1
|
Pozdyshev DV, Leisi EV, Muronetz VI, Golyshev SA, Kurochkina LP. Cytotoxicity of α-synuclein amyloid fibrils generated with phage chaperonin OBP. Biochem Biophys Res Commun 2025; 742:151127. [PMID: 39644608 DOI: 10.1016/j.bbrc.2024.151127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Chaperonins are known to be important players in the conversion of amyloidogenic proteins into amyloid precursors in a variety of neurodegenerative diseases. However, the mechanisms of their action is still poorly understood. In this work, we used a single-ring chaperonin of the bacteriophage OBP, which functions in an ATP-dependent manner but has a simpler structure than other chaperonins. The effect of the chaperonin OBP on the conversion of human α-synuclein mutant A53T into amyloid was studied and the cytotoxicity of the formed fibrils was investigated. The phage chaperonin OBP was expressed in HEK293T cells together with the human α-synuclein mutant A53T. Both proteins showed a diffuse distribution within the cell cytoplasm as determined by fluorescence microscopy using specific antibodies. Separate and co-expression of the two proteins did not result in the formation of distinguishable protein aggregates in the cells, nor did it have any effect on cell viability. However, the co-expression of chaperonin and α-synuclein did result in the appearance of some dimeric and oligomeric forms of α-synuclein in the insoluble fraction of the cell lysate. It can therefore be concluded that chaperonin OBP stimulates the amyloid transformation of α-synuclein A53T when both proteins are co-expressed in eukaryotic cells. A comparison of the cytotoxicity of mutant α-synuclein amyloid forms obtained in vitro, both during spontaneous fibrillation and with the participation of the chaperonin OBP, showed that the maximum effect on HEK293T and SH-SY5Y cells, resulting in the death of more than 50 % of the population, was exerted by α-synuclein fibrils formed under chaperonin action in the presence of ATP. In the context of recent data on the spread of amyloid α-synuclein from the gut to the brain, the role of phage chaperonins in the pathological aggregation of amyloidogenic proteins in the human body and the potential use of the OBP chaperonin in cellular models of synucleinopathies are discussed.
Collapse
Affiliation(s)
- Denis V Pozdyshev
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia.
| | - Evgeniia V Leisi
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia; Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Bld 73, 119991, Moscow, Russia
| | - Vladimir I Muronetz
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia; Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Bld 73, 119991, Moscow, Russia
| | - Sergei A Golyshev
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia
| | - Lidia P Kurochkina
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia
| |
Collapse
|
2
|
Bastian P, Konieczna L, Dulski J, Daca A, Jacewicz D, Płoska A, Knap N, Sławek J, Bączek T, Kalinowski L, Drzeżdżon J, Roszmann A, Belka M, Górska-Ponikowska M. 2-Methoxyestradiol and Hydrogen Peroxide as Promising Biomarkers in Parkinson's Disease. Mol Neurobiol 2024; 61:148-166. [PMID: 37589832 PMCID: PMC10791893 DOI: 10.1007/s12035-023-03575-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Estrogens function in numerous physiological processes including controlling brain cell growth and differentiation. 2-Methoxestradiol (2-ME2), a 17β-estradiol (E2) metabolite, is known for its anticancer effects as observed both in vivo and in vitro. 2-ME2 affects all actively dividing cells, including neurons. The study aimed to determine whether 2-ME2 is a potentially cancer-protective or rather neurodegenerative agent in a specific tissue culture model as well as a clinical setup. In this study, 2-ME2 activity was determined in a Parkinson's disease (PD) in vitro model based on the neuroblastoma SH-SY5Y cell line. The obtained results suggest that 2-ME2 generates nitro-oxidative stress and controls heat shock proteins (HSP), resulting in DNA strand breakage and apoptosis. On the one hand, it may affect intensely dividing cells preventing cancer development; however, on the other hand, this kind of activity within the central nervous system may promote neurodegenerative diseases like PD. Thus, the translational value of 2-ME2's neurotoxic activity in a PD in vitro model was also investigated. LC-MS/MS technique was used to evaluate estrogens and their derivatives, namely, hydroxy and methoxyestrogens, in PD patients' blood, whereas the stopped-flow method was used to assess hydrogen peroxide (H2O2) levels. Methoxyestrogens and H2O2 levels were increased in patients' blood as compared to control subjects, but hydoxyestrogens were simultaneously decreased. From the above, we suggest that the determination of plasma levels of methoxyestrogens and H2O2 may be a novel PD biomarker. The presented research is the subject of the pending patent application "The use of hydrogen peroxide and 17β-estradiol and its metabolites as biomarkers in the diagnosis of neurodegenerative diseases," no. P.441360.
Collapse
Affiliation(s)
- Paulina Bastian
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
| | - Lucyna Konieczna
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Dagmara Jacewicz
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jarosław Sławek
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza Street 11/12, 80-233, Gdansk, Poland
| | - Joanna Drzeżdżon
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Anna Roszmann
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Magdalena Górska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany.
- Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy.
| |
Collapse
|
3
|
Kumari A, Srivastava A, Jagdale P, Ayanur A, Khanna VK. Lambda-cyhalothrin enhances inflammation in nigrostriatal region in rats: Regulatory role of NF-κβ and JAK-STAT signaling. Neurotoxicology 2023; 96:101-117. [PMID: 37060950 DOI: 10.1016/j.neuro.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
The risk to develop neurobehavioural abnormalities in humans on exposure to lambda-cyhalothrin (LCT) - a type II synthetic pyrethroid has enhanced significantly due to its extensive uses in agriculture, homes, veterinary practices and public health programs. Earlier, we found that the brain dopaminergic system is vulnerable to LCT and affects motor functions in rats. In continuation to this, the present study is focused to unravel the role of neuroinflammation in LCT-induced neurotoxicity in substantia nigra and corpus striatum in rats. Increase in the mRNA expression of proinflammatory cytokines (TNF- α, IL-1β, IL-6) and iNOS whereas decrease in anti-inflammatory cytokine (IL-10) was distinct both in substantia nigra and corpus striatum of rats treated with LCT (0.5, 1.0, 3.0 mg/kg body weight, p.o, for 45 days) as compared to control rats. Further, LCT-treated rats exhibited increased levels of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (Iba-1), the glial marker proteins both in substantia nigra and corpus striatum as compared to controls. Exposure of rats to LCT also caused alterations in the levels of heat shock protein 60 (HSP60) and mRNA expression of toll-like receptors (TLR2 and TLR4) in the substantia nigra and corpus striatum. An increase in the phosphorylation of key proteins involved in NF-kβ (P65, Iκβ, IKKα, IKKβ) and JAK/STAT (STAT1, STAT3) signaling and alteration in the protein levels of JAK1 and JAK2 was prominent in LCT-treated rats. Histological studies revealed damage of dopaminergic neurons and reactive gliosis as evidenced by the presence of darkly stained pyknotic neurons and decrease in Nissl substance and an increase in infiltration of immune cells both in substantia nigra and corpus striatum of LCT-treated rats. Presence of reactive microglia and astrocytes in LCT-treated rats was also distinct in ultrastructural studies. The results exhibit that LCT may damage dopaminergic neurons in the substantia nigra and corpus striatum by inducing inflammation as a result of stimulation of neuroglial cells involving activation of NF-κβ and JAK/STAT signaling.
Collapse
Affiliation(s)
- Anima Kumari
- Developmental Toxicology Laboratory, Area - Systems Toxicology & Health Risk Assessment, CSIR - Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anugya Srivastava
- Developmental Toxicology Laboratory, Area - Systems Toxicology & Health Risk Assessment, CSIR - Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pankaj Jagdale
- Central Pathology Laboratory, Area - Regulatory Toxicology, CSIR - Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India
| | - Anjaneya Ayanur
- Central Pathology Laboratory, Area - Regulatory Toxicology, CSIR - Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India
| | - Vinay Kumar Khanna
- Developmental Toxicology Laboratory, Area - Systems Toxicology & Health Risk Assessment, CSIR - Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India.
| |
Collapse
|
4
|
Chronic Treatment with the Probiotics Lacticaseibacillus rhamnosus GG and Bifidobacterium lactis BB12 Attenuates Motor Impairment, Striatal Microglial Activation, and Dopaminergic Loss in Rats with 6-Hydroxydopamine-induced Hemiparkinsonism. Neuroscience 2022; 507:79-98. [PMID: 36370934 DOI: 10.1016/j.neuroscience.2022.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/05/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Gut dysbiosis is considered a risk factor for Parkinson's disease (PD), and chronic treatment with probiotics could prevent it. Here we report the assessment of a probiotic mixture [Lacticaseibacillus rhamnosus GG (LGG), and Bifidobacterium animalis lactis BB-12 (BB-12)] administered to male rats 2 weeks before and 3 weeks after injecting 6-hydroxydopamine (6-OHDA) into the right striatum, a model that mimics the early stages of PD. Before and after lesion, animals were subjected to behavioral tests: narrow beam, cylinder test, and apomorphine (APO)-induced rotations. Dopaminergic (DA) denervation and microglia recruitment were assessed with tyrosine hydroxylase (TH+) and ionized calcium-binding protein-1 adapter (Iba1+) immunostaining, respectively. Post 6-OHDA injury, rats treated with sunflower oil (probiotics vehicle) developed significant decrease in crossing speed and increases in contralateral paw slips (narrow beam), forepaw use asymmetry (cylinder), and APO-induced rotations. In striatum, 6-OHDA eliminated ≈2/3 of TH+ area and caused significant increase of Iba1+ microglia population. Retrograde axonal degeneration suppressed ≈2/5 of TH+ neurons in the substantia nigra pars compacta (SNpc). In hemiparkinsonian rats, probiotics treatment significantly improved the crossing speed, and also reduced paw slips (postlesion days 14 and 21), the loss of TH+ neurons in SNpc, and the loss of TH+ area and of Iba1+ microglia count in striatum, without affecting the proportion of microglia morphological phenotypes. Probiotics treatment did not attenuate forepaw use asymmetry nor APO-induced rotations. These results indicate that the mixture of probiotics LGG and BB-12 protects nigrostriatal DA neurons against 6-OHDA-induced damage, supporting their potential as preventive treatment of PD.
Collapse
|
5
|
Araújo de Lima L, Oliveira Cunha PL, Felicio Calou IB, Tavares Neves KR, Facundo HT, Socorro de Barros Viana G. Effects of vitamin D (VD3) supplementation on the brain mitochondrial function of male rats, in the 6-OHDA-induced model of Parkinson's disease. Neurochem Int 2022; 154:105280. [PMID: 35026378 DOI: 10.1016/j.neuint.2022.105280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/21/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022]
Abstract
Mitochondria dysfunction is an important factor involved in PD pathogenesis. We reported neuroprotective actions of vitamin D (VD3) on a PD model, and now we investigated the VD3 effects on the brain mitochondrial function. We focused on oxygen consumption, respiratory control ratio (RCR), ADP/O ratio, mitochondria swelling, H2O2 production, and SOD activity. Additionally, immunohistochemistry assays for the dopamine system markers (TH and DAT) and mitochondrial markers (VDAC1 and Hsp60) were also carried out in the striata. Young adult male Wistar rats (250 g, 2.5 months age) were anesthetized and subjected to stereotaxic surgery and injection of saline (SO group) or 6-OHDA, into the right striatum. Brain mitochondria were isolated from the groups: sham-operated (SO), 6-OHDA, 6-OHDA pretreated with VD3 for 7, days before the 6-OHDA lesion (6-OHDA+VD3, pre-) or treated with VD3 for 14 days, after the 6-OHDA lesion (6-OHDA+VD3, post-). VD3 prevented decreases in oxygen consumption, RCR, and ADP/O ratio observed after 6-OHDA injury. Noteworthy, a very low (oxygen consumption and RCR) or no improvement (ADP/O) were observed in the 6-OHDA+VD3 post- group. VD3 also prevented the increased mitochondria swelling and H2O2 production and a decrease in SOD activity, respectively, in the 6-OHDA injured mitochondria. Also, VD3 supplementation protected the hemiparkinsonian brain from decreases in TH and DAT expressions and decreased the upregulation of mitochondrial markers, as VDAC 1 and Hsp60. In conclusion, VD3 showed neuroprotective actions on brain mitochondria injured by 6-OHDA and should stimulate translational studies focusing on its use as a therapeutic strategy for the treatment of neurodegenerative diseases as PD.
Collapse
|
6
|
Functions and Therapeutic Potential of Extracellular Hsp60, Hsp70, and Hsp90 in Neuroinflammatory Disorders. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11020736] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is implicated in central nervous system (CNS) diseases, but the molecular mechanisms involved are poorly understood. Progress may be accelerated by developing a comprehensive view of the pathogenesis of CNS disorders, including the immune and the chaperone systems (IS and CS). The latter consists of the molecular chaperones; cochaperones; and chaperone cofactors, interactors, and receptors of an organism and its main collaborators in maintaining protein homeostasis (canonical function) are the ubiquitin–proteasome system and chaperone-mediated autophagy. The CS has also noncanonical functions, for instance, modulation of the IS with induction of proinflammatory cytokines. This deserves investigation because it may be at the core of neuroinflammation, and elucidation of its mechanism will open roads toward developing efficacious treatments centered on molecular chaperones (i.e., chaperonotherapy). Here, we discuss information available on the role of three members of the CS—heat shock protein (Hsp)60, Hsp70, and Hsp90—in IS modulation and neuroinflammation. These three chaperones occur intra- and extracellularly, with the latter being the most likely involved in neuroinflammation because they can interact with the IS. We discuss some of the interactions, their consequences, and the molecules involved but many aspects are still incompletely elucidated, and we hope that this review will encourage research based on the data presented to pave the way for the development of chaperonotherapy. This may consist of blocking a chaperone that promotes destructive neuroinflammation or replacing or boosting a defective chaperone with cytoprotective activity against neurodegeneration.
Collapse
|
7
|
Reyhani S, Abbaspanah B, Mousavi SH. Umbilical cord-derived mesenchymal stem cells in neurodegenerative disorders: from literature to clinical practice. Regen Med 2020; 15:1561-1578. [PMID: 32479211 DOI: 10.2217/rme-2019-0119] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 04/29/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have provided a promising tool for cell therapy. Umbilical cord (UC) is one of the best sources of MSCs since its collection is noninvasive, and effortless, and the cells from this source are more capable and prolific. It has been proven that the differentiation, migration and protective properties of UC-MSCs are superior compared with other kinds of stem cells. Moreover, incurable neurodegenerative diseases, such as Alzheimer's disease, multiple sclerosis, Parkinson's disease and Huntington, encourage scientists to apply UC-MSCs transplantation in order to find a definite treatment. This review will focus on the preclinical and clinical use of mesenchymal stem cells derived from human umbilical cord in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Samira Reyhani
- Department of Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran 14177-44361, Iran
| | - Bahareh Abbaspanah
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran 14177-44361, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran 14177-44361, Iran
| |
Collapse
|
8
|
Kishinevsky S, Wang T, Rodina A, Chung SY, Xu C, Philip J, Taldone T, Joshi S, Alpaugh ML, Bolaender A, Gutbier S, Sandhu D, Fattahi F, Zimmer B, Shah SK, Chang E, Inda C, Koren J, Saurat NG, Leist M, Gross SS, Seshan VE, Klein C, Tomishima MJ, Erdjument-Bromage H, Neubert TA, Henrickson RC, Chiosis G, Studer L. HSP90-incorporating chaperome networks as biosensor for disease-related pathways in patient-specific midbrain dopamine neurons. Nat Commun 2018; 9:4345. [PMID: 30341316 PMCID: PMC6195591 DOI: 10.1038/s41467-018-06486-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Environmental and genetic risk factors contribute to Parkinson’s Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho-STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases. The early molecular events that ultimately lead to neuronal cell death in pathologies such as Parkinson’s disease are poorly understood. Here the authors use pluripotent stem-cell-derived human midbrain neurons and chemical biology tools to gain molecular level insight into the events induced by toxic and genetic stresses that mimic those occurring during neurodegeneration.
Collapse
Affiliation(s)
- Sarah Kishinevsky
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Neuroscience Graduate Program of Weill Cornell Graduate School of Biomedical Sciences, Weill Cornell Medical College, 1300 York Avenue, Box 65, New York, NY, 10065, USA
| | - Tai Wang
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Anna Rodina
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Sun Young Chung
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Chao Xu
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - John Philip
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Tony Taldone
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Suhasini Joshi
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Mary L Alpaugh
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Molecular and Cellular Biosciences, Rowan University, 1275 York Avenue, Glassboro, NJ, 08028, USA
| | - Alexander Bolaender
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Simon Gutbier
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, 78464, Germany
| | - Davinder Sandhu
- Department of Pharmacology, Weill Cornell College of Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Faranak Fattahi
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Bastian Zimmer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Smit K Shah
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Elizabeth Chang
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Carmen Inda
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Hostos Community College, City University of New York, Bronx, NY, 10453, USA
| | - John Koren
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Nathalie G Saurat
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, 78464, Germany
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell College of Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10017, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, 23538, Germany
| | - Mark J Tomishima
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,SKI Stem Cell Research Facility, 1275 York Avenue, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.,Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Ronald C Henrickson
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Gabriela Chiosis
- Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Lorenz Studer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA.,Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 256, New York, NY, 10065, USA
| |
Collapse
|
9
|
Ferreira SA, Romero-Ramos M. Microglia Response During Parkinson's Disease: Alpha-Synuclein Intervention. Front Cell Neurosci 2018; 12:247. [PMID: 30127724 PMCID: PMC6087878 DOI: 10.3389/fncel.2018.00247] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
The discovery of the central role played by the protein alpha-synuclein in Parkinson's disease and other Lewy body brain disorders has had a great relevance in the understanding of the degenerative process occurring in these diseases. In addition, during the last two decades, the evidence suggesting an immune response in Parkinson's disease patients have multiplied. The role of the immune system in the disease is supported by data from genetic studies and patients, as well as from laboratory animal models and cell cultures. In the immune response, the microglia, the immune cell of the brain, will have a determinant role. Interestingly, alpha-synuclein is suggested to have a central function not only in the neuronal events occurring in Parkinson's disease, but also in the immune response during the disease. Numerous studies have shown that alpha-synuclein can act directly on immune cells, such as microglia in brain, initiating a sterile response that will have consequences for the neuronal health and that could also translate in a peripheral immune response. In parallel, microglia should also act clearing alpha-synuclein thus avoiding an overabundance of the protein, which is crucial to the disease progression. Therefore, the microglia response in each moment will have significant consequences for the neuronal fate. Here we will review the literature addressing the microglia response in Parkinson's disease with an especial focus on the protein alpha-synuclein. We will also reflect upon the limitations of the studies carried so far and in the therapeutic possibilities opened based on these recent findings.
Collapse
Affiliation(s)
- Sara A Ferreira
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
10
|
Lizama BN, Palubinsky AM, McLaughlin B. Alterations in the E3 ligases Parkin and CHIP result in unique metabolic signaling defects and mitochondrial quality control issues. Neurochem Int 2018; 117:139-155. [PMID: 28851515 PMCID: PMC5826822 DOI: 10.1016/j.neuint.2017.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 08/11/2017] [Accepted: 08/21/2017] [Indexed: 01/07/2023]
Abstract
E3 ligases are essential scaffold proteins, facilitating the transfer of ubiquitin from E2 enzymes to lysine residues of client proteins via isopeptide bonds. The specificity of substrate binding and the expression and localization of E3 ligases can, however, endow these proteins with unique features with variable effects on mitochondrial, metabolic and CNS function. By comparing and contrasting two E3 ligases, Parkin and C-terminus of HSC70-Interacting protein (CHIP) we seek to highlight the biophysical properties that may promote mitochondrial dysfunction, acute stress signaling and critical developmental periods to cease in response to mutations in these genes. Encoded by over 600 human genes, RING-finger proteins are the largest class of E3 ligases. Parkin contains three RING finger domains, with R1 and R2 separated by an in-between region (IBR) domain. Loss-of-function mutations in Parkin were identified in patients with early onset Parkinson's disease. CHIP is a member of the Ubox family of E3 ligases. It contains an N-terminal TPR domain and forms unique asymmetric homodimers. While CHIP can substitute for mutated Parkin and enhance survival, CHIP also has unique functions. The differences between these proteins are underscored by the observation that unlike Parkin-deficient animals, CHIP-null animals age prematurely and have significantly impaired motor function. These properties make these E3 ligases appealing targets for clinical intervention. In this work, we discuss how biophysical and metabolic properties of these E3 ligases have driven rapid progress in identifying roles for E3 ligases in development, proteostasis, mitochondrial biology, and cell health, as well as new data about how these proteins alter the CNS proteome.
Collapse
Affiliation(s)
- Britney N Lizama
- Neuroscience Graduate Group, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States.
| | - Amy M Palubinsky
- Neuroscience Graduate Group, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States
| | - BethAnn McLaughlin
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Department of Pharmacology, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States
| |
Collapse
|
11
|
Maiti P, Manna J, Dunbar GL. Current understanding of the molecular mechanisms in Parkinson's disease: Targets for potential treatments. Transl Neurodegener 2017; 6:28. [PMID: 29090092 PMCID: PMC5655877 DOI: 10.1186/s40035-017-0099-z] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
Gradual degeneration and loss of dopaminergic neurons in the substantia nigra, pars compacta and subsequent reduction of dopamine levels in striatum are associated with motor deficits that characterize Parkinson’s disease (PD). In addition, half of the PD patients also exhibit frontostriatal-mediated executive dysfunction, including deficits in attention, short-term working memory, speed of mental processing, and impulsivity. The most commonly used treatments for PD are only partially or transiently effective and are available or applicable to a minority of patients. Because, these therapies neither restore the lost or degenerated dopaminergic neurons, nor prevent or delay the disease progression, the need for more effective therapeutics is critical. In this review, we provide a comprehensive overview of the current understanding of the molecular signaling pathways involved in PD, particularly within the context of how genetic and environmental factors contribute to the initiation and progression of this disease. The involvement of molecular chaperones, autophagy-lysosomal pathways, and proteasome systems in PD are also highlighted. In addition, emerging therapies, including pharmacological manipulations, surgical procedures, stem cell transplantation, gene therapy, as well as complementary, supportive and rehabilitation therapies to prevent or delay the progression of this complex disease are reviewed.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA.,Department of Biology, Saginaw Valley State University, Saginaw, MI 48604 USA
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38105 USA
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA
| |
Collapse
|
12
|
Yang MH, Chen KC, Chiang PW, Chung TW, Chen WJ, Chu PY, Chen SCJ, Lu YS, Yuan CH, Wang MC, Lin CY, Huang YF, Jong SB, Lin PC, Tyan YC. Proteomic Profiling of Neuroblastoma Cells Adhesion on Hyaluronic Acid-Based Surface for Neural Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1917394. [PMID: 28053978 PMCID: PMC5174748 DOI: 10.1155/2016/1917394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/10/2016] [Accepted: 10/05/2016] [Indexed: 12/26/2022]
Abstract
The microenvironment of neuron cells plays a crucial role in regulating neural development and regeneration. Hyaluronic acid (HA) biomaterial has been applied in a wide range of medical and biological fields and plays important roles in neural regeneration. PC12 cells have been reported to be capable of endogenous NGF synthesis and secretion. The purpose of this research was to assess the effect of HA biomaterial combining with PC12 cells conditioned media (PC12 CM) in neural regeneration. Using SH-SY5Y cells as an experimental model, we found that supporting with PC12 CM enhanced HA function in SH-SY5Y cell proliferation and adhesion. Through RP-nano-UPLC-ESI-MS/MS analyses, we identified increased expression of HSP60 and RanBP2 in SH-SY5Y cells grown on HA-modified surface with cotreatment of PC12 CM. Moreover, we also identified factors that were secreted from PC12 cells and may promote SH-SY5Y cell proliferation and adhesion. Here, we proposed a biomaterial surface enriched with neurotrophic factors for nerve regeneration application.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ko-Chin Chen
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Pei-Wen Chiang
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tze-Wen Chung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 112, Taiwan
| | - Wan-Jou Chen
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Pei-Yu Chu
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sharon Chia-Ju Chen
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Shan Lu
- Office of Safety, Health and Environment, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Cheng-Hui Yuan
- Mass Spectrometry Laboratory, Chemical, Molecular and Materials Analysis Center, Department of Chemistry, National University of Singapore, Singapore 119077
| | - Ming-Chen Wang
- Department of Biomedical Engineering, Chung Yuan Christian University, Chungli 300, Taiwan
| | - Chia-Yang Lin
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ying-Fong Huang
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Shiang-Bin Jong
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Po-Chiao Lin
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Yu-Chang Tyan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
13
|
Kuter K, Kratochwil M, Marx SH, Hartwig S, Lehr S, Sugawa MD, Dencher NA. Native DIGE proteomic analysis of mitochondria from substantia nigra and striatum during neuronal degeneration and its compensation in an animal model of early Parkinson's disease. Arch Physiol Biochem 2016; 122:238-256. [PMID: 27467289 DOI: 10.1080/13813455.2016.1197948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cause of Parkinson's disease (PD) is still not understood. Motor symptoms are not observed at early stages of disease due to compensatory processes. Dysfunction of mitochondria was indicated already at preclinical PD. Selective toxin 6-OHDA was applied to kill dopaminergic neurons in substantia nigra and disturb neuronal transmission in striatum. Early phase of active degeneration and later stage, when surviving cells adapted to function normally, were analysed. 2D BN/SDS difference gel electrophoresis (DIGE) of mitochondrial proteome enabled to point out crucial processes involved at both time-points in dopaminergic structures. Marker proteins such as DPYSL2, HSP60, ATP1A3, EAAT2 indicated structural remodelling, cytoskeleton rearrangement, organelle trafficking, axon outgrowth and regeneration. Adaptations in dopaminergic and glutamatergic neurotransmission, recycling of synaptic vesicles, along with enlargement of mitochondria mass were proposed as causative for compensation. Changed expression of carbohydrates metabolism and oxidative phosphorylation proteins were described, including their protein-protein interactions and supercomplex assembly.
Collapse
Affiliation(s)
- Katarzyna Kuter
- a Department of Neuropsychopharmacology , Polish Academy of Sciences , Kraków , Poland
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Manuela Kratochwil
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sven-Hendric Marx
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sonja Hartwig
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Stephan Lehr
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Michiru D Sugawa
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
- e Clinical Neurobiology, Charité-Universitätsmedizin , Berlin , Germany
| | - Norbert A Dencher
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| |
Collapse
|
14
|
More SV, Kumar H, Cho DY, Yun YS, Choi DK. Toxin-Induced Experimental Models of Learning and Memory Impairment. Int J Mol Sci 2016; 17:E1447. [PMID: 27598124 PMCID: PMC5037726 DOI: 10.3390/ijms17091447] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 02/07/2023] Open
Abstract
Animal models for learning and memory have significantly contributed to novel strategies for drug development and hence are an imperative part in the assessment of therapeutics. Learning and memory involve different stages including acquisition, consolidation, and retrieval and each stage can be characterized using specific toxin. Recent studies have postulated the molecular basis of these processes and have also demonstrated many signaling molecules that are involved in several stages of memory. Most insights into learning and memory impairment and to develop a novel compound stems from the investigations performed in experimental models, especially those produced by neurotoxins models. Several toxins have been utilized based on their mechanism of action for learning and memory impairment such as scopolamine, streptozotocin, quinolinic acid, and domoic acid. Further, some toxins like 6-hydroxy dopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amyloid-β are known to cause specific learning and memory impairment which imitate the disease pathology of Parkinson's disease dementia and Alzheimer's disease dementia. Apart from these toxins, several other toxins come under a miscellaneous category like an environmental pollutant, snake venoms, botulinum, and lipopolysaccharide. This review will focus on the various classes of neurotoxin models for learning and memory impairment with their specific mechanism of action that could assist the process of drug discovery and development for dementia and cognitive disorders.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Hemant Kumar
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Duk-Yeon Cho
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Yo-Sep Yun
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| |
Collapse
|
15
|
Swaroop S, Sengupta N, Suryawanshi AR, Adlakha YK, Basu A. HSP60 plays a regulatory role in IL-1β-induced microglial inflammation via TLR4-p38 MAPK axis. J Neuroinflammation 2016; 13:27. [PMID: 26838598 PMCID: PMC4736186 DOI: 10.1186/s12974-016-0486-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2016] [Indexed: 01/08/2023] Open
Abstract
Background IL-1β, also known as “the master regulator of inflammation”, is a potent pro-inflammatory cytokine secreted by activated microglia in response to pathogenic invasions or neurodegeneration. It initiates a vicious cycle of inflammation and orchestrates various molecular mechanisms involved in neuroinflammation. The role of IL-1β has been extensively studied in neurodegenerative disorders; however, molecular mechanisms underlying inflammation induced by IL-1β are still poorly understood. The objective of our study is the comprehensive identification of molecular circuitry involved in IL-1β-induced inflammation in microglia through protein profiling. Methods To achieve our aim, we performed the proteomic analysis of N9 microglial cells with and without IL-1β treatment at different time points. Expression of HSP60 in response to IL-1β administration was checked by quantitative real-time PCR, immunoblotting, and immunofluorescence. Interaction of HSP60 with TLR4 was determined by co-immunoprecipitation. Inhibition of TLR4 was done using TLR4 inhibitor to reveal its effect on IL-1β-induced inflammation. Further, effect of HSP60 knockdown and overexpression were assessed on the inflammation in microglia. Specific MAPK inhibitors were used to reveal the downstream MAPK exclusively involved in HSP60-induced inflammation in microglia. Results Total 21 proteins were found to be differentially expressed in response to IL-1β treatment in N9 microglial cells. In silico analysis of these proteins revealed unfolded protein response as one of the most significant molecular functions, and HSP60 turned out to be a key hub molecule. IL-1β induced the expression as well as secretion of HSP60 in extracellular milieu during inflammation of N9 cells. Secreted HSP60 binds to TLR4 and inhibition of TLR4 suppressed IL-1β-induced inflammation to a significant extent. Our knockdown and overexpression studies demonstrated that HSP60 increases the phosphorylation of ERK, JNK, and p38 MAPKs in N9 cells during inflammation. Specific inhibition of p38 by inhibitors suppressed HSP60-induced inflammation, thus pointed towards the major role of p38 MAPK rather than ERK1/2 and JNK in HSP60-induced inflammation. Furthermore, silencing of upstream modulator of p38, i.e., MEK3/6 also reduced HSP60-induced inflammation. Conclusions IL-1β induces expression of HSP60 in N9 microglial cells that further augments inflammation via TLR4-p38 MAPK axis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0486-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shalini Swaroop
- National Brain Research Centre, Manesar, Haryana, 122051, India
| | | | | | - Yogita K Adlakha
- National Brain Research Centre, Manesar, Haryana, 122051, India.
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122051, India.
| |
Collapse
|
16
|
Zhao C, Li H, Zhao XJ, Liu ZX, Zhou P, Liu Y, Feng MJ. Heat shock protein 60 affects behavioral improvement in a rat model of Parkinson’s disease grafted with human umbilical cord mesenchymal stem cell-derived dopaminergic-like neurons. Neurochem Res 2016; 41:1238-49. [DOI: 10.1007/s11064-015-1816-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/19/2015] [Accepted: 12/23/2015] [Indexed: 01/01/2023]
|
17
|
Investigating the role of Sirt1-modulated oxidative stress in relation to benign paroxysmal positional vertigo and Parkinson's disease. Neurobiol Aging 2015; 36:2607-16. [PMID: 26130063 DOI: 10.1016/j.neurobiolaging.2015.05.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/29/2015] [Accepted: 05/20/2015] [Indexed: 01/08/2023]
Abstract
Benign paroxysmal positional vertigo (BPPV) is one of the most frequently encountered primary complaints in dizziness clinics. The incidence of BPPV has been proven to increase with age. The relationship between BPPV and another neurodegenerative disease, Parkinson's disease (PD), has not been previously discussed. This study aimed to investigate the relationship of BPPV and PD with oxidative stress. A total of 30,811 subjects participated in our cohort study. The study cohort comprised 5057 BPPV patients and a comparison cohort of 25,754 nonBPPV patients. SIRT1 axis gene expression was investigated in BPPV patient blood samples and a PD cell model of 6-hydroxydopamine (6-OHDA)-treated PC-12 cells to elucidate the potential in vitro and in vivo mechanisms of degeneration in PD and BPPV. Our data suggest that BPPV patients with histories of head injuries show a significantly higher hazard to develop subsequent PD (hazard ratio, 3.942; confidence interval, 1.523-10.205, p = 0.005). We also observed that oxidative status is increased in blood samples from patients with BPPV. Our in vitro study suggests that SIRT1 function is inhibited by oxidative stress, which thereby promotes 6-hydroxydopamine-induced cell death. We conclude that BPPV is independently associated with an increased risk of PD. This finding may be attributed to oxidative stress-mediated inhibition of SIRT1 expression levels.
Collapse
|
18
|
Gammazza AM, Colangeli R, Orban G, Pierucci M, Di Gennaro G, Bello ML, D'Aniello A, Bucchieri F, Pomara C, Valentino M, Muscat R, Benigno A, Zummo G, de Macario EC, Cappello F, Di Giovanni G, Macario AJL. Hsp60 response in experimental and human temporal lobe epilepsy. Sci Rep 2015; 5:9434. [PMID: 25801186 PMCID: PMC4371150 DOI: 10.1038/srep09434] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/20/2015] [Indexed: 01/01/2023] Open
Abstract
The mitochondrial chaperonin Hsp60 is a ubiquitous molecule with multiple roles, constitutively expressed and inducible by oxidative stress. In the brain, Hsp60 is widely distributed and has been implicated in neurological disorders, including epilepsy. A role for mitochondria and oxidative stress has been proposed in epileptogenesis of temporal lobe epilepsy (TLE). Here, we investigated the involvement of Hsp60 in TLE using animal and human samples. Hsp60 immunoreactivity in the hippocampus, measured by Western blotting and immunohistochemistry, was increased in a rat model of TLE. Hsp60 was also increased in the hippocampal dentate gyrus neurons somata and neuropil and hippocampus proper (CA3, CA1) of the epileptic rats. We also determined the circulating levels of Hsp60 in epileptic animals and TLE patients using ELISA. The epileptic rats showed circulating levels of Hsp60 higher than controls. Likewise, plasma post-seizure Hsp60 levels in patients were higher than before the seizure and those of controls. These results demonstrate that Hsp60 is increased in both animals and patients with TLE in affected tissues, and in plasma in response to epileptic seizures, and point to it as biomarker of hippocampal stress potentially useful for diagnosis and patient management.
Collapse
Affiliation(s)
- Antonella Marino Gammazza
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Roberto Colangeli
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Gergely Orban
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Massimo Pierucci
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | | | - Margherita Lo Bello
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | | | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Cristoforo Pomara
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Richard Muscat
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Arcangelo Benigno
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giovanni Zummo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore; and IMET, Columbus Center, Baltimore, MD, USA
| | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Institute “Paolo Sotgiu” for Research in Quantitative and Quantum Psychiatry and Cardiology, University of Human Sciences and Technology (LUDES), Lugano, Switzerland
| | - Giuseppe Di Giovanni
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Neuroscience Division, School of Bioscience, Cardiff University, Cardiff, UK
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore; and IMET, Columbus Center, Baltimore, MD, USA
| |
Collapse
|