular targets, β-amyloid was much explored with little success and thus created a feel and palpable growing emphasis on discovering new putative and novel targets for AD. This has inspired medicinal chemists to explore new targets, including microglia, triggering receptors expressed on myeloid cells 2 (Trem-2), and notum carboxylesterase, to discover new lead compounds. This review explores the functions, pathophysiological roles, and importance of all AD-related targets that address therapeutic and preventive approaches for the treatment and protection of Alzheimer's disease.
Collapse
Affiliation(s)
- Patil Ashwini
- Department of Pharmaceutical Chemistry, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
| | - Bodhankar Subhash
- Department of Pharmacology, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
| | - Muthal Amol
- Department of Pharmacology, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
- University of California, Davis, CA 95616, USA
| | - Pawar Atmaram
- Department of Pharmaceutics, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
| | - Kulkarni Ravindra
- Department of Pharmaceutical Chemistry, BVDU’S Poona College of Pharmacy, Erandwane Pune-411038, Maharashtra, India
| |
Collapse
|
3
|
Loh YH, Lv J, Goh Y, Sun X, Zhu X, Muheyati M, Luan Y. Remodelling of T-Tubules and Associated Calcium Handling Dysfunction in Heart Failure: Mechanisms and Therapeutic Insights. Can J Cardiol 2024; 40:2569-2588. [PMID: 39455023 DOI: 10.1016/j.cjca.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In cardiomyocytes, transverse tubules (T-tubules) are sarcolemmal invaginations that facilitate excitation-contraction coupling and diastolic function. The clinical significance of T-tubules has become evident in that their remodelling is recognised as a hallmark feature of heart failure (HF) and a key contributor to disrupted Ca2+ homeostasis, compromised cardiac function, and arrhythmogenesis. Further investigations have revealed that T-tubule remodelling is particularly pronounced in HF with reduced ejection fraction (HFrEF), but not in HF with preserved ejection fraction, implying that T-tubule remodelling may play a crucial pathophysiologic role in HFrEF. While research on the functional importance of T-tubules is ongoing, T-tubule remodelling has been found to be reversible. That finding has triggered a surge in studies aimed at identifying specific therapeutic approaches for HFrEF. This review discusses the functional importance of T-tubules and their microdomains, the pathophysiology of T-tubule remodelling, and the potential mechanisms of current HFrEF therapeutic approaches in reversing T-tubule alterations. We also highlight discrepancies regarding the roles of T-tubule proteins in the recovery process across studies to offer valuable insights for future research.
Collapse
Affiliation(s)
- Yi Hao Loh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Jingyi Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yenfang Goh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xiangjie Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xianfeng Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Critical Care Medicine, Hangzhou Ninth People's Hospital, China
| | - Muergen Muheyati
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; School of Medicine, Shaoxing University, China.
| |
Collapse
|
4
|
Giraud Q, Laporte J. Amphiphysin-2 (BIN1) functions and defects in cardiac and skeletal muscle. Trends Mol Med 2024; 30:579-591. [PMID: 38514365 DOI: 10.1016/j.molmed.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
Amphiphysin-2 is a ubiquitously expressed protein also known as bridging integrator 1 (BIN1), playing a critical role in membrane remodeling, trafficking, and cytoskeleton dynamics in a wide range of tissues. Mutations in the gene encoding BIN1 cause centronuclear myopathies (CNM), and recent evidence has implicated BIN1 in heart failure, underlining its crucial role in both skeletal and cardiac muscle. Furthermore, altered expression of BIN1 is linked to an increased risk of late-onset Alzheimer's disease and several types of cancer, including breast, colon, prostate, and lung cancers. Recently, the first proof-of-concept for potential therapeutic strategies modulating BIN1 were obtained for muscle diseases. In this review article, we discuss the similarities and differences in BIN1's functions in cardiac and skeletal muscle, along with its associated diseases and potential therapies.
Collapse
Affiliation(s)
- Quentin Giraud
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch-Graffenstaden, 67400, France
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch-Graffenstaden, 67400, France.
| |
Collapse
|
5
|
Garcia‐Agudo LF, Shi Z, Smith IF, Kramár EA, Tran K, Kawauchi S, Wang S, Collins S, Walker A, Shi K, Neumann J, Liang HY, Da Cunha C, Milinkeviciute G, Morabito S, Miyoshi E, Rezaie N, Gomez‐Arboledas A, Arvilla AM, Ghaemi DI, Tenner AJ, LaFerla FM, Wood MA, Mortazavi A, Swarup V, MacGregor GR, Green KN. BIN1 K358R suppresses glial response to plaques in mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:2922-2942. [PMID: 38460121 PMCID: PMC11032570 DOI: 10.1002/alz.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 03/11/2024]
Abstract
INTRODUCTION The BIN1 coding variant rs138047593 (K358R) is linked to Late-Onset Alzheimer's Disease (LOAD) via targeted exome sequencing. METHODS To elucidate the functional consequences of this rare coding variant on brain amyloidosis and neuroinflammation, we generated BIN1K358R knock-in mice using CRISPR/Cas9 technology. These mice were subsequently bred with 5xFAD transgenic mice, which serve as a model for Alzheimer's pathology. RESULTS The presence of the BIN1K358R variant leads to increased cerebral amyloid deposition, with a dampened response of astrocytes and oligodendrocytes, but not microglia, at both the cellular and transcriptional levels. This correlates with decreased neurofilament light chain in both plasma and brain tissue. Synaptic densities are significantly increased in both wild-type and 5xFAD backgrounds homozygous for the BIN1K358R variant. DISCUSSION The BIN1 K358R variant modulates amyloid pathology in 5xFAD mice, attenuates the astrocytic and oligodendrocytic responses to amyloid plaques, decreases damage markers, and elevates synaptic densities. HIGHLIGHTS BIN1 rs138047593 (K358R) coding variant is associated with increased risk of LOAD. BIN1 K358R variant increases amyloid plaque load in 12-month-old 5xFAD mice. BIN1 K358R variant dampens astrocytic and oligodendrocytic response to plaques. BIN1 K358R variant decreases neuronal damage in 5xFAD mice. BIN1 K358R upregulates synaptic densities and modulates synaptic transmission.
