1
|
In Vitro Antiviral Effect and Potential Neuroprotection of Salvadora persica L. Stem Bark Extract against Lipopolysaccharides-Induced Neuroinflammation in Mice: LC-ESI-MS/MS Analysis of the Methanol Extract. Pharmaceuticals (Basel) 2023; 16:ph16030398. [PMID: 36986497 PMCID: PMC10058283 DOI: 10.3390/ph16030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroinflammation is a serious immunomodulatory complex disorder that causes neurological and somatic ailments. The treatment of brain inflammation with new drugs derived from natural sources is a significant therapeutic goal. Utilizing LC-ESI-MS/MS analysis, the active constituents of Salvadora persica extract (SPE) were identified tentatively as exerting antioxidant and anti-inflammatory effects in natural medicine. Herein, we determined the antiviral potential of SPE against herpes simplex virus type 2 (HSV-2) using the plaque assay. HSV-2 is a neurotropic virus that can cause neurological diseases. SPE exhibited promising antiviral potential with a half-maximal cytotoxic concentration (CC50) of 185.960 ± 0.1 µg/mL and a half-maximal inhibitory concentration (IC50) of 8.946 ± 0.02 µg/mL. The in vivo study of the SPE impact against lipopolysaccharide (LPS)-induced neuroinflammation was performed using 42 mice divided into seven groups. All groups were administered LPS (0.25 mg/kg) intraperitoneally, except for the normal and SPE groups 1 and 2. Groups 5, 6, and 7 received 100, 200, and 300 mg/kg SPE. It was revealed that SPE inhibited acetylcholinesterase in the brain. It increased superoxide dismutase and catalase while decreasing malondialdehyde, which explains its antioxidative stress activity. SPE downregulated the gene expression of the inducible nitric oxide synthase, as well as the apoptotic markers (caspase-3 and c-Jun). In addition, it decreased the expression of the proinflammatory cytokines (interleukin-6 and tumor necrosis factor-alpha). Mice administered SPE (300 mg/kg) with LPS exhibited normal neurons in the cerebral cortices, hippocampus pyramidal layer, and cerebellum, as determined by the histopathological analysis. Therefore, using S. persica to prevent and treat neurodegeneration could be a promising new therapeutic strategy to be explored.
Collapse
|
2
|
Craig RA, Fox BM, Hu C, Lexa KW, Osipov M, Thottumkara AP, Larhammar M, Miyamoto T, Rana A, Kane LA, Yulyaningsih E, Solanoy H, Nguyen H, Chau R, Earr T, Kajiwara Y, Fleck D, Lucas A, Haddick PCG, Takahashi RH, Tong V, Wang J, Canet MJ, Poda SB, Scearce-Levie K, Srivastava A, Sweeney ZK, Xu M, Zhang R, He J, Lei Y, Zhuo Z, de Vicente J. Discovery of Potent and Selective Dual Leucine Zipper Kinase/Leucine Zipper-Bearing Kinase Inhibitors with Neuroprotective Properties in In Vitro and In Vivo Models of Amyotrophic Lateral Sclerosis. J Med Chem 2022; 65:16290-16312. [PMID: 36469401 DOI: 10.1021/acs.jmedchem.2c01056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Dual leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) are regulators of neuronal degeneration and axon growth. Therefore, there is a considerable interest in developing DLK/LZK inhibitors for neurodegenerative diseases. Herein, we use ligand- and structure-based drug design approaches for identifying novel amino-pyrazine inhibitors of DLK/LZK. DN-1289 (14), a potent and selective dual DLK/LZK inhibitor, demonstrated excellent in vivo plasma half-life across species and is anticipated to freely penetrate the central nervous system with no brain impairment based on in vivo rodent pharmacokinetic studies and human in vitro transporter data. Proximal target engagement and disease relevant pathway biomarkers were also favorably regulated in an in vivo model of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Robert A Craig
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Brian M Fox
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Cheng Hu
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Katrina W Lexa
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Maksim Osipov
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Arun P Thottumkara
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Martin Larhammar
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Takashi Miyamoto
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Anil Rana
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Lesley A Kane
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ernie Yulyaningsih
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Hilda Solanoy
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Hoang Nguyen
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Roni Chau
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Timothy Earr
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Yuji Kajiwara
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Daniel Fleck
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Anthony Lucas
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Patrick C G Haddick
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ryan H Takahashi
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Vincent Tong
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Jing Wang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Mark J Canet
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Suresh B Poda
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Kimberly Scearce-Levie
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ankita Srivastava
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Zachary K Sweeney
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Musheng Xu
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Rui Zhang
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Jianrong He
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Yanan Lei
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Zheng Zhuo
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Javier de Vicente
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| |
Collapse
|
3
|
Fucoxanthin Prevents Long-Term Administration l-DOPA-Induced Neurotoxicity through the ERK/JNK-c-Jun System in 6-OHDA-Lesioned Mice and PC12 Cells. Mar Drugs 2022; 20:md20040245. [PMID: 35447917 PMCID: PMC9025159 DOI: 10.3390/md20040245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
As the most abundant marine carotenoid extracted from seaweeds, fucoxanthin is considered to have neuroprotective activity via its excellent antioxidant properties. Oxidative stress is regarded as an important starting factor for neuronal cell loss and necrosis, is one of the causes of Parkinson’s disease (PD), and is considered to be the cause of adverse reactions caused by the current PD commonly used treatment drug levodopa (l-DA). Supplementation with antioxidants early in PD can effectively prevent neurodegeneration and inhibit apoptosis in dopaminergic neurons. At present, the effect of fucoxanthin in improving the adverse effects triggered by long-term l-DA administration in PD patients is unclear. In the present study, we found that fucoxanthin can reduce cytotoxicity and suppress the high concentration of l-DA (200 μM)-mediated cell apoptosis in the 6-OHDA-induced PC12 cells through improving the reduction in mitochondrial membrane potential, suppressing ROS over-expression, and inhibiting active of ERK/JNK-c-Jun system and expression of caspase-3 protein. These results were demonstrated by PD mice with long-term administration of l-DA showing enhanced motor ability after intervention with fucoxanthin. Our data indicate that fucoxanthin may prove useful in the treatment of PD patients with long-term l-DA administration.
Collapse
|
4
|
Methodology and Neuromarkers for Cetaceans’ Brains. Vet Sci 2022; 9:vetsci9020038. [PMID: 35202291 PMCID: PMC8879147 DOI: 10.3390/vetsci9020038] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/01/2023] Open
Abstract
Cetacean brain sampling may be an arduous task due to the difficulty of collecting and histologically preparing such rare and large specimens. Thus, one of the main challenges of working with cetaceans’ brains is to establish a valid methodology for an optimal manipulation and fixation of the brain tissue, which allows the samples to be viable for neuroanatomical and neuropathological studies. With this in view, we validated a methodology in order to preserve the quality of such large brains (neuroanatomy/neuropathology) and at the same time to obtain fresh brain samples for toxicological, virological, and microbiological analysis (neuropathology). A fixation protocol adapted to brains, of equal or even three times the size of human brains, was studied and tested. Finally, we investigated the usefulness of a panel of 20 antibodies (neuromarkers) associated with the normal structure and function of the brain, pathogens, age-related, and/or functional variations. The sampling protocol and some of the 20 neuromarkers have been thought to explore neurodegenerative diseases in these long-lived animals. To conclude, many of the typical measures used to evaluate neuropathological changes do not tell us if meaningful cellular changes have occurred. Having a wide panel of antibodies and histochemical techniques available allows for delving into the specific behavior of the neuronal population of the brain nuclei and to get a “fingerprint” of their real status.
Collapse
|
5
|
Andrés-Benito P, Carmona M, Pirla MJ, Torrejón-Escribano B, Del Rio JA, Ferrer I. Dysregulated Protein Phosphorylation as Main Contributor of Granulovacuolar Degeneration at the First Stages of Neurofibrillary Tangles Pathology. Neuroscience 2021; 518:119-140. [PMID: 34757172 DOI: 10.1016/j.neuroscience.2021.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 02/08/2023]
Abstract
The hippocampus of cases with neurofibrillary tangles (NFT) pathology classified as stages I-II, III-IV, and V-VI without comorbidities, and middle-aged (MA) individuals with no NFT pathology, were examined to learn about the composition of granulovacuolar degeneration (GVD). Our results confirm the presence of CK1-δ, p38-P Thr180/Tyr182, SAPK/JNK-P Thr183/Thr185, GSK-3α/β-P Tyr279/Tyr216, and GSK-3β Ser9 in the cytoplasmic granules in a subset of neurons of the CA1 and CA2 subfields of the hippocampus. Also, we identify the presence of PKA α/β-P Thr197, SRC-P Tyr416, PAK1-P Ser199/Ser204, CAMK2A-P Tyr197, and PKCG-P Thr655 in cytoplasmic granules in cases with NFT pathology, but not in MA cases. Our results also confirm the presence of β-catenin-P Ser45/Thr41, IREα-P Ser274, eIF2α-P Ser51, TDP-43-P Ser403-404 (but absent TDP-43), and ubiquitin in cytoplasmic granules. Other components of the cytoplasmic granules are MAP2-P Thr1620/1623, MAP1B-P Thr1265, ADD1-P Ser726, and ADD1/ADD1-P Ser726/Ser713, in addition to several tau species including 3Rtau, 4Rtau, and tau-P Ser262. The analysis of GVD at progressive stages of NFT pathology reveals the early appearance of phosphorylated kinases and proteins in cytoplasmic granules at stages I-II, before the appearance of pre-tangles and NFTs. Most of these granules are not surrounded by LAMP1-positive membranes. Markers of impaired ubiquitin-protesome system, abnormal reticulum stress response, and altered endocytic and autophagic pathways occur in a subpopulation of neurons containing cytoplasmic granules, and they appear later. These observations suggest early phosphorylation of kinases leading to their activation, and resulting in the abnormal phosphorylation of various substrates, including tau, as a main alteration at the first stages of GVD.
Collapse
Affiliation(s)
- Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| | - Margarita Carmona
- Department of Pathology and Experimental Therapeutics, University of Barcelona, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| | - Mónica Jordán Pirla
- Department of Pathology and Experimental Therapeutics, University of Barcelona, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| | - Benjamín Torrejón-Escribano
- Advanced Light Microscopy Unit (Campus de Bellvitge), Scientific and Technical Facility (CCiTUB), University of Barcelona, Hospitalet de Llobregat, Spain.
| | - José Antonio Del Rio
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology, Science Park Barcelona (PCB), Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Spain.
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
6
|
Abstract
Phosphorylation is a reversible, enzyme-controlled posttranslational process affecting approximately one-third of all proteins in eukaryotic cells at any given time. Any deviation in the degree and/or site of phosphorylation leads to an abnormal conformation of proteins, resulting in a decline or loss of their function. Knowledge of phosphorylation-related pathways is essential for understanding the understanding of the disease pathogenesis and for the design of new therapeutic strategies. Recent availability of various kinases at an affordable price differs in activity, specificity, and stability and provides the opportunity of studying and modulating this reaction in vitro. We can exploit this knowledge for other applications. There is an enormous potential to produce fully decorated and active recombinant proteins, either for biomedical or cosmetic applications. Closely related is the possibility to exploit current achievements and develop new safe and efficacious vaccines, drugs, and immunomodulators. In this review, we outlined the current enzyme-based possibilities for in vitro phosphorylation of peptides and recombinant proteins and the added value that immobilized kinases provide.
Collapse
|
7
|
Thingore C, Kshirsagar V, Juvekar A. Amelioration of oxidative stress and neuroinflammation in lipopolysaccharide-induced memory impairment using Rosmarinic acid in mice. Metab Brain Dis 2021; 36:299-313. [PMID: 33068223 DOI: 10.1007/s11011-020-00629-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022]
Abstract
Oxidative stress plays a pivotal part in the manifestation of neuroinflammation, which further leads to neurodegenerative diseases like Alzheimer's disease (AD). Systemic administration of lipopolysaccharide (LPS) induces neuroinflammation resulting in memory impairment (MI) and cognitive decline. In this study, we evaluated whether prophylactic administration of Rosmarinic acid (RA), a naturally occurring compound, exerts a neuroprotective effect in LPS-induced MI and cognitive decline. Herein, Swiss albino mice were pre-treated with RA (0.5 mg/kg and 1 mg/kg i.p.) for 28 days and were intermittently exposed to LPS (0.25 mg/kg i.p.) for 7 days. LPS caused poor memory retention and increased cognitive decline in Morris water maze (MWM) and Y maze paradigms respectively. Additionally, LPS increased oxidative stress which was denoted by a decrease in superoxide dismutase (SOD) activity, decrease in reduced glutathione (GSH) levels, and increased lipid peroxidation in the brain. Imbalance in the cholinergic system was analyzed by measuring the acetylcholinesterase (AChE) activity. Pre-treatment with RA improved memory and behavioral disturbances by alleviating oxidative stress and AChE activity. LPS augmented levels of tumor necrosis factor (TNF-α), interleukin (IL)-6, caspase-3, and c-Jun. Pre-treatment with RA revitalized the elevated levels of proinflammatory cytokines and apoptotic proteins. In conclusion, this study showcases the amelioration of MI by RA in LPS-challenged memory and cognitive decline, which could be credited to its anti-oxidant effect, inhibitory effect on both proinflammatory cytokines and apoptotic regulators, and reduction in AChE activity.
Collapse
Affiliation(s)
- Chetan Thingore
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India
| | - Viplav Kshirsagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India
| | - Archana Juvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, Maharashtra, India.
| |
Collapse
|
8
|
Zulfiqar Z, Shah FA, Shafique S, Alattar A, Ali T, Alvi AM, Rashid S, Li S. Repurposing FDA Approved Drugs as JNK3 Inhibitor for Prevention of Neuroinflammation Induced by MCAO in Rats. J Inflamm Res 2020; 13:1185-1205. [PMID: 33384558 PMCID: PMC7770337 DOI: 10.2147/jir.s284471] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Background Stress-associated kinases are considered major pathological mediators in several incurable neurological disorders. Importantly, among these stress kinases, the c-Jun NH2-terminal kinase (JNK) has been linked to numerous neuropathological conditions, including oxidative stress, neuroinflammation, and brain degeneration associated with brain injuries such as ischemia/reperfusion injury. In this study, we adopted a drug repurposing/reprofiling approach to explore novel JNK3 inhibitors from FDA-approved medications to supplement existing therapeutic strategies. Materials and Methods We performed in silico docking analysis and molecular dynamics simulation to screen potential candidates from the FDA approved drug library using the standard JNK inhibitor SP600125 as a reference. After the virtual screening, dabigatran, estazolam, leucovorin, and pitavastatin were further examined in ischemic stroke using an animal rodent model of focal cerebral ischemia using transient middle cerebral artery occlusion (t-MCAO). The selected drugs were probed for neuroprotective effectiveness by measuring the infarct area (%) and neurological deficits using a 28-point composite score. Biochemical assays including ELISA and immunohistochemical experiments were performed. Results We obtained structural insights for dabigatran, estazolam, and pitavastatin binding to JNK3, revealing a significant contribution of the hydrophobic regions and significant residues of active site regions. To validate the docking results, the pharmacological effects of dabigatran, estazolam, leucovorin, and pitavastatin on MCAO were tested in parallel with the JNK inhibitor SP600125. After MCAO surgery, severe neurological deficits were detected in the MCAO group compared with the sham controls, which were significantly reversed by dabigatran, estazolam, and pitavastatin treatment. Aberrant morphological features and brain damage were observed in the ipsilateral cortex and striatum of the MCAO groups. The drugs restored the anti-oxidant enzyme activity and reduced the levels of oxidative stress-induced p-JNK and neuroinflammatory mediators such as NF-kB and TNF-ɑ in rats subjected to MCAO. Conclusion Our results demonstrated that the novel FDA-approved medications attenuate ischemic stroke-induced neuronal degeneration, possibly by inhibiting JNK3. Being FDA-approved safe medications, the use of these drugs can be clinically translated for ischemic stroke-associated brain degeneration and other neurodegenerative diseases associated with oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Zikra Zulfiqar
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Shagufta Shafique
- National Center for Bioinformatics, Quaid-I-Azam University, Islamabad, Pakistan
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Tahir Ali
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arooj Mohsin Alvi
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid-I-Azam University, Islamabad, Pakistan
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People's Republic of China
| |
Collapse
|
9
|
Wiersma VI, Hoozemans JJM, Scheper W. Untangling the origin and function of granulovacuolar degeneration bodies in neurodegenerative proteinopathies. Acta Neuropathol Commun 2020; 8:153. [PMID: 32883341 PMCID: PMC7469111 DOI: 10.1186/s40478-020-00996-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
In the brains of tauopathy patients, tau pathology coincides with the presence of granulovacuolar degeneration bodies (GVBs) both at the regional and cellular level. Recently, it was shown that intracellular tau pathology causes GVB formation in experimental models thus explaining the strong correlation between these neuropathological hallmarks in the human brain. These novel models of GVB formation provide opportunities for future research into GVB biology, but also urge reevaluation of previous post-mortem observations. Here, we review neuropathological data on GVBs in tauopathies and other neurodegenerative proteinopathies. We discuss the possibility that intracellular aggregates composed of proteins other than tau are also able to induce GVB formation. Furthermore, the potential mechanisms of GVB formation and the downstream functional implications hereof are outlined in view of the current available data. In addition, we provide guidelines for the identification of GVBs in tissue and cell models that will help to facilitate and streamline research towards the elucidation of the role of these enigmatic and understudied structures in neurodegeneration.
