1
|
Patel R, Cheng R, Cardona CL, Angeles E, Singh G, Miller S, Ashok A, Teich AF, Piriz A, Maldonado A, Jimenez-Velazquez IZ, Mayeux R, Lee JH, Sproul AA. Reduced SH3RF3 may protect against Alzheimer's disease by lowering microglial pro-inflammatory responses via modulation of JNK and NFkB signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.23.600281. [PMID: 38979369 PMCID: PMC11230201 DOI: 10.1101/2024.06.23.600281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding how high-risk individuals are protected from Alzheimer's disease (AD) may illuminate potential therapeutic targets. We identified protective genetic variants in SH3RF3/POSH2 that delayed the onset of AD among individuals carrying the PSEN1 G206A mutation. SH3RF3 acts as a JNK pathway scaffold and activates NFκB signaling. While effects of SH3RF3 knockdown in human neurons were subtle, including decreased ptau S422, knockdown in human microglia significantly reduced inflammatory cytokines in response to either a viral mimic or oAβ42. This was associated with reduced activation of JNK and NFκB pathways in response to these stimuli. Pharmacological inhibition of JNK or NFκB signaling phenocopied SH3RF3 knockdown. We also found PSEN1 G206A microglia had reduced inflammatory response to oAβ42. Thus, further reduction of microglial inflammatory responses in PSEN1 G206A mutant carriers by protective variants in SH3RF3 might reduce the link between amyloid and neuroinflammation to subsequently delay the onset of AD.
Collapse
|
2
|
MicroRNAs and MAPKs: Evidence of These Molecular Interactions in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054736. [PMID: 36902178 PMCID: PMC10003111 DOI: 10.3390/ijms24054736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder known to be the leading cause of dementia worldwide. Many microRNAs (miRNAs) were found deregulated in the brain or blood of AD patients, suggesting a possible key role in different stages of neurodegeneration. In particular, mitogen-activated protein kinases (MAPK) signaling can be impaired by miRNA dysregulation during AD. Indeed, the aberrant MAPK pathway may facilitate the development of amyloid-beta (Aβ) and Tau pathology, oxidative stress, neuroinflammation, and brain cell death. The aim of this review was to describe the molecular interactions between miRNAs and MAPKs during AD pathogenesis by selecting evidence from experimental AD models. Publications ranging from 2010 to 2023 were considered, based on PubMed and Web of Science databases. According to obtained data, several miRNA deregulations may regulate MAPK signaling in different stages of AD and conversely. Moreover, overexpressing or silencing miRNAs involved in MAPK regulation was seen to improve cognitive deficits in AD animal models. In particular, miR-132 is of particular interest due to its neuroprotective functions by inhibiting Aβ and Tau depositions, as well as oxidative stress, through ERK/MAPK1 signaling modulation. However, further investigations are required to confirm and implement these promising results.
Collapse
|
3
|
Interaction between TRPML1 and p62 in Regulating Autophagosome-Lysosome Fusion and Impeding Neuroaxonal Dystrophy in Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8096009. [PMID: 35116093 PMCID: PMC8807035 DOI: 10.1155/2022/8096009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022]
Abstract
The loss of transient receptor potential mucolipin 1 (TRPML1), an endosomal and lysosomal Ca2+-releasing channel, has been implicated in neurodegenerative disorders. Mounting evidence have shown that TRPML1 could clear intraneuronal amyloid-β (Aβ), which triggers a hypothesis that TRPML1 activation may be beneficial for axonal transport in Alzheimer's disease (AD). In this work, the functional roles of TRPML1 were studied in the APP/PS1 transgenic mice and Aβ1-42-stimulated hippocampal neurons HT22. We found that lentivirus-mediated overexpression of TRPML1 was shown to promote an accumulation of autolysosomes and increase brain-derived neurotrophic factor (BDNF) transportation to the nucleus, suggesting an axon-protective function. More importantly, we found that TRPML1 also increased p62 that interacted with dynein. Lentivirus-mediated knockdown of p62 or inhibition of dynein by ciliobrevin D stimulation was found to reduce autolysosome formation and nuclear accumulation of BDNF in HT22 cells with Aβ1-42 stimulation. Inhibition of p62 by XRK3F2 stimulation was observed to promote the death of hippocampal neurons of the APP/PS1 transgenic mice. TRPML1 recruited dynein by interacting with p62 to promote the autophagosome-lysosome fusion to mediate BDNF nuclear translocation to impede axon dystrophy in mice with Alzheimer-like phenotypes. In summary, these results demonstrate the presence of a TRPML1/p62/dynein regulatory network in AD, and activation of TRPML1 is required for axon protection to prevent neuroaxonal dystrophy.
Collapse
|
4
|
Liu X, Chu W, Shang S, Ma L, Jiang C, Ding Y, Wang J, Zhang S, Shao B. Preliminary study on the anti-apoptotic mechanism of Astragaloside IV on radiation-induced brain cells. Int J Immunopathol Pharmacol 2021; 34:2058738420954594. [PMID: 32902354 PMCID: PMC7485151 DOI: 10.1177/2058738420954594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With multiple targets and low cytotoxicity, natural medicines can be used as potential neuroprotective agents. The increase in oxidative stress levels and inflammatory responses in the brain caused by radiation affects cognitive function and neuronal structure, and ultimately leads to abnormal changes in neurogenesis, differentiation, and apoptosis. Astragaloside Ⅳ (AS-Ⅳ), one of the main active constituents of astragalus, is known for its antioxidant, antihypertensive, antidiabetic, anti-infarction, anti-inflammatory, anti-apoptotic and wound healing, angiogenesis, and other protective effects. In this study, the mechanism of AS-IV against radiation-induced apoptosis of brain cells in vitro and in vivo was explored by radiation modeling, which provided a theoretical basis for the development of anti-radiation Chinese herbal active molecules and brain health products. In order to study the protective mechanism of AS-IV on radiation-induced brain cell apoptosis in mice, the paper constructed a radiation-induced brain cell apoptosis model, using TUNEL staining, flow cytometry, Western blotting to analyze AS-IV resistance mechanism to radiation-induced brain cell apoptosis. The results of TUNEL staining and flow cytometry showed that the apoptosis rate of radiation group was significantly increased. The results of Western blotting indicated that the expression levels of p-JNK, p-p38, p53, Caspase-9 and Caspase-3 protein, and the ratio of Bax to Bcl-2 in radiation group were significantly increased. There was no significant difference in the expression levels of JNK and p38. After AS-IV treatment, the apoptosis was reduced and the expression of apoptosis related proteins was changed. These data suggested that AS-IV can effectively reduce radiation-induced apoptosis of brain cells, and its mechanism may be related to the phosphorylation regulation of JNK-p38.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Weiwei Chu
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Shuying Shang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Liang Ma
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Chenxin Jiang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yanping Ding
- School wof Life Sciences, Northwest Normal University, Lanzhou, Gansu Province, China
| | - Jianlin Wang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Shengxiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Baoping Shao
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
5
|
Rehman IU, Ahmad R, Khan I, Lee HJ, Park J, Ullah R, Choi MJ, Kang HY, Kim MO. Nicotinamide Ameliorates Amyloid Beta-Induced Oxidative Stress-Mediated Neuroinflammation and Neurodegeneration in Adult Mouse Brain. Biomedicines 2021; 9:biomedicines9040408. [PMID: 33920212 PMCID: PMC8070416 DOI: 10.3390/biomedicines9040408] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) is the most predominant age-related neurodegenerative disease, pathologically characterized by the accumulation of aggregates of amyloid beta Aβ1–42 and tau hyperphosphorylation in the brain. It is considered to be the primary cause of cognitive dysfunction. The aggregation of Aβ1–42 leads to neuronal inflammation and apoptosis. Since vitamins are basic dietary nutrients that organisms need for their growth, survival, and other metabolic functions, in this study, the underlying neuroprotective mechanism of nicotinamide (NAM) Vitamin B3 against Aβ1–42 -induced neurotoxicity was investigated in mouse brains. Intracerebroventricular (i.c.v.) Aβ1–42 injection elicited neuronal dysfunctions that led to memory impairment and neurodegeneration in mouse brains. After 24 h after Aβ1–42 injection, the mice were treated with NAM (250 mg/kg intraperitoneally) for 1 week. For biochemical and Western blot studies, the mice were directly sacrificed, while for confocal and “immunohistochemical staining”, mice were perfused transcardially with 4% paraformaldehyde. Our biochemical, immunofluorescence, and immunohistochemical results showed that NAM can ameliorate neuronal inflammation and apoptosis by reducing oxidative stress through lowering malondialdehyde and 2,7-dichlorofluorescein levels in an Aβ1–42-injected mouse brains, where the regulation of p-JNK further regulated inflammatory marker proteins (TNF-α, IL-1β, transcription factor NF-kB) and apoptotic marker proteins (Bax, caspase 3, PARP1). Furthermore, NAM + Aβ treatment for 1 week increased the amount of survival neurons and reduced neuronal cell death in Nissl staining. We also analyzed memory dysfunction via behavioral studies and the analysis showed that NAM could prevent Aβ1–42 -induced memory deficits. Collectively, the results of this study suggest that NAM may be a potential preventive and therapeutic candidate for Aβ1–42 -induced reactive oxygen species (ROS)-mediated neuroinflammation, neurodegeneration, and neurotoxicity in an adult mouse model.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Jungsung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Myeong Jun Choi
- Research and Development Center, Axceso Bio-pharma co, Anyang 14056, Korea;
| | - Hee Young Kang
- Department of Neurology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52828, Korea;
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
6
|
Saha P, Guha S, Biswas SC. P38K and JNK pathways are induced by amyloid-β in astrocyte: Implication of MAPK pathways in astrogliosis in Alzheimer's disease. Mol Cell Neurosci 2020; 108:103551. [PMID: 32896578 DOI: 10.1016/j.mcn.2020.103551] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocyte activation is one of the crucial hallmarks of Alzheimer's disease (AD) along with amyloid-β (Aβ) plaques, neurofibrillary tangles and neuron death. Glial scar and factors secreted from activated astrocytes have important contribution on neuronal health in AD. In this study, we investigated the mechanisms of astrocyte activation both in in vitro and in vivo models of AD. In this regard, mitogen activated protein kinase (MAPK) signalling cascades that control several fundamental and stress related cellular events, has been implicated in astrocyte activation in various neurological diseases. We checked activation of different MAPKs by western blot and immunocytochemistry and found that both JNK and p38K, but not ERK pathways are activated in Aβ-treated astrocytes in culture and in Aβ-infused rat brain cortex. Next, to investigate the downstream consequences of these two MAPKs (JNK and p38K) in Aβ-induced astrocyte activation, we individually blocked these pathways by specific inhibitors in presence and absence of Aβ and checked Aβ-induced cellular proliferation, morphological changes and glial fibrillary acidic protein (GFAP) upregulation. We found that activation of both JNK and p38K signalling cascades are involved in astrocyte proliferation evoked by Aβ, whereas only p38K pathway is implicated in morphological changes and GFAP upregulation in astrocytes exposed to Aβ. To further validate the implication of p38K pathway in Aβ-induced astrocyte activation, we also observed that transcription factor ATF2, a downstream phosphorylation substrate of p38, is phosphorylated upon Aβ treatment. Taken together, our study indicates that p38K and JNK pathways mediate astrocyte activation and both the pathways are involved in cellular proliferation but only p38K pathway contributes in morphological changes triggered by Aβ.