Collapse
Affiliation(s)
| | - Zechuan Shi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Ian F. Smith
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Enikö A. Kramár
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Kai‐Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
| | - Heidi Yahan Liang
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Samuel Morabito
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Emily Miyoshi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Narges Rezaie
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Angela Gomez‐Arboledas
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Adrian Mendoza Arvilla
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Daryan Iman Ghaemi
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Department of Molecular Biology & BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Frank M. LaFerla
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| | - Ali Mortazavi
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Vivek Swarup
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
- Center for Complex Biological Systems, University of CaliforniaIrvineCaliforniaUSA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, ULAR, Office of Research, University of CaliforniaIrvineCaliforniaUSA
- Department of Developmental and Cell BiologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kim N. Green
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological Disorders, University of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
6
|
Tripathi S, Sharma Y, Rane R, Kumar D. CRISPR/Cas9 Gene Editing: A Novel Approach Towards Alzheimer's Disease Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1405-1424. [PMID: 38716549 DOI: 10.2174/0118715273283786240408034408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 10/22/2024]
Abstract
In defiance of the vast amount of information regarding Alzheimer's disease (AD) that has been learned over the past thirty years, progress toward developing an effective therapy has been difficult. A neurological ailment that progresses and cannot be reversed is Alzheimer's disease, which shows neurofibrillary tangles, beta-amyloid plaque, and a lack of cognitive processes that is created by tau protein clumps with hyperphosphorylation that finally advances to neuronal damage without a recognized treatment, which has stimulated research into new therapeutic strategies. The protein CAS9 is linked to CRISPR, which is a clustered Regularly Interspaced Short Palindromic Repeat that inactivates or corrects a gene by recognizing a gene sequence that produces a doublestranded break has enchanted a whole amount of interest towards its potency to cure gene sequences in AD. The novel CRISPR-Cas9 applications for developing in vitro and in vivo models to the benefit of AD investigation and therapies are thoroughly analyzed in this work. The discussion will also touch on the creation of delivery methods, which is a significant obstacle to the therapeutic use of CRISPR/Cas9 technology. By concentrating on specific genes, such as those that are significant early- onset AD risk factors and late-onset AD risk factors, like the apolipoprotein E4 (APOE4) gene, this study aims to evaluate the potential application of CRISPR/Cas9 as a possible treatment for AD.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Yashika Sharma
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Rajesh Rane
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Dileep Kumar
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| |
Collapse
|
7
|
Tamkus G, Uchida K, Lopatin AN. T-tubule recovery after detubulation in isolated mouse cardiomyocytes. Physiol Rep 2023; 11:e15779. [PMID: 37537144 PMCID: PMC10400551 DOI: 10.14814/phy2.15779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023] Open
Abstract
Remodeling of cardiac t-tubules in normal and pathophysiological conditions is an important process contributing to the functional performance of the heart. While it is well documented that deterioration of t-tubule network associated with various pathological conditions can be reversed under certain conditions, the mechanistic understanding of the recovery process is essentially lacking. Accordingly, in this study we investigated some aspects of the recovery of t-tubules after experimentally-induced detubulation. T-tubules of isolated mouse ventricular myocytes were first sealed using osmotic shock approach, and their recovery under various experimental conditions was then characterized using electrophysiologic and imaging techniques. The data show that t-tubule recovery is a strongly temperature-dependent process involving reopening of previously collapsed t-tubular segments. T-tubule recovery is slowed by (1) metabolic inhibition of cells, (2) reducing influx of extracellular Ca2+ as well as by (3) both stabilization and disruption of microtubules. Overall, the data show that t-tubule recovery is a highly dynamic process involving several central intracellular structures and processes and lay the basis for more detailed investigations in this area.
Collapse
Affiliation(s)
- Greta Tamkus
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Present address:
John T. Milliken Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Keita Uchida
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Present address:
Department of PhysiologyPennsylvania Muscle Institute, University of Pennsylvania, Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Anatoli N. Lopatin
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
8
|
Young-Pearse TL, Lee H, Hsieh YC, Chou V, Selkoe DJ. Moving beyond amyloid and tau to capture the biological heterogeneity of Alzheimer's disease. Trends Neurosci 2023; 46:426-444. [PMID: 37019812 PMCID: PMC10192069 DOI: 10.1016/j.tins.2023.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023]
Abstract
Alzheimer's disease (AD) manifests along a spectrum of cognitive deficits and levels of neuropathology. Genetic studies support a heterogeneous disease mechanism, with around 70 associated loci to date, implicating several biological processes that mediate risk for AD. Despite this heterogeneity, most experimental systems for testing new therapeutics are not designed to capture the genetically complex drivers of AD risk. In this review, we first provide an overview of those aspects of AD that are largely stereotyped and those that are heterogeneous, and we review the evidence supporting the concept that different subtypes of AD are important to consider in the design of agents for the prevention and treatment of the disease. We then dive into the multifaceted biological domains implicated to date in AD risk, highlighting studies of the diverse genetic drivers of disease. Finally, we explore recent efforts to identify biological subtypes of AD, with an emphasis on the experimental systems and data sets available to support progress in this area.
Collapse
Affiliation(s)
- Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yi-Chen Hsieh
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vicky Chou
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Caporizzo MA, Prosser BL. The microtubule cytoskeleton in cardiac mechanics and heart failure. Nat Rev Cardiol 2022; 19:364-378. [PMID: 35440741 PMCID: PMC9270871 DOI: 10.1038/s41569-022-00692-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 12/13/2022]
Abstract
The microtubule network of cardiac muscle cells has unique architectural and biophysical features to accommodate the demands of the working heart. Advances in live-cell imaging and in deciphering the 'tubulin code' have shone new light on this cytoskeletal network and its role in heart failure. Microtubule-based transport orchestrates the growth and maintenance of the contractile apparatus through spatiotemporal control of translation, while also organizing the specialized membrane systems required for excitation-contraction coupling. To withstand the high mechanical loads of the working heart, microtubules are post-translationally modified and physically reinforced. In response to stress to the myocardium, the microtubule network remodels, typically through densification, post-translational modification and stabilization. Under these conditions, physically reinforced microtubules resist the motion of the cardiomyocyte and increase myocardial stiffness. Accordingly, modified microtubules have emerged as a therapeutic target for reducing stiffness in heart failure. In this Review, we discuss the latest evidence on the contribution of microtubules to cardiac mechanics, the drivers of microtubule network remodelling in cardiac pathologies and the therapeutic potential of targeting cardiac microtubules in acquired heart diseases.
Collapse
Affiliation(s)
- Matthew A Caporizzo
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT, USA
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Abstract
Microtubules are essential cytoskeletal elements found in all eukaryotic cells. The structure and composition of microtubules regulate their function, and the dynamic remodeling of the network by posttranslational modifications and microtubule-associated proteins generates diverse populations of microtubules adapted for various contexts. In the cardiomyocyte, the microtubules must accommodate the unique challenges faced by a highly contractile, rigidly structured, and long-lasting cell. Through their canonical trafficking role and positioning of mRNA, proteins, and organelles, microtubules regulate essential cardiomyocyte functions such as electrical activity, calcium handling, protein translation, and growth. In a more specialized role, posttranslationally modified microtubules form load-bearing structures that regulate myocyte mechanics and mechanotransduction. Modified microtubules proliferate in cardiovascular diseases, creating stabilized resistive elements that impede cardiomyocyte contractility and contribute to contractile dysfunction. In this review, we highlight the most exciting new concepts emerging from recent studies into canonical and noncanonical roles of cardiomyocyte microtubules.
Collapse
Affiliation(s)
- Keita Uchida
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Emily A Scarborough
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
11
|
G N S HS, Marise VLP, Satish KS, Yergolkar AV, Krishnamurthy M, Ganesan Rajalekshmi S, Radhika K, Burri RR. Untangling huge literature to disinter genetic underpinnings of Alzheimer's Disease: A systematic review and meta-analysis. Ageing Res Rev 2021; 71:101421. [PMID: 34371203 DOI: 10.1016/j.arr.2021.101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/25/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Drug discovery for Alzheimer's Disease (AD) is channeled towards unravelling key disease specific drug targets/genes to predict promising therapeutic candidates. Though enormous literature on AD genetics is available, there exists dearth in data pertinent to drug targets and crucial pathological pathways intertwined in disease progression. Further, the research findings revealing genetic associations failed to demonstrate consistency across different studies. This scenario prompted us to initiate a systematic review and meta-analysis with an aim of unearthing significant genetic hallmarks of AD. Initially, a Boolean search strategy was developed to retrieve case-control studies from PubMed, Cochrane, ProQuest, Europe PMC, grey literature and HuGE navigator. Subsequently, certain inclusion and exclusion criteria were framed to shortlist the relevant studies. These studies were later critically appraised using New Castle Ottawa Scale and Q-Genie followed by data extraction. Later, meta-analysis was performed only for those Single Nucleotide Polymorphisms (SNPs) which were evaluated in at least two different ethnicities from two different reports. Among, 204,351 studies retrieved, 820 met our eligibility criteria and 117 were processed for systematic review after critical appraisal. Ultimately, meta-analysis was performed for 23 SNPs associated with 15 genes which revealed significant associations of rs3865444 (CD33), rs7561528 (BIN1) and rs1801133 (MTHFR) with AD risk.
Collapse
|
12
|
Franzmeier N, Ossenkoppele R, Brendel M, Rubinski A, Smith R, Kumar A, Mattsson-Carlgren N, Strandberg O, Duering M, Buerger K, Dichgans M, Hansson O, Ewers M. The BIN1 rs744373 Alzheimer's disease risk SNP is associated with faster Aβ-associated tau accumulation and cognitive decline. Alzheimers Dement 2021; 18:103-115. [PMID: 34060233 DOI: 10.1002/alz.12371] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/16/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The BIN1 rs744373 single nucleotide polymorphism (SNP) is a key genetic risk locus for Alzheimer's disease (AD) associated with tau pathology. Because tau typically accumulates in response to amyloid beta (Aβ), we tested whether BIN1 rs744373 accelerates Aβ-related tau accumulation. METHODS We included two samples (Alzheimer's Disease Neuroimaging Initiative [ADNI], n = 153; Biomarkers for Identifying Neurodegenerative Disorders Early and Reliably [BioFINDER], n = 63) with longitudinal 18 F-Flortaucipir positron emission tomography (PET), Aβ biomarkers, and longitudinal cognitive assessments. We assessed whether BIN1 rs744373 was associated with faster tau-PET accumulation at a given level of Aβ and whether faster BIN1 rs744373-associated tau-PET accumulation mediated cognitive decline. RESULTS BIN1 rs744373 risk-allele carriers showed faster global tau-PET accumulation (ADNI/BioFINDER, P < .001/P < .001). We found significant Aβ by rs744373 interactions on global tau-PET change (ADNI: β/standard error [SE] = 0.42/0.14, P = 0.002; BioFINDER: β/SE = -0.35/0.15, P = .021), BIN1 risk-allele carriers showed accelerated tau-PET accumulation at higher Aβ levels. In ADNI, rs744373 effects on cognitive decline were mediated by faster global tau-PET accumulation (β/SE = 0.20/0.07, P = .005). DISCUSSION BIN1-associated AD risk is potentially driven by accelerated tau accumulation in the face of Aβ.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anna Rubinski
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Atul Kumar
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Marco Duering
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.,Medical Image Analysis Center (MIAC AG), Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | |
Collapse
|
13
|
Mechanisms and Regulation of Cardiac Ca V1.2 Trafficking. Int J Mol Sci 2021; 22:ijms22115927. [PMID: 34072954 PMCID: PMC8197997 DOI: 10.3390/ijms22115927] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/05/2023] Open
Abstract
During cardiac excitation contraction coupling, the arrival of an action potential at the ventricular myocardium triggers voltage-dependent L-type Ca2+ (CaV1.2) channels in individual myocytes to open briefly. The level of this Ca2+ influx tunes the amplitude of Ca2+-induced Ca2+ release from ryanodine receptors (RyR2) on the junctional sarcoplasmic reticulum and thus the magnitude of the elevation in intracellular Ca2+ concentration and ultimately the downstream contraction. The number and activity of functional CaV1.2 channels at the t-tubule dyads dictates the amplitude of the Ca2+ influx. Trafficking of these channels and their auxiliary subunits to the cell surface is thus tightly controlled and regulated to ensure adequate sarcolemmal expression to sustain this critical process. To that end, recent discoveries have revealed the existence of internal reservoirs of preformed CaV1.2 channels that can be rapidly mobilized to enhance sarcolemmal expression in times of acute stress when hemodynamic and metabolic demand increases. In this review, we provide an overview of the current thinking on CaV1.2 channel trafficking dynamics in the heart. We highlight the numerous points of control including the biosynthetic pathway, the endosomal recycling pathway, ubiquitination, and lysosomal and proteasomal degradation pathways, and discuss the effects of β-adrenergic and angiotensin receptor signaling cascades on this process.
Collapse
|
14
|
Caporizzo MA, Chen CY, Prosser BL. Cardiac microtubules in health and heart disease. Exp Biol Med (Maywood) 2019; 244:1255-1272. [PMID: 31398994 DOI: 10.1177/1535370219868960] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes are large (∼40,000 µm3), rod-shaped muscle cells that provide the working force behind each heartbeat. These highly structured cells are packed with dense cytoskeletal networks that can be divided into two groups—the contractile (i.e. sarcomeric) cytoskeleton that consists of filamentous actin-myosin arrays organized into myofibrils, and the non-sarcomeric cytoskeleton, which is composed of β- and γ-actin, microtubules, and intermediate filaments. Together, microtubules and intermediate filaments form a cross-linked scaffold, and these networks are responsible for the delivery of intracellular cargo, the transmission of mechanical signals, the shaping of membrane systems, and the organization of myofibrils and organelles. Microtubules are extensively altered as part of both adaptive and pathological cardiac remodeling, which has diverse ramifications for the structure and function of the cardiomyocyte. In heart failure, the proliferation and post-translational modification of the microtubule network is linked to a number of maladaptive processes, including the mechanical impediment of cardiomyocyte contraction and relaxation. This raises the possibility that reversing microtubule alterations could improve cardiac performance, yet therapeutic efforts will strongly benefit from a deeper understanding of basic microtubule biology in the heart. The aim of this review is to summarize the known physiological roles of the cardiomyocyte microtubule network, the consequences of its pathological remodeling, and to highlight the open and intriguing questions regarding cardiac microtubules. Impact statement Advancements in cell biological and biophysical approaches and super-resolution imaging have greatly broadened our view of tubulin biology over the last decade. In the heart, microtubules and microtubule-based transport help to organize and maintain key structures within the cardiomyocyte, including the sarcomere, intercalated disc, protein clearance machinery and transverse-tubule and sarcoplasmic reticulum membranes. It has become increasingly clear that post translational regulation of microtubules is a key determinant of their sub-cellular functionality. Alterations in microtubule network density, stability, and post-translational modifications are hallmarks of pathological cardiac remodeling, and modified microtubules can directly impede cardiomyocyte contractile function in various forms of heart disease. This review summarizes the functional roles and multi-leveled regulation of the cardiac microtubule cytoskeleton and highlights how refined experimental techniques are shedding mechanistic clarity on the regionally specified roles of microtubules in cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Matthew A Caporizzo
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Christina Yingxian Chen
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.,Penn Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Basheer WA, Shaw RM. Connexin 43 and CaV1.2 Ion Channel Trafficking in Healthy and Diseased Myocardium. Circ Arrhythm Electrophysiol 2018; 9:e001357. [PMID: 27266274 DOI: 10.1161/circep.115.001357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/29/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Wassim A Basheer
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.)
| | - Robin M Shaw
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.).
| |
Collapse
|
16
|
Abstract
Alzheimer's disease (AD), the main form of dementia in the elderly, is the most common progressive neurodegenerative disease characterized by rapidly progressive cognitive dysfunction and behavior impairment. AD exhibits a considerable heritability and great advances have been made in approaches to searching the genetic etiology of AD. In AD genetic studies, methods have developed from classic linkage-based and candidate-gene-based association studies to genome-wide association studies (GWAS) and next generation sequencing (NGS). The identification of new susceptibility genes has provided deeper insights to understand the mechanisms underlying AD. In addition to searching novel genes associated with AD in large samples, the NGS technologies can also be used to shed light on the 'black matter' discovery even in smaller samples. The shift in AD genetics between traditional studies and individual sequencing will allow biomaterials of each patient as the central unit of genetic studies. This review will cover genetic findings in AD and consequences of AD genetic findings. Firstly, we will discuss the discovery of mutations in APP, PSEN1, PSEN2, APOE, and ADAM10. Then we will summarize and evaluate the information obtained from GWAS of AD. Finally, we will outline the efforts to identify rare variants associated with AD using NGS.
Collapse
|
17
|
Sun Q, Xie N, Tang B, Li R, Shen Y. Alzheimer's Disease: From Genetic Variants to the Distinct Pathological Mechanisms. Front Mol Neurosci 2017; 10:319. [PMID: 29056900 PMCID: PMC5635057 DOI: 10.3389/fnmol.2017.00319] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/20/2017] [Indexed: 11/13/2022] Open
Abstract
Being the most common cause of dementia, AD is a polygenic and neurodegenerative disease. Complex and multiple factors have been shown to be involved in its pathogenesis, of which the genetics play an indispensable role. It is widely accepted that discovery of potential genes related to the pathogenesis of AD would be of great help for the understanding of neurodegeneration and thus further promote molecular diagnosis in clinic settings. Generally, AD could be clarified into two types according to the onset age, the early-onset AD (EOAD) and the late-onset AD (LOAD). Progresses made by genetic studies on both EOAD and LOAD are believed to be essential not only for the revolution of conventional ideas but also for the revelation of new pathological mechanisms underlying AD pathogenesis. Currently, albeit the genetics of LOAD is much less well-understood compared to EOAD due to its complicated and multifactorial essence, Genome-wide association studies (GWASs) and next generation sequencing (NGS) approaches have identified dozens of novel genes that may provide insight mechanism of LOAD. In this review, we analyze functions of the genes and summarize the distinct pathological mechanisms of how these genes would be involved in the pathogenesis of AD.
Collapse
Affiliation(s)
- Qiying Sun
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China.,Center for Advanced Therapeutic Strategies for Brain Disorders and Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL, United States
| | - Nina Xie
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Beisha Tang
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rena Li
- Center for Advanced Therapeutic Strategies for Brain Disorders and Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL, United States.,National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yong Shen
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China.,Center for Advanced Therapeutic Strategies for Brain Disorders and Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL, United States.,Neurodegenerative Disorder Research Center, University of Science and Technology of China School of Life Sciences, Hefei, China.,Hefei Material Science at Microscale National Laboratory, Hefei, China
| |
Collapse
|
18
|
D'Alessandro M, Hnia K, Gache V, Koch C, Gavriilidis C, Rodriguez D, Nicot AS, Romero NB, Schwab Y, Gomes E, Labouesse M, Laporte J. Amphiphysin 2 Orchestrates Nucleus Positioning and Shape by Linking the Nuclear Envelope to the Actin and Microtubule Cytoskeleton. Dev Cell 2016; 35:186-98. [PMID: 26506308 DOI: 10.1016/j.devcel.2015.09.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 12/31/2022]
Abstract
Nucleus positioning is key for intracellular organization, cell differentiation, and organ development and is affected in many diseases, including myopathies due to alteration in amphiphysin-2 (BIN1). The actin and microtubule cytoskeletons are essential for nucleus positioning, but their crosstalk in this process is sparsely characterized. Here, we report that impairment of amphiphysin/BIN1 in Caenorhabditis elegans, mammalian cells, or muscles from patients with centronuclear myopathy alters nuclear position and shape. We show that AMPH-1/BIN1 binds to nesprin and actin, as well as to the microtubule-binding protein CLIP170 in both species. Expression of the microtubule-anchoring CAP-GLY domain of CLIP170 fused to the nuclear-envelope-anchoring KASH domain of nesprin rescues nuclear positioning defects of amph-1 mutants. Amphiphysins thus play a central role in linking the nuclear envelope with the actin and microtubule cytoskeletons. We propose that BIN1 has a direct and evolutionarily conserved role in nuclear positioning, altered in myopathies.
Collapse
Affiliation(s)
- Manuela D'Alessandro
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France; University Claude Bernard Lyon 1, CGphiMC UMR CNRS 5534, 69622 Villeurbanne, France.
| | - Karim Hnia
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France
| | - Vincent Gache
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, 75634 Paris, France; CNRS UMR5239, Laboratoire de Biologie Moléculaire de la Cellule (LBMC), Ecole Normale Supérieure de Lyon, 69364 Lyon Cedex 07, France
| | - Catherine Koch
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France
| | | | - David Rodriguez
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France
| | - Anne-Sophie Nicot
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France
| | - Norma B Romero
- Morphology Neuromuscular Unit of the Myology Institute, GHU Pitié-Salpêtrière, 75013 Paris, France
| | - Yannick Schwab
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France; Cell Biology and Biophysics Unit, EMBL Heidelberg, 69117 Heidelberg, Germany
| | - Edgar Gomes
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, 75634 Paris, France; Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Michel Labouesse
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France; UMR7622, IBPS, UPMC, 75252 Paris, France
| | - Jocelyn Laporte
- IGBMC, INSERM U964, CNRS UMR7104, Strasbourg University, 67404 Illkirch, France.
| |
Collapse
|
19
|
Zhu R, Liu X, He Z. The Bridging Integrator 1 Gene Polymorphism rs744373 and the Risk of Alzheimer's Disease in Caucasian and Asian Populations: An Updated Meta-Analysis. Mol Neurobiol 2016; 54:1419-1428. [PMID: 26846281 DOI: 10.1007/s12035-016-9760-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 01/26/2016] [Indexed: 01/02/2023]
Abstract
Recent genome-wide association studies have identified an association between the bridging integrator 1 gene (BIN1) rs744373 polymorphism and late-onset Alzheimer's disease (LOAD) in individuals of European ancestry. Additionally, a number of studies have focused on the association between rs744373 and Alzheimer's disease in Caucasian and East Asian populations. However, these results remain inconclusive because of the relatively small sample sizes investigated. Here, we reevaluated this association using samples from seven articles including 22 independent studies comprising 11,832 LOAD patients and 18,133 controls identified by searching PubMed, MEDLINE, and AlzGene databases up to December 2015. We observed no significant heterogeneity between Asian and Caucasian populations. Additive, dominant, and recessive models revealed a significant association between rs744373 and LOAD in the pooled population, while subgroup analysis also identified significant findings in the East Asian population under the additive model (odds ratio (OR) = 1.10, 95 % confidence interval (CI) 1.02-1.19, P = 0.01) and dominant model (OR = 1.13, 95 % CI 1.03-1.25, P = 0.01), but not under the recessive model. The current meta-analysis further supports previous findings that the rs744373 polymorphism may be associated with LOAD risk in Caucasian and Asian populations. To our knowledge, this is the first large meta-analysis to investigate the association between the rs744373 polymorphism and LOAD in East Asian, American, and European populations.
Collapse
Affiliation(s)
- Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, China
| | - Xu Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Shenyang, 110001, China.
| |
Collapse
|
20
|
Lazaris A, Hwang KS, Goukasian N, Ramirez LM, Eastman J, Blanken AE, Teng E, Gylys K, Cole G, Saykin AJ, Shaw LM, Trojanowski JQ, Jagust WJ, Weiner MW, Apostolova LG. Alzheimer risk genes modulate the relationship between plasma apoE and cortical PiB binding. NEUROLOGY-GENETICS 2015; 1:e22. [PMID: 27066559 PMCID: PMC4809461 DOI: 10.1212/nxg.0000000000000022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 08/13/2015] [Indexed: 01/28/2023]
Abstract
Objective: We investigated the association between apoE protein plasma levels and brain amyloidosis and the effect of the top 10 Alzheimer disease (AD) risk genes on this association. Methods: Our dataset consisted of 18 AD, 52 mild cognitive impairment, and 3 cognitively normal Alzheimer's Disease Neuroimaging Initiative 1 (ADNI1) participants with available [11C]-Pittsburgh compound B (PiB) and peripheral blood protein data. We used cortical pattern matching to study associations between plasma apoE and cortical PiB binding and the effect of carrier status for the top 10 AD risk genes. Results: Low plasma apoE was significantly associated with high PiB SUVR, except in the sensorimotor and entorhinal cortex. For BIN1 rs744373, the association was observed only in minor allele carriers. For CD2AP rs9349407 and CR1 rs3818361, the association was preserved only in minor allele noncarriers. We did not find evidence for modulation by CLU, PICALM, ABCA7, BIN1, and MS4A6A. Conclusions: Our data show that BIN1 rs744373, CD2AP rs9349407, and CR1 rs3818361 genotypes modulate the association between apoE protein plasma levels and brain amyloidosis, implying a potential epigenetic/downstream interaction.
Collapse
Affiliation(s)
- Andreas Lazaris
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Kristy S Hwang
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Naira Goukasian
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Leslie M Ramirez
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Jennifer Eastman
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Anna E Blanken
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Edmond Teng
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Karen Gylys
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Greg Cole
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Andrew J Saykin
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Leslie M Shaw
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - John Q Trojanowski
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - William J Jagust
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Michael W Weiner
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | - Liana G Apostolova
- University of California Berkeley (A.L.), Berkeley; Oakland University William Beaumont School of Medicine (K.S.H.), Rochester, MI; Department of Neurology (K.S.H., N.G., A.E.B., E.T., G.C., L.G.A.), David Geffen School of Medicine at UCLA, Los Angeles, CA; Drexel University College of Medicine (L.M.R.), Philadelphia, PA; Northwestern University Feinberg School of Medicine (J.E.), Chicago, IL; Veterans Affairs Greater Los Angeles Healthcare System (E.T., G.C.), Los Angeles, CA; School of Nursing (K.G.), UCLA, Los Angeles, CA; Department of Radiology and Imaging Sciences, Center for Neuroimaging (A.J.S., L.G.A.), Department of Neurology (L.G.A.), and Department of Medical and Molecular Genetics (L.G.A.), School of Medicine, Indiana University, Indianapolis; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), University of Pennsylvania School of Medicine, Philadelphia; Department of Public Health and Neuroscience (W.J.J.), UC Berkeley, CA; and Department of Veterans' Affairs Medical Center (M.W.W.), San Francisco, CA
| | | |
Collapse
|
21
|
Tau phosphorylation regulates the interaction between BIN1's SH3 domain and Tau's proline-rich domain. Acta Neuropathol Commun 2015; 3:58. [PMID: 26395440 PMCID: PMC4580349 DOI: 10.1186/s40478-015-0237-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/07/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The application of high-throughput genomic approaches has revealed 24 novel risk loci for Alzheimer's disease (AD). We recently reported that the bridging integrator 1 (BIN1) risk gene is linked to Tau pathology. RESULTS We used glutathione S-transferase pull-down assays and nuclear magnetic resonance (NMR) experiments to demonstrate that BIN1 and Tau proteins interact directly and then map the interaction between BIN1's SH3 domain and Tau's proline-rich domain (PRD) . Our NMR data showed that Tau phosphorylation at Thr231 weakens the SH3-PRD interaction. Using primary neurons, we found that BIN1-Tau complexes partly co-localize with the actin cytoskeleton; however, these complexes were not observed with Thr231-phosphorylated Tau species. CONCLUSION Our results show that (i) BIN1 and Tau bind through an SH3-PRD interaction and (ii) the interaction is downregulated by phosphorylation of Tau Thr231 (and potentially other residues). Our study sheds new light on regulation of the BIN1/Tau interaction and opens up new avenues for exploring its complex's role in the pathogenesis of AD.
Collapse
|
22
|
Holler CJ, Davis PR, Beckett TL, Platt TL, Webb RL, Head E, Murphy MP. Bridging integrator 1 (BIN1) protein expression increases in the Alzheimer's disease brain and correlates with neurofibrillary tangle pathology. J Alzheimers Dis 2015; 42:1221-7. [PMID: 25024306 DOI: 10.3233/jad-132450] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent genome wide association studies have implicated bridging integrator 1 (BIN1) as a late-onset Alzheimer's disease (AD) susceptibility gene. There are at least 15 different known isoforms of BIN1, with many being expressed in the brain including the longest isoform (iso1), which is brain-specific and localizes to axon initial segments and nodes of Ranvier. It is currently unknown what role BIN1 plays in AD. We analyzed BIN1 protein expression from a large number (n = 71) of AD cases and controls from five different brain regions (hippocampus, inferior parietal cortex, inferior temporal cortex, frontal cortex (BA9), and superior and middle temporal gyri). We found that the amount of the largest isoform of BIN1 was significantly reduced in the AD brain compared to age-matched controls, and smaller BIN1 isoforms were significantly increased. Further, BIN1 was significantly correlated with the amount of neurofibrillary tangle (NFT) pathology but not with either diffuse or neuritic plaques, or with the amount of amyloid-β peptide. BIN1 is known to be abnormally expressed in another human disease, myotonic dystrophy, which also features prominent NFT pathology. These data suggest that BIN1 is likely involved in AD as a modulator of NFT pathology, and that this role may extend to other human diseases that feature tau pathology.
Collapse
Affiliation(s)
- Christopher J Holler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Paulina R Davis
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Thomas L Platt
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Robin L Webb
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA University of Kentucky Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
23
|
Day CA, Baetz NW, Copeland CA, Kraft LJ, Han B, Tiwari A, Drake KR, De Luca H, Chinnapen DJF, Davidson MW, Holmes RK, Jobling MG, Schroer TA, Lencer WI, Kenworthy AK. Microtubule motors power plasma membrane tubulation in clathrin-independent endocytosis. Traffic 2015; 16:572-90. [PMID: 25690058 PMCID: PMC4440230 DOI: 10.1111/tra.12269] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 12/13/2022]
Abstract
How the plasma membrane is bent to accommodate clathrin-independent endocytosis remains uncertain. Recent studies suggest Shiga and cholera toxin induce membrane curvature required for their uptake into clathrin-independent carriers by binding and cross-linking multiple copies of their glycosphingolipid receptors on the plasma membrane. But it remains unclear if toxin-induced sphingolipid crosslinking provides sufficient mechanical force for deforming the plasma membrane, or if host cell factors also contribute to this process. To test this, we imaged the uptake of cholera toxin B-subunit into surface-derived tubular invaginations. We found that cholera toxin mutants that bind to only one glycosphingolipid receptor accumulated in tubules, and that toxin binding was entirely dispensable for membrane tubulations to form. Unexpectedly, the driving force for tubule extension was supplied by the combination of microtubules, dynein and dynactin, thus defining a novel mechanism for generating membrane curvature during clathrin-independent endocytosis.
Collapse
Affiliation(s)
- Charles A Day
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Current address: Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Nicholas W Baetz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Courtney A Copeland
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lewis J Kraft
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA
| | - Bing Han
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kimberly R Drake
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Heidi De Luca
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Daniel J-F Chinnapen
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Michael W Davidson
- National High Magnetic Field Laboratory, The Florida State University, Tallahassee, FL, USA
| | - Randall K Holmes
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael G Jobling
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Trina A Schroer
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Wayne I Lencer
- GI Cell Biology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School and the Harvard Digestive Diseases Center, Boston, MA, USA
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Epithelial Biology Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
24
|
Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 2015; 77:43-51. [PMID: 24951455 PMCID: PMC4234692 DOI: 10.1016/j.biopsych.2014.05.006] [Citation(s) in RCA: 948] [Impact Index Per Article: 94.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 01/18/2023]
Abstract
We review the genetic risk factors for late-onset Alzheimer's disease (AD) and their role in AD pathogenesis. More recent advances in understanding of the human genome-technologic advances in methods to analyze millions of polymorphisms in thousands of subjects-have revealed new genes associated with AD risk, including ABCA7, BIN1, CASS4, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA-DRB5-DBR1, INPP5D, MS4A, MEF2C, NME8, PICALM, PTK2B, SLC24H4-RIN3, SORL1, and ZCWPW1. Emerging technologies to analyze the entire genome in large data sets have also revealed coding variants that increase AD risk: PLD3 and TREM2. We review the relationship between these AD risk genes and the cellular and neuropathologic features of AD. Understanding the mechanisms underlying the association of these genes with risk for disease will provide the most meaningful targets for therapeutic development to date.
Collapse
Affiliation(s)
| | - Alison M. Goate
- Corresponding author Contact information: Department of Psychiatry, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8134, St. Louis, MO 63110, phone: 314-362-8691, fax: 314-747-2983,
| |
Collapse
|
25
|
Amphiphysin 2 (BIN1) in physiology and diseases. J Mol Med (Berl) 2014; 92:453-63. [DOI: 10.1007/s00109-014-1138-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 02/11/2014] [Accepted: 02/17/2014] [Indexed: 12/15/2022]
|
26
|
Federighi G, Traina G, Macchi M, Ciampini C, Bernardi R, Baldi E, Bucherelli C, Brunelli M, Scuri R. Modulation of gene expression in contextual fear conditioning in the rat. PLoS One 2013; 8:e80037. [PMID: 24278235 PMCID: PMC3837011 DOI: 10.1371/journal.pone.0080037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/27/2013] [Indexed: 01/30/2023] Open
Abstract
In contextual fear conditioning (CFC) a single training leads to long-term memory of context-aversive electrical foot-shocks association. Mid-temporal regions of the brain of trained and naive rats were obtained 2 days after conditioning and screened by two-directional suppression subtractive hybridization. A pool of differentially expressed genes was identified and some of them were randomly selected and confirmed with qRT-PCR assay. These transcripts showed high homology for rat gene sequences coding for proteins involved in different cellular processes. The expression of the selected transcripts was also tested in rats which had freely explored the experimental apparatus (exploration) and in rats to which the same number of aversive shocks had been administered in the same apparatus, but temporally compressed so as to make the association between painful stimuli and the apparatus difficult (shock-only). Some genes resulted differentially expressed only in the rats subjected to CFC, others only in exploration or shock-only rats, whereas the gene coding for translocase of outer mitochondrial membrane 20 protein and nardilysin were differentially expressed in both CFC and exploration rats. For example, the expression of stathmin 1 whose transcripts resulted up regulated was also tested to evaluate the transduction and protein localization after conditioning.
Collapse
Affiliation(s)
- Giuseppe Federighi
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Giovanna Traina
- Dipartimento di Scienze Economico-Estimative e degli Alimenti, Sezione di Chimica Bromatologica, Biochimica, Fisiologia e Nutrizione, Università degli Studi di Perugia, Perugia, Italy
| | - Monica Macchi
- Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Cristina Ciampini
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Rodolfo Bernardi
- Dipartimento di Scienze Agrarie, Genetica Alimentari e Agro-Ambientali, Università di Pisa, Pisa, Italy
| | - Elisabetta Baldi
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | - Corrado Bucherelli
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | | | - Rossana Scuri
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| |
Collapse
|
27
|
Increased expression of BIN1 mediates Alzheimer genetic risk by modulating tau pathology. Mol Psychiatry 2013; 18:1225-34. [PMID: 23399914 PMCID: PMC3807661 DOI: 10.1038/mp.2013.1] [Citation(s) in RCA: 337] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 10/26/2012] [Accepted: 12/10/2012] [Indexed: 12/24/2022]
Abstract
Genome-wide association studies (GWAS) have identified a region upstream the BIN1 gene as the most important genetic susceptibility locus in Alzheimer's disease (AD) after APOE. We report that BIN1 transcript levels were increased in AD brains and identified a novel 3 bp insertion allele ∼28 kb upstream of BIN1, which increased (i) transcriptional activity in vitro, (ii) BIN1 expression levels in human brain and (iii) AD risk in three independent case-control cohorts (Meta-analysed Odds ratio of 1.20 (1.14-1.26) (P=3.8 × 10(-11))). Interestingly, decreased expression of the Drosophila BIN1 ortholog Amph suppressed Tau-mediated neurotoxicity in three different assays. Accordingly, Tau and BIN1 colocalized and interacted in human neuroblastoma cells and in mouse brain. Finally, the 3 bp insertion was associated with Tau but not Amyloid loads in AD brains. We propose that BIN1 mediates AD risk by modulating Tau pathology.
Collapse
|
28
|
Gibbs EM, Davidson AE, Telfer WR, Feldman EL, Dowling JJ. The myopathy-causing mutation DNM2-S619L leads to defective tubulation in vitro and in developing zebrafish. Dis Model Mech 2013; 7:157-61. [PMID: 24135484 PMCID: PMC3882057 DOI: 10.1242/dmm.012286] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
DNM2 is a ubiquitously expressed GTPase that regulates multiple subcellular processes. Mutations in DNM2 are a common cause of centronuclear myopathy, a severe disorder characterized by altered skeletal muscle structure and function. The precise mechanisms underlying disease-associated DNM2 mutations are unresolved. We examined the common DNM2-S619L mutation using both in vitro and in vivo approaches. Expression of DNM2-S619L in zebrafish led to the accumulation of aberrant vesicular structures and to defective excitation-contraction coupling. Expression of DNM2-S619L in COS7 cells resulted in defective BIN1-dependent tubule formation. These data suggest that DNM2-S619L causes disease, in part, by interfering with membrane tubulation.
Collapse
Affiliation(s)
- Elizabeth M Gibbs
- Department of Neuroscience, University of Michigan Medical Center, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | |
Collapse
|
29
|
Tan MS, Yu JT, Tan L. Bridging integrator 1 (BIN1): form, function, and Alzheimer's disease. Trends Mol Med 2013; 19:594-603. [PMID: 23871436 DOI: 10.1016/j.molmed.2013.06.004] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/24/2013] [Accepted: 06/21/2013] [Indexed: 12/13/2022]
Abstract
The bridging integrator 1 (BIN1) gene, also known as amphiphysin 2, has recently been identified as the most important risk locus for late onset Alzheimer's disease (LOAD), after apolipoprotein E (APOE). Here, we summarize the known functions of BIN1 and discuss the polymorphisms associated with LOAD, as well as their possible physiological effects. Emerging data suggest that BIN1 affects AD risk primarily by modulating tau pathology, but other affected cellular functions are discussed, including endocytosis/trafficking, inflammation, calcium homeostasis, and apoptosis. Epigenetic modifications are important for AD pathogenesis, and we review data that suggests the possible DNA methylation of the BIN1 promoter. Finally, given the potential contributions of BIN1 to AD pathogenesis, targeting BIN1 might present novel opportunities for AD therapy.
Collapse
Affiliation(s)
- Meng-Shan Tan
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266003, China; Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China
| | | | | |
Collapse
|
30
|
Trafford AW, Clarke JD, Richards MA, Eisner DA, Dibb KM. Calcium signalling microdomains and the t-tubular system in atrial mycoytes: potential roles in cardiac disease and arrhythmias. Cardiovasc Res 2013; 98:192-203. [PMID: 23386275 DOI: 10.1093/cvr/cvt018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The atria contribute 25% to ventricular stroke volume and are the site of the commonest cardiac arrhythmia, atrial fibrillation (AF). The initiation of contraction in the atria is similar to that in the ventricle involving a systolic rise of intracellular Ca(2+) concentration ([Ca(2+)](i)). There are, however, substantial inter-species differences in the way systolic Ca(2+) is regulated in atrial cells. These differences are a consequence of a well-developed and functionally relevant transverse (t)-tubule network in the atria of large mammals, including humans, and its virtual absence in smaller laboratory species such as the rat. Where T-tubules are absent, the systolic Ca(2+) transient results from a 'fire-diffuse-fire' sequential recruitment of Ca(2+) release sites from the cell edge to the centre and hence marked spatiotemporal heterogeneity of systolic Ca(2+). Conversely, the well-developed T-tubule network in large mammals ensures a near synchronous rise of [Ca(2+)](i). In addition to synchronizing the systolic rise of [Ca(2+)](i), the presence of T-tubules in the atria of large mammals, by virtue of localization of the L-type Ca(2+) channels and Na(+)-Ca(2+) exchanger antiporters on the T-tubules, may serve to, respectively, accelerate changes in the amplitude of the systolic Ca(2+) transient during inotropic manoeuvres and lower diastolic [Ca(2+)](i). On the other hand, the presence of T-tubules and hence wider cellular distribution of the Na(+)-Ca(2+) exchanger may predispose the atria of large mammals to Ca(2+)-dependent delayed afterdepolarizations (DADs); this may be a determining factor in why the atria of large mammals spontaneously develop and maintain AF.
Collapse
Affiliation(s)
- Andrew W Trafford
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, Institute of Cardiovascular Science, University of Manchester, 3.23 Core Technology Facility, 46 Grafton Street, Manchester M13 9PT, UK
| | | | | | | | | |
Collapse
|
31
|
Further insights into Alzheimer's disease. QUALITY IN AGEING AND OLDER ADULTS 2012. [DOI: 10.1108/14717791211264043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
Abstract
Genetic factors are now recognized to play an important role in most age-related dementias. Although other factors, including aging itself, contribute to dementia, in this review the authors focus on the role of specific disease-causing genes and genetic factors in the most common age-related dementias. They review each dementia within the context of a genes/environment continuum, with varying levels of genetic versus environmental influence. All major classes of dementia will be discussed but greatest attention will be given to the most common dementia, Alzheimer's disease, for which several new genetic factors were recently identified.
Collapse
Affiliation(s)
- Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | |
Collapse
|
33
|
de Kreuk BJ, Nethe M, Fernandez-Borja M, Anthony EC, Hensbergen PJ, Deelder AM, Plomann M, Hordijk PL. The F-BAR domain protein PACSIN2 associates with Rac1 and regulates cell spreading and migration. J Cell Sci 2011; 124:2375-88. [PMID: 21693584 DOI: 10.1242/jcs.080630] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Rac1 GTPase controls cytoskeletal dynamics and is a key regulator of cell spreading and migration mediated by signaling through effector proteins, such as the PAK kinases and the Scar and WAVE proteins. We previously identified a series of regulatory proteins that associate with Rac1 through its hypervariable C-terminal domain, including the Rac1 activator β-Pix (also known as Rho guanine-nucleotide-exchange factor 7) and the membrane adapter caveolin-1. Here, we show that Rac1 associates, through its C-terminus, with the F-BAR domain protein PACSIN2, an inducer of membrane tubulation and a regulator of endocytosis. We show that Rac1 localizes with PACSIN2 at intracellular tubular structures and on early endosomes. Active Rac1 induces a loss of PACSIN2-positive tubular structures. By contrast, Rac1 inhibition results in an accumulation of PACSIN2-positive tubules. In addition, PACSIN2 appears to regulate Rac1 signaling; siRNA-mediated loss of PACSIN2 increases the levels of Rac1-GTP and promotes cell spreading and migration in a wound healing assay. Moreover, ectopic expression of PACSIN2 reduces Rac1-GTP levels in a fashion that is dependent on the PACSIN2-Rac1 interaction, on the membrane-tubulating capacity of PACSIN2 and on dynamin. These data identify the BAR-domain protein PACSIN2 as a Rac1 interactor that regulates Rac1-mediated cell spreading and migration.
Collapse
Affiliation(s)
- Bart-Jan de Kreuk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Quiñones GA, Oro AE. BAR domain competition during directional cellular migration. Cell Cycle 2010; 9:2522-8. [PMID: 20581461 PMCID: PMC2990792 DOI: 10.4161/cc.9.13.12123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 04/19/2010] [Indexed: 01/08/2023] Open
Abstract
While directed cellular migration facilitates the coordinated movement of cells during development and tissue repair, the precise mechanisms regulating the interplay between the extracellular environment, the actin cytoskeleton, and the overlying plasma membrane remain inadequately understood. The BAR domain family of lipid binding, actin cytoskeletal regulators are gaining greater appreciation for their role in these critical processes. BAR domain proteins are involved as both positive and negative regulators of endocytosis, membrane plasticity, and directional cell migration. This review focuses on the functional relationship between different classes of BAR domain proteins and their role in guiding cell migration through regulation of the endocytic machinery. Competition for key signaling substrates by positive and negative BAR domain endocytic regulators appears to mediate control of directional cell migration, and may have wider applicability to other trafficking functions associated with development and carcinogenesis.
Collapse
Affiliation(s)
- Gabriel A Quiñones
- Program in Epithelial Biology and Cancer Biology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
35
|
Hong TT, Smyth JW, Gao D, Chu KY, Vogan JM, Fong TS, Jensen BC, Colecraft HM, Shaw RM. BIN1 localizes the L-type calcium channel to cardiac T-tubules. PLoS Biol 2010; 8:e1000312. [PMID: 20169111 PMCID: PMC2821894 DOI: 10.1371/journal.pbio.1000312] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 01/12/2010] [Indexed: 01/15/2023] Open
Abstract
The BAR domain protein superfamily is involved in membrane invagination and endocytosis, but its role in organizing membrane proteins has not been explored. In particular, the membrane scaffolding protein BIN1 functions to initiate T-tubule genesis in skeletal muscle cells. Constitutive knockdown of BIN1 in mice is perinatal lethal, which is associated with an induced dilated hypertrophic cardiomyopathy. However, the functional role of BIN1 in cardiomyocytes is not known. An important function of cardiac T-tubules is to allow L-type calcium channels (Cav1.2) to be in close proximity to sarcoplasmic reticulum-based ryanodine receptors to initiate the intracellular calcium transient. Efficient excitation-contraction (EC) coupling and normal cardiac contractility depend upon Cav1.2 localization to T-tubules. We hypothesized that BIN1 not only exists at cardiac T-tubules, but it also localizes Cav1.2 to these membrane structures. We report that BIN1 localizes to cardiac T-tubules and clusters there with Cav1.2. Studies involve freshly acquired human and mouse adult cardiomyocytes using complementary immunocytochemistry, electron microscopy with dual immunogold labeling, and co-immunoprecipitation. Furthermore, we use surface biotinylation and live cell confocal and total internal fluorescence microscopy imaging in cardiomyocytes and cell lines to explore delivery of Cav1.2 to BIN1 structures. We find visually and quantitatively that dynamic microtubules are tethered to membrane scaffolded by BIN1, allowing targeted delivery of Cav1.2 from the microtubules to the associated membrane. Since Cav1.2 delivery to BIN1 occurs in reductionist non-myocyte cell lines, we find that other myocyte-specific structures are not essential and there is an intrinsic relationship between microtubule-based Cav1.2 delivery and its BIN1 scaffold. In differentiated mouse cardiomyocytes, knockdown of BIN1 reduces surface Cav1.2 and delays development of the calcium transient, indicating that Cav1.2 targeting to BIN1 is functionally important to cardiac calcium signaling. We have identified that membrane-associated BIN1 not only induces membrane curvature but can direct specific antegrade delivery of microtubule-transported membrane proteins. Furthermore, this paradigm provides a microtubule and BIN1-dependent mechanism of Cav1.2 delivery to T-tubules. This novel Cav1.2 trafficking pathway should serve as an important regulatory aspect of EC coupling, affecting cardiac contractility in mammalian hearts.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/genetics
- Calcium Signaling/physiology
- Cell Line
- Cells, Cultured
- HeLa Cells
- Humans
- Immunohistochemistry
- Immunoprecipitation
- Male
- Mice
- Mice, Knockout
- Microscopy, Electron, Transmission
- Myocardial Contraction/genetics
- Myocardial Contraction/physiology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Myocytes, Cardiac/ultrastructure
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Sarcoplasmic Reticulum/metabolism
- Sarcoplasmic Reticulum/ultrastructure
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Ting-Ting Hong
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - James W. Smyth
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Danchen Gao
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Kevin Y. Chu
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Jacob M. Vogan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Tina S. Fong
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Brian C. Jensen
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Henry M. Colecraft
- Department of Physiology, Columbia University, New York, New York, United States of America
| | - Robin M. Shaw
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
36
|
Waite AL, Schaner P, Richards N, Balci-Peynircioglu B, Masters SL, Brydges SD, Fox M, Hong A, Yilmaz E, Kastner DL, Reinherz EL, Gumucio DL. Pyrin Modulates the Intracellular Distribution of PSTPIP1. PLoS One 2009; 4:e6147. [PMID: 19584923 PMCID: PMC2702820 DOI: 10.1371/journal.pone.0006147] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 06/03/2009] [Indexed: 01/23/2023] Open
Abstract
PSTPIP1 is a cytoskeleton-associated adaptor protein that links PEST-type phosphatases to their substrates. Mutations in PSTPIP1 cause PAPA syndrome (Pyogenic sterile Arthritis, Pyoderma gangrenosum, and Acne), an autoinflammatory disease. PSTPIP1 binds to pyrin and mutations in pyrin result in familial Mediterranean fever (FMF), a related autoinflammatory disorder. Since disease-associated mutations in PSTPIP1 enhance pyrin binding, PAPA syndrome and FMF are thought to share a common pathoetiology. The studies outlined here describe several new aspects of PSTPIP1 and pyrin biology. We document that PSTPIP1, which has homology to membrane-deforming BAR proteins, forms homodimers and generates membrane-associated filaments in native and transfected cells. An extended FCH (Fes-Cip4 homology) domain in PSTPIP1 is necessary and sufficient for its self-aggregation. We further show that the PSTPIP1 filament network is dependent upon an intact tubulin cytoskeleton and that the distribution of this network can be modulated by pyrin, indicating that this is a dynamic structure. Finally, we demonstrate that pyrin can recruit PSTPIP1 into aggregations (specks) of ASC, another pyrin binding protein. ASC specks are associated with inflammasome activity. PSTPIP1 molecules with PAPA-associated mutations are recruited by pyrin to ASC specks with particularly high efficiency, suggesting a unique mechanism underlying the robust inflammatory phenotype of PAPA syndrome.
Collapse
Affiliation(s)
- Andrea L. Waite
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Philip Schaner
- Division of Radiology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Neil Richards
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Seth L. Masters
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Susannah D. Brydges
- Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Disease, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Michelle Fox
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Arthur Hong
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Engin Yilmaz
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Daniel L. Kastner
- Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Disease, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Ellis L. Reinherz
- Harvard Medical School, Laboratory of Immunology, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Deborah L. Gumucio
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|