Collapse
|
10
|
Cores Á, Piquero M, Villacampa M, León R, Menéndez JC. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020; 10:E904. [PMID: 32545924 PMCID: PMC7356958 DOI: 10.3390/biom10060904] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
NRF2 acts by controlling gene expression, being the master regulator of the Phase II antioxidant response, and also being key to the control of neuroinflammation. NRF2 activity is regulated at several levels, including protein degradation by the proteasome, transcription, and post-transcription. The purpose of this review is to offer a concise and critical overview of the main mechanisms of NRF2 regulation and their actual or potential use as targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Marta Piquero
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Mercedes Villacampa
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| |
Collapse
|
11
|
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
|
12
|
Su R, Su W, Jiao Q. NGF protects neuroblastoma cells against β-amyloid-induced apoptosis via the Nrf2/HO-1 pathway. FEBS Open Bio 2019; 9:2063-2071. [PMID: 31605506 PMCID: PMC6886293 DOI: 10.1002/2211-5463.12742] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/21/2019] [Accepted: 10/09/2019] [Indexed: 01/18/2023] Open
Abstract
As one of the main neurotrophic factors, nerve growth factor (NGF) participates in various processes related to viability, plasticity, and neuronal growth. NGF is known to protect against cell death and toxicity triggered by β-amyloid (Aβ), but the underlying mechanism remains unclear. Here, we investigated this process in SKNSH neuroblastoma, in which NGF reduced cell death induced by Aβ25-35. Furthermore, NGF suppressed the production of reactive oxygen species (ROS) and promoted antioxidant function via Aβ25-35. Additionally, we demonstrated that NGF impaired the activation of the JNK/c-Jun signaling pathway and significantly increased Nrf2 nuclear translocation and HO-1 expression. Nrf2 elimination abolished the protective effect of NGF-1 on Aβ25-35-induced ROS generation, apoptosis, and activation of the JNK/c-Jun pathway. The results of our study indicate that NGF protects neuroblastoma against injury triggered by Aβ25-35 via suppression of ROS-JNK/c-Jun pathway stimulation through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Rui Su
- Department of NeurosurgeryLuoyang Central Hospital Affiliated to Zhengzhou UniversityChina
| | - Wei Su
- Department of Intensive Care UnitSir Run Run Shaw Hospital Affiliated by Zhejiang University School of MedicineHangzhouChina
| | - Qian Jiao
- Department of Anesthesia SurgerySanmenxia Central HospitalChina
| |
Collapse
|
13
|
Li HQ, Ip SP, Yuan QJ, Zheng GQ, Tsim KKW, Dong TTX, Lin G, Han Y, Liu Y, Xian YF, Lin ZX. Isorhynchophylline ameliorates cognitive impairment via modulating amyloid pathology, tau hyperphosphorylation and neuroinflammation: Studies in a transgenic mouse model of Alzheimer's disease. Brain Behav Immun 2019; 82:264-278. [PMID: 31476414 DOI: 10.1016/j.bbi.2019.08.194] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/14/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Isorhynchophylline (IRN) has been demonstrated to have distinct anti-Alzheimer's disease (AD) activity in several animal models of AD. In this study, we aimed at evaluating the preventive effect of IRN on the cognitive deficits and amyloid pathology in TgCRND8 mice. Male TgCRND8 mice were administered with IRN (20 or 40 mg/kg) by oral gavage daily for 4 months, followed by assessing the spatial learning and memory functions with the Radial Arm Maze (RAM) test. Brain tissues were determined immunohistochemically or biochemically for changes in amyloid pathology, tau hyperphosphorylation and neuroinflammation. Our results revealed that IRN (40 mg/kg) significantly ameliorated cognitive deficits in TgCRND8 mice. In addition, IRN (40 mg/kg) markedly reduced the levels of Aβ40, Aβ42 and tumor necrosis factor (TNF-α), interleukin 6 (IL-6) and IL-1β, and modulated the amyloid precursor protein (APP) processing and phosphorylation by altering the protein expressions of β-site APP cleaving enzyme-1 (BACE-1), phosphorylated APP (Thr668), presenilin-1 (PS-1) and anterior pharynx-defective-1 (APH-1), as well as insulin degrading enzyme (IDE), a major Aβ-degrading enzyme. IRN was also found to inhibit the phosphorylation of tau at the sites of Thr205 and Ser396. Immunofluorescence showed that IRN reduced the Aβ deposition, and suppressed the activation of microglia (Iba-1) and astrocytes (GFAP) in the cerebral cortex and hippocampus of TgCRND8 mice. Furthermore, IRN was able to attenuate the ratios of p-c-Jun/c-Jun and p-JNK/JNK in the brains of TgCRND8 mice. IRN also showed marked inhibitory effect on JNK signaling pathway in the Aβ-treated rat primary hippocampus neurons. We conclude that IRN improves cognitive impairment in TgCRND8 transgenic mice via reducing Aβ generation and deposition, tau hyperphosphorylation and neuroinflammation through inhibiting the activation of JNK signaling pathway, and has good potential for further development into pharmacological treatment for AD.
Collapse
Affiliation(s)
- Hui-Qin Li
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Qiu-Ju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Province, PR China.
| | - Karl K W Tsim
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region.
| | - Tina T X Dong
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region.
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Yifan Han
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region.
| | - Yue Liu
- Cardiovascular Disease Centre, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, PR China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
14
|
Le Pichon CE, Meilandt WJ, Dominguez S, Solanoy H, Lin H, Ngu H, Gogineni A, Sengupta Ghosh A, Jiang Z, Lee SH, Maloney J, Gandham VD, Pozniak CD, Wang B, Lee S, Siu M, Patel S, Modrusan Z, Liu X, Rudhard Y, Baca M, Gustafson A, Kaminker J, Carano RAD, Huang EJ, Foreman O, Weimer R, Scearce-Levie K, Lewcock JW. Loss of dual leucine zipper kinase signaling is protective in animal models of neurodegenerative disease. Sci Transl Med 2018; 9:9/403/eaag0394. [PMID: 28814543 DOI: 10.1126/scitranslmed.aag0394] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 11/23/2016] [Accepted: 03/27/2017] [Indexed: 12/16/2022]
Abstract
Hallmarks of chronic neurodegenerative disease include progressive synaptic loss and neuronal cell death, yet the cellular pathways that underlie these processes remain largely undefined. We provide evidence that dual leucine zipper kinase (DLK) is an essential regulator of the progressive neurodegeneration that occurs in amyotrophic lateral sclerosis and Alzheimer's disease. We demonstrate that DLK/c-Jun N-terminal kinase signaling was increased in mouse models and human patients with these disorders and that genetic deletion of DLK protected against axon degeneration, neuronal loss, and functional decline in vivo. Furthermore, pharmacological inhibition of DLK activity was sufficient to attenuate the neuronal stress response and to provide functional benefit even in the presence of ongoing disease. These findings demonstrate that pathological activation of DLK is a conserved mechanism that regulates neurodegeneration and suggest that DLK inhibition may be a potential approach to treat multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Claire E Le Pichon
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - William J Meilandt
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Sara Dominguez
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hilda Solanoy
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Han Lin
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hai Ngu
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alvin Gogineni
- Department of Biomedical Imaging, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Zhiyu Jiang
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Seung-Hye Lee
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Janice Maloney
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vineela D Gandham
- Department of Biomedical Imaging, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christine D Pozniak
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bei Wang
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Sebum Lee
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael Siu
- Department of Discovery Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Snahel Patel
- Department of Discovery Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Xingrong Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - York Rudhard
- In Vitro Pharmacology, Evotec AG, Manfred Eigen Campus, 22419 Hamburg, Germany
| | - Miriam Baca
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Amy Gustafson
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Josh Kaminker
- Department of Bioinformatics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Richard A D Carano
- Department of Biomedical Imaging, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA.,Pathology Service 113B, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Oded Foreman
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robby Weimer
- Department of Biomedical Imaging, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kimberly Scearce-Levie
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph W Lewcock
- Department of Neuroscience, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
15
|
Ibrahim OHM, Hassan MA. The Use of Anti-Diabetic Drugs in Alzheimer’s Disease, New Therapeutic Options and Future Perspective. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/pp.2018.96013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
16
|
Kalathur RKR, Pedro Pinto J, Sahoo B, Chaurasia G, Futschik ME. HDNetDB: A Molecular Interaction Database for Network-Oriented Investigations into Huntington's Disease. Sci Rep 2017; 7:5216. [PMID: 28701700 PMCID: PMC5507972 DOI: 10.1038/s41598-017-05224-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Huntington’s disease (HD) is a progressive and fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene. Although HD is monogenic, its molecular manifestation appears highly complex and involves multiple cellular processes. The recent application of high throughput platforms such as microarrays and mass-spectrometry has indicated multiple pathogenic routes. The massive data generated by these techniques together with the complexity of the pathogenesis, however, pose considerable challenges to researchers. Network-based methods can provide valuable tools to consolidate newly generated data with existing knowledge, and to decipher the interwoven molecular mechanisms underlying HD. To facilitate research on HD in a network-oriented manner, we have developed HDNetDB, a database that integrates molecular interactions with many HD-relevant datasets. It allows users to obtain, visualize and prioritize molecular interaction networks using HD-relevant gene expression, phenotypic and other types of data obtained from human samples or model organisms. We illustrated several HDNetDB functionalities through a case study and identified proteins that constitute potential cross-talk between HD and the unfolded protein response (UPR). HDNetDB is publicly accessible at http://hdnetdb.sysbiolab.eu.
Collapse
Affiliation(s)
- Ravi Kiran Reddy Kalathur
- SysBioLab, Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal. .,Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - José Pedro Pinto
- SysBioLab, Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | | | - Gautam Chaurasia
- Institute for Theoretical Biology, Charité, Humboldt-University, Berlin, Germany
| | - Matthias E Futschik
- SysBioLab, Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal. .,Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Algarve, Portugal. .,School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, Devon, United Kingdom.
| |
Collapse
|
17
|
Rehman SU, Ahmad A, Yoon GH, Khan M, Abid MN, Kim MO. Inhibition of c-Jun N-Terminal Kinase Protects Against Brain Damage and Improves Learning and Memory After Traumatic Brain Injury in Adult Mice. Cereb Cortex 2017; 28:2854-2872. [DOI: 10.1093/cercor/bhx164] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/12/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
- Shafiq Ur Rehman
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Ashfaq Ahmad
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Gwang-Ho Yoon
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Mehtab Khan
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Muhammad Noman Abid
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Myeong Ok Kim
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
18
|
Lachen-Montes M, Zelaya MV, Segura V, Fernández-Irigoyen J, Santamaría E. Progressive modulation of the human olfactory bulb transcriptome during Alzheimer´s disease evolution: novel insights into the olfactory signaling across proteinopathies. Oncotarget 2017; 8:69663-69679. [PMID: 29050232 PMCID: PMC5642507 DOI: 10.18632/oncotarget.18193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/07/2017] [Indexed: 01/01/2023] Open
Abstract
Alzheimer´s disease (AD) is characterized by progressive dementia, initially presenting olfactory dysfunction. Despite the olfactory bulb (OB) is the first central structure of the olfactory pathway, we lack a complete molecular characterization of the transcriptional events that occurs in this olfactory area during AD progression. To address this gap in knowledge, we have assessed the genome-wide expression in postmortem OBs from subjects with varying degree of AD pathology. A stage-dependent deregulation of specific pathways was observed, revealing transmembrane transport, and neuroinflammation as part of the functional modules that are disrupted across AD grading. Potential drivers of neurodegeneration predicted by network-driven transcriptomics were monitored across different types of dementia, including progressive supranuclear palsy (PSP), mixed dementia, and frontotemporal lobar degeneration (FTLD). Epidermal growth factor receptor (EGFR) expression was significantly increased in the OB of AD and mixed dementia subjects. Moreover, a significant increment in the activation of signal transducer and activator of transcription 3 (STAT3) was exclusively detected in advanced AD stages, whereas total STAT3 levels were specifically overexpressed in mixed dementia. Furthermore, transcription factors deregulated in the OB of mixed dementia subjects such as cAMP Responsive Element Binding Protein 1 (CREB1) and AP-1 Transcription Factor Subunit (c-Jun) were not differentially modulated at olfactory level across AD grading. On the other hand, olfactory expression of this signal transducer panel was unchanged in PSP and FTLD subjects. Taken together, this study unveils cross-disease similarities and differences for specific signal transducers, providing mechanistic clues to the intriguing divergence of AD pathology across proteinopathies.
Collapse
Affiliation(s)
- Mercedes Lachen-Montes
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - María Victoria Zelaya
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Pathological Anatomy Department, Navarra Hospital Complex, Pamplona, Spain
| | - Víctor Segura
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| |
Collapse
|
19
|
Yao Z, Yang W, Gao Z, Jia P. Nicotinamide mononucleotide inhibits JNK activation to reverse Alzheimer disease. Neurosci Lett 2017; 647:133-140. [PMID: 28330719 DOI: 10.1016/j.neulet.2017.03.027] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/11/2017] [Accepted: 03/15/2017] [Indexed: 01/19/2023]
Abstract
Amyloid-β (Aβ) oligomers have been accepted as major neurotoxic agents in the therapy of Alzheimer's disease (AD). It has been shown that the activity of nicotinamide adenine dinucleotide (NAD+) is related with the decline of Aβ toxicity in AD. Nicotinamide mononucleotide (NMN), the important precursor of NAD+, is produced during the reaction of nicotinamide phosphoribosyl transferase (Nampt). This study aimed to figure out the potential therapeutic effects of NMN and its underlying mechanisms in APPswe/PS1dE9 (AD-Tg) mice. We found that NMN gave rise to a substantial improvement in behavioral measures of cognitive impairments compared to control AD-Tg mice. In addition, NMN treatment significantly decreased β-amyloid production, amyloid plaque burden, synaptic loss, and inflammatory responses in transgenic animals. Mechanistically, NMN effectively controlled JNK activation. Furthermore, NMN potently progressed nonamyloidogenic amyloid precursor protein (APP) and suppressed amyloidogenic APP by mediating the expression of APP cleavage secretase in AD-Tg mice. Based on our findings, it was suggested that NMN substantially decreases multiple AD-associated pathological characteristically at least partially by the inhibition of JNK activation.
Collapse
Affiliation(s)
- Zhiwen Yao
- Department of Neurology, YangPu Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenhao Yang
- Department of Neurology, YangPu Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhiqiang Gao
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Peng Jia
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
20
|
Granulovacuolar degeneration: a neurodegenerative change that accompanies tau pathology. Acta Neuropathol 2016; 132:339-59. [PMID: 27062260 DOI: 10.1007/s00401-016-1562-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/08/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
Abstract
Granule-containing vacuoles in the cytoplasm of hippocampal neurons are a neuropathological feature of Alzheimer's disease. Granulovacuolar degeneration (GVD) is not disease-specific and can be observed in other neurodegenerative disorders and even in the brains of non-demented elderly people. However, several studies have reported much higher numbers of neurons undergoing GVD in the hippocampus of Alzheimer's disease cases. Recently, a neuropathological staging system for GVD has facilitated neuropathological assessment. Data obtained by electron microscopy and immunolabeling suggest that GVD inclusions are a special form of autophagic vacuole. GVD frequently occurs together with pathological changes of the microtubule-associated protein tau, but to date, the relationship between the two lesions remains elusive. Originally identified in hematoxylin- and silver-stained sections, immunolabeling has shown that the granules are composed of a variety of proteins, including those related to tau pathology, autophagy, diverse signal transduction pathways, cell stress and apoptosis. Several of these proteins serve as markers of GVD. Most researchers and authors have interpreted the sequestration of proteins into GVD inclusions as either a cellular defense mechanism or one that leads to the impairment of important cellular functions. This review provides a detailed overview of the various aspects of GVD and focuses on the relationship between tau pathology and GVD.
Collapse
|
21
|
Mittal K, Katare DP. Shared links between type 2 diabetes mellitus and Alzheimer's disease: A review. Diabetes Metab Syndr 2016; 10:S144-S149. [PMID: 26907971 DOI: 10.1016/j.dsx.2016.01.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/09/2016] [Indexed: 10/22/2022]
Abstract
Epidemiological studies have proved that, there are pathophysiological connections between Type 2 Diabetes Mellitus (T2DM) and Alzheimer's disease (AD). Diabetic patients have higher incidences of cognitive impairment and hence they are more at the risk of developing AD. Some of the recent evidences have majorly stated the effects of insulin resistance in the disturbance of various biological processes and signaling pathways. Both hyperglycemia and hypoglycemic conditions contributes in dysfunctioning of cognitive abilities and functions. The present review summarizes the evidences which establish the possible links between the two pathologies on the account of molecular, biochemical and at histopathological level. The information regarding their interactions was collected from different databases and journals. The gathered information will clearly establish the link among the two pathologies and will be helpful in future for the development of drugs for Type 3 Diabetes.
Collapse
Affiliation(s)
- Khyati Mittal
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Deepshikha Pande Katare
- Proteomics and Translational Research Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
22
|
L-DOPA modulates cell viability through the ERK-c-Jun system in PC12 and dopaminergic neuronal cells. Neuropharmacology 2016; 101:87-97. [DOI: 10.1016/j.neuropharm.2015.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 08/25/2015] [Accepted: 09/03/2015] [Indexed: 01/20/2023]
|
23
|
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]
|
24
|
Akhter R, Sanphui P, Das H, Saha P, Biswas SC. The regulation of p53 up-regulated modulator of apoptosis by JNK/c-Jun pathway in β-amyloid-induced neuron death. J Neurochem 2015; 134:1091-103. [PMID: 25891762 DOI: 10.1111/jnc.13128] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/10/2015] [Accepted: 03/31/2015] [Indexed: 12/29/2022]
Abstract
Neuronal loss in selective areas of brain underlies the pathology of Alzheimer's disease (AD). Recent evidences place oligomeric β-amyloid (Aβ) central to the disease. However, mechanism of neuron death in response to Aβ remains elusive. Activation of the c-Jun N-terminal kinase (JNK) pathway and induction of the AP-1 transcription factor c-Jun are reported in AD. However, targets of JNK/c-Jun in Aβ-induced neuron death are mostly unknown. Our study shows that pro-apoptotic proteins, Bim (Bcl-2 interacting mediator of cell death) and Puma (p53 up-regulated modulator of apoptosis) are targets of c-Jun in Aβ-treated neurons. We demonstrate that the JNK/c-Jun pathway is activated, in cultures of cortical neurons following treatment with oligomeric Aβ and in AD transgenic mice, and that inhibition of this pathway by selective inhibitor blocks induction of Puma by Aβ. We also find that both JNK and p53 pathways co-operatively regulate Puma expression in Aβ-treated neurons. Moreover, we identified a novel AP1-binding site on rat puma gene which is necessary for direct binding of c-Jun with Puma promoter. Finally, we find that knocking down of c-Jun by siRNA provides significant protection from Aβ toxicity and that induction of Bim and Puma by Aβ in neurons requires c-Jun. Taken together, our results suggest that both Bim and Puma are target of c-Jun and elucidate the intricate regulation of Puma expression by JNK/c-Jun and p53 pathways in neurons upon Aβ toxicity. JNK/c-Jun pathway is shown to be activated in neurons of the Alzheimer's disease (AD) brain and plays a vital role in neuron death in AD models. However, downstream targets of c-Jun in this disease have not been thoroughly elucidated. Our study shows that two important pro-apoptotic proteins, Bim (Bcl-2 interacting mediator of cell death) and Puma (p53 up-regulated modulator of apoptosis) are targets of c-Jun in Aβ-treated neurons. We demonstrate that the JNK/c-jun pathway is activated, in cultures of cortical neurons following treatment with oligomeric Aβ and in AD transgenic mice, and that inhibition of this pathway by selective inhibitor blocks induction of Puma by Aβ. We have also observed functional co-operation of both JNK and p53 pathway in regulation of Puma under Aβ toxicity. Most importantly, we identified a novel AP1-binding site on rat puma gene which is necessary for direct binding of c-Jun with Puma promoter. Thus, our results suggest that both Bim and Puma are target of c-Jun and elucidate the intricate regulation of Puma expression by JNK/c-Jun and p53 pathways in neurons upon Aβ toxicity.
Collapse
Affiliation(s)
- Rumana Akhter
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Priyankar Sanphui
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Hrishita Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Pampa Saha
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Subhas Chandra Biswas
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
25
|
Liu C, Tan FCK, Xiao ZC, Dawe GS. Amyloid precursor protein enhances Nav1.6 sodium channel cell surface expression. J Biol Chem 2015; 290:12048-57. [PMID: 25767117 DOI: 10.1074/jbc.m114.617092] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Amyloid precursor protein (APP) is commonly associated with Alzheimer disease, but its physiological function remains unknown. Nav1.6 is a key determinant of neuronal excitability in vivo. Because mouse models of gain of function and loss of function of APP and Nav1.6 share some similar phenotypes, we hypothesized that APP might be a candidate molecule for sodium channel modulation. Here we report that APP colocalized and interacted with Nav1.6 in mouse cortical neurons. Knocking down APP decreased Nav1.6 sodium channel currents and cell surface expression. APP-induced increases in Nav1.6 cell surface expression were Go protein-dependent, enhanced by a constitutively active Go protein mutant, and blocked by a dominant negative Go protein mutant. APP also regulated JNK activity in a Go protein-dependent manner. JNK inhibition attenuated increases in cell surface expression of Nav1.6 sodium channels induced by overexpression of APP. JNK, in turn, phosphorylated APP. Nav1.6 sodium channel surface expression was increased by T668E and decreased by T668A, mutations of APP695 mimicking and preventing Thr-668 phosphorylation, respectively. Phosphorylation of APP695 at Thr-668 enhanced its interaction with Nav1.6. Therefore, we show that APP enhances Nav1.6 sodium channel cell surface expression through a Go-coupled JNK pathway.
Collapse
Affiliation(s)
- Chao Liu
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Francis Chee Kuan Tan
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Zhi-Cheng Xiao
- the Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical College, Kunming 650031, China, and the Shunxi-Monash Immune Regeneration and Neuroscience Laboratories, Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Gavin S Dawe
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456,
| |
Collapse
|
26
|
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
|
27
|
Jimenez-Del-Rio M, Velez-Pardo C. Alzheimer’s Disease, Drosophila melanogaster and Polyphenols. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 863:21-53. [DOI: 10.1007/978-3-319-18365-7_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
28
|
Patel S, Cohen F, Dean BJ, De La Torre K, Deshmukh G, Estrada AA, Ghosh AS, Gibbons P, Gustafson A, Huestis MP, Le Pichon CE, Lin H, Liu W, Liu X, Liu Y, Ly CQ, Lyssikatos JP, Ma C, Scearce-Levie K, Shin YG, Solanoy H, Stark KL, Wang J, Wang B, Zhao X, Lewcock JW, Siu M. Discovery of Dual Leucine Zipper Kinase (DLK, MAP3K12) Inhibitors with Activity in Neurodegeneration Models. J Med Chem 2014; 58:401-18. [DOI: 10.1021/jm5013984] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Changyou Ma
- Department
of Chemistry, WuXi AppTec Co., Ltd., 288 Fute Zhonglu, Wai Gao Qiao Free
Trade Zone, Shanghai, 200131, P. R. China
| | | | | | | | | | - Jian Wang
- Department
of Chemistry, WuXi AppTec Co., Ltd., 288 Fute Zhonglu, Wai Gao Qiao Free
Trade Zone, Shanghai, 200131, P. R. China
| | | | | | | | | |
Collapse
|
29
|
Evaluation of traditional medicines for neurodegenerative diseases using Drosophila models. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:967462. [PMID: 24790636 PMCID: PMC3984789 DOI: 10.1155/2014/967462] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/17/2014] [Accepted: 02/24/2014] [Indexed: 12/19/2022]
Abstract
Drosophila is one of the oldest and most powerful genetic models and has led to novel insights into a variety of biological processes. Recently, Drosophila has emerged as a model system to study human diseases, including several important neurodegenerative diseases. Because of the genomic similarity between Drosophila and humans, Drosophila neurodegenerative disease models exhibit a variety of human-disease-like phenotypes, facilitating fast and cost-effective in vivo genetic modifier screening and drug evaluation. Using these models, many disease-associated genetic factors have been identified, leading to the identification of compelling drug candidates. Recently, the safety and efficacy of traditional medicines for human diseases have been evaluated in various animal disease models. Despite the advantages of the Drosophila model, its usage in the evaluation of traditional medicines is only nascent. Here, we introduce the Drosophila model for neurodegenerative diseases and some examples demonstrating the successful application of Drosophila models in the evaluation of traditional medicines.
Collapse
|
30
|
Kamal MA, Priyamvada S, Anbazhagan AN, Jabir NR, Tabrez S, Greig NH. Linking Alzheimer's disease and type 2 diabetes mellitus via aberrant insulin signaling and inflammation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2014; 13:338-46. [PMID: 24074448 PMCID: PMC5947865 DOI: 10.2174/18715273113126660137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 03/16/2013] [Accepted: 04/02/2013] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two progressive and devastating health disorders afflicting millions of people worldwide. The probability and incidence of both have increased considerably in recent years consequent to increased longevity and population growth. Progressively more links are being continuously found between inflammation and central nervous system disorders like AD, Parkinson's disease, Huntington's disease, motor neuron disease, multiple sclerosis, stroke, traumatic brain injury and even cancers of the nervous tissue. The depth of the relationship depends on the timing and extent of anti- or pro-inflammatory gene expression. Inflammation has also been implicated in T2DM. Misfolding and fibrillization (of tissue specific and/or non-specific proteins) are features common to both AD and T2DM and are induced by as well as contribute to inflammation and stress (oxidative/ glycation). This review appraises the roles of inflammation and abnormalities in the insulin signaling system as important shared features of T2DM and AD. The capacity of anti-cholinesterases in reducing the level of certain common inflammatory markers in particular if they may provide therapeutic potential to mitigate awry mechanisms leading to AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Nigel H Greig
- Metabolomics & Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
31
|
Caspase-2 is essential for c-Jun transcriptional activation and Bim induction in neuron death. Biochem J 2013; 455:15-25. [PMID: 23815625 DOI: 10.1042/bj20130556] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, β-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1β-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.
Collapse
|
32
|
Pozniak CD, Sengupta Ghosh A, Gogineni A, Hanson JE, Lee SH, Larson JL, Solanoy H, Bustos D, Li H, Ngu H, Jubb AM, Ayalon G, Wu J, Scearce-Levie K, Zhou Q, Weimer RM, Kirkpatrick DS, Lewcock JW. Dual leucine zipper kinase is required for excitotoxicity-induced neuronal degeneration. ACTA ACUST UNITED AC 2013; 210:2553-67. [PMID: 24166713 PMCID: PMC3832926 DOI: 10.1084/jem.20122832] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Loss of dual leucine zipper kinase results in attenuated JNK/c-Jun stress response pathway activation and reduced neuronal degeneration after kainic acid–induced excitotoxic seizures. Excessive glutamate signaling is thought to underlie neurodegeneration in multiple contexts, yet the pro-degenerative signaling pathways downstream of glutamate receptor activation are not well defined. We show that dual leucine zipper kinase (DLK) is essential for excitotoxicity-induced degeneration of neurons in vivo. In mature neurons, DLK is present in the synapse and interacts with multiple known postsynaptic density proteins including the scaffolding protein PSD-95. To examine DLK function in the adult, DLK-inducible knockout mice were generated through Tamoxifen-induced activation of Cre-ERT in mice containing a floxed DLK allele, which circumvents the neonatal lethality associated with germline deletion. DLK-inducible knockouts displayed a modest increase in basal synaptic transmission but had an attenuation of the JNK/c-Jun stress response pathway activation and significantly reduced neuronal degeneration after kainic acid–induced seizures. Together, these data demonstrate that DLK is a critical upstream regulator of JNK-mediated neurodegeneration downstream of glutamate receptor hyper-activation and represents an attractive target for the treatment of indications where excitotoxicity is a primary driver of neuronal loss.
Collapse
Affiliation(s)
- Christine D Pozniak
- Department of Neuroscience, 2 Department of Biomedical Imaging, 3 Department of Bioinformatics and Computational Biology, 4 Department of Protein Chemistry, 5 Department of Pathology, Genentech, Inc., South San Francisco, CA 94080
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Schmit TL, Dowell JA, Maes ME, Wilhelm M. c-Jun N-terminal kinase regulates mGluR-dependent expression of post-synaptic FMRP target proteins. J Neurochem 2013; 127:772-81. [PMID: 24047560 PMCID: PMC3992883 DOI: 10.1111/jnc.12453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 12/01/2022]
Abstract
Fragile X syndrome (FXS) is caused by the loss of functional fragile X mental retardation protein (FMRP). Loss of FMRP results in an elevated basal protein expression profile of FMRP targeted mRNAs, a loss of local metabotropic glutamate receptor (mGluR)-regulated protein synthesis, exaggerated long-term depression and corresponding learning and behavioral deficits. Evidence shows that blocking mGluR signaling in FXS models ameliorates these deficits. Therefore, understanding the signaling mechanisms downstream of mGluR stimulation may provide additional therapeutic targets for FXS. Kinase cascades are an integral mechanism regulating mGluR-dependent protein translation. The c-Jun N-terminal kinase (JNK) pathway has been shown to regulate mGluR-dependent nuclear transcription; however, the involvement of JNK in local, synaptic signaling has not been explored. Here, we show that JNK is both necessary and sufficient for mGluR-dependent expression of a subset of FMRP target proteins. In addition, JNK activity is basally elevated in fmr1 knockout mouse synapses, and blocking JNK activity reduces the over-expression of post-synaptic proteins in these mice. Together, these data suggest that JNK may be an important signaling mechanism downstream of mGluR stimulation, regulating FMRP-dependent protein synthesis. Furthermore, local, post-synaptic dysregulation of JNK activity may provide a viable target to ameliorate the deficits involved in FXS. Expression of many FMRP target proteins is enhanced in FXS. Here, we evaluated the role of JNKs in FXS. We found that JNK signaling is activated upon mGluR stimulation in wild-type neurons. Conversely, JNK activity is basally elevated in fmr1 knockout. Inhibiting JNK reduced the expression of FMRP target proteins and driving JNK activity increased the expression of these proteins.
Collapse
Affiliation(s)
- Travis L Schmit
- Department of Pediatrics and the Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
34
|
Huntwork-Rodriguez S, Wang B, Watkins T, Ghosh AS, Pozniak CD, Bustos D, Newton K, Kirkpatrick DS, Lewcock JW. JNK-mediated phosphorylation of DLK suppresses its ubiquitination to promote neuronal apoptosis. ACTA ACUST UNITED AC 2013; 202:747-63. [PMID: 23979718 PMCID: PMC3760612 DOI: 10.1083/jcb.201303066] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neuronal injury induces JNK phosphorylation of DLK, which reduces DLK ubiquitination and creates a positive feedback loop to enhance JNK signaling and increase apoptosis. Neurons are highly polarized cells that often project axons a considerable distance. To respond to axonal damage, neurons must transmit a retrograde signal to the nucleus to enable a transcriptional stress response. Here we describe a mechanism by which this signal is propagated through injury-induced stabilization of dual leucine zipper-bearing kinase (DLK/MAP3K12). After neuronal insult, specific sites throughout the length of DLK underwent phosphorylation by c-Jun N-terminal kinases (JNKs), which have been shown to be downstream targets of DLK pathway activity. These phosphorylation events resulted in increased DLK abundance via reduction of DLK ubiquitination, which was mediated by the E3 ubiquitin ligase PHR1 and the de-ubiquitinating enzyme USP9X. Abundance of DLK in turn controlled the levels of downstream JNK signaling and apoptosis. Through this feedback mechanism, the ubiquitin–proteasome system is able to provide an additional layer of regulation of retrograde stress signaling to generate a global cellular response to localized external insults.
Collapse
Affiliation(s)
- Sarah Huntwork-Rodriguez
- Department of Neuroscience, 2 Department of Microchemical Proteomics, and 3 Department of Physiological Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Koike Y, Kondo H, Kondo S, Takagi M, Kano Y. Effect of a steam foot spa on geriatric inpatients with cognitive impairment: a pilot study. Clin Interv Aging 2013; 8:543-8. [PMID: 23717038 PMCID: PMC3663437 DOI: 10.2147/cia.s44005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate whether a steam foot spa improves cognitive impairment in geriatric inpatients. Methods Geriatric inpatients with cognitive impairment were given a steam foot spa treatment at 42°C for 20 minutes for 2 weeks (5 days/week). Physiological indicators such as blood pressure, percutaneous oxygen saturation, pulse, tympanic temperature, and sleep time and efficiency were assessed. Cognitive function and behavioral and psychological symptoms of dementia were assessed using the Mini-Mental State Examination, Dementia Mood Assessment Scale, and Dementia Behavior Disturbance scale. Results Significant decreases in systolic (P < 0.01) and diastolic blood pressure (P < 0.05) along with a significant increase in tympanic temperature (P < 0.01) were observed after the steam foot spas. A significant improvement was seen in the Mini-Mental State Examination score (P < 0.01) and the overall dementia severity items in Dementia Mood Assessment Scale (P < 0.05). Limitations Japanese people are very fond of foot baths. However, it is difficult to understand why inpatients cannot receive steam foot baths. In this study, a control group was not used. Raters and enforcers were not blinded. Conclusion The results of this pilot study suggest that steam foot spas mitigate cognitive impairment in geriatric inpatients.
Collapse
Affiliation(s)
- Yoshihisa Koike
- Department of Occupational Therapy, Prefectural University of Hiroshima, Mihara, Japan.
| | | | | | | | | |
Collapse
|
36
|
Cheng C, Li W, Zhang Z, Yoshimura S, Hao Q, Zhang C, Wang Z. MicroRNA-144 is regulated by activator protein-1 (AP-1) and decreases expression of Alzheimer disease-related a disintegrin and metalloprotease 10 (ADAM10). J Biol Chem 2013; 288:13748-61. [PMID: 23546882 DOI: 10.1074/jbc.m112.381392] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND MicroRNA (miR) dysregulation is found in Alzheimer disease (AD). A disintegrin and metalloprotease 10 (ADAM10) prevents generation of amyloid β (Aβ) and decrease AD pathology. RESULTS miR-144 suppresses ADAM10 expression and is up-regulated by activator protein-1. CONCLUSION miR-144 is a negative regulator of ADAM10 and may be involved in AD pathogenesis. SIGNIFICANCE The first work to demonstrate the function of miRNA-144 and its regulation in the pathogenesis of AD. Amyloid β-peptide (Aβ) accumulating in the brain of Alzheimer disease (AD) patients is believed to be the main pathophysiologcal cause of the disease. Proteolytic processing of the amyloid precursor protein by α-secretase ADAM10 (a disintegrin and metalloprotease 10) protects the brain from the production of the Aβ. Meanwhile, dysregulation or aberrant expression of microRNAs (miRNAs) has been widely documented in AD patients. In this study, we demonstrated that overexpression of miR-144, which was previously reported to be increased in elderly primate brains and AD patients, significantly decreased activity of the luciferase reporter containing the ADAM10 3'-untranslated region (3'-UTR) and suppressed the ADAM10 protein level, whereas the miR-144 inhibitor led to an increase of the luciferase activity. The negative regulation caused by miR-144 was strictly dependent on the binding of the miRNA to its recognition element in the ADAM10 3'-UTR. Moreover, we also showed that activator protein-1 regulates the transcription of miR-144 and the up-regulation of miR-144 at least partially induces the suppression of the ADAM10 protein in the presence of Aβ. In addition, we found that miR-451, a miRNA processed from a single gene locus with miR-144, is also involved in the regulation of ADAM10 expression. Taken together, our data therefore demonstrate miR-144/451 is a negative regulator of the ADAM10 protein and suggest a mechanistic role for miR-144/451 in AD pathogenesis.
Collapse
Affiliation(s)
- Cong Cheng
- Protein Science Key Laboratory of the Ministry of Education, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | | | | | | | | | | | | |
Collapse
|
37
|
Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 2013; 1:a006189. [PMID: 22229116 DOI: 10.1101/cshperspect.a006189] [Citation(s) in RCA: 2207] [Impact Index Per Article: 183.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The neuropathological hallmarks of Alzheimer disease (AD) include "positive" lesions such as amyloid plaques and cerebral amyloid angiopathy, neurofibrillary tangles, and glial responses, and "negative" lesions such as neuronal and synaptic loss. Despite their inherently cross-sectional nature, postmortem studies have enabled the staging of the progression of both amyloid and tangle pathologies, and, consequently, the development of diagnostic criteria that are now used worldwide. In addition, clinicopathological correlation studies have been crucial to generate hypotheses about the pathophysiology of the disease, by establishing that there is a continuum between "normal" aging and AD dementia, and that the amyloid plaque build-up occurs primarily before the onset of cognitive deficits, while neurofibrillary tangles, neuron loss, and particularly synaptic loss, parallel the progression of cognitive decline. Importantly, these cross-sectional neuropathological data have been largely validated by longitudinal in vivo studies using modern imaging biomarkers such as amyloid PET and volumetric MRI.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Alzheimer Research Unit of the MassGeneral Institute for Neurodegenerative Disease, Department of Neurology of the Massachusetts General Hospital, and Harvard Medical School, Charlestown, Massachusetts, USA, 02129-4404
| | | | | | | |
Collapse
|
38
|
Park SH, Lee S, Hong YK, Hwang S, Lee JH, Bang SM, Kim YK, Koo BS, Lee IS, Cho KS. Suppressive Effects of SuHeXiang Wan on Amyloid-β42-Induced Extracellular Signal-Regulated Kinase Hyperactivation and Glial Cell Proliferation in a Transgenic Drosophila Model of Alzheimer’s Disease. Biol Pharm Bull 2013; 36:390-8. [DOI: 10.1248/bpb.b12-00792] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Soojin Lee
- Department of Biological Sciences, Konkuk University
| | - Yoon Ki Hong
- Department of Biological Sciences, Konkuk University
| | - Soojin Hwang
- Department of Biological Sciences, Konkuk University
| | - Jang Ho Lee
- Department of Biological Sciences, Konkuk University
| | - Se Min Bang
- Department of Biological Sciences, Konkuk University
| | - Young-Kyoon Kim
- Department of Forest Products & Biotechnology, Kookmin University
| | - Byung-Soo Koo
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University
| | - Im-Soon Lee
- Department of Biological Sciences, Konkuk University
| | | |
Collapse
|
39
|
Tamagno E, Guglielmotto M, Monteleone D, Vercelli A, Tabaton M. Transcriptional and post-transcriptional regulation of β-secretase. IUBMB Life 2012. [DOI: 10.1002/iub.1099] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
40
|
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
|
41
|
The bad, the good, and the ugly about oxidative stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:163913. [PMID: 22619696 PMCID: PMC3350994 DOI: 10.1155/2012/163913] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 01/16/2012] [Accepted: 02/07/2012] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), and cancer (e.g., leukemia) are the most devastating disorders affecting millions of people worldwide. Except for some kind of cancers, no effective and/or definitive therapeutic treatment aimed to reduce or to retard the clinic and pathologic symptoms induced by AD and PD is presently available. Therefore, it is urgently needed to understand the molecular basis of these disorders. Since oxidative stress (OS) is an important etiologic factor of the pathologic process of AD, PD, and cancer, understanding how intracellular signaling pathways respond to OS will have a significant implication in the therapy of these diseases. Here, we propose a model of minimal completeness of cell death signaling induced by OS as a mechanistic explanation of neuronal and cancer cell demise. This mechanism might provide the basis for therapeutic design strategies. Finally, we will attempt to associate PD, cancer, and OS. This paper critically analyzes the evidence that support the “oxidative stress model” in neurodegeneration and cancer.
Collapse
|
42
|
Hong YK, Lee S, Park SH, Lee JH, Han SY, Kim ST, Kim YK, Jeon S, Koo BS, Cho KS. Inhibition of JNK/dFOXO pathway and caspases rescues neurological impairments in Drosophila Alzheimer’s disease model. Biochem Biophys Res Commun 2012; 419:49-53. [DOI: 10.1016/j.bbrc.2012.01.122] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 01/25/2012] [Indexed: 01/10/2023]
|
43
|
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
|
44
|
Akter K, Lanza EA, Martin SA, Myronyuk N, Rua M, Raffa RB. Diabetes mellitus and Alzheimer's disease: shared pathology and treatment? Br J Clin Pharmacol 2011; 71:365-76. [PMID: 21284695 DOI: 10.1111/j.1365-2125.2010.03830.x] [Citation(s) in RCA: 237] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Epidemiological and basic science evidence suggest a possible shared pathophysiology between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD). It has even been hypothesized that AD might be 'type 3 diabetes'. The present review summarizes some of the evidence for the possible link, putative biochemical pathways and ongoing clinical trials of antidiabetic drugs in AD patients. The primary and review literature were searched for articles published in peer-reviewed sources that were related to a putative connection between T2DM and AD. In addition, public sources of clinical trials were searched for the relevant information regarding the testing of antidiabetic drugs in AD patients. The evidence for a connection between T2DM and AD is based upon a variety of diverse studies, but definitive biochemical mechanisms remain unknown. Additional study is needed to prove the existence or the extent of a link between T2DM and AD, but sufficient evidence exists to warrant further study. Presently, AD patients might benefit from treatment with pharmacotherapy currently used to treat T2DM and clinical trials of such therapy are currently underway.
Collapse
Affiliation(s)
- Kawser Akter
- Temple University School of Pharmacy, Philadelphia, PA19140, USA
| | | | | | | | | | | |
Collapse
|
45
|
Moh C, Kubiak JZ, Bajic VP, Zhu X, Smith MA, Lee HG. Cell cycle deregulation in the neurons of Alzheimer's disease. Results Probl Cell Differ 2011; 53:565-76. [PMID: 21630160 PMCID: PMC5925746 DOI: 10.1007/978-3-642-19065-0_23] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cell cycle consists of four main phases: G(1), S, G(2), and M. Most cells undergo these cycles up to 40-60 times in their life. However, neurons remain in a nondividing, nonreplicating phase, G(0). Neurons initiate but do not complete cell division, eventually entering apoptosis. Research has suggested that like cancer, Alzheimer's disease (AD) involves dysfunction in neuronal cell cycle reentry, leading to the development of the two-hit hypothesis of AD. The first hit is abnormal cell cycle reentry, which typically results in neuronal apoptosis and prevention of AD. However, with the second hit of chronic oxidative damage preventing apoptosis, neurons gain "immortality" analogous to tumor cells. Once both of these hits are activated, AD can develop and produce senile plaques and neurofibrillary tangles throughout brain tissue. In this review, we propose a mechanism for neuronal cell cycle reentry and the development of AD.
Collapse
Affiliation(s)
- Calvin Moh
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
46
|
Design and synthesis of disubstituted thiophene and thiazole based inhibitors of JNK. Bioorg Med Chem Lett 2010; 20:7303-7. [DOI: 10.1016/j.bmcl.2010.10.066] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 10/11/2010] [Accepted: 10/14/2010] [Indexed: 11/24/2022]
|
47
|
Shen S, Callaghan D, Juzwik C, Xiong H, Huang P, Zhang W. ABCG2 reduces ROS-mediated toxicity and inflammation: a potential role in Alzheimer's disease. J Neurochem 2010; 114:1590-604. [PMID: 20626554 DOI: 10.1111/j.1471-4159.2010.06887.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is characterized by accumulation and deposition of Aβ peptides in the brain. Aβ deposition generates reactive-oxygen species (ROS), which are involved in Alzheimer's inflammatory and neurodegenerative pathology. We have previously observed that, in Alzheimer's disease brain, ABCG2 is up-regulated and AP-1 is activated, but NF-κB is not activated. In the present study, we examine the roles and mechanism of ABCG2 on ROS generation, inflammatory gene expression and signaling, heme homeostasis and Aβ production in cell models and on inflammatory signaling and Aβ deposition in Abcg2-knockout and wild-type mice. Our results show that ABCG2 plays a protective role against oxidative stress by decreasing ROS generation, enhancing antioxidant capacity, regulating heme level, and inhibiting inflammatory response in cell models. ABCG2 inhibits NF-κB activation but has less effect on AP-1 activation induced by ROS. This results in inhibition of interleukin-8 and growth-related oncogene (GRO) expression induced by ROS via NF-κB pathway. Abcg2 deficiency increased Aβ deposition and NF-κB activation in the brains of Abcg2-knockout mice compared with controls. These findings suggest that ABCG2 may relieve oxidative stress and inflammatory response via inhibiting NF-κB signaling pathway in cell models and brain tissues and thus may play a potential protective role in Alzheimer's neuroinflammatory response.
Collapse
Affiliation(s)
- Shanshan Shen
- Department of Clinical Medicine, Southeast University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
48
|
Jing Z, Caltagarone J, Bowser R. Altered subcellular distribution of c-Abl in Alzheimer's disease. J Alzheimers Dis 2009; 17:409-22. [PMID: 19363261 DOI: 10.3233/jad-2009-1062] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
c-Abl is a non-receptor tyrosine kinase that participates in multiple signaling pathways linking the cell surface, cytoskeleton, and the nucleus. Recent in vitro studies have also linked c-Abl to amyloid-beta-induced toxicity and tau phosphorylation. To further characterize a potential role of c-Abl in Alzheimer's disease (AD), we examined the expression and distribution of total and phosphorylated forms of c-Abl in the hippocampus of AD and control subjects. Laser scanning confocal microscopy was used to examine the colocalization of c-Abl with AD pathology. Our results demonstrate alterations in the presence and distribution of c-Abl and phosphorylated isoforms of c-Abl within the hippocampus during AD. Total unphosphorylated c-Abl was highest in non-demented control hippocampus. Activated isoforms of c-Abl were most abundant in AD hippocampus and co-localized with AD pathology, including granulovacuolar degeneration bodies, c-Abl interacts with phosphorylated tau in AD brain and may contribute to the formation of tau pathology. These studies demonstrate altered activation and distribution of c-Abl during AD, suggesting a role for c-Abl in Abeta signal transduction and generation of tau pathology in AD.
Collapse
Affiliation(s)
- Zheng Jing
- Department of Pathology, University of Pittsburgh, School of Medicine, BST S-420, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
49
|
Tamagno E, Guglielmotto M, Giliberto L, Vitali A, Borghi R, Autelli R, Danni O, Tabaton M. JNK and ERK1/2 pathways have a dual opposite effect on the expression of BACE1. Neurobiol Aging 2009; 30:1563-73. [DOI: 10.1016/j.neurobiolaging.2007.12.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 12/17/2007] [Accepted: 12/19/2007] [Indexed: 12/29/2022]
|
50
|
Sun KH, Lee HG, Smith MA, Shah K. Direct and indirect roles of cyclin-dependent kinase 5 as an upstream regulator in the c-Jun NH2-terminal kinase cascade: relevance to neurotoxic insults in Alzheimer's disease. Mol Biol Cell 2009; 20:4611-9. [PMID: 19776350 DOI: 10.1091/mbc.e09-05-0433] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Significant increase in JNK, c-Jun, and Cdk5 activities are reported in Alzheimer's disease (AD). Inhibition of c-Jun prevents neuronal cell death in in vivo AD models, highlighting it as a major JNK effector. Both JNK and Cdk5 promote neurodegeneration upon deregulation; however, Cdk5 has not been mechanistically linked to JNK or c-Jun. This study presents the first mechanism showing Cdk5 as a major regulator of the JNK cascade. Deregulated Cdk5 induces biphasic activation of JNK pathway. The first phase revealed c-Jun as a direct substrate of Cdk5, whose activation is independent of reactive oxygen species (ROS) and JNK. In the second phase, Cdk5 activates c-Jun via ROS-mediated activation of JNK. Rapid c-Jun activation is supported by in vivo data showing c-Jun phosphorylation in cerebral cortex upon p25 induction in transgenic mice. Cdk5-mediated biphasic activation of c-Jun highlights c-Jun, rather than JNK, as an important therapeutic target, which was confirmed in neuronal cells. Finally, Cdk5 inhibition endows superior protection against neurotoxicity, suggesting that Cdk5 is a preferable therapeutic target for AD relative to JNK and c-Jun.
Collapse
Affiliation(s)
- Kai-Hui Sun
- Department of Chemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|