1
|
Ansari MM, Sahu SK, Singh TG, Singh SRJ, Kaur P. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. Eur J Pharmacol 2024; 979:176816. [PMID: 39038637 DOI: 10.1016/j.ejphar.2024.176816] [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: 03/23/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.
Collapse
Affiliation(s)
- Md Mustafiz Ansari
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | | | - Sovia R J Singh
- University Language Centre- Chitkara Business School, Chitkara University, Punjab, India
| | - Paranjeet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
2
|
Chakraborty J, Chakraborty S, Chakraborty S, Narayan MN. Entanglement of MAPK pathways with gene expression and its omnipresence in the etiology for cancer and neurodegenerative disorders. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194988. [PMID: 37739217 DOI: 10.1016/j.bbagrm.2023.194988] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Mitogen Activated Protein Kinase (MAPK) is one of the most well characterized cellular signaling pathways that controls fundamental cellular processes including proliferation, differentiation, and apoptosis. These cellular functions are consequences of transcription of regulatory genes that are influenced and regulated by the MAP-Kinase signaling cascade. MAP kinase components such as Receptor Tyrosine Kinases (RTKs) sense external cues or ligands and transmit these signals via multiple protein complexes such as RAS-RAF, MEK, and ERKs and eventually modulate the transcription factors inside the nucleus to induce transcription and other regulatory functions. Aberrant activation, dysregulation of this signaling pathway, and genetic alterations in any of these components results in the developmental disorders, cancer, and neurodegenerative disorders. Over the years, the MAPK pathway has been a prime pharmacological target, to treat complex human disorders that are genetically linked such as cancer, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. The current review re-visits the mechanism of MAPK pathways in gene expression regulation. Further, a current update on the progress of the mechanistic understanding of MAPK components is discussed from a disease perspective.
Collapse
Affiliation(s)
- Joydeep Chakraborty
- Institute for Advancing Health through Agriculture, Texas A&M Agrilife, College Station, TX, USA
| | - Sayan Chakraborty
- Department of Anesthesiology, Weill Cornell School of Medicine, New York, USA
| | - Sohag Chakraborty
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, USA
| | - Mahesh N Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
3
|
Singh R, Kaur N, Dhingra N, Kaur T. Protein misfolding, ER Stress and Chaperones: An approach to develop chaperone-based therapeutics for Alzheimer's Disease. Int J Neurosci 2021:1-21. [PMID: 34402740 DOI: 10.1080/00207454.2021.1968859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disorder with complex etiology that eventually leads to dementia. The main culprit of AD is the extracellular deposition of β-amyloid (Aβ) and intracellular neurofibrillary tangles. The protein conformational change and protein misfolding are the key events of AD pathophysiology, therefore endoplasmic reticulum (ER) stress is an apparent consequence. ER, stress-induced unfolded protein response (UPR) mediators (viz. PERK, IRE1, and ATF6) have been reported widely in the AD brain. Considering these factors, preventing proteins misfolding or aggregation of tau or amyloidogenic proteins appears to be the best approach to halt its pathogenesis. Therefore, therapies through chemical and pharmacological chaperones came to light as an alternative for the treatment of AD. Diverse studies have demonstrated 4-phenylbutyric acid (4-PBA) as a potential therapeutic agent in AD. The current review outlined the mechanism of protein misfolding, different etiological features behind the progression of AD, the significance of ER stress in AD, and the potential therapeutic role of different chaperones to counter AD. The study also highlights the gaps in current knowledge of the chaperones-based therapeutic approach and the possibility of developing chaperones as a potential therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Rimaljot Singh
- Department of Biophysics, Panjab University Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| |
Collapse
|
4
|
Hepp Rehfeldt SC, Majolo F, Goettert MI, Laufer S. c-Jun N-Terminal Kinase Inhibitors as Potential Leads for New Therapeutics for Alzheimer's Diseases. Int J Mol Sci 2020; 21:E9677. [PMID: 33352989 PMCID: PMC7765872 DOI: 10.3390/ijms21249677] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's Disease (AD) is becoming more prevalent as the population lives longer. For individuals over 60 years of age, the prevalence of AD is estimated at 40.19% across the world. Regarding the cognitive decline caused by the disease, mitogen-activated protein kinases (MAPK) pathways such as the c-Jun N-terminal kinase (JNK) pathway are involved in the progressive loss of neurons and synapses, brain atrophy, and augmentation of the brain ventricles, being activated by synaptic dysfunction, oxidative stress, and excitotoxicity. Nowadays, AD symptoms are manageable, but the disease itself remains incurable, thus the inhibition of JNK3 has been explored as a possible therapeutic target, considering that JNK is best known for its involvement in propagating pro-apoptotic signals. This review aims to present biological aspects of JNK, focusing on JNK3 and how it relates to AD. It was also explored the recent development of inhibitors that could be used in AD treatment since several drugs/compounds in phase III clinical trials failed. General aspects of the MAPK family, therapeutic targets, and experimental treatment in models are described and discussed throughout this review.
