1
|
Baskar D, Ganji SR, Thomas A, Polavarapu K, Nandeesh BN, Sanka SB, Srivastava K, Kotambail A, Arunachal G, Boddu VK, Nashi S, Nalini A, Vengalil S. MICU1 related myopathy - a rare report from India. J Neuromuscul Dis 2024; 11:1295-1299. [PMID: 39973469 DOI: 10.1177/22143602241288400] [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] [Indexed: 02/21/2025]
Abstract
Mitochondrial Calcium Uptake 1 (MICU1) is an important component of mitochondrial calcium channel regulator. Mutations in MICU1 result in a rare syndrome of myopathy with extrapyramidal features. Here we report a rare case of MICU1 related myopathy from India. A 23 years old male presented with 10 years history of proximal muscle weakness with exertional myalgia and fatigue. Examination showed facial dysmorphism with facial weakness and mildly reduced visual acuity. Limb girdle pattern of weakness with hypoactive tendon reflexes were noted without extrapyramidal signs. He had elevated serum creatine level of 1542 IU/L. Muscle MRI had novel findings of selective fatty infiltration of hamstrings, medial gastrocnemius and soleus. Muscle biopsy showed myopathic with secondary neurogenic changes along with few COX deficient fibres. Genetic analysis showed compound heterozygous pathogenic variants in MICU1 gene at intron 9 (c.1072-1 G > C) - splice site variant and exon 5 (c.513T > A) - stop gained variant, both resulting in loss of function of the protein. The variants were segregating in unaffected parents in heterozygous state with variant c.1072-1 G > C in the unaffected father and variant c.513T > A (p. Tyr171*) in the unaffected mother confirming the diagnosis. This report highlights the phenotype of limb girdle weakness with facial dysmorphism and optic atrophy expanding the spectrum of MICU1 related syndrome with novel MRI muscle and histopathological findings.
Collapse
Affiliation(s)
- Dipti Baskar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Suma Reddy Ganji
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Aneesha Thomas
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Bevinahalli N Nandeesh
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Sai Bhargava Sanka
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Kosha Srivastava
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | | | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Vijay Kumar Boddu
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), India
| |
Collapse
|
2
|
Moawad MHED, Serag I, Alkhawaldeh IM, Abbas A, Sharaf A, Alsalah S, Sadeq MA, Shalaby MMM, Hefnawy MT, Abouzid M, Meshref M. Exploring the Mechanisms and Therapeutic Approaches of Mitochondrial Dysfunction in Alzheimer's Disease: An Educational Literature Review. Mol Neurobiol 2024:10.1007/s12035-024-04468-y. [PMID: 39254911 DOI: 10.1007/s12035-024-04468-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Alzheimer's disease (AD) presents a significant challenge to global health. It is characterized by progressive cognitive deterioration and increased rates of morbidity and mortality among older adults. Among the various pathophysiologies of AD, mitochondrial dysfunction, encompassing conditions such as increased reactive oxygen production, dysregulated calcium homeostasis, and impaired mitochondrial dynamics, plays a pivotal role. This review comprehensively investigates the mechanisms of mitochondrial dysfunction in AD, focusing on aspects such as glucose metabolism impairment, mitochondrial bioenergetics, calcium signaling, protein tau and amyloid-beta-associated synapse dysfunction, mitophagy, aging, inflammation, mitochondrial DNA, mitochondria-localized microRNAs, genetics, hormones, and the electron transport chain and Krebs cycle. While lecanemab is the only FDA-approved medication to treat AD, we explore various therapeutic modalities for mitigating mitochondrial dysfunction in AD, including antioxidant drugs, antidiabetic agents, acetylcholinesterase inhibitors (FDA-approved to manage symptoms), nutritional supplements, natural products, phenylpropanoids, vaccines, exercise, and other potential treatments.
Collapse
Affiliation(s)
- Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Abdulrahman Sharaf
- Department of Clinical Pharmacy, Salmaniya Medical Complex, Government Hospital, Manama, Bahrain
| | - Sumaya Alsalah
- Ministry of Health, Primary Care, Governmental Health Centers, Manama, Bahrain
| | | | | | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Bojang KP, Manchana V. Impact of vegetarianism on cognition and neuropsychological status among urban community-dwelling adults in Telangana, South India: a cross-sectional study. Eur J Nutr 2024; 63:1089-1101. [PMID: 38305863 DOI: 10.1007/s00394-024-03328-8] [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: 09/05/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
PURPOSE The potential benefits of vegetarian diets in reducing cognitive impairment have garnered attention due to existing mixed results; hence, our study aims to examine the impact of vegetarianism on cognitive function and neuropsychological status among urban community-dwelling adults from Telangana. METHODS The dietary patterns were self-reported and dietary data collected using a Food Frequency Questionnaire while cognitive function was assessed using Mini-Mental State Examination (MMSE) and depression, anxiety, and stress (DASS-21) questionnaire for psychological measures. Adults (N = 304) aged 40 years and above, who followed either a vegetarian or non-vegetarian diet for at least 6 months prior to data collection, scored MMSE ≥ 19 indicating mild cognitive impairment, and were recruited using convenience sampling. RESULTS Among the participants, vegetarians (n = 155) exhibited significantly better mood states compared to non-vegetarians (n = 149), as indicated by lower scores on the DASS subscales for depression (10.0 ± 0.06 vs. 17.0 ± 0.07, p = < 0.001), anxiety (4.0 ± 0.05 vs. 6.0 ± 0.07, p = 0.005), and stress (8.0 ± 0.02 vs. 10.0 ± 0.05, p = 0.007). Vegetarians also demonstrated superior cognitive functioning compared to non-vegetarians, as indicated by higher MMSE scores (26.0 ± 0.04 vs. 24.0 ± 0.03, p = < 0.001). MMSE scores were inversely correlated with depression, anxiety, and stress for vegetarians (ρ = - 0.371, p = 0.000; ρ = - 0.027, p = 0.734; and ρ = - 0.105, p = 0.914), respectively. Similar to the vegetarians group, MMSE scores were negatively correlated with depression (ρ = - 0.059), but the correlation is not significant. CONCLUSION Vegetarianism exerted a positive influence on the cognitive and neuropsychological status of the investigated population. Nevertheless, additional research is required to comprehend the underlying mechanisms that elucidate the long-term effects of vegetarianism and plant-based nutritional interventions on brain health.
Collapse
Affiliation(s)
| | - Varalakshmi Manchana
- School of Medical Sciences, University of Hyderabad, Hyderabad, Telangana, India.
| |
Collapse
|
4
|
Sánchez JC, Alemán A, Henao JF, Olaya JC, Ehrlich BE. NCS-1 protein regulates TRPA1 channel through the PI3K pathway in breast cancer and neuronal cells. J Physiol Biochem 2024; 80:451-463. [PMID: 38564162 PMCID: PMC11074019 DOI: 10.1007/s13105-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
The physical and functional interaction between transient receptor potential channel ankyrin 1 (TRPA1) and neuronal calcium sensor 1 (NCS-1) was assessed. NCS-1 is a calcium (Ca2+) sensor found in many tissues, primarily neurons, and TRPA1 is a Ca2+ channel involved not only in thermal and pain sensation but also in conditions such as cancer and chemotherapy-induced peripheral neuropathy, in which NCS-1 is also a regulatory component.We explored the interactions between these two proteins by employing western blot, qRT-PCR, co-immunoprecipitation, Ca2+ transient monitoring with Fura-2 spectrophotometry, and electrophysiology assays in breast cancer cells (MDA-MB-231) with different levels of NCS-1 expression and neuroblastoma cells (SH-SY5Y).Our findings showed that the expression of TRPA1 was directly correlated with NCS-1 levels at both the protein and mRNA levels. Additionally, we found a physical and functional association between these two proteins. Physically, the NCS-1 and TRPA1 co-immunoprecipitate. Functionally, NCS-1 enhanced TRPA1-dependent Ca2+ influx, current density, open probability, and conductance, where the functional effects depended on PI3K. Conclusion: NCS-1 appears to act not only as a Ca2+ sensor but also modulates TRPA1 protein expression and channel function in a direct fashion through the PI3K pathway. These results contribute to understanding how Ca2+ homeostasis is regulated and provides a mechanism underlying conditions where Ca2+ dynamics are compromised, including breast cancer. With a cellular pathway identified, targeted treatments can be developed for breast cancer and neuropathy, among other related diseases.
Collapse
Affiliation(s)
- Julio C Sánchez
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia.
| | - Alexander Alemán
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan F Henao
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan C Olaya
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Barbara E Ehrlich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
5
|
Kwon HJ, Jung HY, Choi SY, Hwang IK, Kim DW, Shin MJ. Protective effect of Tat fused HPCA protein on neuronal cell death caused by ischemic injury. Heliyon 2024; 10:e23488. [PMID: 38192804 PMCID: PMC10772100 DOI: 10.1016/j.heliyon.2023.e23488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Background Bain ischemia is a disease that occurs for various reasons, induces reactive oxygen species (ROS), and causes fatal damage to the nervous system. Protective effect of HPCA on ischemic injury has not been extensively studied despite its significance in regulating calcium homeostasis and promoting neuronal survival in CA1 region of the brain. Objective We investigate the role of HPCA in ischemic injury using a cell-permeable Tat peptide fused HPCA protein (Tat-HPCA). Methods Western blot analysis determined the penetration of Tat-HPCA into HT-22 cells and apoptotic signaling pathways. 5-CFDA, AM, DCF-DA, and TUNEL staining confirmed intracellular ROS production and DNA damage. The intracellular Ca2+ was measured in primary cultured neurons treated with H2O2. Protective effects were examined using immunohistochemistry and cognitive function tests by passive avoidance test and 8-arm radial maze test. Results Tat-HPCA effectively penetrated into HT-22 cells and inhibited H2O2-induced apoptosis, oxidative stress, and DNA fragmentation. It also effectively inhibited phosphorylation of JNK and regulated the activation of Caspase, Bax, Bcl-2, and PARP, leading to inhibition of apoptosis. Moreover, Ca2+ concentration decreased in cells treated with Tat-HPCA in primary cultured neurons. In an animal model of ischemia, Tat-HPCA effectively penetrated the hippocampus, inhibited cell death, and regulated activities of astrocytes and microglia. Additionally, Cognitive function tests show that Tat-HPCA improves neurobehavioral outcomes after cerebral ischemic injury. Conclusion These results suggest that Tat-HPCA might have potential as a therapeutic agent for treating oxidative stress-related diseases induced by ischemic injury, including ischemia.
Collapse
Affiliation(s)
- Hyun Jung Kwon
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyo Young Jung
- Department of Veterinary Medicine, Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
6
|
Tran CM, Ra JS, Rhyu DY, Kim KT. Transcriptome analysis reveals differences in developmental neurotoxicity mechanism of methyl-, ethyl-, and propyl- parabens in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 268:115704. [PMID: 37979356 DOI: 10.1016/j.ecoenv.2023.115704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/26/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Studies on the comparison of developmental (neuro) toxicity of parabens are currently limited, and unharmonized concentrations between phenotypic observations and transcriptome analysis hamper the understanding of their differential molecular mechanisms. Thus, developmental toxicity testing was conducted herein using the commonly used methyl- (MtP), ethyl- (EtP), and propyl-parabens (PrP) in zebrafish embryos. With a benchmark dose of 5%, embryonic-mortality-based point-of-departure (M-POD) values of the three parabens were determined, and changes in locomotor behavior were evaluated at concentrations of 0, M-POD/50, M-POD/10, and M-POD, where transcriptome analysis was conducted to explore the underlying neurotoxicity mechanism. Higher long-chained parabens were more toxic than short-chained parabens, as determined by the M-POD values of 154.1, 72.6, and 24.2 µM for MtP, EtP, and PrP, respectively. Meanwhile, exposure to EtP resulted in hyperactivity, whereas no behavioral effect was observed with MtP and PrP. Transcriptome analysis revealed that abnormal behaviors in the EtP-exposed group were associated with distinctly enriched pathways in signaling, transport, calcium ion binding, and metal binding. In contrast, exposure to MtP and PrP mainly disrupted membranes and transmembranes, which are closely linked to abnormal embryonic development rather than neurobehavioral changes. According to the changes in the expressions of signature mRNAs, tentative transcriptome-based POD values for each paraben were determined as MtP (2.68 µM), EtP (3.85 µM), and PrP (1.4 µM). This suggests that different molecular perturbations initiated at similar concentrations determined the extent and toxicity outcome differently. Our findings provide insight into better understanding the differential developmental neurotoxicity mechanisms of parabens.
Collapse
Affiliation(s)
- Cong Minh Tran
- Department of Energy and Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Jin-Sung Ra
- Eco-testing and Risk Assessment Center, Korea Institute of Industrial Technology (KITECH), Ansan 15588, Republic of Korea
| | - Dong Young Rhyu
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 FOUR, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Ki-Tae Kim
- Department of Energy and Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; Department of Environmental Engineering, Seoul National University of Sciences and Technology, Seoul 01811, Republic of Korea.
| |
Collapse
|
7
|
Muñoz-Reyes D, McClelland LJ, Arroyo-Urea S, Sánchez-Yepes S, Sabín J, Pérez-Suárez S, Menendez M, Mansilla A, García-Nafría J, Sprang S, Sanchez-Barrena MJ. The neuronal calcium sensor NCS-1 regulates the phosphorylation state and activity of the Gα chaperone and GEF Ric-8A. eLife 2023; 12:e86151. [PMID: 38018500 PMCID: PMC10732572 DOI: 10.7554/elife.86151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023] Open
Abstract
The neuronal calcium sensor 1 (NCS-1), an EF-hand Ca2+ binding protein, and Ric-8A coregulate synapse number and probability of neurotransmitter release. Recently, the structures of Ric-8A bound to Gα have revealed how Ric-8A phosphorylation promotes Gα recognition and activity as a chaperone and guanine nucleotide exchange factor. However, the molecular mechanism by which NCS-1 regulates Ric-8A activity and its interaction with Gα subunits is not well understood. Given the interest in the NCS-1/Ric-8A complex as a therapeutic target in nervous system disorders, it is necessary to shed light on this molecular mechanism of action at atomic level. We have reconstituted NCS-1/Ric-8A complexes to conduct a multimodal approach and determine the sequence of Ca2+ signals and phosphorylation events that promote the interaction of Ric-8A with Gα. Our data show that the binding of NCS-1 and Gα to Ric-8A are mutually exclusive. Importantly, NCS-1 induces a structural rearrangement in Ric-8A that traps the protein in a conformational state that is inaccessible to casein kinase II-mediated phosphorylation, demonstrating one aspect of its negative regulation of Ric-8A-mediated G-protein signaling. Functional experiments indicate a loss of Ric-8A guanine nucleotide exchange factor (GEF) activity toward Gα when complexed with NCS-1, and restoration of nucleotide exchange activity upon increasing Ca2+ concentration. Finally, the high-resolution crystallographic data reported here define the NCS-1/Ric-8A interface and will allow the development of therapeutic synapse function regulators with improved activity and selectivity.
Collapse
Affiliation(s)
- Daniel Muñoz-Reyes
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| | - Levi J McClelland
- Center for Biomolecular Structure and Dynamics, and Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Sandra Arroyo-Urea
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Laboratorio de Microscopías Avanzadas (LMA), University of ZaragozaZaragozaSpain
| | - Sonia Sánchez-Yepes
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadridSpain
| | - Juan Sabín
- AFFINImeter Scientific & Development team, Software 4 Science DevelopmentsSantiago de CompostelaSpain
- Departamento de Física Aplicada, Universidad de Santiago de CompostelaSantiago de CompostelaSpain
| | - Sara Pérez-Suárez
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| | - Margarita Menendez
- Department of Biological Physical-Chemisty, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
- Ciber of Respiratory Diseases, ISCIIIMadridSpain
| | - Alicia Mansilla
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadridSpain
- Department of Systems Biology, Universidad de AlcalaMadridSpain
| | - Javier García-Nafría
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Laboratorio de Microscopías Avanzadas (LMA), University of ZaragozaZaragozaSpain
| | - Stephen Sprang
- Center for Biomolecular Structure and Dynamics, and Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Maria Jose Sanchez-Barrena
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| |
Collapse
|
8
|
Alluri R, Kilari EK, Pasala PK, Kopalli SR, Koppula S. Repurposing Diltiazem for Its Neuroprotective Anti-Dementia Role against Intra-Cerebroventricular Streptozotocin-Induced Sporadic Alzheimer's Disease-Type Rat Model. Life (Basel) 2023; 13:1688. [PMID: 37629545 PMCID: PMC10455909 DOI: 10.3390/life13081688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neuropsychiatric disorder and a common cause of progressive dementia. Diltiazem (DTZ), the non-dihydropyridine benzothiazepine class of calcium channel blocker (CCB), used clinically in angina and other cardiovascular disorders, has proven neurological benefits. In the present study, the neuroprotective anti-dementia effects of DTZ against intra-cerebroventricular-streptozotocin (ICV-STZ)-induced sporadic AD (SAD)-type rat model was investigated. ICV-STZ-induced cognitive impairments were measured via passive avoidance and Morris water maze tasks. Anti-oxidative enzyme status, pro-inflammatory markers, and amyloid-beta (Aβ) protein expression in rat brain tissues were measured using ELISA kits, Western blotting, and immunostaining techniques. The data revealed that ICV-STZ injection in rats significantly induced cognitive deficits and altered the levels of oxidative and pro-inflammatory markers (p < 0.05~p < 0.001). Treatment with DTZ (10 mg/kg, 20 mg/kg, and 40 mg/kg, p.o.) daily for twenty-one days, 1 h before a single ICV-STZ (3 mg/kg) injection, significantly improved cognitive impairments and ameliorated the ICV-STZ-induced altered nitrite, pro-inflammatory cytokines (TNF-α, and IL-1β) and anti-oxidative enzyme levels (superoxide dismutase, lipid peroxidation, and glutathione). Further, DTZ restored the increased Aβ protein expression in ICV-STZ-induced brain tissue. Considering the results obtained, DTZ might have a potential therapeutic role in treating and managing AD and related dementia pathologies due to its anti-dementia activity in SAD-type conditions in rats induced by ICV-STZ.
Collapse
Affiliation(s)
- Ramesh Alluri
- Cognitive Science Research Initiative Lab., Department of Pharmacology, Vishnu Institute of Pharmaceutical Education and Research, Medak Dist., Narsapur 502313, India
| | - Eswar Kumar Kilari
- Department of Pharmacology, University College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, India
| | - Praveen Kumar Pasala
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Jawaharlal Nehru Technological University Anantapur—JNTUA, Anantapur 515721, India
| | - Spandana Rajendra Kopalli
- Department of Integrated Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Science, Konkuk University, Chungju-si 380-701, Republic of Korea
| |
Collapse
|
9
|
Babić Leko M, Mihelčić M, Jurasović J, Nikolac Perković M, Španić E, Sekovanić A, Orct T, Zubčić K, Langer Horvat L, Pleić N, Kiđemet-Piskač S, Vogrinc Ž, Pivac N, Diana A, Borovečki F, Hof PR, Šimić G. Heavy Metals and Essential Metals Are Associated with Cerebrospinal Fluid Biomarkers of Alzheimer's Disease. Int J Mol Sci 2022; 24:467. [PMID: 36613911 PMCID: PMC9820819 DOI: 10.3390/ijms24010467] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Various metals have been associated with the pathogenesis of Alzheimer's disease (AD), principally heavy metals that are environmental pollutants (such as As, Cd, Hg, and Pb) and essential metals whose homeostasis is disturbed in AD (such as Cu, Fe, and Zn). Although there is evidence of the involvement of these metals in AD, further research is needed on their mechanisms of toxicity. To further assess the involvement of heavy and essential metals in AD pathogenesis, we compared cerebrospinal fluid (CSF) AD biomarkers to macro- and microelements measured in CSF and plasma. We tested if macro- and microelements' concentrations (heavy metals (As, Cd, Hg, Ni, Pb, and Tl), essential metals (Na, Mg, K, Ca, Fe, Co, Mn, Cu, Zn, and Mo), essential non-metals (B, P, S, and Se), and other non-essential metals (Al, Ba, Li, and Sr)) are associated with CSF AD biomarkers that reflect pathological changes in the AD brain (amyloid β1-42, total tau, phosphorylated tau isoforms, NFL, S100B, VILIP-1, YKL-40, PAPP-A, and albumin). We used inductively coupled plasma mass spectroscopy (ICP-MS) to determine macro- and microelements in CSF and plasma, and enzyme-linked immunosorbent assays (ELISA) to determine protein biomarkers of AD in CSF. This study included 193 participants (124 with AD, 50 with mild cognitive impairment, and 19 healthy controls). Simple correlation, as well as machine learning algorithms (redescription mining and principal component analysis (PCA)), demonstrated that levels of heavy metals (As, Cd, Hg, Ni, Pb, and Tl), essential metals (Ca, Co, Cu, Fe, Mg, Mn, Mo, Na, K, and Zn), and essential non-metals (P, S, and Se) are positively associated with CSF phosphorylated tau isoforms, VILIP-1, S100B, NFL, and YKL-40 in AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Medical Biology, University of Split School of Medicine, 21000 Split, Croatia
| | - Matej Mihelčić
- Department of Mathematics, University of Zagreb Faculty of Science, 10000 Zagreb, Croatia
| | - Jasna Jurasović
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | | | - Ena Španić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ankica Sekovanić
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Tatjana Orct
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Klara Zubčić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Nikolina Pleić
- Department of Medical Biology, University of Split School of Medicine, 21000 Split, Croatia
| | | | - Željka Vogrinc
- Laboratory for Neurobiochemistry, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Nela Pivac
- Ruđer Bošković Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia
| | - Andrea Diana
- Laboratory of Neurogenesis and Neuropoiesis, Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Fran Borovečki
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb Medical School, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
10
|
Tan YZ, Abbas YM, Wu JZ, Wu D, Keon KA, Hesketh GG, Bueler SA, Gingras AC, Robinson CV, Grinstein S, Rubinstein JL. CryoEM of endogenous mammalian V-ATPase interacting with the TLDc protein mEAK-7. Life Sci Alliance 2022; 5:e202201527. [PMID: 35794005 PMCID: PMC9263379 DOI: 10.26508/lsa.202201527] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/18/2022] Open
Abstract
V-ATPases are rotary proton pumps that serve as signaling hubs with numerous protein binding partners. CryoEM with exhaustive focused classification allowed detection of endogenous proteins associated with porcine kidney V-ATPase. An extra C subunit was found in ∼3% of complexes, whereas ∼1.6% of complexes bound mEAK-7, a protein with proposed roles in dauer formation in nematodes and mTOR signaling in mammals. High-resolution cryoEM of porcine kidney V-ATPase with recombinant mEAK-7 showed that mEAK-7's TLDc domain interacts with V-ATPase's stator, whereas its C-terminal α helix binds V-ATPase's rotor. This crosslink would be expected to inhibit rotary catalysis. However, unlike the yeast TLDc protein Oxr1p, exogenous mEAK-7 does not inhibit V-ATPase and mEAK-7 overexpression in cells does not alter lysosomal or phagosomal pH. Instead, cryoEM suggests that the mEAK-7:V-ATPase interaction is disrupted by ATP-induced rotation of the rotor. Comparison of Oxr1p and mEAK-7 binding explains this difference. These results show that V-ATPase binding by TLDc domain proteins can lead to effects ranging from strong inhibition to formation of labile interactions that are sensitive to the enzyme's activity.
Collapse
Affiliation(s)
- Yong Zi Tan
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Yazan M Abbas
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Jing Ze Wu
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Di Wu
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Kristine A Keon
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Geoffrey G Hesketh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Stephanie A Bueler
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Carol V Robinson
- Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
Putscher E, Hecker M, Fitzner B, Boxberger N, Schwartz M, Koczan D, Lorenz P, Zettl UK. Genetic risk variants for multiple sclerosis are linked to differences in alternative pre-mRNA splicing. Front Immunol 2022; 13:931831. [PMID: 36405756 PMCID: PMC9670805 DOI: 10.3389/fimmu.2022.931831] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/12/2022] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic immune-mediated disease of the central nervous system to which a genetic predisposition contributes. Over 200 genetic regions have been associated with increased disease risk, but the disease-causing variants and their functional impact at the molecular level are mostly poorly defined. We hypothesized that single-nucleotide polymorphisms (SNPs) have an impact on pre-mRNA splicing in MS. METHODS Our study focused on 10 bioinformatically prioritized SNP-gene pairs, in which the SNP has a high potential to alter alternative splicing events (ASEs). We tested for differential gene expression and differential alternative splicing in B cells from MS patients and healthy controls. We further examined the impact of the SNP genotypes on ASEs and on splice isoform expression levels. Novel genotype-dependent effects on splicing were verified with splicing reporter minigene assays. RESULTS We were able to confirm previously described findings regarding the relation of MS-associated SNPs with the ASEs of the pre-mRNAs from GSDMB and SP140. We also observed an increased IL7R exon 6 skipping when comparing relapsing and progressive MS patients to healthy subjects. Moreover, we found evidence that the MS risk alleles of the SNPs rs3851808 (EFCAB13), rs1131123 (HLA-C), rs10783847 (TSFM), and rs2014886 (TSFM) may contribute to a differential splicing pattern. Of particular interest is the genotype-dependent exon skipping of TSFM due to the SNP rs2014886. The minor allele T creates a donor splice site, resulting in the expression of the exon 3 and 4 of a short TSFM transcript isoform, whereas in the presence of the MS risk allele C, this donor site is absent, and thus the short transcript isoform is not expressed. CONCLUSION In summary, we found that genetic variants from MS risk loci affect pre-mRNA splicing. Our findings substantiate the role of ASEs with respect to the genetics of MS. Further studies on how disease-causing genetic variants may modify the interactions between splicing regulatory sequence elements and RNA-binding proteins can help to deepen our understanding of the genetic susceptibility to MS.
Collapse
Affiliation(s)
- Elena Putscher
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Michael Hecker
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Brit Fitzner
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Nina Boxberger
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Margit Schwartz
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Dirk Koczan
- Rostock University Medical Center, Institute of Immunology, Rostock, Germany
| | - Peter Lorenz
- Rostock University Medical Center, Institute of Immunology, Rostock, Germany
| | - Uwe Klaus Zettl
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| |
Collapse
|
12
|
IQM-PC332, a Novel DREAM Ligand with Antinociceptive Effect on Peripheral Nerve Injury-Induced Pain. Int J Mol Sci 2022; 23:ijms23042142. [PMID: 35216258 PMCID: PMC8876042 DOI: 10.3390/ijms23042142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
Neuropathic pain is a form of chronic pain arising from damage of the neural cells that sense, transmit or process sensory information. Given its growing prevalence and common refractoriness to conventional analgesics, the development of new drugs with pain relief effects constitutes a prominent clinical need. In this respect, drugs that reduce activity of sensory neurons by modulating ion channels hold the promise to become effective analgesics. Here, we evaluated the mechanical antinociceptive effect of IQM-PC332, a novel ligand of the multifunctional protein downstream regulatory element antagonist modulator (DREAM) in rats subjected to chronic constriction injury of the sciatic nerve as a model of neuropathic pain. IQM-PC332 administered by intraplantar (0.01–10 µg) or intraperitoneal (0.02–1 µg/kg) injection reduced mechanical sensitivity by ≈100% of the maximum possible effect, with ED50 of 0.27 ± 0.05 µg and 0.09 ± 0.01 µg/kg, respectively. Perforated-patch whole-cell recordings in isolated dorsal root ganglion (DRG) neurons showed that IQM-PC332 (1 and 10 µM) reduced ionic currents through voltage-gated K+ channels responsible for A-type potassium currents, low, T-type, and high voltage-activated Ca2+ channels, and transient receptor potential vanilloid-1 (TRPV1) channels. Furthermore, IQM-PC332 (1 µM) reduced electrically evoked action potentials in DRG neurons from neuropathic animals. It is suggested that by modulating multiple DREAM–ion channel signaling complexes, IQM-PC332 may serve a lead compound of novel multimodal analgesics.
Collapse
|
13
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Bitarafan F, Khodaeian M, Amjadi Sardehaei E, Darvishi FZ, Almadani N, Nilipour Y, Garshasbi M. Identification of a novel MICU1 nonsense variant causes myopathy with extrapyramidal signs in an Iranian consanguineous family. Mol Cell Pediatr 2021; 8:6. [PMID: 33969448 PMCID: PMC8107061 DOI: 10.1186/s40348-021-00116-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ca2+ as a universal second messenger regulates basic biological functions including cell cycle, cell proliferation, cell differentiation, and cell death. Lack of the protein mitochondrial calcium uptake1 (MICU1), which has been regarded as a gatekeeper of Ca ions, leads to the abnormal mitochondrial Ca2+ handling, excessive production of reactive oxygen species (ROS), and increased cell death. Mutations in MICU1 gene causes a very rare neuromuscular disease, myopathy with extrapyramidal signs (MPXPS), due to primary alterations in mitochondrial calcium signaling which demonstrates the key role of mitochondrial Ca2+ uptake. To date, 13 variants have been reported in MICU1 gene in 44 patients presented with the vast spectrum of symptoms. CASE PRESENTATION Here, we report a 44-year-old Iranian patient presented with learning disability, muscle weakness, easy fatigability, reduced tendon reflexes, ataxia, gait disturbance, elevated hepatic transaminases, elevated serum creatine kinase (CK), and elevated lactate dehydrogenase (LDH). We identified a novel nonsense variant c.385C>T; p.(R129*) in MICU1 gene by whole exome sequencing (WES) and segregation analysis. CONCLUSIONS Our finding along with previous studies provides more evidence on the clinical presentation of the disease caused by pathogenic mutations in MICU1. Finding more variants and expanding the spectrum of the disease increases the diagnostic rate of molecular testing in screening of this kind of diseases and in turn improves the quality of counseling for at risk couples and helps them to minimize the risks of having affected children.
Collapse
Affiliation(s)
- Fatemeh Bitarafan
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | | | | | - Navid Almadani
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Yalda Nilipour
- Pediatric Pathology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
15
|
Liu T, Song Y, Hu A. Neuroprotective mechanisms of mangiferin in neurodegenerative diseases. Drug Dev Res 2021; 82:494-502. [PMID: 33458836 DOI: 10.1002/ddr.21783] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022]
Abstract
The central nervous system (CNS) regulates and coordinates an extensive array of complex processes requiring harmonious regulation of specific genes. CNS disorders represent a large burden on society and cause enormous disability and economic losses. Traditional Chinese medicine (TCM) has been used for many years in the treatment of neurological illnesses, such as Alzheimer's disease, Parkinson's disease, stroke, and depression, as the combination of TCM and Western medicine has superior therapeutic efficacy and minimal toxic side effects. Mangiferin (MGF) is an active compound of the traditional Chinese herb rhizome anemarrhenae, which has antioxidant, anti-inflammation, anti-lipid peroxidation, immunomodulatory, and anti-apoptotic functions in the CNS. MGF has been demonstrated to have therapeutic effects in CNS diseases through a multitude of mechanisms. This review outlines the latest research on the neuroprotective ability of MGF and the diverse molecular mechanisms involved.
Collapse
Affiliation(s)
- Tingjun Liu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| | - Yuanjian Song
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China.,Department of Genetics, Xuzhou Engineering Research Center of Medical Genetics and Transformation, Xuzhou Medical University, Xuzhou, China
| | - Ankang Hu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| |
Collapse
|
16
|
Disorder in a two-domain neuronal Ca 2+-binding protein regulates domain stability and dynamics using ligand mimicry. Cell Mol Life Sci 2020; 78:2263-2278. [PMID: 32936312 PMCID: PMC7966663 DOI: 10.1007/s00018-020-03639-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/08/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022]
Abstract
Understanding the interplay between sequence, structure and function of proteins has been complicated in recent years by the discovery of intrinsically disordered proteins (IDPs), which perform biological functions in the absence of a well-defined three-dimensional fold. Disordered protein sequences account for roughly 30% of the human proteome and in many proteins, disordered and ordered domains coexist. However, few studies have assessed how either feature affects the properties of the other. In this study, we examine the role of a disordered tail in the overall properties of the two-domain, calcium-sensing protein neuronal calcium sensor 1 (NCS-1). We show that loss of just six of the 190 residues at the flexible C-terminus is sufficient to severely affect stability, dynamics, and folding behavior of both ordered domains. We identify specific hydrophobic contacts mediated by the disordered tail that may be responsible for stabilizing the distal N-terminal domain. Moreover, sequence analyses indicate the presence of an LSL-motif in the tail that acts as a mimic of native ligands critical to the observed order-disorder communication. Removing the disordered tail leads to a shorter life-time of the ligand-bound complex likely originating from the observed destabilization. This close relationship between order and disorder may have important implications for how investigations into mixed systems are designed and opens up a novel avenue of drug targeting exploiting this type of behavior.
Collapse
|
17
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
18
|
Baksheeva VE, Nemashkalova EL, Firsov AM, Zalevsky AO, Vladimirov VI, Tikhomirova NK, Philippov PP, Zamyatnin AA, Zinchenko DV, Antonenko YN, Permyakov SE, Zernii EY. Membrane Binding of Neuronal Calcium Sensor-1: Highly Specific Interaction with Phosphatidylinositol-3-Phosphate. Biomolecules 2020; 10:biom10020164. [PMID: 31973069 PMCID: PMC7072451 DOI: 10.3390/biom10020164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neuronal calcium sensors are a family of N-terminally myristoylated membrane-binding proteins possessing a different intracellular localization and thereby targeting unique signaling partner(s). Apart from the myristoyl group, the membrane attachment of these proteins may be modulated by their N-terminal positively charged residues responsible for specific recognition of the membrane components. Here, we examined the interaction of neuronal calcium sensor-1 (NCS-1) with natural membranes of different lipid composition as well as individual phospholipids in form of multilamellar liposomes or immobilized monolayers and characterized the role of myristoyl group and N-terminal lysine residues in membrane binding and phospholipid preference of the protein. NCS-1 binds to photoreceptor and hippocampal membranes in a Ca2+-independent manner and the binding is attenuated in the absence of myristoyl group. Meanwhile, the interaction with photoreceptor membranes is less dependent on myristoylation and more sensitive to replacement of K3, K7, and/or K9 of NCS-1 by glutamic acid, reflecting affinity of the protein to negatively charged phospholipids. Consistently, among the major phospholipids, NCS-1 preferentially interacts with phosphatidylserine and phosphatidylinositol with micromolar affinity and the interaction with the former is inhibited upon mutating of N-terminal lysines of the protein. Remarkably, NCS-1 demonstrates pronounced specific binding to phosphoinositides with high preference for phosphatidylinositol-3-phosphate. The binding does not depend on myristoylation and, unexpectedly, is not sensitive to the charge inversion mutations. Instead, phosphatidylinositol-3-phosphate can be recognized by a specific site located in the N-terminal region of the protein. These data provide important novel insights into the general mechanism of membrane binding of NCS-1 and its targeting to specific phospholipids ensuring involvement of the protein in phosphoinositide-regulated signaling pathways.
Collapse
Affiliation(s)
- Viktoriia E. Baksheeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, 142290 Moscow Region, Russia; (E.L.N.); (S.E.P.)
| | - Alexander M. Firsov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
| | - Arthur O. Zalevsky
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia;
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Vasily I. Vladimirov
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences in Pushchino, Pushchino, 142290 Moscow Region, Russia; (V.I.V.); (D.V.Z.)
| | - Natalia K. Tikhomirova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
| | - Pavel P. Philippov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
| | - Andrey A. Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Dmitry V. Zinchenko
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences in Pushchino, Pushchino, 142290 Moscow Region, Russia; (V.I.V.); (D.V.Z.)
| | - Yuri N. Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
| | - Sergey E. Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, 142290 Moscow Region, Russia; (E.L.N.); (S.E.P.)
| | - Evgeni Yu. Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (A.M.F.); (N.K.T.); (P.P.P.); (Y.N.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-495-939-2344
| |
Collapse
|
19
|
Dean B, Parkin GM, Gibbons AS. Associations between catechol-O-methyltransferase (COMT) genotypes at rs4818 and rs4680 and gene expression in human dorsolateral prefrontal cortex. Exp Brain Res 2020; 238:477-486. [DOI: 10.1007/s00221-020-05730-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022]
|
20
|
Abstract
Synaptic plasticity is a fundamental property of neurons referring to the activity-dependent changes in the strength and efficacy of synaptic transmission at preexisting synapses. Such changes can last from milliseconds to hours, days, or even longer and are involved in learning and memory as well as in development and response of the brain to injuries. Several types of synaptic plasticity have been described across neuronal types, brain regions, and species, but all of them share in one way or another capital importance of Ca2+-mediated processes. In this chapter, we will focus on the Ca2+-dependent events necessary for the induction and expression of multiple forms of synaptic plasticity.
Collapse
|
21
|
Simons C, Benkert J, Deuter N, Poetschke C, Pongs O, Schneider T, Duda J, Liss B. NCS-1 Deficiency Affects mRNA Levels of Genes Involved in Regulation of ATP Synthesis and Mitochondrial Stress in Highly Vulnerable Substantia nigra Dopaminergic Neurons. Front Mol Neurosci 2019; 12:252. [PMID: 31827421 PMCID: PMC6890851 DOI: 10.3389/fnmol.2019.00252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 09/27/2019] [Indexed: 12/20/2022] Open
Abstract
Neuronal Ca2+ sensor proteins (NCS) transduce changes in Ca2+ homeostasis into altered signaling and neuronal function. NCS-1 activity has emerged as important for neuronal viability and pathophysiology. The progressive degeneration of dopaminergic (DA) neurons, particularly within the Substantia nigra (SN), is the hallmark of Parkinson's disease (PD), causing its motor symptoms. The activity-related Ca2+ homeostasis of SN DA neurons, mitochondrial dysfunction, and metabolic stress promote neurodegeneration and PD. In contrast, NCS-1 in general has neuroprotective effects. The underlying mechanisms are unclear. We analyzed transcriptional changes in SN DA neurons upon NCS-1 loss by combining UV-laser microdissection and RT-qPCR-approaches to compare expression levels of a panel of PD and/or Ca2+-stress related genes from wildtype and NCS-1 KO mice. In NCS-1 KO, we detected significantly lower mRNA levels of mitochondrially coded ND1, a subunit of the respiratory chain, and of the neuron-specific enolase ENO2, a glycolytic enzyme. We also detected lower levels of the mitochondrial uncoupling proteins UCP4 and UCP5, the PARK7 gene product DJ-1, and the voltage-gated Ca2+ channel Cav2.3 in SN DA neurons from NCS-1 KO. Transcripts of other analyzed uncoupling proteins (UCPs), mitochondrial Ca2+ transporters, PARK genes, and ion channels were not altered. As Cav channels are linked to regulation of gene expression, metabolic stress and degeneration of SN DA neurons in PD, we analyzed Cav2.3 KO mice, to address if the transcriptional changes in NCS-1 KO were also present in Cav.2.3 KO, and thus probably correlated with lower Cav2.3 transcripts. However, in SN DA neurons from Cav2.3 KO mice, ND1 mRNA as well as genomic DNA levels were elevated, while ENO2, UCP4, UCP5, and DJ-1 transcript levels were not altered. In conclusion, our data indicate a possible novel function of NCS-1 in regulating gene transcription or stabilization of mRNAs in SN DA neurons. Although we do not provide functional data, our findings at the transcript level could point to impaired ATP production (lower ND1 and ENO2) and elevated metabolic stress (lower UCP4, UCP5, and DJ-1 levels) in SN DA neurons from NCS-1 KO mice. We speculate that NCS-1 is involved in stimulating ATP synthesis, while at the same time controlling mitochondrial metabolic stress, and in this way could protect SN DA neurons from degeneration.
Collapse
Affiliation(s)
- Carsten Simons
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Julia Benkert
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Nora Deuter
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | - Olaf Pongs
- Institute of Physiology, Center for Integrative Physiology and Molecular Medicine, University of the Saarland, Homburg, Germany
| | - Toni Schneider
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Johanna Duda
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,New College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Abstract
Kv channel-interacting proteins (KChIPs) belong to the neuronal calcium sensor (NCS) family of Ca2+-binding EF-hand proteins. KChIPs constitute a group of specific auxiliary β-subunits for Kv4 channels, the molecular substrate of transient potassium currents in both neuronal and non-neuronal tissues. Moreover, KChIPs can interact with presenilins to control ER calcium signaling and apoptosis, and with DNA to control gene transcription. Ca2+ binding via their EF-hands, with the consequence of conformational changes, is well documented for KChIPs. Moreover, the Ca2+ dependence of the presenilin/KChIP complex may be related to Alzheimer’s disease and the Ca2+ dependence of the DNA/KChIP complex to pain sensing. However, only in few cases could the Ca2+ binding to KChIPs be directly linked to the control of excitability in nerve and muscle cells known to express Kv4/KChIP channel complexes. This review summarizes current knowledge about the Ca2+ binding properties of KChIPs and the Ca2+ dependencies of macromolecular complexes containing KChIPs, including those with presenilins, DNA and especially Kv4 channels. The respective physiological or pathophysiolgical roles of Ca2+ binding to KChIPs are discussed.
Collapse
Affiliation(s)
- Robert Bähring
- a Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin , Universitätsklinikum Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
23
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
24
|
Chen KF, Lowe S, