1
|
Sjekloća L, Buratti E. Conserved region of human TDP-43 is structurally similar to membrane binding protein FARP1 and protein chaperons BAG6 and CYP33. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001388. [PMID: 39583578 PMCID: PMC11582883 DOI: 10.17912/micropub.biology.001388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Transactive response DNA-binding protein of 43 KDa (TDP-43) is important for RNA metabolism in all animals and in humans is involved in neuromuscular diseases. Full-length TDP-43 is prone to oligomerization and misfolding what renders difficult its characterization. We report that TDP-43 domains are structurally similar to lipid binding protein FARP1 and protein chaperons BAG6 and CYP33. Sequence analysis suggests putative lipid binding sites throughout TDP-43 and in vitro thioflavin T fluorescence assays show that cholesterol and phosphatidylcholine affect fibrillation of recombinant TDP-43 fragments. Our findings suggest that TDP-43 can bind lipids directly and it may contribute to its own chaperoning.
Collapse
Affiliation(s)
- Ljiljana Sjekloća
- Molecular Pathology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Emanuele Buratti
- Molecular Pathology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
2
|
Afsar A, Zhang L. Putative Molecular Mechanisms Underpinning the Inverse Roles of Mitochondrial Respiration and Heme Function in Lung Cancer and Alzheimer's Disease. BIOLOGY 2024; 13:185. [PMID: 38534454 DOI: 10.3390/biology13030185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Mitochondria are the powerhouse of the cell. Mitochondria serve as the major source of oxidative stress. Impaired mitochondria produce less adenosine triphosphate (ATP) but generate more reactive oxygen species (ROS), which could be a major factor in the oxidative imbalance observed in Alzheimer's disease (AD). Well-balanced mitochondrial respiration is important for the proper functioning of cells and human health. Indeed, recent research has shown that elevated mitochondrial respiration underlies the development and therapy resistance of many types of cancer, whereas diminished mitochondrial respiration is linked to the pathogenesis of AD. Mitochondria govern several activities that are known to be changed in lung cancer, the largest cause of cancer-related mortality worldwide. Because of the significant dependence of lung cancer cells on mitochondrial respiration, numerous studies demonstrated that blocking mitochondrial activity is a potent strategy to treat lung cancer. Heme is a central factor in mitochondrial respiration/oxidative phosphorylation (OXPHOS), and its association with cancer is the subject of increased research in recent years. In neural cells, heme is a key component in mitochondrial respiration and the production of ATP. Here, we review the role of impaired heme metabolism in the etiology of AD. We discuss the numerous mitochondrial effects that may contribute to AD and cancer. In addition to emphasizing the significance of heme in the development of both AD and cancer, this review also identifies some possible biological connections between the development of the two diseases. This review explores shared biological mechanisms (Pin1, Wnt, and p53 signaling) in cancer and AD. In cancer, these mechanisms drive cell proliferation and tumorigenic functions, while in AD, they lead to cell death. Understanding these mechanisms may help advance treatments for both conditions. This review discusses precise information regarding common risk factors, such as aging, obesity, diabetes, and tobacco usage.
Collapse
Affiliation(s)
- Atefeh Afsar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
3
|
Neațu M, Covaliu A, Ioniță I, Jugurt A, Davidescu EI, Popescu BO. Monoclonal Antibody Therapy in Alzheimer's Disease. Pharmaceutics 2023; 16:60. [PMID: 38258071 PMCID: PMC11154277 DOI: 10.3390/pharmaceutics16010060] [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: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative condition marked by the progressive deterioration of cognitive abilities, memory impairment, and the accumulation of abnormal proteins, specifically beta-amyloid plaques and tau tangles, within the brain. Despite extensive research efforts, Alzheimer's disease remains without a cure, presenting a significant global healthcare challenge. Recently, there has been an increased focus on antibody-based treatments as a potentially effective method for dealing with Alzheimer's disease. This paper offers a comprehensive overview of the current status of research on antibody-based molecules as therapies for Alzheimer's disease. We will briefly mention their mechanisms of action, therapeutic efficacy, and safety profiles while addressing the challenges and limitations encountered during their development. We also highlight some crucial considerations in antibody-based treatment development, including patient selection criteria, dosing regimens, or safety concerns. In conclusion, antibody-based therapies present a hopeful outlook for addressing Alzheimer's disease. While challenges remain, the accumulating evidence suggests that these therapies may offer substantial promise in ameliorating or preventing the progression of this debilitating condition, thus potentially enhancing the quality of life for the millions of individuals and families affected by Alzheimer's disease worldwide.
Collapse
Affiliation(s)
- Monica Neațu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Anca Covaliu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Iulia Ioniță
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Ana Jugurt
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Eugenia Irene Davidescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.N.); (A.C.); (I.I.); (A.J.); (B.O.P.)
- Department of Neurology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Department of Cell Biology, Neurosciences and Experimental Myology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
4
|
Stallings NR, O'Neal MA, Hu J, Shen ZJ, Malter JS. Long-term normalization of calcineurin activity in model mice rescues Pin1 and attenuates Alzheimer's phenotypes without blocking peripheral T cell IL-2 response. Alzheimers Res Ther 2023; 15:179. [PMID: 37849016 PMCID: PMC10580561 DOI: 10.1186/s13195-023-01323-5] [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: 02/16/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Current treatments for Alzheimer's disease (AD) have largely failed to yield significant therapeutic benefits. Novel approaches are desperately needed to help address this immense public health issue. Data suggests that early intervention at the first stages of mild cognitive impairment may have a greater chance for success. The calcineurin (CN)-Pin1 signaling cascade can be selectively targeted with tacrolimus (FK506), a highly specific, FDA-approved CN inhibitor used safely for > 20 years in solid organ transplant recipients. AD prevalence was significantly reduced in solid organ recipients treated with FK506. METHODS Time release pellets were used to deliver constant FK506 dosage to APP/PS1 mice without deleterious manipulation or handling. Immunofluorescence, histology, molecular biology, and behavior were used to evaluate changes in AD pathology. RESULTS FK506 can be safely and consistently delivered into juvenile APP/PS1 mice via time-release pellets to levels roughly seen in transplant patients, leading to the normalization of CN activity and reduction or elimination of AD pathologies including synapse loss, neuroinflammation, and cognitive impairment. Pin1 activity and function were rescued despite the continuing presence of high levels of transgenic Aβ42. Indicators of neuroinflammation including Iba1 positivity and IL-6 production were also reduced to normal levels. Peripheral blood mononuclear cells (PBMC) obtained during treatment or splenocytes isolated at euthanasia activated normally after mitogens. CONCLUSIONS Low-dose, constant FK506 can normalize CNS CN and Pin1 activity, suppress neuroinflammation, and attenuate AD-associated pathology without blocking peripheral IL-2 responses making repurposed FK506 a viable option for early, therapeutic intervention in AD.
Collapse
Affiliation(s)
- Nancy R Stallings
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Melissa A O'Neal
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Jie Hu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines, Dallas, TX, 75390, USA.
| |
Collapse
|
5
|
Chakari-Khiavi F, Mirzaie A, Khalilzadeh B, Yousefi H, Abolhasan R, Kamrani A, Pourakbari R, Shahpasand K, Yousefi M, Rashidi MR. Application of Pt@ZIF-8 nanocomposite-based electrochemical biosensor for sensitive diagnosis of tau protein in Alzheimer's disease patients. Sci Rep 2023; 13:16163. [PMID: 37758805 PMCID: PMC10533502 DOI: 10.1038/s41598-023-43180-0] [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: 06/27/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive brain disorder characterized by the ongoing decline of brain functions. Studies have revealed the detrimental effects of hyperphosphorylated tau (p-tau) protein fibrils in AD pathogenesis, highlighting the importance of this factor in the early-stage detection of AD conditions. We designed an electrochemical immunosensor for quantitative detection of the cis conformation of the p-tau protein (cis-p-tau) employing platinum nanoparticles (Pt NPs) supported on zeolitic imidazolate frameworks (ZIF) for modifying the glassy carbon electrode (GCE) surface. Under optimum conditions, the immunosensor selectively and sensitively detected cis-p-tau within the broad linear range of 1 fg mL-1 to 10 ng mL-1 and the low limit of detection (LOD) of 1 fg mL-1 with desired reproducibility and stability. Furthermore, the fabricated immunosensor's performance was examined for the cis-p-tau analysis in the serum of AD patients, indicating its accuracy and feasibility for real-sample analysis. Notably, this is the first application of Pt@ZIF-8 nanocomposite in fabricating a valid immunosensor for selective cis-p-tau detection, even in the presence of trans-p-tau. It is worth mentioning that the enzyme-linked immunosorbent assay (ELISA) reference technique is not able to evaluate pico- or femtomolar concentrations of cis-p-tau, making the fabricated immunosensor superior for early-stage measurement and screening of AD.
Collapse
Affiliation(s)
- Forough Chakari-Khiavi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, PO Box: 6446-14155, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Mirzaie
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran.
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Rozita Abolhasan
- Department of Immunology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Kamrani
- Department of Immunology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Pourakbari
- Department of Immunology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, 1665659911, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran
| | - Mohammad-Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, PO Box: 6446-14155, Tabriz, Iran.
- Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Roqanian S, Ahmadian S, Nabavi SM, Pakdaman H, Shafiezadeh M, Goudarzi G, Shahpasand K. Tau nuclear translocation is a leading step in tau pathology process through P53 stabilization and nucleolar dispersion. J Neurosci Res 2022; 100:1084-1104. [PMID: 35170061 DOI: 10.1002/jnr.25024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022]
Abstract
Tau protein abnormalities are associated with various neurodegenerative disorders, including Alzheimer's disease (AD) and traumatic brain injury (TBI). In tau-overexpressing SHSY5Y cells and iPSC-derived neuron models of frontotemporal dementia (FTD), axonal tau translocates into the nuclear compartment, resulting in neuronal dysfunction. Despite extensive research, the mechanisms by which tau translocation results in neurodegeneration remain elusive thus far. We studied the nuclear displacement of different P-tau species [Cis phosphorylated Thr231-tau (cis P-tau), phosphorylated Ser202/Thr205-tau (AT8 P-tau), and phosphorylated Thr212/Ser214-tau (AT100 P-tau)] at various time points using starvation in primary cortical neurons and single severe TBI (ssTBI) in male mouse cerebral cortices as tauopathy models. While all P-tau species translocated into the somatodendritic compartment in response to stress, cis P-tau did so more rapidly than the other species. Notably, nuclear localization of P-tau was associated with p53 apoptotic stabilization and nucleolar stress, both of which resulted in neurodegeneration. In summary, our findings indicate that P-tau nuclear translocation results in p53-dependent apoptosis and nucleolar dispersion, which is consistent with neurodegeneration.
Collapse
Affiliation(s)
- Shaqayeq Roqanian
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahin Ahmadian
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Seyed Masood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Brain Mapping Research Center, Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Shafiezadeh
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ghazaleh Goudarzi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
7
|
Wei Y, Liu M, Wang D. The propagation mechanisms of extracellular tau in Alzheimer's disease. J Neurol 2021; 269:1164-1181. [PMID: 33913022 DOI: 10.1007/s00415-021-10573-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 01/07/2023]
Abstract
Tubulin-associated unit (tau) is an important microtubule-associated protein. The abnormal intracellular aggregation of tau has been strongly associated with Alzheimer's disease (AD). Accumulating evidence has conclusively demonstrated that tau is present in the cytoplasm of neurons and is also actively released into the extracellular space. However, the types of tau species that are released are unclear, as is the mechanism of their release by donor neurons and subsequent uptake by recipient neurons in AD. Understanding the underlying mechanisms of abnormal tau cell-to-cell transmission can provide novel insights into the etiology and pathogenesis of AD and can help identify new targets for the development of AD therapies focused on counteracting neurodegeneration or even preventing it. From this perspective, the present review focuses on recent advances in understanding the mechanisms regulating the levels of extracellular tau and discusses the role of such mechanisms in the propagation of tau-associated pathology.
Collapse
Affiliation(s)
- Yun Wei
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing city, 100091, China.
| | - Meixia Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing city, 100091, China
| | - Dongxin Wang
- Jining Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shandong province, 272000, China
| |
Collapse
|
8
|
Nakhjiri E, Vafaee MS, Hojjati SMM, Shahabi P, Shahpasand K. Tau Pathology Triggered by Spinal Cord Injury Can Play a Critical Role in the Neurotrauma Development. Mol Neurobiol 2020; 57:4845-4855. [PMID: 32808121 DOI: 10.1007/s12035-020-02061-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Traumatic spinal cord injury (SCI) can result in substantial neurological impairment along with significant emotional and psychological distress. It is clear that there is profound neurodegeneration upon SCI, gradually spread to other spinal cord regions and brain areas. Despite extensive considerations, it remains uncertain how pathogenicity diffuses in the cord. It has been reported that tau protein abnormal hyperphosphorylation plays a central role in neurodegeneration triggered by traumatic brain injury (TBI). Tau is a microtubule-associated protein, heavily implicated in neurodegenerative diseases. Importantly, tau pathology spreads in a traumatic brain in a timely manner. In particular, we have recently demonstrated that phosphorylated tau at Thr231 exists in two distinct cis and trans conformations, in which that cis P-tau is extremely neurotoxic, has a prion nature, and spreads to various brain areas and cerebrospinal fluid (CSF) upon trauma. On the other hand, tau pathology, in particular hyperphosphorylation at Thr231, has been observed upon SCI. Taken these together, we conclude that cis pT231-tau may accumulate and spread in the spinal cord as well as CSF and diffuse tau pathology in the central nervous system (CNS). Moreover, antibody against cis P-tau can target intracellular cis P-tau and protect pathology spreading. Thus, considering cis P-tau as a driver of tau pathology and neurodegeneration upon SCI would open new windows toward understanding the disease development and early biomarkers. Furthermore, it would help us develop effective therapies for SCI patients.
Collapse
Affiliation(s)
- Elnaz Nakhjiri
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manuchehr S Vafaee
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | | | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Makinwa Y, Musich PR, Zou Y. Phosphorylation-Dependent Pin1 Isomerization of ATR: Its Role in Regulating ATR's Anti-apoptotic Function at Mitochondria, and the Implications in Cancer. Front Cell Dev Biol 2020; 8:281. [PMID: 32426354 PMCID: PMC7203486 DOI: 10.3389/fcell.2020.00281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/31/2020] [Indexed: 12/11/2022] Open
Abstract
Peptidyl-prolyl isomerization is an important post-translational modification of protein because proline is the only amino acid that can stably exist as cis and trans, while other amino acids are in the trans conformation in protein backbones. This makes prolyl isomerization a unique mechanism for cells to control many cellular processes. Isomerization is a rate-limiting process that requires a peptidyl-prolyl cis/trans isomerase (PPIase) to overcome the energy barrier between cis and trans isomeric forms. Pin1, a key PPIase in the cell, recognizes a phosphorylated Ser/Thr-Pro motif to catalyze peptidyl-prolyl isomerization in proteins. The significance of the phosphorylation-dependent Pin1 activity was recently highlighted for isomerization of ATR (ataxia telangiectasia- and Rad3-related). ATR, a PIKK protein kinase, plays a crucial role in DNA damage responses (DDR) by phosphorylating hundreds of proteins. ATR can form cis or trans isomers in the cytoplasm depending on Pin1 which isomerizes cis-ATR to trans-ATR. Trans-ATR functions primarily in the nucleus. The cis-ATR, containing an exposed BH3 domain, is anti-apoptotic at mitochondria by binding to tBid, preventing activation of pro-apoptotic Bax. Given the roles of apoptosis in many human diseases, particularly cancer, we propose that cytoplasmic cis-ATR enables cells to evade apoptosis, thus addicting cancer cells to cis-ATR formation for survival. But in normal DDR, a predominance of trans-ATR in the nucleus coordinates with a minimal level of cytoplasmic cis-ATR to promote DNA repair while preventing cell death; however, cells can die when DNA repair fails. Therefore, a delicate balance/equilibrium of the levels of cis- and trans-ATR is required to ensure the cellular homeostasis. In this review, we make a case that this anti-apoptotic role of cis-ATR supports oncogenesis, while Pin1 that drives the formation of trans-ATR suppresses tumor growth. We offer a potential, novel target that can be specifically targeted in cancer cells, without killing normal cells, to significantly reduce the adverse effects usually seen in cancer treatment. We also raise important issues regarding the roles of phosphorylation-dependent Pin1 isomerization of ATR in diseases and propose areas of future studies that would shed more understanding on this important cellular mechanism.
Collapse
Affiliation(s)
- Yetunde Makinwa
- Department of Cancer Biology, University of Toledo College of Medicine, Toledo, OH, United States
| | - Phillip R Musich
- Department of Biomedical Sciences, JH Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yue Zou
- Department of Cancer Biology, University of Toledo College of Medicine, Toledo, OH, United States.,Department of Biomedical Sciences, JH Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
10
|
Hadi F, Akrami H, Shahpasand K, Fattahi MR. Wnt signalling pathway and tau phosphorylation: A comprehensive study on known connections. Cell Biochem Funct 2020; 38:686-694. [PMID: 32232872 DOI: 10.1002/cbf.3530] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/01/2020] [Accepted: 03/13/2020] [Indexed: 12/31/2022]
Abstract
The Wnt pathway is the most important cascade in the nervous system; evidence has indicated that deregulation of the Wnt pathway induced pathogenic hallmarks of neurodegenerative diseases. Glycogen synthase kinase-3β (GSK-3β) as the main member of the Wnt pathway increases tau inclusions, the main marker in the neurodegenerative diseases. Phosphorylated tau is observed in the pre-tangle of the neurons in the early stage of neurodegenerative diseases. The researchers always try to improve pharmacological approaches of new therapeutic strategies to the treatment of neurodegenerative diseases that are required to represent a significant entry point by understanding the theoretical interactions of the molecular pathways. In this review, we have discussed the recent knowledge about the canonical and non-canonical Wnt signalling pathway, GSK-3β, Wnt/β-catenin antagonists, tau phosphorylation, and their important roles in the neurodegenerative diseases.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Hassan Akrami
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad R Fattahi
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Chen D, Zhou XZ, Lee TH. Death-Associated Protein Kinase 1 as a Promising Drug Target in Cancer and Alzheimer's Disease. Recent Pat Anticancer Drug Discov 2020; 14:144-157. [PMID: 30569876 PMCID: PMC6751350 DOI: 10.2174/1574892814666181218170257] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/23/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023]
Abstract
Background: Death-Associated Protein Kinase 1 (DAPK1) plays an important role in apopto-sis, tumor suppression and neurodegeneration including Alzheimer’s Disease (AD). Objective: This review will describe the diverse roles of DAPK1 in the development of cancer and AD, and the current status of drug development targeting DAPK1-based therapies. Methods: Reports of DAPK1 regulation, function and substrates were analyzed using genetic DAPK1 manipulation and chemical DAPK1 modulators. Results: DAPK1 expression and activity are deregulated in cancer and AD. It is down-regulated and/or inactivated by multiple mechanisms in many human cancers, and elicits a protective effect to counteract numerous death stimuli in cancer, including activation of the master regulator Pin1. Moreover, loss of DAPK1 expression has correlated strongly with tumor recurrence and metastasis, suggesting that lack of sufficient functional DAPK1 might contribute to cancer. In contrast, DAPK1 is highly expressed in the brains of most human AD patients and has been identified as one of the genetic factors affecting suscepti-bility to late-onset AD. The absence of DAPK1 promotes efficient learning and better memory in mice and prevents the development of AD by acting on many key proteins including Pin1 and its downstream tar-gets tau and APP. Recent patents show that DAPK1 modulation might be used to treat both cancer and AD. Conclusion: DAPK1 plays a critical role in diverse physiological processes and importantly, its deregula-tion is implicated in the pathogenesis of either cancer or AD. Therefore, manipulating DAPK1 activity and/or expression may be a promising therapeutic option for cancer or AD.
Collapse
Affiliation(s)
- Dongmei Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Xiao Z Zhou
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Tae H Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| |
Collapse
|
12
|
Bittar A, Bhatt N, Kayed R. Advances and considerations in AD tau-targeted immunotherapy. Neurobiol Dis 2019; 134:104707. [PMID: 31841678 PMCID: PMC6980703 DOI: 10.1016/j.nbd.2019.104707] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/27/2019] [Accepted: 12/08/2019] [Indexed: 12/18/2022] Open
Abstract
The multifactorial and complex nature of Alzheimer’s disease (AD) has made it difficult to identify therapeutic targets that are causally involved in the disease process. However, accumulating evidence from experimental and clinical studies that investigate the early disease process point towards the required role of tau in AD etiology. Importantly, a large number of studies investigate and characterize the plethora of pathological forms of tau protein involved in disease onset and propagation. Immunotherapy is one of the most clinical approaches anticipated to make a difference in the field of AD therapeutics. Tau –targeted immunotherapy is the new direction after the failure of amyloid beta (Aß)-targeted immunotherapy and the growing number of studies that highlight the Aß-independent disease process. It is now well established that immunotherapy alone will most likely be insufficient as a monotherapy. Therefore, this review discusses updates on tau-targeted immunotherapy studies, AD-relevant tau species, updates on promising biomarkers and a prospect on combination therapies to surround the disease propagation in an efficient and timely manner.
Collapse
Affiliation(s)
- Alice Bittar
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| | - Nemil Bhatt
- Department of Neuroscience, Cell Biology and Anatomy, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| | - Rakez Kayed
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| |
Collapse
|
13
|
Baquero J, Varriano S, Ordonez M, Kuczaj P, Murphy MR, Aruggoda G, Lundine D, Morozova V, Makki AE, Alonso ADC, Kleiman FE. Nuclear Tau, p53 and Pin1 Regulate PARN-Mediated Deadenylation and Gene Expression. Front Mol Neurosci 2019; 12:242. [PMID: 31749682 PMCID: PMC6843027 DOI: 10.3389/fnmol.2019.00242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
While nuclear tau plays a role in DNA damage response (DDR) and chromosome relaxation, the mechanisms behind these functions are not fully understood. Here, we show that tau forms complex(es) with factors involved in nuclear mRNA processing such as tumor suppressor p53 and poly(A)-specific ribonuclease (PARN) deadenylase. Tau induces PARN activity in different cellular models during DDR, and this activation is further increased by p53 and inhibited by tau phosphorylation at residues implicated in neurological disorders. Tau's binding factor Pin1, a mitotic regulator overexpressed in cancer and depleted in Alzheimer's disease (AD), also plays a role in the activation of nuclear deadenylation. Tau, Pin1 and PARN target the expression of mRNAs deregulated in AD and/or cancer. Our findings identify novel biological roles of tau and toxic effects of hyperphosphorylated-tau. We propose a model in which factors involved in cancer and AD regulate gene expression by interactions with the mRNA processing machinery, affecting the transcriptome and suggesting insights into alternative mechanisms for the initiation and/or developments of these diseases.
Collapse
Affiliation(s)
- Jorge Baquero
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Sophia Varriano
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Martha Ordonez
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Pawel Kuczaj
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Michael R. Murphy
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Gamage Aruggoda
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Devon Lundine
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Viktoriya Morozova
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Ali Elhadi Makki
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Alejandra del C. Alonso
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, Graduate Center, The City University of New York, Staten Island, NY, United States
| | - Frida E. Kleiman
- Chemistry Department, Hunter College and Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| |
Collapse
|
14
|
Tapia-Rojas C, Cabezas-Opazo F, Deaton CA, Vergara EH, Johnson GVW, Quintanilla RA. It's all about tau. Prog Neurobiol 2018; 175:54-76. [PMID: 30605723 DOI: 10.1016/j.pneurobio.2018.12.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 12/07/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
Abstract
Tau is a protein that is highly enriched in neurons and was originally defined by its ability to bind and stabilize microtubules. However, it is now becoming evident that the functions of tau extend beyond its ability to modulate microtubule dynamics. Tau plays a role in mediating axonal transport, synaptic structure and function, and neuronal signaling pathways. Although tau plays important physiological roles in neurons, its involvement in neurodegenerative diseases, and most prominently in the pathogenesis of Alzheimer disease (AD), has directed the majority of tau studies. However, a thorough knowledge of the physiological functions of tau and its post-translational modifications under normal conditions are necessary to provide the foundation for understanding its role in pathological settings. In this review, we will focus on human tau, summarizing tau structure and organization, as well as its posttranslational modifications associated with physiological processes. We will highlight possible mechanisms involved in mediating the turnover of tau and finally discuss newly elucidated tau functions in a physiological context.
Collapse
Affiliation(s)
- Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Santiago, Chile
| | - Fabian Cabezas-Opazo
- Laboratory of Neurodegenerative Diseases, Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile
| | - Carol A Deaton
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, NY, USA
| | - Erick H Vergara
- Laboratory of Neurodegenerative Diseases, Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, NY, USA
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile; Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIIA), Santiago, Chile.
| |
Collapse
|
15
|
Clark IA, Vissel B. Therapeutic implications of how TNF links apolipoprotein E, phosphorylated tau, α-synuclein, amyloid-β and insulin resistance in neurodegenerative diseases. Br J Pharmacol 2018; 175:3859-3875. [PMID: 30097997 PMCID: PMC6151331 DOI: 10.1111/bph.14471] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 12/24/2022] Open
Abstract
While cytokines such as TNF have long been recognized as essential to normal cerebral physiology, the implications of their chronic excessive production within the brain are now also increasingly appreciated. Syndromes as diverse as malaria and lead poisoning, as well as non‐infectious neurodegenerative diseases, illustrate this. These cytokines also orchestrate changes in tau, α‐synuclein, amyloid‐β levels and degree of insulin resistance in most neurodegenerative states. New data on the effects of salbutamol, an indirect anti‐TNF agent, on α‐synuclein and Parkinson's disease, APOE4 and tau add considerably to the rationale of the anti‐TNF approach to understanding, and treating, these diseases. Therapeutic advances being tested, and arguably useful for a number of the neurodegenerative diseases, include a reduction of excess cerebral TNF, whether directly, with a specific anti‐TNF biological agent such as etanercept via Batson's plexus, or indirectly via surgically implanting stem cells. Inhaled salbutamol also warrants investigating further across the neurodegenerative disease spectrum. It is now timely to integrate this range of new information across the neurodegenerative disease spectrum, rather than keep seeing it through the lens of individual disease states.
Collapse
Affiliation(s)
- I A Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, NSW, Australia.,St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
16
|
Albayram O, Angeli P, Bernstein E, Baxley S, Gao Z, Lu KP, Zhou XZ. Targeting Prion-like Cis Phosphorylated Tau Pathology in Neurodegenerative Diseases. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2018; 8:443. [PMID: 30197831 PMCID: PMC6122852 DOI: 10.4172/2161-0460.1000443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tau is a microtubule-associated protein heavily implicated in neurodegenerative diseases collectively known as tauopathies, including Alzheimer's disease and chronic traumatic encephalopathy. Phosphorylation of tau at Thr231 allows for the isomerization of phosphorylated tau (p-tau) into distinct cis and trans conformations. Cis, but not trans, p-tau is detectable not only in Alzheimer's disease and chronic traumatic encephalopathy, but also right after traumatic brain injury depending on injury severity and frequency both in humans and animal models. Cis p-tau is not only neurotoxic but also spreads from a neuron to another in a prion-like fashion, functioning as a primary driver of neurodegeneration, which can be effectively neutralized by cis p-tau antibody. This represents an exciting new opportunity for understanding disease development and developing early biomarkers and effective therapies of tauopathies.
Collapse
Affiliation(s)
- Onder Albayram
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| | - Peter Angeli
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| | - Elizabeth Bernstein
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| | - Sean Baxley
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| | - Ziang Gao
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine Fujian Medical University, Fuzhou, Fujian, China
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA 02215, USA
| |
Collapse
|
17
|
Moszczynski AJ, Strong W, Xu K, McKee A, Brown A, Strong MJ. Pathologic Thr 175 tau phosphorylation in CTE and CTE with ALS. Neurology 2018; 90:e380-e387. [PMID: 29298849 PMCID: PMC5791789 DOI: 10.1212/wnl.0000000000004899] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/22/2017] [Indexed: 12/14/2022] Open
Abstract
Objective To investigate whether chronic traumatic encephalopathy (CTE) and CTE with amyotrophic lateral sclerosis (CTE-ALS) exhibit features previously observed in other tauopathies of pathologic phosphorylation of microtubule-associated protein tau at Thr175 (pThr175 tau) and Thr231 (pThr231 tau), and glycogen synthase kinase–3β (GSK3β) activation, and whether these pathologic features are a consequence of traumatic brain injury (TBI). Methods Tau isoform expression was assayed by western blot in 6 stage III CTE cases. We also used immunohistochemistry to analyze 5 cases each of CTE, CTE-ALS, and 5 controls for expression of activated GSK3β, pThr175 tau, pThr231 tau, and oligomerized tau within spinal cord tissue and hippocampus. Using a rat model of moderate TBI, we assessed tau pathology and phospho-GSK3β expression at 3 months postinjury. Results CTE and CTE-ALS are characterized by the presence of all 6 tau isoforms in both soluble and insoluble tau isolates. Activated GSK3β, pThr175 tau, pThr231 tau, and oligomerized tau protein expression was observed in hippocampal neurons and spinal motor neurons. We observed tau neuronal pathology (fibrillar inclusions and axonal damage) and increased levels of pThr175 tau and activated GSK3β in moderate TBI rats. Conclusions Pathologic phosphorylation of tau at Thr175 and Thr231 and activation of GSK3β are features of the tauopathy of CTE and CTE-ALS. These features can be replicated in an animal model of moderate TBI.
Collapse
Affiliation(s)
- Alexander J Moszczynski
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Wendy Strong
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Kathy Xu
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Ann McKee
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Arthur Brown
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Michael J Strong
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA.
| |
Collapse
|
18
|
Shahpasand K, Sepehri Shamloo A, Nabavi SM, Ping Lu K, Zhen Zhou X. "Tau immunotherapy: Hopes and hindrances". Hum Vaccin Immunother 2017; 14:277-284. [PMID: 29049003 DOI: 10.1080/21645515.2017.1393594] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder having two major pathological hallmarks: the extracellular senile plaques and intracellular neurofibrillary tangles composed of amyloid beta protein and hyperphosphorylated tau respectively. Removal of protein deposits from AD brains are the newer attempts for treating AD. The major developments in this direction have been the amyloid and tau based therapeutics. While senile plaque removal employing monoclonal antibodies (mAbs) restore brain function in mouse models of AD, tau has been recently introduced as the major neurodegenerative factor mediating neural cell death. So, several research groups have focused on tau therapy. So far, the outcome of tau immunotherapy has been promising and clearance of hyperphosphorylated tau has been shown to restore the brain function in animal models. But the point is which phosphorylated tau is the most critical form to be removed from the brain, especially because removal of physiologic tau can cause neurodegenerative consequence. Recently, we have shown that phosphorylated tau at Thr231 in the cis conformation is a very early driver of neurodegeneration and cis mAb treatment efficiently restores brain structure and function in TBI models. Because of efficient therapeutic effects in mice model of TBI and considering cis pT231-tau accumulation in AD brains, it could be a very good candidate for tau immunotherapy upon several tauopathy disorders including AD.
Collapse
Affiliation(s)
- Koorosh Shahpasand
- a Department of Brain and Cognitive Sciences , Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Alireza Sepehri Shamloo
- a Department of Brain and Cognitive Sciences , Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Seyed Massood Nabavi
- a Department of Brain and Cognitive Sciences , Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Kun Ping Lu
- b Division of Translational Therapeutics, Department of Medicine , Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA.,c Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| | - Xiao Zhen Zhou
- b Division of Translational Therapeutics, Department of Medicine , Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA.,c Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
19
|
Ojo JO, Mouzon B, Algamal M, Leary P, Lynch C, Abdullah L, Evans J, Mullan M, Bachmeier C, Stewart W, Crawford F. Chronic Repetitive Mild Traumatic Brain Injury Results in Reduced Cerebral Blood Flow, Axonal Injury, Gliosis, and Increased T-Tau and Tau Oligomers. J Neuropathol Exp Neurol 2016; 75:636-55. [PMID: 27251042 DOI: 10.1093/jnen/nlw035] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Indexed: 12/14/2022] Open
Abstract
Exposure to repetitive mild traumatic brain injury (mTBI) is a risk factor for chronic traumatic encephalopathy, which is characterized by patchy deposition of hyperphosphorylated tau aggregates in neurons and astrocytes at the depths of cortical sulci. We developed an mTBI paradigm to explore effects of repetitive concussive-type injury over several months in mice with a human tau genetic background (hTau). Two injuries were induced in the hTau mice weekly over a period of 3 or 4 months and the effects were compared with those in noninjured sham animals. Behavioral and in vivo measures and detailed neuropathological assessments were conducted 6 months after the first injury. Our data confirm impairment in cerebral blood flow and white matter damage. This was accompanied by a 2-fold increase in total tau levels and mild increases in tau oligomers/conformers and pTau (Thr231) species in brain gray matter. There was no evidence of neurofibrillary/astroglial tangles, neuropil threads, or perivascular foci of tau immunoreactivity. There were neurobehavioral deficits (ie, disinhibition and impaired cognitive performance) in the mTBI animals. These data support the relevance of this new mTBI injury model for studying the consequences of chronic repetitive mTBI in humans, and the role of tau in TBI.
Collapse
Affiliation(s)
- Joseph O Ojo
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS).
| | - Benoit Mouzon
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Moustafa Algamal
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Paige Leary
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Cillian Lynch
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Laila Abdullah
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - James Evans
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Michael Mullan
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Corbin Bachmeier
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - William Stewart
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| | - Fiona Crawford
- From the Roskamp Institute, Sarasota, Florida (JOO, BM, MA, PL, CL, LA, JE, MM, CB, FC); James A. Haley Veterans' Hospital, Tampa, Florida (BM, LA, CB, FC); Open University, Milton Keynes, UK (BM, MA, CL, CB, FC); Bay Pines VA Healthcare System, Bay Pines, Florida (CB); Queen Elizabeth University Hospital, Glasgow, UK (WS); University of Glasgow, Glasgow, UK (WS); and University of Pennsylvania, Philadelphia, Pennsylvania (WS)
| |
Collapse
|
20
|
Pan J, Connolly ID, Dangelmajer S, Kintzing J, Ho AL, Grant G. Sports-related brain injuries: connecting pathology to diagnosis. Neurosurg Focus 2016; 40:E14. [DOI: 10.3171/2016.1.focus15607] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain injuries are becoming increasingly common in athletes and represent an important diagnostic challenge. Early detection and management of brain injuries in sports are of utmost importance in preventing chronic neurological and psychiatric decline. These types of injuries incurred during sports are referred to as mild traumatic brain injuries, which represent a heterogeneous spectrum of disease. The most dramatic manifestation of chronic mild traumatic brain injuries is termed chronic traumatic encephalopathy, which is associated with profound neuropsychiatric deficits. Because chronic traumatic encephalopathy can only be diagnosed by postmortem examination, new diagnostic methodologies are needed for early detection and amelioration of disease burden. This review examines the pathology driving changes in athletes participating in high-impact sports and how this understanding can lead to innovations in neuroimaging and biomarker discovery.
Collapse
Affiliation(s)
| | | | | | - James Kintzing
- 3Bioengineering, Stanford University School of Medicine, Stanford, California
| | | | | |
Collapse
|
21
|
Khan SS, Bloom GS. Tau: The Center of a Signaling Nexus in Alzheimer's Disease. Front Neurosci 2016; 10:31. [PMID: 26903798 PMCID: PMC4746348 DOI: 10.3389/fnins.2016.00031] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/25/2016] [Indexed: 12/04/2022] Open
Abstract
Tau is a microtubule-associated protein whose misfolding, hyper-phosphorylation, loss of normal function and toxic gain of function are linked to several neurodegenerative disorders, including Alzheimer's disease (AD). This review discusses the role of tau in amyloid-β (Aβ) induced toxicity in AD. The consequences of tau dysfunction, starting from the axon and concluding at somadendritic compartments, will be highlighted.
Collapse
Affiliation(s)
- Shahzad S Khan
- Department of Biology, University of Virginia Charlottesville, VA, USA
| | - George S Bloom
- Department of Biology, University of Virginia Charlottesville, VA, USA
| |
Collapse
|
22
|
Meehan W, Mannix R, Zafonte R, Pascual-Leone A. Chronic traumatic encephalopathy and athletes. Neurology 2015; 85:1504-11. [PMID: 26253448 DOI: 10.1212/wnl.0000000000001893] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/20/2015] [Indexed: 12/13/2022] Open
Abstract
Recent case reports have described athletes previously exposed to repetitive head trauma while participating in contact sports who later in life developed mood disorders, headaches, cognitive difficulties, suicidal ideation, difficulties with speech, and aggressive behavior. Postmortem discoveries show that some of these athletes have pathologic findings that are collectively termed chronic traumatic encephalopathy (CTE). Current hypotheses suggest that concussions or perhaps blows to the head that do not cause the signs and symptoms necessary for making the diagnosis of concussion, so-called subconcussive blows, cause both the clinical and pathologic findings. There are, however, some athletes who participate in contact sports who do not develop the findings ascribed to CTE. Furthermore, there are people who have headaches, mood disorders, cognitive difficulties, suicidal ideation, and other clinical problems who have neither been exposed to repeated head trauma nor possessed the pathologic postmortem findings of those currently diagnosed with CTE. The current lack of prospective data and properly designed case-control studies limits the current understanding of CTE, leading to debate about the causes of the neuropathologic findings and the clinical observations. Given the potential for referral and recall bias in available studies, it remains unclear whether or not the pathologic findings made postmortem cause the presumed neurobehavioral sequela and whether the presumed risk factors, such as sports activity, cerebral concussions, and subconcussive blows, are solely causative of the clinical signs and symptoms. This article discusses the current evidence and the associated limitations.
Collapse
Affiliation(s)
- William Meehan
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA.
| | - Rebekah Mannix
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Ross Zafonte
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Alvaro Pascual-Leone
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
23
|
Pedersen JT, Sigurdsson EM. Tau immunotherapy for Alzheimer's disease. Trends Mol Med 2015; 21:394-402. [DOI: 10.1016/j.molmed.2015.03.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 11/27/2022]
|
24
|
Blair LJ, Baker JD, Sabbagh JJ, Dickey CA. The emerging role of peptidyl-prolyl isomerase chaperones in tau oligomerization, amyloid processing, and Alzheimer's disease. J Neurochem 2015; 133:1-13. [PMID: 25628064 DOI: 10.1111/jnc.13033] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/12/2014] [Accepted: 01/05/2015] [Indexed: 12/20/2022]
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases), a unique family of molecular chaperones, regulate protein folding at proline residues. These residues are abundant within intrinsically disordered proteins, like the microtubule-associated protein tau. Tau has been shown to become hyperphosphorylated and accumulate as one of the two main pathological hallmarks in Alzheimer's disease, the other being amyloid beta (Ab). PPIases, including Pin1, FK506-binding protein (FKBP) 52, FKBP51, and FKBP12, have been shown to interact with and regulate tau biology. This interaction is particularly important given the numerous proline-directed phosphorylation sites found on tau and the role phosphorylation has been found to play in pathogenesis. This regulation then affects downstream aggregation and oligomerization of tau. However, many PPIases have yet to be explored for their effects on tau biology, despite the high likelihood of interaction based on proline content. Moreover, Pin1, FKBP12, FKBP52, cyclophilin (Cyp) A, CypB, and CypD have been shown to also regulate Ab production or the toxicity associated with Ab pathology. Therefore, PPIases directly and indirectly regulate pathogenic protein multimerization in Alzheimer's disease and represent a family rich in targets for modulating the accumulation and toxicity.
Collapse
Affiliation(s)
- Laura J Blair
- Department of Molecular Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida, USA
| | | | | | | |
Collapse
|
25
|
Wang JZ, Zhang Y. Configuration-specific immunotherapy targeting cis pThr231-Pro232 tau for Alzheimer disease. J Neurol Sci 2014; 348:253-5. [PMID: 25467137 DOI: 10.1016/j.jns.2014.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 11/18/2022]
Abstract
Tau pathology is the main pathological characteristic of mild cognitive impairment (MCI) and Alzheimer disease (AD), and tau-based therapeutic strategies have great implications in the prevention of MCI and AD. The phosphorylation of threonine 231 preceding proline 232 (pThr231-Pro232) triggers tau hyperphosphorylation, tau aggregation, and tau pathology. Interestingly, the pThr231-Pro232 motif may be in a cis or trans configuration, but several recent studies have firstly indicated that cis, but not trans, pThr231-Pro232 tau is a striking therapeutic target for MCI and AD. Cis pThr231-Pro232 tau appears firstly in MCI and accumulates exclusively in the development of AD. Moreover, cis pThr231-Pro232 tau has low affinity to microtubules, high resistance to dephosphorylation and degradation, and a potent tendency to aggregate. On the contrary, trans pThr231-Pro232 tau has normal physiological activity in vivo. Fortunately, Pin1 is the only known isomerase that catalyzes pThr231-Pro232 tau from the neurotoxic cis to nontoxic trans configuration, which prevents MCI and AD. Nonetheless, as we have mentioned before, Pin1 is frequently inactivated under abnormal physiological conditions in vivo. Therefore, it is necessary to clear cis pThr231-Pro232 tau by immunotherapy when Pin1 is insufficient, in order to avoid the occurrence of MCI and AD.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan, China.
| | - Yong Zhang
- Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan, China
| |
Collapse
|
26
|
Moore JD, Potter A. Pin1 inhibitors: Pitfalls, progress and cellular pharmacology. Bioorg Med Chem Lett 2013; 23:4283-91. [PMID: 23796453 DOI: 10.1016/j.bmcl.2013.05.088] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/24/2013] [Accepted: 05/28/2013] [Indexed: 01/12/2023]
Abstract
Compelling data supports the hypothesis that Pin1 inhibitors will be useful for the therapy of cancer: Pin1 deficient mice resist the induction of breast cancers normally evoked by expression of MMTV-driven Ras or Erb2 alleles. While Pin1 poses challenges for drug discovery, several groups have identified potent antagonists by structure based drug design, significant progress has been made designing peptidic inhibitors and a number of natural products have been found that blockade Pin1, notably epigallocatchechin gallate (EGCG), a major flavonoid in green tea. Here we critically discuss the modes of action and likely specificity of these compounds, concluding that a suitable chemical biology tool for probing the function of Pin1 has yet to be found. We conclude by outlining some open questions regarding the target validation of Pin1 and the prospects for identification of improved inhibitors in the future.
Collapse
Affiliation(s)
- Jonathan D Moore
- Horizon Discovery, Cambridge Research Park, Cambridge CB25 9TL, UK.
| | | |
Collapse
|