Collapse
Affiliation(s)
- Stephanie Cristine Hepp Rehfeldt
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Fernanda Majolo
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre CEP 90619-900, Rio Grande do Sul, Brazil
| | - Márcia Inês Goettert
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Stefan Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Faculty of Sciences, University of Tuebingen, D-72076 Tuebingen, Germany
| |
Collapse
|
5
|
Tripathy D, Migazzi A, Costa F, Roncador A, Gatto P, Fusco F, Boeri L, Albani D, Juárez-Hernández JL, Musio C, Colombo L, Salmona M, Wilhelmus MMM, Drukarch B, Pennuto M, Basso M. Increased transcription of transglutaminase 1 mediates neuronal death in in vitro models of neuronal stress and Aβ1-42-mediated toxicity. Neurobiol Dis 2020; 140:104849. [PMID: 32222473 DOI: 10.1016/j.nbd.2020.104849] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/01/2020] [Accepted: 03/24/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. At the pre-symptomatic phase of the disease, the processing of the amyloid precursor protein (APP) produces toxic peptides, called amyloid-β 1-42 (Aβ 1-42). The downstream effects of Aβ 1-42 production are not completely uncovered. Here, we report the involvement of transglutaminase 1 (TG1) in in vitro AD models of neuronal toxicity. TG1 was increased at late stages of the disease in the hippocampus of a mouse model of AD and in primary cortical neurons undergoing stress. Silencing of TGM1 gene was sufficient to prevent Aβ-mediated neuronal death. Conversely, its overexpression enhanced cell death. TGM1 upregulation was mediated at the transcriptional level by an activator protein 1 (AP1) binding site that when mutated halted TGM1 promoter activation. These results indicate that TG1 acts downstream of Aβ-toxicity, and that its stress-dependent increase makes it suitable for pharmacological intervention.
Collapse
Affiliation(s)
- Debasmita Tripathy
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy
| | - Alice Migazzi
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy
| | - Federica Costa
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy
| | - Alessandro Roncador
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy
| | - Pamela Gatto
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy
| | - Federica Fusco
- Department of Neuroscience, Laboratory of Genetics of Neurodegenerative Disorders, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Laboratory of Genetics of Neurodegenerative Disorders, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - J Leon Juárez-Hernández
- Institute of Biophysics, Trento Unit, National Research Council (IBF-CNR), Bruno Kessler Foundation (FBK), LabSSAH, Via alla Cascata 56/C, 38123 Trento, Italy
| | - Carlo Musio
- Institute of Biophysics, Trento Unit, National Research Council (IBF-CNR), Bruno Kessler Foundation (FBK), LabSSAH, Via alla Cascata 56/C, 38123 Trento, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Laboratory of Biochemistry and Protein Chemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Laboratory of Biochemistry and Protein Chemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - M M Micha Wilhelmus
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, the Netherlands
| | - Benjamin Drukarch
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, the Netherlands
| | - Maria Pennuto
- Dulbecco Telethon Institute Lab of Neurodegenerative Diseases, Centre for Integrative Biology (CIBIO), University of Trento, Italy; Department of Biomedical sciences, via Ugo Bassi 58/B, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, 35100 Padova, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, TN, Italy.
| |
Collapse
|
6
|
Colombo A, Hsia HE, Wang M, Kuhn PH, Brill MS, Canevazzi P, Feederle R, Taveggia C, Misgeld T, Lichtenthaler SF. Non-cell-autonomous function of DR6 in Schwann cell proliferation. EMBO J 2018; 37:embj.201797390. [PMID: 29459438 DOI: 10.15252/embj.201797390] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 12/20/2017] [Accepted: 01/16/2018] [Indexed: 12/21/2022] Open
Abstract
Death receptor 6 (DR6) is an orphan member of the TNF receptor superfamily and controls cell death and differentiation in a cell-autonomous manner in different cell types. Here, we report an additional non-cell-autonomous function for DR6 in the peripheral nervous system (PNS). DR6-knockout (DR6 KO) mice showed precocious myelination in the PNS Using an in vitro myelination assay, we demonstrate that neuronal DR6 acts in trans on Schwann cells (SCs) and reduces SC proliferation and myelination independently of its cytoplasmic death domain. Mechanistically, DR6 was found to be cleaved in neurons by "a disintegrin and metalloprotease 10" (ADAM10), releasing the soluble DR6 ectodomain (sDR6). Notably, in the in vitro myelination assay, sDR6 was sufficient to rescue the DR6 KO phenotype. Thus, in addition to the cell-autonomous receptor function of full-length DR6, the proteolytically released sDR6 can unexpectedly also act as a paracrine signaling factor in the PNS in a non-cell-autonomous manner during SC proliferation and myelination. This new mode of DR6 signaling will be relevant in future attempts to target DR6 in disease settings.
Collapse
Affiliation(s)
- Alessio Colombo
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Hung-En Hsia
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technical University Munich, Munich, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technical University Munich, Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Paolo Canevazzi
- Division of Neuroscience, INSPE at San Raffaele Scientific Institute, Milan, Italy
| | - Regina Feederle
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Diabetes and Obesity, Monoclonal Antibody Research Group, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany.,Munich Center for Systems Neurology (SyNergy), Munich, Germany
| | - Carla Taveggia
- Division of Neuroscience, INSPE at San Raffaele Scientific Institute, Milan, Italy
| | - Thomas Misgeld
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,Munich Center for Systems Neurology (SyNergy), Munich, Germany.,Center for Integrated Protein Sciences (CIPSM), Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany .,Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technical University Munich, Munich, Germany.,Munich Center for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
7
|
Gourmaud S, Mouton-Liger F, Abadie C, Meurs EF, Paquet C, Hugon J. Dual Kinase Inhibition Affords Extended in vitro Neuroprotection in Amyloid-β Toxicity. J Alzheimers Dis 2018; 54:1659-1670. [PMID: 27636848 DOI: 10.3233/jad-160509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In Alzheimer's disease (AD), the amyloid cascade hypothesis proposes that amyloid-beta (Aβ) neurotoxicity leads to neuroinflammation, synaptic loss, and neuronal degeneration. In AD patients, anti-amyloid immunotherapies did not succeed because they were possibly administered late in AD progression. Modulating new targets associated with Aβ toxicity, such as PKR (double-stranded RNA dependent kinase), and JNK (c-Jun N-terminal kinase) is a major goal for neuroprotection. These two pro-apoptotic kinases are activated in AD brains and involved in Aβ production, tau phosphorylation, neuroinflammation, and neuronal death. In HEK cells transfected with siRNA directed against PKR, and in PKR knockout (PKR-/-) mice neurons, we showed that PKR triggers JNK activation. Aβ-induced neuronal apoptosis, measured by cleaved PARP (Poly ADP-ribose polymerase) and cleaved caspase 3 levels, was reduced in PKR-/- neurons. Two selective JNK inhibitory peptides also produced a striking reduction of Aβ toxicity. Finally, the dual inhibition of PKR and JNK nearly abolished Aβ toxicity in primary cultured neurons. These results reveal that dual kinase inhibition can afford neuroprotection and this approach is worth being tested in in vivo AD and oxidative stress models.