Collapse
Affiliation(s)
- Pampa Saha
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India
| | - Subhalakshmi Guha
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India
| | - Subhas Chandra Biswas
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India.
| |
Collapse
|
7
|
Hu YD, Tang CL, Jiang JZ, Lv HY, Wu YB, Qin XD, Shi S, Zhao B, Zhu XN, Xia ZY. Neuroprotective Effects of Dexmedetomidine Preconditioning on Oxygen-glucose Deprivation-reoxygenation Injury in PC12 Cells via Regulation of Ca 2+-STIM1/Orai1 Signaling. Curr Med Sci 2020; 40:699-707. [PMID: 32862381 DOI: 10.1007/s11596-020-2201-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/30/2020] [Indexed: 12/26/2022]
Abstract
Dexmedetomidine (DEX), a potent and highly selective agonist for α2-adrenergic receptors (α2AR), exerts neuroprotective effects by reducing apoptosis through decreased neuronal Ca2+ influx. However, the exact action mechanism of DEX and its effects on oxygen-glucose deprivation-reoxygenation (OGD/R) injury in vitro are unknown. We demonstrate that DEX pretreatment reduced OGD/R injury in PC12 cells, as evidenced by decreased oxidative stress, autophagy, and neuronal apoptosis. Specifically, DEX pretreatment decreased the expression levels of stromal interaction molecule 1 (STIM1) and calcium release-activated calcium channel protein 1 (Orai1), and reduced the concentration of intracellular calcium pools. In addition, variations in cytosolic calcium concentration altered apoptosis rate of PC12 cells after exposure to hypoxic conditions, which were modulated through STIM1/Orai1 signaling. Moreover, DEX pretreatment decreased the expression levels of Beclin-1 and microtubule-associated protein 1A/1B-light chain 3 (LC3), hallmark markers of autophagy, and the formation of autophagosomes. In conclusion, these results suggested that DEX exerts neuroprotective effects against oxidative stress, autophagy, and neuronal apoptosis after OGD/R injury via modulation of Ca2+-STIM1/Orai1 signaling. Our results offer insights into the molecular mechanisms of DEX in protecting against neuronal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yi-da Hu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chao-Liang Tang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Jia-Zhen Jiang
- Department of Emergency, Huashan Hospital North, Fudan University, Shanghai, 201907, China
| | - Hai-Yan Lv
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Yuan-Bo Wu
- Department of Neurology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xiu-de Qin
- Department of Neurology, Shenzhen Traditional Chinese Medicine Hospital, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Si Shi
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao-Nan Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
8
|
Falcicchia C, Tozzi F, Arancio O, Watterson DM, Origlia N. Involvement of p38 MAPK in Synaptic Function and Dysfunction. Int J Mol Sci 2020; 21:ijms21165624. [PMID: 32781522 PMCID: PMC7460549 DOI: 10.3390/ijms21165624] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer’s disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aβ peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy;
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA;
| | | | - Nicola Origlia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-3153193
| |
Collapse
|
9
|
Ullah R, Khan M, Shah SA, Saeed K, Kim MO. Natural Antioxidant Anthocyanins-A Hidden Therapeutic Candidate in Metabolic Disorders with Major Focus in Neurodegeneration. Nutrients 2019; 11:E1195. [PMID: 31141884 PMCID: PMC6628002 DOI: 10.3390/nu11061195] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
All over the world, metabolic syndrome constitutes severe health problems. Multiple factors have been reported in the pathogenesis of metabolic syndrome. Metabolic disorders result in reactive oxygen species (ROS) induced oxidative stress, playing a vital role in the development and pathogenesis of major health issues, including neurological disorders Alzheimer's disease (AD) Parkinson's disease (PD). Considerable increasing evidence indicates the substantial contribution of ROS-induced oxidative stress in neurodegenerative diseases. An imbalanced metabolism results in a defective antioxidant defense system, free radicals causing inflammation, cellular apoptosis, and tissue damage. Due to the annual increase in financial and social burdens, in addition to the adverse effects associated with available synthetic agents, treatment diversion from synthetic to natural approaches has occurred. Antioxidants are now being considered as convincing therapeutic agents against various neurodegenerative disorders. Therefore, medicinal herbs and fruits currently receive substantially more attention as commercial sources of antioxidants. In this review, we argue that ROS-targeted therapeutic interventions with naturally occurring antioxidant flavonoid, anthocyanin, and anthocyanin-loaded nanoparticles might be the ultimate treatment against devastating illnesses. Furthermore, we elucidate the hidden potential of the neuroprotective role of anthocyanins and anthocyanin-loaded nanoparticles in AD and PD neuropathies, which lack sufficient attention compared with other polyphenols, despite their strong antioxidant potential. Moreover, we address the need for future research studies of native anthocyanins and nano-based-anthocyanins, which will be helpful in developing anthocyanin treatments as therapeutic mitochondrial antioxidant drug-like regimens to delay or prevent the progression of neurodegenerative diseases, such as AD and PD.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Mehtab Khan
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Shahid Ali Shah
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
- Department of Chemistry, Sarhad University of Science & Information Technology (SUIT), Peshawar Khyber Pakhtunkhwa 25000, Pakistan.
| | - Kamran Saeed
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| |
Collapse
|
10
|
Morroniside prevents H2O2 or Aβ1–42-induced apoptosis via attenuating JNK and p38 MAPK phosphorylation. Eur J Pharmacol 2018; 834:295-304. [DOI: 10.1016/j.ejphar.2018.07.047] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/13/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
|
11
|
Son HJ, Jeong YJ, Yoon HJ, Lee SY, Choi GE, Park JA, Kim MH, Lee KC, Lee YJ, Kim MK, Cho K, Kang DY. Assessment of brain beta-amyloid deposition in transgenic mouse models of Alzheimer's disease with PET imaging agents 18F-flutemetamol and 18F-florbetaben. BMC Neurosci 2018; 19:45. [PMID: 30053803 PMCID: PMC6063010 DOI: 10.1186/s12868-018-0447-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although amyloid beta (Aβ) imaging is widely used for diagnosing and monitoring Alzheimer's disease in clinical fields, paralleling comparison between 18F-flutemetamol and 18F-florbetaben was rarely attempted in AD mouse model. We performed a comparison of Aβ PET images between 18F-flutemetamol and 18F-florbetaben in a recently developed APPswe mouse model, C57BL/6-Tg (NSE-hAPPsw) Korl. RESULTS After an injection (0.23 mCi) of 18F-flutemetamol and 18F-florbetaben at a time interval of 2-3 days, we compared group difference of SUVR and kinetic parameters between the AD (n = 7) and control (n = 7) mice, as well as between 18F-flutemetamol and 18F-florbetaben image. In addition, bio-distribution and histopathology were conducted. With visual image and VOI-based SUVR analysis, the AD group presented more prominent uptake than did the control group in both the 18F-florbetaben and 18F-flutemetamol images. With kinetic analysis, the 18F-florbetaben images showed differences in K1 and k4 between the AD and control groups, although 18F-flutemetamol images did not show significant difference. 18F-florbetaben images showed more prominent cortical uptake and matched well to the thioflavin S staining images than did the 18F-flutemetamol image. In contrast, 18F-flutemetamol images presented higher K1, k4, K1/k2 values than those of 18F-florbetaben images. Also, 18F-flutemetamol images presented prominent uptake in the bowel and bladder, consistent with higher bio-distribution in kidney, lung, blood and heart. CONCLUSIONS Compared with 18F-flutemetamol images, 18F-florbetaben images showed prominent visual uptake intensity, SUVR, and higher correlations with the pathology. In contrast, 18F-flutemetamol was more actively metabolized than was 18F-florbetaben (Son et al. in J Nucl Med 58(Suppl 1):S278, 2017].
Collapse
Affiliation(s)
- Hye Joo Son
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Young Jin Jeong
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Hyun Jin Yoon
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Sang Yoon Lee
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Go-Eun Choi
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Ji-Ae Park
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Min Hwan Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yong Jin Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Mun Ki Kim
- Pohang Center of Evolution of Biomaterials, Pohang Technopark, Pohang, Korea
| | - Kook Cho
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Do-Young Kang
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| |
Collapse
|
12
|
Kiyota T, Machhi J, Lu Y, Dyavarshetty B, Nemati M, Zhang G, Mosley RL, Gelbard HA, Gendelman HE. URMC-099 facilitates amyloid-β clearance in a murine model of Alzheimer's disease. J Neuroinflammation 2018; 15:137. [PMID: 29729668 PMCID: PMC5935963 DOI: 10.1186/s12974-018-1172-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/23/2018] [Indexed: 01/19/2023] Open
Abstract
Background The mixed lineage kinase type 3 inhibitor URMC-099 facilitates amyloid-beta (Aβ) clearance and degradation in cultured murine microglia. One putative mechanism is an effect of URMC-099 on Aβ uptake and degradation. As URMC-099 promotes endolysosomal protein trafficking and reduces Aβ microglial pro-inflammatory activities, we assessed whether these responses affect Aβ pathobiogenesis. To this end, URMC-099’s therapeutic potential, in Aβ precursor protein/presenilin-1 (APP/PS1) double-transgenic mice, was investigated in this model of Alzheimer’s disease (AD). Methods Four-month-old APP/PS1 mice were administered intraperitoneal URMC-099 injections at 10 mg/kg daily for 3 weeks. Brain tissues were examined by biochemical, molecular and immunohistochemical tests. Results URMC-099 inhibited mitogen-activated protein kinase 3/4-mediated activation and attenuated β-amyloidosis. Microglial nitric oxide synthase-2 and arginase-1 were co-localized with lysosomal-associated membrane protein 1 (Lamp1) and Aβ. Importatly, URMC-099 restored synaptic integrity and hippocampal neurogenesis in APP/PS1 mice. Conclusions URMC-099 facilitates Aβ clearance in the brain of APP/PS1 mice. The multifaceted immune modulatory and neuroprotective roles of URMC-099 make it an attractive candidate for ameliorating the course of AD. This is buttressed by removal of pathologic Aβ species and restoration of the brain’s microenvironment during disease.
Collapse
Affiliation(s)
- Tomomi Kiyota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhagyalaxmi Dyavarshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maryam Nemati
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gang Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
13
|
Ning C, Wang HMD, Gao R, Chang YC, Hu F, Meng X, Huang SY. Marine-derived protein kinase inhibitors for neuroinflammatory diseases. Biomed Eng Online 2018; 17:46. [PMID: 29690896 PMCID: PMC5916827 DOI: 10.1186/s12938-018-0477-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/17/2018] [Indexed: 12/29/2022] Open
Abstract
Neuroinflammation is primarily characterized by overexpression of proinflammatory mediators produced by glial activation or immune cell infiltration. Several kinases have been shown to be critical mediators in neuroinflammation. One of the largest groups of kinases is protein kinases, which have been the second most studied group of drug targets after G-protein-coupled receptors. Thus far, most of the approved kinase inhibitor drugs are adenosine triphosphate-competitive inhibitors with various off-target liabilities because of cross-reactivities; however, marine-derived compounds provide opportunities for discovering allosteric kinase inhibitors. This review summarizes the potential of marine-derived protein kinase inhibitors in the field of neuroinflammatory diseases, such as Parkinson disease, Alzheimer disease, multiple sclerosis, and pain. The previous studies from 1990 to 2017 in this review have shown that marine-derived protein kinase inhibitors have great potential to elicit anti-neuroinflammatory or neuroprotective responses in in vitro and in vivo models of neuroinflammatory diseases. This suggests that further exploration and investigation of these marine-derived protein kinase inhibitors on neuroinflammatory diseases are warranted. Therefore, this review may inspire further discovery of new protein kinase inhibitors from a marine origin and additional neuroscience studies focusing on these valuable marine-derived protein kinase inhibitors.
Collapse
Affiliation(s)
- Chong Ning
- College of Light Industry, Liaoning University, Shenyang, 110036, China
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 40227, Taiwan.,College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Rong Gao
- Yangtze Delta Region Institute of Tsinghua University, Zhejiang, 314006, China.,Jiaxing Deqin Biotechnology Co., Ltd, Zhejiang, 314006, China
| | - Yu-Chia Chang
- Greenhouse Systems Technology Center, Central Region Campus, Industrial Technology Research Institute, Nantou, 540, Taiwan
| | - Fengqing Hu
- College of Light Industry, Liaoning University, Shenyang, 110036, China
| | - Xianjun Meng
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Shi-Ying Huang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China. .,Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou, 362000, China. .,Key Laboratory of Inshore Resources Biotechnology (Quanzhou Normal University) Fujian Province University, Quanzhou, 362000, China.
| |
Collapse
|
14
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
15
|
Abdullah M, Takase H, Nunome M, Enomoto H, Ito JI, Gong JS, Michikawa M. Amyloid-β Reduces Exosome Release from Astrocytes by Enhancing JNK Phosphorylation. J Alzheimers Dis 2018; 53:1433-41. [PMID: 27392863 DOI: 10.3233/jad-160292] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Exosomes are small extracellular vesicles secreted by variety of cell types such as neurons, astrocytes, and oligodendrocytes. It is suggested that exosomes play essential role in the maintenance of the neuronal functions and also in the clearance of amyloid-β (Aβ) from the brain. Aβ is well known to cause neuronal cell death, whereas little is known about its effect on astrocytes. In this study, we examined the effect of Aβ on release of exosomes from astrocytes in culture. We analyzed release of exosomes and apoE, both of which are known to remove/clear Aβ from the brain, in the culture medium of astrocytes. We found that exosome and apoE-HDL were successfully separated by density gradient ultracentrifugation demonstrated by distribution of their specific markers, flotillin and HSP90, and cholesterol, and morphological analysis using electron microscopy. Exosome release was significantly reduced by Aβ1-42 treatment in cultured astrocytes accompanied by an increased JNK phosphorylation. Whereas, apoE-HDL release remained unchanged. A JNK inhibitor restored the decreased levels of exosome release induced by Aβ treatment to levels similar to those of control, suggesting that Aβ1-42 inhibits exosome release via stimulation of JNK signal pathway. Because exosomes are shown to remove Aβ in the brain, our findings suggest that increased Aβ levels in the brain may impair the exosome-mediated Aβ clearance pathway.
Collapse
Affiliation(s)
- Mohammad Abdullah
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Hiroshi Takase
- Core Laboratory, Graduate School of Medical Sciences, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Mari Nunome
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Hiroyuki Enomoto
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Jin-Ichi Ito
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Jian-Sheng Gong
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Nagoya City University, Mizuho-cyo, Mizuho-ku, Nagoya, Aichi, Japan
| |
Collapse
|
16
|
Alzheimer's disease pathology and the unfolded protein response: prospective pathways and therapeutic targets. Behav Pharmacol 2018; 28:161-178. [PMID: 28252521 DOI: 10.1097/fbp.0000000000000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many vital interdependent cellular functions including proteostasis, lipogenesis and Ca homeostasis are executed by the endoplasmic reticulum (ER). Exogenous insults can impair ER performance: this must be rapidly corrected or cell death will ensue. Protective adaptations can boost the functional capacity of the ER and form the basis of the unfolded protein response (UPR). Activated in response to the accumulation of misfolded proteins, the UPR can halt protein translation while increasing protein-handling chaperones and the degradation of erroneous proteins through a conserved three-tier molecular cascade. However, prolonged activation of the UPR can result in the maladaptation of the system, resulting in the activation of inflammatory and apoptotic effectors. Recently, UPR and its involvement in neurodegenerative disease has attracted much interest and numerous potentially 'drugable' points of crosstalk are now emerging. Here, we summarize the functions of the ER and UPR, and highlight evidence for its potential role in the pathogenesis of Alzheimer's disease, before discussing several key targets with therapeutic potential.
Collapse
|
17
|
Gu Y, Ma LJ, Bai XX, Jie J, Zhang XF, Chen D, Li XP. Mitogen-activated protein kinase phosphatase 1 protects PC12 cells from amyloid beta-induced neurotoxicity. Neural Regen Res 2018; 13:1842-1850. [PMID: 30136701 PMCID: PMC6128043 DOI: 10.4103/1673-5374.238621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in the regulation of cell growth, proliferation, differentiation, transformation and death. Mitogen-activated protein kinase phosphatase 1 (MKP1) has an inhibitory effect on the p38MAPK and JNK pathways, but it is unknown whether it plays a role in Aβ-induced oxidative stress and neuronal inflammation. In this study, PC12 cells were infected with MKP1 shRNA, MKP1 lentivirus or control lentivirus for 12 hours, and then treated with 0.1, 1, 10 or 100 μM amyloid beta 42 (Aβ42). The cell survival rate was measured using the cell counting kit-8 assay. MKP1, tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) mRNA expression levels were analyzed using quantitative real time-polymerase chain reaction. MKP1 and phospho-c-Jun N-terminal kinase (JNK) expression levels were assessed using western blot assay. Reactive oxygen species (ROS) levels were detected using 2′,7′-dichlorofluorescein diacetate. Mitochondrial membrane potential was measured using flow cytometry. Superoxide dismutase activity and malondialdehyde levels were evaluated using the colorimetric method. Lactate dehydrogenase activity was measured using a microplate reader. Caspase-3 expression levels were assessed by enzyme-linked immunosorbent assay. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase dUTP nick end labeling method. MKP1 overexpression inhibited Aβ-induced JNK phosphorylation and the increase in ROS levels. It also suppressed the Aβ-induced increase in TNF-α and IL-1β levels as well as apoptosis in PC12 cells. In contrast, MKP1 knockdown by RNA interference aggravated Aβ-induced oxidative stress, inflammation and cell damage in PC12 cells. Furthermore, the JNK-specific inhibitor SP600125 abolished this effect of MKP1 knockdown on Aβ-induced neurotoxicity. Collectively, these results show that MKP1 mitigates Aβ-induced apoptosis, oxidative stress and neuroinflammation by inhibiting the JNK signaling pathway, thereby playing a neuroprotective role.
Collapse
Affiliation(s)
- Yue Gu
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lian-Jun Ma
- Endoscopy Center, the China-Japan Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Xue Bai
- Cadre's Wards, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jing Jie
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiu-Fang Zhang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Dong Chen
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Ping Li
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
18
|
Zhang L, Fang Y, Cheng X, Lian YJ, Xu HL, Zeng ZS, Zhu HC. Curcumin Exerts Effects on the Pathophysiology of Alzheimer's Disease by Regulating PI(3,5)P2 and Transient Receptor Potential Mucolipin-1 Expression. Front Neurol 2017; 8:531. [PMID: 29062301 PMCID: PMC5640884 DOI: 10.3389/fneur.2017.00531] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 09/22/2017] [Indexed: 12/14/2022] Open
Abstract
Background To validate our speculation that curcumin may ameliorate Alzheimer’s disease (AD) pathogenesis by regulating PI(3,5)P2 and transient receptor potential mucolipin-1 (TRPML1) expression levels. Methods We developed an animal model presenting AD by APP/PS1 transgenes. The mouse clonal hippocampal neuronal cell line HT-22 was treated with amyloid-β1-42 (Aβ1-42). Curcumin was administrated both in vivo and in vitro. MTS assay was used to detect cell viability, and the lysosomal [Ca2+] ion concentration was detected. The number of autophagosomes was detected by the transmission electron microscopic examination. Illumina RNA-seq was used to analyze the different expression patterns between Aβ1-42-treated cells without and with curcumin treatment. The protein level was analyzed by the Western blotting analysis. PI(3,5)P2 or TRPML1 was knocked down in HT-22 cells or in APP/PS1 transgenic mice. Morris water maze and recognition task were performed to trace the cognitive ability. Results Curcumin increased cell viability, decreased the number of autophagosomes, and increased lysosomal Ca2+ levels in Aβ1-42-treated HT-22 cells. Sequencing analysis identified TRPLML1 as the most significantly upregulated gene after curcumin treatment. Western blotting results also showed that TRPML1 was upregulated and mTOR/S6K signaling pathway was activated and markers of the autophagy–lysosomal system were downregulated after curcumin use in Aβ1-42-treated HT-22 cells. Knockdown of PI (3,5)P2 or TRPML1 increased the protein levels of markers of the autophagy–lysosomal system after curcumin use in Aβ1-42-treated HT-22 cells, inhibited mTOR/S6K signaling pathway, increased the protein levels of markers of the autophagy–lysosomal system after curcumin use in APP/PS1 mice. Besides, knockdown of PI(3,5)P2 or TRPML1 reversed the protective role of curcumin on memory and recognition impairments in mice with APP/PS1 transgenes. Conclusion To some extent, it suggested that the effects of curcumin on AD pathogenesis were, at least partially, associated with PI(3,5)P2 and TRPML1 expression levels.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Fang
- Department of Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya-Jun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong-Liang Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhao-Shu Zeng
- Department of Legal Medicine, The College of Basic Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Hong-Can Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the world. The "amyloid hypothesis" is one of the predominant hypotheses for the pathogenesis of AD. Besides, tau protein accumulation, calcium homeostasis disruption, and glial cell activation are also remarkable features in AD. Recently, there are some reports showing that TRPC channels may function in AD development, especially TRPC6. In this chapter, we will discuss the evidence for the involvement of TRPC channels in Alzheimer's disease and the potential of therapeutics for AD based on TRPC channels.
Collapse
|
20
|
Liu Y, Jiang L, Li X. κ-carrageenan-derived pentasaccharide attenuates Aβ25–35-induced apoptosis in SH-SY5Y cells via suppression of the JNK signaling pathway. Mol Med Rep 2016; 15:285-290. [DOI: 10.3892/mmr.2016.6006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 09/15/2016] [Indexed: 11/05/2022] Open
|
21
|
Mao Y, Guo Z, Zheng T, Jiang Y, Yan Y, Yin X, Chen Y, Zhang B. Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging Cell 2016; 15:893-902. [PMID: 27457264 PMCID: PMC5013027 DOI: 10.1111/acel.12498] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2016] [Indexed: 11/26/2022] Open
Abstract
Brain insulin signaling deficits contribute to multiple pathological features of Alzheimer's disease (AD). Although intranasal insulin has shown efficacy in patients with AD, the underlying mechanisms remain largely unillustrated. Here, we demonstrate that intranasal insulin improves cognitive deficits, ameliorates defective brain insulin signaling, and strongly reduces β‐amyloid (Aβ) production and plaque formation after 6 weeks of treatment in 4.5‐month‐old APPswe/PS1dE9 (APP/PS1) mice. Furthermore, c‐Jun N‐terminal kinase activation, which plays a pivotal role in insulin resistance and AD pathologies, is significantly inhibited. The alleviation of amyloid pathology by intranasal insulin results mainly from enhanced nonamyloidogenic processing and compromised amyloidogenic processing of amyloid precursor protein (APP), and from a reduction in apolipoprotein E protein which is involved in Aβ metabolism. In addition, intranasal insulin effectively promotes hippocampal neurogenesis in APP/PS1 mice. This study, exploring the mechanisms underlying the beneficial effects of intranasal insulin on Aβ pathologies in vivo for the first time, highlights important preclinical evidence that intranasal insulin is potentially an effective therapeutic method for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yan‐Fang Mao
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Zhangyu Guo
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Tingting Zheng
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yasi Jiang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yaping Yan
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Xinzhen Yin
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yanxing Chen
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Baorong Zhang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| |
Collapse
|
22
|
Protein Kinases and Parkinson's Disease. Int J Mol Sci 2016; 17:ijms17091585. [PMID: 27657053 PMCID: PMC5037850 DOI: 10.3390/ijms17091585] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/09/2016] [Accepted: 09/01/2016] [Indexed: 01/09/2023] Open
Abstract
Currently, the lack of new drug candidates for the treatment of major neurological disorders such as Parkinson’s disease has intensified the search for drugs that can be repurposed or repositioned for such treatment. Typically, the search focuses on drugs that have been approved and are used clinically for other indications. Kinase inhibitors represent a family of popular molecules for the treatment and prevention of various cancers, and have emerged as strong candidates for such repurposing because numerous serine/threonine and tyrosine kinases have been implicated in the pathobiology of Parkinson’s disease. This review focuses on various kinase-dependent pathways associated with the expression of Parkinson’s disease pathology, and evaluates how inhibitors of these pathways might play a major role as effective therapeutic molecules.
Collapse
|
23
|
Liu K, Chojnacki JE, Wade EE, Saathoff JM, Lesnefsky EJ, Chen Q, Zhang S. Bivalent Compound 17MN Exerts Neuroprotection through Interaction at Multiple Sites in a Cellular Model of Alzheimer's Disease. J Alzheimers Dis 2016; 47:1021-33. [PMID: 26401780 DOI: 10.3233/jad-150242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple pathogenic factors have been suggested to play a role in the development of Alzheimer's disease (AD). The multifactorial nature of AD also suggests the potential use of compounds with polypharmacology as effective disease-modifying agents. Recently, we have developed a bivalent strategy to include cell membrane anchorage into the molecular design. Our results demonstrated that the bivalent compounds exhibited multifunctional properties and potent neuroprotection in a cellular AD model. Herein, we report the mechanistic exploration of one of the representative bivalent compounds, 17MN, in MC65 cells. Our results established that MC65 cells die through a necroptotic mechanism upon the removal of tetracycline (TC). Furthermore, we have shown that mitochondrial membrane potential and cytosolic Ca2+ levels are increased upon removal of TC. Our bivalent compound 17MN can reverse such changes and protect MC65 cells from TC removal induced cytotoxicity. The results also suggest that 17MN may function between the Aβ species and RIPK1 in producing its neuroprotection. Colocalization studies employing a fluorescent analog of 17MN and confocal microscopy demonstrated the interactions of 17MN with both mitochondria and endoplasmic reticulum, thus suggesting that 17MN exerts its neuroprotection via a multiple-site mechanism in MC65 cells. Collectively, these results strongly support our original design rationale of bivalent compounds and encourage further optimization of this bivalent strategy to develop more potent analogs as novel disease-modifying agents for AD.
Collapse
Affiliation(s)
- Kai Liu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Jeremy E Chojnacki
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Emily E Wade
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - John M Saathoff
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward J Lesnefsky
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA, USA.,Biochemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Qun Chen
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
24
|
p53-dependent SIRT6 expression protects Aβ42-induced DNA damage. Sci Rep 2016; 6:25628. [PMID: 27156849 PMCID: PMC4860716 DOI: 10.1038/srep25628] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/20/2016] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and age-related neurodegenerative disease. Elucidating the cellular changes that occur during ageing is an important step towards understanding the pathogenesis and progression of neurodegenerative disorders. SIRT6 is a member of the mammalian sirtuin family of anti-aging genes. However, the relationship between SIRT6 and AD has not yet been elucidated. Here, we report that SIRT6 protein expression levels are reduced in the brains of both the 5XFAD AD mouse model and AD patients. Aβ42, a major component of senile plaques, decreases SIRT6 expression, and Aβ42-induced DNA damage is prevented by the overexpression of SIRT6 in HT22 mouse hippocampal neurons. Also, there is a strong negative correlation between Aβ42-induced DNA damage and p53 levels, a protein involved in DNA repair and apoptosis. In addition, upregulation of p53 protein by Nutlin-3 prevents SIRT6 reduction and DNA damage induced by Aβ42. Taken together, this study reveals that p53-dependent SIRT6 expression protects cells from Aβ42-induced DNA damage, making SIRT6 a promising new therapeutic target for the treatment of AD.
Collapse
|
25
|
Kim H, Youn K, Ahn MR, Kim OY, Jeong WS, Ho CT, Jun M. Neuroprotective effect of loganin against Aβ25-35-induced injury via the NF-κB-dependent signaling pathway in PC12 cells. Food Funct 2016; 6:1108-16. [PMID: 25778782 DOI: 10.1039/c5fo00055f] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amyloid-beta (Aβ) protein, the main constituent of senile plaques, is believed to play a pivotal role in the pathogenesis of Alzheimer's disease (AD). AD is closely associated with inflammatory reactions which are considered to be responses to Aβ deposition. The present study investigated the effect of loganin on Aβ25-35-induced inflammatory damage and the underlying molecular mechanism of its neuroprotective action. Loganin predominantly prevented Aβ25-35-stimulated cell death through suppressing ROS generation, and attenuating apoptosis by inhibiting caspase-3 activity and regulating cell cycle. Furthermore, loganin suppressed the level of TNF-α and protein expression of iNOS and COX-2 in Aβ25-35-injured PC12 cells. These inhibitions appeared to correlate with the suppression of NF-κB activation by loganin, as pre-treating cells with loganin blocked the translocation of NF-κB into the nuclear compartment and degradation of the inhibitory subunit IκB. Loganin substantially inhibited phosphorylation of MAPKs including ERK1/2, p38 and JNK, which are closely related to regulation of NF-κB activation. Taken together, the results implied that loganin attenuated neuroinflammatory responses through the inactivation of NF-κB by NF-κB dependent inflammatory pathways and phosphorylation of MAPK in Aβ25-35-induced PC12 cells.
Collapse
Affiliation(s)
- Hyeri Kim
- Department of Food Science and Nutrition, Dong-A University, Busan 604-714, Korea.
| | | | | | | | | | | | | |
Collapse
|
26
|
Mohammadi M, Guan J, Khodagholi F, Yans A, Khalaj S, Gholami M, Taghizadeh GH, Aliaghaei A, Abdollahi M, Ghahremani MH, Sharifzadeh M. Reduction of autophagy markers mediated protective effects of JNK inhibitor and bucladesine on memory deficit induced by Aβ in rats. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:501-10. [PMID: 26899864 DOI: 10.1007/s00210-016-1222-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/10/2016] [Indexed: 12/15/2022]
Abstract
Autophagy, the process of self-degradation of cellular components, has an important role in neurodegenerative diseases, such as Alzheimer's disease. In this study, we investigated the effects of SP600125 as c-Jun N-terminal kinase (JNK) inhibitor and bucladesine as a cyclic adenosine 3',5'-monophosphate (cAMP) analog on spatial memory and expression of autophagic factors in Aβ-injected rats. Male Wistar rats were used. Rats were randomly allocated into five groups as following: amyloid beta (Aβ)-only group, Aβ + SP600125 (30 μg/1 μ/side, n = 7) and/or bucladesine (100 μM/1 μl/side, n = 7), and the normal control (vehicle only) group. The treatments were administered bilaterally to the CA1 sub-region of the hippocampus stereotaxically. Spatial reference memory was performed using Morris Water Maze 21 days later. The expression of authophagy markers (beclin1, Atg7, Atg12, and LC3 II/LC3 I) in the hippocampus was evaluated using western blotting. Compared to the vehicle group, Aβ administration reduced spatial reference learning (P < 0.001) and memory (P < 0.01) and upregulated the expression of beclin1, Atg7, Atg12, and LC3 II/I (P < 0.0001). Compare to Aβ-only group, the administration of SP600125 and/or bucladesine improved spatial reference learning (P < 0.001) and memory (P < 0.01). Compared to the Aβ-only group, the treatment with SP600125 and/or bucladesine also reduced beclin1, Atg7, Atg12, and LC3 II/I (P < 0.0001) which was similar to amount of normal rats. In summary, it seems that the improvement of spatial memory by SP600125 and/or bucladesine in Aβ-injected rats is in relation with normalizing of autophagy to the physiologic level, possibly through neuroprotection and/or neuroplasticity.
Collapse
Affiliation(s)
- M Mohammadi
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - J Guan
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medicine and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, New Zealand.,Gravida National Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - F Khodagholi
- Neuroscience Research Centre, ShahidBeheshti University of Medical Sciences, Tehran, Iran.,Neurobiology Research Centre, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - A Yans
- Neuroscience Research Centre, ShahidBeheshti University of Medical Sciences, Tehran, Iran.,Neurobiology Research Centre, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - S Khalaj
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - M Gholami
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - G H Taghizadeh
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran.,Department of Neuroscience, Faculty of Advanced Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A Aliaghaei
- Department of Anatomy and Cell Biology, School of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - M Abdollahi
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - M H Ghahremani
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran
| | - M Sharifzadeh
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran. .,Department of Neuroscience, Faculty of Advanced Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Yarza R, Vela S, Solas M, Ramirez MJ. c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for Alzheimer's Disease. Front Pharmacol 2016; 6:321. [PMID: 26793112 PMCID: PMC4709475 DOI: 10.3389/fphar.2015.00321] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/28/2015] [Indexed: 01/08/2023] Open
Abstract
c-Jun N-terminal kinases (JNKs) are a family of protein kinases that play a central role in stress signaling pathways implicated in gene expression, neuronal plasticity, regeneration, cell death, and regulation of cellular senescence. It has been shown that there is a JNK pathway activation after exposure to different stressing factors, including cytokines, growth factors, oxidative stress, unfolded protein response signals or Aβ peptides. Altogether, JNKs have become a focus of screening strategies searching for new therapeutic approaches to diabetes, cancer or liver diseases. In addition, activation of JNK has been identified as a key element responsible for the regulation of apoptosis signals and therefore, it is critical for pathological cell death associated with neurodegenerative diseases and, among them, with Alzheimer’s disease (AD). In addition, in vitro and in vivo studies have reported alterations of JNK pathways potentially associated with pathogenesis and neuronal death in AD. JNK’s, particularly JNK3, not only enhance Aβ production, moreover it plays a key role in the maturation and development of neurofibrillary tangles. This review aims to explain the rationale behind testing therapies based on inhibition of JNK signaling for AD in terms of current knowledge about the pathophysiology of the disease. Keeping in mind that JNK3 is specifically expressed in the brain and activated by stress-stimuli, it is possible to hypothesize that inhibition of JNK3 might be considered as a potential target for treating neurodegenerative mechanisms associated with AD.
Collapse
Affiliation(s)
- Ramon Yarza
- Department of Pharmacology and Toxicology, University of Navarra Pamplona, Spain
| | - Silvia Vela
- Department of Pharmacology and Toxicology, University of Navarra Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of NavarraPamplona, Spain; Navarra Institute for Health ResearchPamplona, Spain
| | - Maria J Ramirez
- Department of Pharmacology and Toxicology, University of NavarraPamplona, Spain; Navarra Institute for Health ResearchPamplona, Spain
| |
Collapse
|
28
|
Qin Y, Chen Z, Han X, Wu H, Yu Y, Wu J, Liu S, Hou Y. Progesterone attenuates Aβ(25-35)-induced neuronal toxicity via JNK inactivation and progesterone receptor membrane component 1-dependent inhibition of mitochondrial apoptotic pathway. J Steroid Biochem Mol Biol 2015; 154:302-11. [PMID: 25576906 DOI: 10.1016/j.jsbmb.2015.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/25/2014] [Accepted: 01/05/2015] [Indexed: 11/21/2022]
Abstract
Progesterone, which acts as a neurosteroid in nervous system, has been shown to have neuroprotective effects in different experiments in vitro and in vivo. Our previous study demonstrates that progesterone exerts neuroprotections in Alzheimer's disease-like rats. Present study attempted to evaluate the protective effects of progesterone on Aβ-treated neurons and potential mechanisms involved in neuroprotection. Results showed that treatment with progesterone protected primary cultured rat cortical neurons against Aβ(25-35)-induced apoptosis. Furthermore, we observed that progesterone alleviated mitochondrial dysfunction by rescuing mitochondrial membrane potential under Aβ challenge. Moreover, progesterone could also attenuate Bax/Bcl-2 proteins ratio upregulation and inhibit the activation of caspase-3 in Aβ-treated neurons. These indicate that progesterone attenuates Aβ(25-35)-induced neuronal toxicity by inhibiting mitochondria-associated apoptotic pathway. Both classic progesterone receptors (classic PR) and progesterone receptor membrane component 1 (PGRMC1), a special progesterone membrane receptor, are broadly expressed throughout the brain. The protective effect of progesterone was partially abolished by PGRMC1 inhibitor AG205 rather than classic PR antagonist RU486 in this study. Additionally, progesterone protected neurons by inhibiting Aβ-induced activation of JNK, which was an upstream signaling component in Aβ-induced mitochondria-associated apoptotic pathway. But this process was independent of PGRMC1. Taken together, these results suggest that progesterone exerts a protective effect against Aβ(25-35)-induced insults at least in part by two complementary pathways: (1) progesterone receptor membrane component 1-dependent inhibition of mitochondrial apoptotic pathway, and (2) blocking Aβ-induced JNK activation. The present study provides new insights into the mechanism by which progesterone brings neuroprotection. This article is part of a Special Issue entitled 'Steroids & Nervous System'.
Collapse
Affiliation(s)
- Yabin Qin
- Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Zesha Chen
- Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Xiaolei Han
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang 050082, Hebei Province, China
| | - Honghai Wu
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang 050082, Hebei Province, China
| | - Yang Yu
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang 050082, Hebei Province, China
| | - Jie Wu
- Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Sha Liu
- Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yanning Hou
- Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang 050082, Hebei Province, China.
| |
Collapse
|
29
|
Jiang F, Mao Y, Liu H, Xu P, Zhang L, Qian X, Sun X. Magnesium Lithospermate B Protects Neurons Against Amyloid β (1–42)-Induced Neurotoxicity Through the NF-κB Pathway. Neurochem Res 2015; 40:1954-65. [DOI: 10.1007/s11064-015-1691-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/20/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022]
|
30
|
Jean YY, Baleriola J, Fà M, Hengst U, Troy CM. Stereotaxic Infusion of Oligomeric Amyloid-beta into the Mouse Hippocampus. J Vis Exp 2015:e52805. [PMID: 26132278 DOI: 10.3791/52805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease is a neurodegenerative disease affecting the aging population. A key neuropathological feature of the disease is the over-production of amyloid-beta and the deposition of amyloid-beta plaques in brain regions of the afflicted individuals. Throughout the years scientists have generated numerous Alzheimer's disease mouse models that attempt to replicate the amyloid-beta pathology. Unfortunately, the mouse models only selectively mimic the disease features. Neuronal death, a prominent effect in the brains of Alzheimer's disease patients, is noticeably lacking in these mice. Hence, we and others have employed a method of directly infusing soluble oligomeric species of amyloid-beta - forms of amyloid-beta that have been proven to be most toxic to neurons - stereotaxically into the brain. In this report we utilize male C57BL/6J mice to document this surgical technique of increasing amyloid-beta levels in a select brain region. The infusion target is the dentate gyrus of the hippocampus because this brain structure, along with the basal forebrain that is connected by the cholinergic circuit, represents one of the areas of degeneration in the disease. The results of elevating amyloid-beta in the dentate gyrus via stereotaxic infusion reveal increases in neuron loss in the dentate gyrus within 1 week, while there is a concomitant increase in cell death and cholinergic neuron loss in the vertical limb of the diagonal band of Broca of the basal forebrain. These effects are observed up to 2 weeks. Our data suggests that the current amyloid-beta infusion model provides an alternative mouse model to address region specific neuron death in a short-term basis. The advantage of this model is that amyloid-beta can be elevated in a spatial and temporal manner.
Collapse
Affiliation(s)
- Ying Y Jean
- Department of Pathology and Cell Biology, Columbia University Medical Center;
| | - Jimena Baleriola
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center
| | - Mauro Fà
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center
| | - Ulrich Hengst
- Department of Pathology and Cell Biology, Columbia University Medical Center; The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center
| | - Carol M Troy
- Department of Pathology and Cell Biology, Columbia University Medical Center; The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center; Department of Neurology, Columbia University Medical Center
| |
Collapse
|
31
|
Adjunctive and long-acting nanoformulated antiretroviral therapies for HIV-associated neurocognitive disorders. Curr Opin HIV AIDS 2015; 9:585-90. [PMID: 25226025 DOI: 10.1097/coh.0000000000000111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW We are pleased to review current and future strategies being developed to modulate neuroinflammation while reducing residual viral burden in the central nervous system. This has been realized by targeted long-acting antiretroviral nano and adjunctive therapies being developed for HIV-infected people. Our ultimate goal is to eliminate virus from its central nervous system reservoirs and, in so doing, reverse the cognitive and motor dysfunctions. RECENT FINDINGS Herein, we highlight our laboratories' development of adjunctive and nanomedicine therapies for HIV-associated neurocognitive disorders. An emphasis is placed on drug-drug interactions that target both the viral life cycle and secretory proinflammatory neurotoxic factors and signaling pathways. SUMMARY Antiretroviral therapy has improved the quality and duration of life for people living with HIV-1. A significant long-term comorbid illness is HIV-associated neurocognitive disorders. Symptoms, although reduced in severity, are common. Disease occurs, in part, through continued low-level viral replication, inducing secondary glial neuroinflammatory activities. Our recent works and those of others have seen disease attenuated in animal models through the use of adjunctive and long-acting reservoir-targeted nanoformulated antiretroviral therapy. The translation of these inventions from animals to humans is the focus of this review.
Collapse
|
32
|
Xu N, Xiao Z, Zou T, Huang Z. Induction of GADD34 Regulates the Neurotoxicity of Amyloid β. Am J Alzheimers Dis Other Demen 2015; 30:313-9. [PMID: 25204313 PMCID: PMC10852579 DOI: 10.1177/1533317514545616] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The possible roles played by growth arrest and DNA damage-inducible gene 34 (GADD34) in Alzheimer's disease (AD) are so far less understood. In this study, we found that GADD34 was increased in the brains of AD transgenic J20 mice. The deposition of β-amyloid (Aβ) peptide is the main component of neurotic plaques in AD brain. Thus, we examined the effect of Aβ in the expression of GADD34 in human SH-SY5Y cells in vitro. Amyloid β (Aβ1-42) treatment led to increased expression of GADD34. Pretreatment with 50 nmol/L of c-Jun N-terminal kinases (JNK) inhibitor SP600125 abolished the upregulation of GADD34. c-Jun silencing by transfection with c-Jun small-interfering RNA abolished the effects of Aβ1-42 on the expression of GADD34. Importantly, chromatin immunoprecipitation studies verified the ability of c-Jun to bind to the GADD34 promoter, and this ability was increased more than 3-fold by Aβ1-42. These data suggest that the induction of GADD34 by Aβ is mediated by JNK/c-Jun pathway. Finally, depletion of GADD34 significantly rescued Aβ-induced cell apoptosis as evidenced by a marked decrease in the number of terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling (TUNEL)-positive cells. Consistently, knockdown of GADD34 attenuated caspase 3 activation induced by Aβ1-42.
Collapse
Affiliation(s)
- Niangui Xu
- Department of Neurology, The Second Xiangya Hospital of the Central South University, Changsha, China
| | - Zhijie Xiao
- Department of Neurology, The Second Xiangya Hospital of the Central South University, Changsha, China
| | - Ting Zou
- Department of Neurology, The Second Xiangya Hospital of the Central South University, Changsha, China
| | - Zhiling Huang
- Department of Neurology, The Second Xiangya Hospital of the Central South University, Changsha, China
| |
Collapse
|
33
|
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: 67] [Impact Index Per Article: 6.7] [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
|
34
|
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]
|
35
|
RAGE inhibition in microglia prevents ischemia-dependent synaptic dysfunction in an amyloid-enriched environment. J Neurosci 2014; 34:8749-60. [PMID: 24966375 DOI: 10.1523/jneurosci.0141-14.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ischemia is known to increase the deleterious effect of β-amyloid (Aβ), contributing to early cognitive impairment in Alzheimer's disease. Here, we investigated whether transient ischemia may function as a trigger for Aβ-dependent synaptic impairment in the entorhinal cortex (EC), acting through specific cellular signaling. We found that synaptic depression induced by oxygen glucose deprivation (OGD) was enhanced in EC slices either in presence of synthetic oligomeric Aβ or in slices from mutant human amyloid precursor protein transgenic mice (mhAPP J20). OGD-induced synaptic depression was ameliorated by functional suppression of RAGE. In particular, overexpression of the dominant-negative form of RAGE targeted to microglia (DNMSR) protects against OGD-induced synaptic impairment in an amyloid-enriched environment, reducing the activation of stress-related kinases (p38MAPK and JNK) and the release of IL-1β. Our results demonstrate a prominent role for the RAGE-dependent neuroinflammatory pathway in the synaptic failure induced by Aβ and triggered by transient ischemia.
Collapse
|
36
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014; 10:1054-1070. [PMID: 24879153 PMCID: PMC4091168 DOI: 10.4161/auto.28528] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 03/09/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
- Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|
37
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014. [PMID: 24879153 DOI: 10.4161/auto.28528.erratum.in:autophagy.2017;13(3):637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA; Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|
38
|
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
|
39
|
Chou CT, Lin WF, Kong ZL, Chen SY, Hwang DF. Taurine prevented cell cycle arrest and restored neurotrophic gene expression in arsenite-treated SH-SY5Y cells. Amino Acids 2013; 45:811-9. [PMID: 23744399 DOI: 10.1007/s00726-013-1524-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
The study investigated the effect of taurine on cell viability and neurotrophic gene expression in arsenite-treated human neuroblastoma SH-SY5Y cells. Arsenite-induced intracellular reactive oxygen species (ROS) and interrupted cell cycle in SH-SY5Y cells. In addition, arsenite reduced mitochondria membrane potential (MMP) and decreased neurotrophic gene expressions such as n-myc downstream-regulated gene 4 (NDRG-4), brain-derived neurotrophic factor (BDNF) and sirtuin-1 (SIRT-1) in SH-SY5Y cells. In parallel, taurine prevented cell cycle, restored MMP and reduced the intracellular ROS level, and taurine recovered NDRG-4, BDNF and SIRT-1 gene expressions in arsenite-treated SH-SY5Y cells while taurine alone has no effect on these parameters.
Collapse
Affiliation(s)
- Chien-Te Chou
- Department of Food Science, National Taiwan Ocean University, 2 Pei-Ning Road, Keelung 202, Taiwan, ROC
| | | | | | | | | |
Collapse
|
40
|
Ma Q, Gelbard HA, Maggirwar SB, Dewhurst S, Gendelman HE, Peterson DR, DiFrancesco R, Hochreiter JS, Morse GD, Schifitto G. Pharmacokinetic interactions of CEP-1347 and atazanavir in HIV-infected patients. J Neurovirol 2013; 19:254-60. [PMID: 23737347 DOI: 10.1007/s13365-013-0172-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/01/2022]
Abstract
CEP-1347 is a potent inhibitor of mixed lineage kinase (MLK), which was investigated for ameliorating HIV-associated neurocognitive disorders. CEP-1347 and atazanavir pharmacokinetics were determined when CEP-1347 50 mg twice daily was administered to HIV-infected patients (n = 20) receiving combination antiretroviral therapy including atazanavir and ritonavir (ATV/RTV, 300/100 mg) once daily continuously. Co-administration of CEP-1347 and ATV/RTV resulted with significant changes in pharmacokinetics of ATV but not RTV. Specifically, an increase in ATV accumulation ratio of 15 % (p = 0.007) and a prolongation of T(½) from 12.7 to 15.9 h (p = 0.002) were observed. The results suggested that co-administration of CEP-1347 with ATV/RTV in HIV-infected patients might result in limited impact on ATV but not on RTV pharmacokinetics.
Collapse
Affiliation(s)
- Qing Ma
- Center for Human Experimental Therapeutics, Clinical and Translational Sciences Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tang Y, Cui YC, Wang XJ, Wu AL, Hu GF, Luo FL, Sun JK, Sun J, Wu LK. Neural progenitor cells derived from adult bone marrow mesenchymal stem cells promote neuronal regeneration. Life Sci 2012; 91:951-8. [PMID: 23000028 DOI: 10.1016/j.lfs.2012.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 08/18/2012] [Accepted: 09/11/2012] [Indexed: 01/17/2023]
Abstract
AIM It is well known that neural stem/progenitor cells (NS/PC) are an ideal cell type for the treatment of central nervous system (CNS) disease. However, ethical problems have severely hampered fetal NS/PC from being widely used as a source for stem cell therapy. Recently, it has been demonstrated that autologous bone marrow mesenchymal stem cells (BMSC) can transdifferentiate into neural progenitor cells (NPC). The biological function of BMSC derived NPC (MDNPC) in neuronal systems remains unknown. In the present study, we aimed to investigate whether MDNPC can promote in vitro neural regeneration, a process comprising mainly the generation of neurons and neurotransmitters. MAIN METHODS We co-cultured BMSC, MDNPC or fetal NS/PC with PC12 cells and studied their roles on proliferation, neurite formation and dopamine release from PC12 cells. Furthermore, we also explored the mechanisms by which MDNPC regulate dopamine secretion from PC12 derived neural cells using Western blot. KEY FINDINGS We found that both MDNPC and NS/PC had similar morphologies and there were no significant differences between MDNPC and NS/PC in promoting PC12 cell proliferation, neurite outgrowth, and dopamine release. We also demonstrated that NS/PC induced dopamine secretion was associated with an upregulation of dopamine transporter (DAT) levels. SIGNIFICANCE In summary, MDNPC were comparable to NS/PC in promoting neural regeneration, indicating that MDNPC are a promising candidate source of neural stem cells for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yue Tang
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yenki P, Khodagholi F, Shaerzadeh F. Inhibition of phosphorylation of JNK suppresses Aβ-induced ER stress and upregulates prosurvival mitochondrial proteins in rat hippocampus. J Mol Neurosci 2012; 49:262-9. [PMID: 22706709 DOI: 10.1007/s12031-012-9837-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 06/06/2012] [Indexed: 12/27/2022]
Abstract
A growing body of evidence indicates that c-Jun N-terminal kinases (JNKs) is activated in Alzheimer's disease. Herein, we examine the effect of the JNK specific inhibitor, SP600125, on the level of functional proteins or transcription factors related to endoplasmic reticulum (ER) and oxidative stress induced by amyloid beta (Aβ). Our results clearly showed the ability of SP600125 to decrease the levels of caspase 12 and calpain 2, two important enzymes involved in ER stress. Aβ has been suggested to be able to decrease the phosphorylation level of cAMP response element-binding (CREB) through mitogen-activated protein kinase pathway. We observed that JNK inhibition in Aβ-injected rats can restore the activation of CREB through increasing its phosphorylation level. This effect may explain the increase observed in c-fos level, as a CREB downstream factor under JNK inhibition in Aβ-injected rats. Following Aβ injection, the levels of pro-survival mitochondrial proteins including nuclear respiratory factor-1 (NRF-1), peroxisome proliferator-activated receptor gamma co-activator 1-alpha, and mitochondrial transcription factor A (TFAM) significantly decreased, which could be returned to control level with JNK inhibition. We suggest that the elevation in the level of PGC1-alpha and other mitochondrial proteins is the result of an increase in CREB activation as the upstream factor of PGC1-alpha. Also, we observed that pretreatment with SP600125 leads to a greater increase of nuclear related factor-2 (Nrf2) level compared with the Aβ-injected group. Nrf2 has been shown to bind to CREB-binding factor leading to their contribution in Nrf2 target genes expression. Besides, NRF-1 and TFAM are reported as Nrf2 targets. Based on our data, we can conclude that JNK carry out partial destructive effects of Aβ in rat brain.
Collapse
Affiliation(s)
- Parvin Yenki
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
43
|
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: 47] [Impact Index Per Article: 3.6] [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
|
44
|
Wilhelm M, Kukekov NV, Schmit TL, Biagas KV, Sproul AA, Gire S, Maes ME, Xu Z, Greene LA. Sh3rf2/POSHER protein promotes cell survival by ring-mediated proteasomal degradation of the c-Jun N-terminal kinase scaffold POSH (Plenty of SH3s) protein. J Biol Chem 2012; 287:2247-56. [PMID: 22128169 PMCID: PMC3265902 DOI: 10.1074/jbc.m111.269431] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 11/26/2011] [Indexed: 12/18/2022] Open
Abstract
We report that Sh3rf2, a homologue of the pro-apoptotic scaffold POSH (Plenty of SH3s), acts as an anti-apoptotic regulator for the c-Jun N-terminal kinase (JNK) pathway. siRNA-mediated knockdown of Sh3rf2 promotes apoptosis of neuronal PC12 cells, cultured cortical neurons, and C6 glioma cells. This death appears to result from activation of JNK signaling. Loss of Sh3rf2 triggers activation of JNK and its target c-Jun. Also, apoptosis promoted by Sh3rf2 knockdown is inhibited by dominant-negative c-Jun as well as by a JNK inhibitor. Investigation of the mechanism by which Sh3rf2 regulates cell survival implicates POSH, a scaffold required for activation of pro-apoptotic JNK/c-Jun signaling. In cells lacking POSH, Sh3rf2 knockdown is unable to activate JNK. We further find that Sh3rf2 binds POSH to reduce its levels by a mechanism that requires the RING domains of both proteins and that appears to involve proteasomal POSH degradation. Conversely, knockdown of Sh3rf2 promotes the stabilization of POSH protein and activation of JNK signaling. Finally, we show that endogenous Sh3rf2 protein rapidly decreases following several different apoptotic stimuli and that knockdown of Sh3rf2 activates the pro-apoptotic JNK pathway in neuronal cells. These findings support a model in which Sh3rf2 promotes proteasomal degradation of pro-apoptotic POSH in healthy cells and in which apoptotic stimuli lead to rapid loss of Sh3rf2 expression, and consequently to stabilization of POSH and JNK activation and cell death. On the basis of these observations, we propose the alternative name POSHER (POSH-eliminating RING protein) for the Sh3rf2 protein.
Collapse
Affiliation(s)
- Michael Wilhelm
- Departments of Pediatrics, Columbia University Health Sciences, New York, New York10032, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Anand R, Kaushal A, Wani WY, Gill KD. Road to Alzheimer's disease: the pathomechanism underlying. Pathobiology 2011; 79:55-71. [PMID: 22205086 DOI: 10.1159/000332218] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/23/2011] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, results from the interplay of various deregulated mechanisms triggering a complex pathophysiology. The neurons suffer from and slowly succumb to multiple irreversible damages, resulting in cell death and thus memory deficits that characterize AD. In spite of our vast knowledge, it is still unclear as to when the disease process starts and how long the perturbations continue before the disease manifests. Recent studies provide sufficient evidence to prove amyloid β (Aβ) as the primary cause initiating secondary events, but Aβ is also known to be produced under normal conditions and to possess physiological roles, hence, the questions that remain are: What are the factors that lead to abnormal Aβ production? When does Aβ turn into a pathological molecule? What is the chain of events that follows Aβ? The answers are still under debate, and further insight may help us in creating better diagnostic and therapeutic options in AD. The present article attempts to review the current literature regarding AD pathophysiology and proposes a pathophysiologic cascade in AD.
Collapse
Affiliation(s)
- R Anand
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | |
Collapse
|
46
|
Mice lacking caspase-2 are protected from behavioral changes, but not pathology, in the YAC128 model of Huntington disease. Mol Neurodegener 2011; 6:59. [PMID: 21854568 PMCID: PMC3180273 DOI: 10.1186/1750-1326-6-59] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 08/19/2011] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Huntington Disease (HD) is a neurodegenerative disorder in which caspase activation and cleavage of substrates, including the huntingtin protein, has been invoked as a pathological mechanism. Specific changes in caspase-2 (casp2) activity have been suggested to contribute to the pathogenesis of HD, however unique casp2 cleavage substrates have remained elusive. We thus utilized mice completely lacking casp2 (casp2-/-) to examine the role played by casp2 in the progression of HD. This 'substrate agnostic' approach allows us to query the effect of casp2 on HD progression without pre-defining proteolytic substrates of interest. RESULTS YAC128 HD model mice lacking casp2 show protection from well-validated motor and cognitive features of HD, including performance on rotarod, swimming T-maze, pre-pulse inhibition, spontaneous alternation and locomotor tasks. However, the specific pathological features of the YAC128 mice including striatal volume loss and testicular degeneration are unaltered in mice lacking casp2. The application of high-resolution magnetic resonance imaging (MRI) techniques validates specific neuropathology in the YAC128 mice that is not altered by ablation of casp2. CONCLUSIONS The rescue of behavioral phenotypes in the absence of pathological improvement suggests that different pathways may be operative in the dysfunction of neural circuitry in HD leading to behavioral changes compared to the processes leading to cell death and volume loss. Inhibition of caspase-2 activity may be associated with symptomatic improvement in HD.
Collapse
|
47
|
Neuregulin-1 prevents amyloid β-induced impairment of long-term potentiation in hippocampal slices via ErbB4. Neurosci Lett 2011; 505:6-9. [PMID: 21787838 DOI: 10.1016/j.neulet.2011.05.246] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/12/2011] [Accepted: 05/12/2011] [Indexed: 12/20/2022]
Abstract
Neuregulin-1 (NRG1) participates in numerous neurodevelopmental processes and plasticity of the brain. Despite this, little is known about its role in Alzheimer's disease (AD). Amyloid β (Aβ) peptide is generally believed to play a critical role in the pathogenesis of AD. The present study examined the effect of synthetic Aβ₁₋₄₂ peptides on long-term potentiation (LTP) in the CA1 region of mice hippocampal slices, a cellular model of learning and memory. We found that application of a test dose of Aβ₁₋₄₂ (200 nM) significantly inhibited the development of LTP without affecting basal synaptic transmission. Pretreatment with NRG1 effectively prevented Aβ₁₋₄₂-induced impairment of LTP, an effect that was dose-dependent. This LTP-restoring action of NRG1 was almost completely abolished by blocking ErbB4, a key NRG1 receptor, suggesting that NRG1 acts through ErbB4 to exert its protective action on LTP. The present study thus provides the first demonstration that NRG1/ErbB4 protects against Aβ-induced hippocampal LTP impairment, suggesting that NRG1 may be a promising candidate for the treatment of early-stage AD.
Collapse
|
48
|
Ferreira DMS, Castro RE, Machado MV, Evangelista T, Silvestre A, Costa A, Coutinho J, Carepa F, Cortez-Pinto H, Rodrigues CMP. Apoptosis and insulin resistance in liver and peripheral tissues of morbidly obese patients is associated with different stages of non-alcoholic fatty liver disease. Diabetologia 2011; 54:1788-98. [PMID: 21455726 DOI: 10.1007/s00125-011-2130-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 03/02/2011] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Non-alcoholic fatty liver disease (NAFLD) is associated with insulin resistance and characterised by different degrees of hepatic lesion. Its pathogenesis and correlation with apoptosis and insulin resistance in insulin target tissues remains incompletely understood. We investigated how insulin signalling, caspase activation and apoptosis correlate with different NAFLD stages in liver, muscle and visceral adipose tissues. METHODS Liver, muscle and adipose tissue biopsies from 26 morbidly obese patients undergoing bariatric surgery were grouped according to the Kleiner-Brunt scoring system into simple steatosis, and less severe and more severe non-alcoholic steatohepatitis (NASH). Apoptosis was assessed by DNA fragmentation, and caspase-2 and -3 activation. Insulin signalling and c-Jun NH(2)-terminal kinase (JNK) proteins were evaluated by western blot. RESULTS Caspase-3 and -2 activation, and DNA fragmentation were markedly increased in the liver of patients with severe NASH vs in that of those with simple steatosis (p < 0.01). Muscle tissue, and to a lesser extent the liver, had decreased tyrosine phosphorylated insulin receptor and insulin receptor substrate in patients with severe NASH, compared with those with simple steatosis (p < 0.01 muscle; p < 0.05 liver). Concomitantly, Akt phosphorylation decreased in muscle, liver and visceral adipose tissues in patients with severe NASH (at least p < 0.05). Finally, JNK phosphorylation was significantly increased in muscle (p < 0.01) and liver (p < 0.05) from NASH patients, compared with tissue from those with simple steatosis. CONCLUSIONS/INTERPRETATION Our results demonstrate a link between apoptosis, insulin resistance and different NAFLD stages, where JNK and caspase-2 may play a key regulatory role.
Collapse
Affiliation(s)
- D M S Ferreira
- Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Park SH, Kim JH, Bae SS, Hong KW, Choi BT, Shin HK. Phosphodiesterase III Inhibitor Cilostazol Protects Amyloid β-Induced Neuronal Cell Injury via Peroxisome Proliferator-Activated Receptor-γ Activation. ACTA ACUST UNITED AC 2011. [DOI: 10.5352/jls.2011.21.5.647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
50
|
Inhibition of JNK phosphorylation reverses memory deficit induced by β-amyloid (1–42) associated with decrease of apoptotic factors. Behav Brain Res 2011; 217:424-31. [DOI: 10.1016/j.bbr.2010.11.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/25/2010] [Accepted: 11/05/2010] [Indexed: 01/11/2023]
|