Collapse
Affiliation(s)
| | | | | | - Eliane F Meurs
- Institut Pasteur, Hepacivirus and Innate Immunity Unit, Paris, France
| | - Claire Paquet
- Inserm UMR-S 942, Paris, France.,Research Memory Centre, Paris Nord Ile de France Saint Louis Lariboisière Fernand Widal Hospital, Paris, France
| | - Jacques Hugon
- Inserm UMR-S 942, Paris, France.,Research Memory Centre, Paris Nord Ile de France Saint Louis Lariboisière Fernand Widal Hospital, Paris, France
| |
Collapse
|
8
|
Emerging Roles of Sirtuin 6 in Alzheimer's Disease. J Mol Neurosci 2017; 64:157-161. [PMID: 29260452 DOI: 10.1007/s12031-017-1005-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is imposing an increasing burden on society. Currently, AD is the leading cause of senile dementia worldwide. Despite the long existence of AD, there is lack of therapies for AD, suggesting that new and effective treatment strategy must be explored. At present, sirtuin pathway has attracted attention from the researchers due to its promising results in laboratory models of aging. In addition, our understanding in the roles of sirtuin 6 in AD has expanded. It has been identified to be involved in telomere maintenance, DNA repair, genome integrity, energy metabolism, and inflammation, which ultimately regulate life span. Recent findings also demonstrate that sirtuin 6 is lacking in AD patients, proposing that it can be a new potential therapeutic target in AD. Therefore, exploring on how sirtuin 6 is related in AD manifestation may accelerate the research of AD further and benefits future AD patients. Keeping that in mind, this review aims to highlight the possible roles of sirtuin 6 in AD manifestation.
Collapse
|
9
|
Brown JC, Petersen A, Zhong L, Himelright ML, Murphy JA, Walikonis RS, Gerges NZ. Bidirectional regulation of synaptic transmission by BRAG1/IQSEC2 and its requirement in long-term depression. Nat Commun 2016; 7:11080. [PMID: 27009485 PMCID: PMC4820844 DOI: 10.1038/ncomms11080] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/16/2016] [Indexed: 01/09/2023] Open
Abstract
Dysfunction of the proteins regulating synaptic function can cause synaptic plasticity imbalance that underlies neurological disorders such as intellectual disability. A study found that four distinct mutations within BRAG1, an Arf-GEF synaptic protein, each led to X-chromosome-linked intellectual disability (XLID). Although the physiological functions of BRAG1 are poorly understood, each of these mutations reduces BRAG1's Arf-GEF activity. Here we show that BRAG1 is required for the activity-dependent removal of AMPA receptors in rat hippocampal pyramidal neurons. Moreover, we show that BRAG1 bidirectionally regulates synaptic transmission. On one hand, BRAG1 is required for the maintenance of synaptic transmission. On the other hand, BRAG1 expression enhances synaptic transmission, independently of BRAG1 Arf-GEF activity or neuronal activity, but dependently on its C-terminus interactions. This study demonstrates a dual role of BRAG1 in synaptic function and highlights the functional relevance of reduced BRAG1 Arf-GEF activity as seen in the XLID-associated human mutations.
Collapse
Affiliation(s)
- Joshua C Brown
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Amber Petersen
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Ling Zhong
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| | - Miranda L Himelright
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Jessica A Murphy
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Randall S Walikonis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269 USA
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin 53132 USA
| |
Collapse
|
10
|
Tackling amyloidogenesis in Alzheimer's disease with A2V variants of Amyloid-β. Sci Rep 2016; 6:20949. [PMID: 26864599 PMCID: PMC4750079 DOI: 10.1038/srep20949] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 02/05/2023] Open
Abstract
We developed a novel therapeutic strategy for Alzheimer’s disease (AD) exploiting the properties of a natural variant of Amyloid-β (Aβ) carrying the A2V substitution, which protects heterozygous carriers from AD by its ability to interact with wild-type Aβ, hindering conformational changes and assembly thereof. As prototypic compound we designed a six-mer mutated peptide (Aβ1-6A2V), linked to the HIV-related TAT protein, which is widely used for brain delivery and cell membrane penetration of drugs. The resulting molecule [Aβ1-6A2VTAT(D)] revealed strong anti-amyloidogenic effects in vitro and protected human neuroblastoma cells from Aβ toxicity. Preclinical studies in AD mouse models showed that short-term treatment with Aβ1-6A2VTAT(D) inhibits Aβ aggregation and cerebral amyloid deposition, but a long treatment schedule unexpectedly increases amyloid burden, although preventing cognitive deterioration. Our data support the view that the AβA2V-based strategy can be successfully used for the development of treatments for AD, as suggested by the natural protection against the disease in human A2V heterozygous carriers. The undesirable outcome of the prolonged treatment with Aβ1-6A2VTAT(D) was likely due to the TAT intrinsic attitude to increase Aβ production, avidly bind amyloid and boost its seeding activity, warning against the use of the TAT carrier in the design of AD therapeutics.
Collapse
|
11
|
Evaluation of the Role of JNK1 in the Hippocampus in an Experimental Model of Familial Alzheimer’s Disease. Mol Neurobiol 2015; 53:6183-6193. [DOI: 10.1007/s12035-015-9522-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/29/2015] [Indexed: 01/18/2023]
|
12
|
Meloni BP, Milani D, Edwards AB, Anderton RS, O'Hare Doig RL, Fitzgerald M, Palmer TN, Knuckey NW. Neuroprotective peptides fused to arginine-rich cell penetrating peptides: Neuroprotective mechanism likely mediated by peptide endocytic properties. Pharmacol Ther 2015; 153:36-54. [PMID: 26048328 DOI: 10.1016/j.pharmthera.2015.06.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/29/2015] [Indexed: 12/22/2022]
Abstract
Several recent studies have demonstrated that TAT and other arginine-rich cell penetrating peptides (CPPs) have intrinsic neuroprotective properties in their own right. Examples, we have demonstrated that in addition to TAT, poly-arginine peptides (R8 to R18; containing 8-18 arginine residues) as well as some other arginine-rich peptides are neuroprotective in vitro (in neurons exposed to glutamic acid excitotoxicity and oxygen glucose deprivation) and in the case of R9 in vivo (after permanent middle cerebral artery occlusion in the rat). Based on several lines of evidence, we propose that this neuroprotection is related to the peptide's endocytosis-inducing properties, with peptide charge and arginine residues being critical factors. Specifically, we propose that during peptide endocytosis neuronal cell surface structures such as ion channels and transporters are internalised, thereby reducing calcium influx associated with excitotoxicity and other receptor-mediated neurodamaging signalling pathways. We also hypothesise that a peptide cargo can act synergistically with TAT and other arginine-rich CPPs due to potentiation of the CPPs endocytic traits rather than by the cargo-peptide acting directly on its supposedly intended intracellular target. In this review, we systematically consider a number of studies that have used CPPs to deliver neuroprotective peptides to the central nervous system (CNS) following stroke and other neurological disorders. Consequently, we critically review evidence that supports our hypothesis that neuroprotection is mediated by carrier peptide endocytosis. In conclusion, we believe that there are strong grounds to regard arginine-rich peptides as a new class of neuroprotective molecules for the treatment of a range of neurological disorders.
Collapse
Affiliation(s)
- Bruno P Meloni
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia.
| | - Diego Milani
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Adam B Edwards
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Ryan S Anderton
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Nedlands, Australia; School of Animal Biology, The University of Western Australia, Nedlands, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Nedlands, Australia; School of Animal Biology, The University of Western Australia, Nedlands, Australia
| | - T Norman Palmer
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia
| | - Neville W Knuckey
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia
| |
Collapse
|
13
|
Zhou Q, Wang M, Du Y, Zhang W, Bai M, Zhang Z, Li Z, Miao J. Inhibition of c-Jun N-terminal kinase activation reverses Alzheimer disease phenotypes in APPswe/PS1dE9 mice. Ann Neurol 2015; 77:637-54. [PMID: 25611954 DOI: 10.1002/ana.24361] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/02/2015] [Accepted: 01/08/2015] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Growing evidence indicates that the activation of c-Jun N-terminal kinase (JNK) is implicated in the multiple major pathological features of Alzheimer disease (AD). However, whether specific inhibition of JNK activation could prevent disease progression in adult transgenic AD models at moderate stage remains unknown. Here we first investigated the potential disease-modifying therapeutic effect of systemic administration of SP600125, a small-molecule JNK-specific inhibitor, in middle-aged APPswe/PS1dE9 mice. METHODS Using behavioral, histological, and biochemical methods, outcomes of SP600125 treatment on neuropathology and cognitive deficits were studied in APPswe/PS1dE9 mice. RESULTS Compared with vehicle-treated APPswe/PS1dE9 mice, chronic treatment of SP600125 for 12 weeks potently inhibited JNK activation, which resulted in a marked improvement of behavioral measures of cognitive deficits and a dramatic reduction in amyloid plaque burden, β-amyloid production, tau hyperphosphorylation, inflammatory responses, and synaptic loss in these transgenic animals. In particular, we found that SP600125 treatment strongly promoted nonamyloidogenic amyloid precursor protein (APP) processing and inhibited amyloidogenic APP processing via regulating APP-cleavage secretase expression (ie, ADAM10, BACE1, and PS1) in APPswe/PS1dE9 mice. INTERPRETATION Our findings demonstrate that chronic SP600125 treatment is powerfully effective in slowing down disease progression by markedly reducing multiple pathological features and ameliorating cognitive deficits associated with AD. This study highlights the concept that active JNK actually contributes to the development of the disease, and provides critical preclinical evidence that specific inhibition of JNK activation by SP600125 treatment may be a novel promising disease-modifying therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Qiong Zhou
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Jadhav SP, Kamath SP, Choolani M, Lu J, Dheen ST. microRNA-200b modulates microglia-mediated neuroinflammation via the cJun/MAPK pathway. J Neurochem 2014; 130:388-401. [PMID: 24749688 DOI: 10.1111/jnc.12731] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/11/2014] [Accepted: 04/01/2014] [Indexed: 12/29/2022]
Abstract
Chronic activation of microglia, the macrophages of the CNS, has been shown to enhance neuronal damage because of excessive release of proinflammatory cytokines and neurotoxic molecules in a number of neurodegenerative diseases. Recent reports showed altered microRNA (miRNA) expression in immune-mediated pathologies, thus suggesting that miRNAs modulate expression of genes involving immune responses. This study demonstrates that miRNA-200b is expressed in microglia and modulates inflammatory response of microglia by regulating mitogen-activated protein kinase pathway. miRNA-200b expression was found to be down-regulated in activated microglia in vivo (traumatic brain injury rat model) and in vitro. A luciferase assay and loss- and gain-of-function studies revealed c-Jun, the transcription factor of cJun-N terminal kinase (JNK) mitogen-activated protein kinase pathway to be the target of miR-200b. Knockdown of miR-200b in microglia increased JNK activity along with an increase in pro-inflammatory cytokines, inducible nitric oxide synthase expression and nitric oxide (NO) production. Conversely, over-expression of miRNA-200b in microglia resulted in a decrease in JNK activity, inducible nitric oxide synthase expression, NO production and migratory potential of activated microglia. Furthermore, miR-200b inhibition resulted in increased neuronal apoptosis after treatment of neuronal cells with conditioned medium obtained from microglial culture. Taken together, these results indicate that miRNA-200b modulates microglial inflammatory process including cytokine secretion, NO production, migration and neuronal survival.
Collapse
Affiliation(s)
- Shweta P Jadhav
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | |
Collapse
|
15
|
Davoli E, Sclip A, Cecchi M, Cimini S, Carrà A, Salmona M, Borsello T. Determination of tissue levels of a neuroprotectant drug: the cell permeable JNK inhibitor peptide. J Pharmacol Toxicol Methods 2014; 70:55-61. [PMID: 24814549 DOI: 10.1016/j.vascn.2014.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/11/2014] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Cell permeable peptides (CPPs) represent a novel tool for the delivery of bioactive molecules into scarcely accessible organs, such as the brain. CPPs have been successfully used in pre-clinical studies for a variety of diseases, ranging from cancer to neurological disorders. However, the mechanisms by which CPPs cross biological membranes, as well as their pharmacokinetic properties, have been poorly explored due to the lack of specific and sensitive analytical methods. METHODS In this paper we describe a protocol to quantitatively determine the amount of CPPs in in vitro and in vivo experimental models. To this end we selected the peptide D-JNKI1 that was shown to prevent neurodegeneration in both acute and chronic degenerative disorders. This method allows an accurate quantitative analysis of D-JNKI1 in both neuronal lysates and tissue homogenates using mass spectrometry and stable isotope dilution approach. RESULTS We found that D-JNKI1 crosses cellular membranes with fast kinetics, through an active and passive mechanism. After acute intraperitoneal (ip) administration of D-JNKI1 in mice, the peptide was found in the main organs with particular regard to the liver and kidney. Interestingly, D-JNKI1 crosses the blood brain barrier (BBB) and reaches the brain, where it remains for one week. DISCUSSION The challenge lies in developing the clinical application of therapeutic cell permeable peptides. Discerning pharmacokinetic properties is a high priority to produce a powerful therapeutic strategy. Overall, our data shed light on the pharmacokinetic properties of D-JNKI1 and supports its powerful neuroprotective effect.
Collapse
Affiliation(s)
- Enrico Davoli
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Alessandra Sclip
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Matteo Cecchi
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Sara Cimini
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Andrea Carrà
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Mario Salmona
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy
| | - Tiziana Borsello
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| |
Collapse
|
16
|
Meloni BP, Craig AJ, Milech N, Hopkins RM, Watt PM, Knuckey NW. The neuroprotective efficacy of cell-penetrating peptides TAT, penetratin, Arg-9, and Pep-1 in glutamic acid, kainic acid, and in vitro ischemia injury models using primary cortical neuronal cultures. Cell Mol Neurobiol 2014; 34:173-81. [PMID: 24213248 PMCID: PMC11488960 DOI: 10.1007/s10571-013-9999-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 10/17/2013] [Indexed: 02/04/2023]
Abstract
Cell-penetrating peptides (CPPs) are small peptides (typically 5-25 amino acids), which are used to facilitate the delivery of normally non-permeable cargos such as other peptides, proteins, nucleic acids, or drugs into cells. However, several recent studies have demonstrated that the TAT CPP has neuroprotective properties. Therefore, in this study, we assessed the TAT and three other CPPs (penetratin, Arg-9, Pep-1) for their neuroprotective properties in cortical neuronal cultures following exposure to glutamic acid, kainic acid, or in vitro ischemia (oxygen-glucose deprivation). Arg-9, penetratin, and TAT-D displayed consistent and high level neuroprotective activity in both the glutamic acid (IC50: 0.78, 3.4, 13.9 μM) and kainic acid (IC50: 0.81, 2.0, 6.2 μM) injury models, while Pep-1 was ineffective. The TAT-D isoform displayed similar efficacy to the TAT-L isoform in the glutamic acid model. Interestingly, Arg-9 was the only CPP that displayed efficacy when washed-out prior to glutamic acid exposure. Neuroprotection following in vitro ischemia was more variable with all peptides providing some level of neuroprotection (IC50; Arg-9: 6.0 μM, TAT-D: 7.1 μM, penetratin/Pep-1: >10 μM). The positive control peptides JNKI-1D-TAT (JNK inhibitory peptide) and/or PYC36L-TAT (AP-1 inhibitory peptide) were neuroprotective in all models. Finally, in a post-glutamic acid treatment experiment, Arg-9 was highly effective when added immediately after, and mildly effective when added 15 min post-insult, while the JNKI-1D-TAT control peptide was ineffective when added post-insult. These findings demonstrate that different CPPs have the ability to inhibit neurodamaging events/pathways associated with excitotoxic and ischemic injuries. More importantly, they highlight the need to interpret neuroprotection studies when using CPPs as delivery agents with caution. On a positive note, the cytoprotective properties of CPPs suggests they are ideal carrier molecules to deliver neuroprotective drugs to the CNS following injury and/or potential neuroprotectants in their own right.
Collapse
Affiliation(s)
- Bruno P Meloni
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia and Australian Neuromuscular Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia,
| | | | | | | | | | | |
Collapse
|
17
|
Clarke M, Pentz R, Bobyn J, Hayley S. Stressor-like effects of c-Jun N-terminal kinase (JNK) inhibition. PLoS One 2012; 7:e44073. [PMID: 22952879 PMCID: PMC3430637 DOI: 10.1371/journal.pone.0044073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 07/30/2012] [Indexed: 11/18/2022] Open
Abstract
There is an urgent need for novel treatment strategies for stressor related disorders, particularly depression and anxiety disorders. Indeed, existing drug treatments are only clinically successful in a subset of patients and relapse is common. This likely stems from the fact that stressor disorders are heterogeneous with multiple biological pathways being affected. To this end, the present investigation sought to assess in mice the contribution of the c-Jun N terminal kinase (JNK) pathway to the behavioral, hormonal and neurochemical effects of an acute stressor. Indeed, although JNK has been shown to modulate glucocorticoid receptors in vitro, virtually nothing is known of the role for JNK in affecting stressor induced pathology. We presently found that the JNK antagonist, SP600125, (but not the p38 antagonist, SB203580) increased plasma corticosterone levels under resting conditions and in the context of an acute stressor (wet bedding + restraint). SP600125 also reduced exploration in an open field arena, but prevented the stressor induced increase in open arm exploration in an elevated plus maze. Finally, SP600125 affected noradrenergic activity in the central amygdala and locus coruleus under resting condition, but prevented the noradrenergic effects within the paraventricular nucleus of the hypothalamus that were induced by the acute stressor exposure. These data suggest inhibiting endogenous JNK can have stressor-like corticoid, behavioral and central monoamine effects under basal conditions, but can actually reverse some behavioral and neurochemical effects of an acute stressor. Thus, endogenous JNK appears to affect stress relevant processes in a context-dependent manner.
Collapse
Affiliation(s)
- Melanie Clarke
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Rowan Pentz
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Jessica Bobyn
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
18
|
Constitutive α- and β-secretase cleavages of the amyloid precursor protein are partially coupled in neurons, but not in frequently used cell lines. Neurobiol Dis 2012; 49:137-47. [PMID: 22940630 DOI: 10.1016/j.nbd.2012.08.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 07/20/2012] [Accepted: 08/16/2012] [Indexed: 11/21/2022] Open
Abstract
Proteolytic cleavage of the amyloid precursor protein (APP) by the two proteases α- and β-secretases controls the generation of the amyloid β peptide (Aβ), a key player in Alzheimer's disease pathogenesis. The α-secretase ADAM10 and the β-secretase BACE1 have opposite effects on Aβ generation and are assumed to compete for APP as a substrate, such that their cleavages are inversely coupled. This concept was mainly demonstrated in studies using activation or overexpression of α- and β-secretases. Here, we report that this inverse coupling is not seen to the same extent upon inhibition of the endogenous proteases. Genetic and pharmacological inhibition of ADAM10 and BACE1 revealed that the endogenous, constitutive α-secretase cleavage of APP is largely uncoupled from β-secretase cleavage and Aβ generation in neuroglioma H4 cells and in neuronally differentiated SH-SY5Y cells. In contrast, inverse coupling was observed in primary cortical neurons. However, this coupling was not bidirectional. Inhibition of BACE1 increased ADAM10 cleavage of APP, but a reduction of ADAM10 activity did not increase the BACE1 cleavage of APP in the neurons. Our analysis shows that the inverse coupling of the endogenous α- and β-secretase cleavages depends on the cellular model and suggests that a reduction of ADAM10 activity is unlikely to increase the AD risk through increased β-secretase cleavage.
Collapse
|
19
|
Sabapathy K. Role of the JNK pathway in human diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 106:145-69. [PMID: 22340717 DOI: 10.1016/b978-0-12-396456-4.00013-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The c-Jun-NH(2)-terminal kinase (JNK) signaling pathway plays a critical role in regulating cell fate, being implicated in a multitude of diseases ranging from cancer to neurological and immunological/inflammatory conditions. Not surprisingly, therefore, it has been sought after for therapeutic intervention, and its inhibition has been shown to ameliorate many pathological conditions in experimental systems, paving the way for initial clinical trials. However, the fundamental problem in fully harnessing the potential provided by the JNK pathway has been the lack of specificity, due to the multiple JNK forms that are involved in multiple cellular processes in various cell types. Moreover, lack of sufficient knowledge of all JNK-interacting proteins and substrates has also hindered progress. This review will therefore focus on the role of the JNKs in human diseases and appraise the efforts to inhibit JNK signaling to ameliorate disease conditions, assessing potential challenges and providing insights into possible future directions to efficiently target this pathway for therapeutic use.
Collapse
Affiliation(s)
- Kanaga Sabapathy
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| |
Collapse
|
20
|
Viana RJS, Nunes AF, Rodrigues CMP. Endoplasmic reticulum enrollment in Alzheimer's disease. Mol Neurobiol 2012; 46:522-34. [PMID: 22815194 DOI: 10.1007/s12035-012-8301-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/05/2012] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) poses a huge challenge for society and health care worldwide as molecular pathogenesis of the disease is poorly understood and curative treatment does not exist. The mechanisms leading to accelerated neuronal cell death in AD are still largely unknown, but accumulation of misfolded disease-specific proteins has been identified as potentially involved. In the present review, we describe the essential role of endoplasmic reticulum (ER) in AD. Despite the function that mitochondria may play as the central major player in the apoptotic process, accumulating evidence highlights ER as a critical organelle in AD. Stress that impairs ER physiology leads to accumulation of unfolded or misfolded proteins, such as amyloid β (Aβ) peptide, the major component of amyloid plaques. In an attempt to ameliorate the accumulation of unfolded proteins, ER stress triggers a protective cellular mechanism, which includes the unfolded protein response (UPR). However, when activation of the UPR is severe or prolonged enough, the final cellular outcome is pathologic apoptotic cell death. Distinct pathways can be activated in this process, involving stress sensors such as the JNK pathway or ER chaperones such as Bip/GRP94, stress modulators such as Bcl-2 family proteins, or even stress effectors such as caspase-12. Here, we detail the involvement of the ER and associated stress pathways in AD and discuss potential therapeutic strategies targeting ER stress.
Collapse
Affiliation(s)
- Ricardo J S Viana
- Research Institute for Medicines and Pharmaceutical Sciences, University of Lisbon, Lisbon 1649-003, Portugal
| | | | | |
Collapse
|
21
|
Inhibition of JNK by a peptide inhibitor reduces traumatic brain injury-induced tauopathy in transgenic mice. J Neuropathol Exp Neurol 2012; 71:116-29. [PMID: 22249463 DOI: 10.1097/nen.0b013e3182456aed] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a major environmental risk factor for subsequent development of Alzheimer disease (AD). Pathological features that are common to AD and many tauopathies are neurofibrillary tangles (NFTs) and neuropil threads composed of hyperphosphorylated tau. Axonal accumulations of total and phospho-tau have been observed within hours to weeks, and intracytoplasmic NFTs have been documented years after severe TBI in humans. We previously reported that controlled cortical impact TBI accelerated tau pathology in young 3xTg-AD mice. Here, we used this TBI mouse model to investigate mechanisms responsible for increased tau phosphorylation and accumulation after brain trauma. We found that TBI resulted in abnormal axonal accumulation of several kinases that phosphorylate tau. Notably, c-Jun N-terminal kinase (JNK) was markedly activated in injured axons and colocalized with phospho-tau. We found that moderate reduction of JNK activity (40%) by a peptide inhibitor, D-JNKi1, was sufficient to reduce total and phospho-tau accumulations in axons of these mice with TBI. Longer-term studies will be required to determine whether reducing acute tau pathology proves beneficial in brain trauma.
Collapse
|
22
|
Sclip A, Antoniou X, Colombo A, Camici GG, Pozzi L, Cardinetti D, Feligioni M, Veglianese P, Bahlmann FH, Cervo L, Balducci C, Costa C, Tozzi A, Calabresi P, Forloni G, Borsello T. c-Jun N-terminal kinase regulates soluble Aβ oligomers and cognitive impairment in AD mouse model. J Biol Chem 2011; 286:43871-43880. [PMID: 22033930 PMCID: PMC3243502 DOI: 10.1074/jbc.m111.297515] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/17/2011] [Indexed: 01/02/2023] Open
Abstract
Alzheimer disease (AD) is characterized by cognitive impairment that starts with memory loss to end in dementia. Loss of synapses and synaptic dysfunction are closely associated with cognitive impairment in AD patients. Biochemical and pathological evidence suggests that soluble Aβ oligomers correlate with cognitive impairment. Here, we used the TgCRND8 AD mouse model to investigate the role of JNK in long term memory deficits. TgCRND8 mice were chronically treated with the cell-penetrating c-Jun N-terminal kinase inhibitor peptide (D-JNKI1). D-JNKI1, preventing JNK action, completely rescued memory impairments (behavioral studies) as well as the long term potentiation deficits of TgCRND8 mice. Moreover, D-JNKI1 inhibited APP phosphorylation in Thr-668 and reduced the amyloidogenic cleavage of APP and Aβ oligomers in brain parenchyma of treated mice. In conclusion, by regulating key pathogenic mechanisms of AD, JNK might hold promise as innovative therapeutic target.
Collapse
Affiliation(s)
- Alessandra Sclip
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Xanthi Antoniou
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Alessio Colombo
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Giovanni G Camici
- Cardiovascular Research Laboratory, Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Laura Pozzi
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Daniele Cardinetti
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Marco Feligioni
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Pietro Veglianese
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Ferdinand H Bahlmann
- Department of Internal Medicine IV, Saarland University Medical Centre, 66421 Homburg/Saar, Germany
| | - Luigi Cervo
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Claudia Balducci
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Cinzia Costa
- Clinica Neurologica Division, Università di Perugia, Ospedale S. Maria della Misericordia, Perugia 06156, Italy
| | - Alessandro Tozzi
- Clinica Neurologica Division, Università di Perugia, Ospedale S. Maria della Misericordia, Perugia 06156, Italy
| | - Paolo Calabresi
- Clinica Neurologica Division, Università di Perugia, Ospedale S. Maria della Misericordia, Perugia 06156, Italy; Fondazione Santa Lucia, Istituto di Ricovero e Cura a Carattere Scientifico, Rome 00143, Italy
| | - Gianluigi Forloni
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Tiziana Borsello
- Neuronal Death and Neuroprotection Laboratory, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano 20156, Italy.
| |
Collapse
|
23
|
Amyloid-β Production: Major Link Between Oxidative Stress and BACE1. Neurotox Res 2011; 22:208-19. [DOI: 10.1007/s12640-011-9283-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 09/28/2011] [Accepted: 09/30/2011] [Indexed: 12/20/2022]
|
24
|
SP600125, a competitive inhibitor of JNK attenuates streptozotocin induced neurocognitive deficit and oxidative stress in rats. Pharmacol Biochem Behav 2010; 96:386-94. [DOI: 10.1016/j.pbb.2010.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Revised: 05/26/2010] [Accepted: 06/20/2010] [Indexed: 02/04/2023]
|
25
|
Mehan S, Meena H, Sharma D, Sankhla R. JNK: A Stress-Activated Protein Kinase Therapeutic Strategies and Involvement in Alzheimer’s and Various Neurodegenerative Abnormalities. J Mol Neurosci 2010; 43:376-90. [DOI: 10.1007/s12031-010-9454-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 09/16/2010] [Indexed: 01/26/2023]
|
26
|
Braithwaite SP, Schmid RS, He DN, Sung MLA, Cho S, Resnick L, Monaghan MM, Hirst WD, Essrich C, Reinhart PH, Lo DC. Inhibition of c-Jun kinase provides neuroprotection in a model of Alzheimer's disease. Neurobiol Dis 2010; 39:311-7. [PMID: 20451607 PMCID: PMC2910136 DOI: 10.1016/j.nbd.2010.04.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 04/13/2010] [Accepted: 04/26/2010] [Indexed: 11/22/2022] Open
Abstract
The c-Jun N-terminal kinase (JNK) pathway potentially links together the three major pathological hallmarks of Alzheimer's disease (AD): development of amyloid plaques, neurofibrillary tangles, and brain atrophy. As activation of the JNK pathway has been observed in amyloid models of AD in association with peri-plaque regions and neuritic dystrophy, as we confirm here for Tg2576/PS(M146L) transgenic mice, we directly tested whether JNK inhibition could provide neuroprotection in a novel brain slice model for amyloid precursor protein (APP)-induced neurodegeneration. We found that APP/amyloid beta (Abeta)-induced neurodegeneration is blocked by both small molecule and peptide inhibitors of JNK, and provide evidence that this neuroprotection occurs downstream of APP/Abeta production and processing. Our findings demonstrate that Abeta can induce neurodegeneration, at least in part, through the JNK pathway and suggest that inhibition of JNK may be of therapeutic utility in the treatment of AD.
Collapse
Affiliation(s)
| | - Ralf S. Schmid
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, 4321 Medical Park Dr., Suite 200, Durham, NC 27704
| | - Dong Ning He
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, 4321 Medical Park Dr., Suite 200, Durham, NC 27704
| | - Mei-Li A. Sung
- Discovery Neuroscience, Wyeth Research, CN8000, Princeton, NJ 08534
| | - Seongeon Cho
- Discovery Neuroscience, Wyeth Research, CN8000, Princeton, NJ 08534
| | - Lynn Resnick
- Discovery Neuroscience, Wyeth Research, CN8000, Princeton, NJ 08534
| | | | - Warren D. Hirst
- Discovery Neuroscience, Wyeth Research, CN8000, Princeton, NJ 08534
| | - Christian Essrich
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, 4321 Medical Park Dr., Suite 200, Durham, NC 27704
| | | | - Donald C. Lo
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, 4321 Medical Park Dr., Suite 200, Durham, NC 27704
| |
Collapse
|
27
|
Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta Mol Basis Dis 2010; 1802:396-405. [DOI: 10.1016/j.bbadis.2009.12.009] [Citation(s) in RCA: 1795] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 11/26/2009] [Accepted: 12/01/2009] [Indexed: 12/13/2022]
|
28
|
Ploia C, Sclip A, Colombo A, Repici M, Gardoni F, Di Luca M, Forloni G, Antoniou X, Borsello T. Role of Glycogen Synthase Kinase-3β in APP Hyperphosphorylation Induced by NMDA Stimulation in Cortical Neurons. Pharmaceuticals (Basel) 2010; 3:42-58. [PMID: 27713242 PMCID: PMC3991020 DOI: 10.3390/ph3010042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 12/03/2009] [Accepted: 01/05/2010] [Indexed: 01/24/2023] Open
Abstract
The phosphorylation of Amyloid Precursor Protein (APP) at Thr668 plays a key role in APP metabolism that is highly relevant to AD. The c-Jun-N-terminal kinase (JNK), glycogen synthase kinase-3β (GSK-3β) and cyclin-dependent kinase 5 (Cdk5) can all be responsible for this phosphorylation. These kinases are activated by excitotoxic stimuli fundamental hallmarks of AD. The exposure of cortical neurons to a high dose of NMDA (100 μM) for 30’-45’ led to an increase of P-APP Thr668. During NMDA stimulation APP hyperphosphorylation has to be assigned to GSK-3β activity, since addition of L803-mts, a substrate competitive inhibitor of GSK-3β reduced APP phosphorylation induced by NMDA. On the contrary, inhibition of JNK and Cdk5 with D-JNKI1 and Roscovitine respectively did not prevent NMDA-induced P-APP increase. These data show a tight connection, in excitotoxic conditions, between APP metabolism and the GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Cristina Ploia
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| | - Alessandra Sclip
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| | - Alessio Colombo
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| | - Mariaelena Repici
- UMR 7102 Neurobiologie des Processus Adaptatifs, Universite P. et M. Curie, 9 quai St Bernard, 75005, Paris, France.
| | - Fabrizio Gardoni
- Dipartimento Scienze Farmacologiche, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy.
| | - Monica Di Luca
- Dipartimento Scienze Farmacologiche, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy.
| | - Gianluigi Forloni
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| | - Xanthi Antoniou
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| | - Tiziana Borsello
- Istituto di ricerche Farmacologiche "Mario Negri", Via La Masa 19, 20156 Milano, Italy.
| |
Collapse
|
29
|
Haeusgen W, Boehm R, Zhao Y, Herdegen T, Waetzig V. Specific activities of individual c-Jun N-terminal kinases in the brain. Neuroscience 2009; 161:951-9. [DOI: 10.1016/j.neuroscience.2009.04.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/06/2009] [Accepted: 04/06/2009] [Indexed: 12/31/2022]
|
30
|
JNK regulates APP cleavage and degradation in a model of Alzheimer's disease. Neurobiol Dis 2009; 33:518-25. [DOI: 10.1016/j.nbd.2008.12.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 12/05/2008] [Accepted: 12/20/2008] [Indexed: 11/23/2022] Open
|
31
|
Meade AJ, Meloni BP, Mastaglia FL, Knuckey NW. The application of cell penetrating peptides for the delivery of neuroprotective peptides/proteins in experimental cerebral ischaemia studies. ACTA ACUST UNITED AC 2009. [DOI: 10.6030/1939-067x-2.1.21] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|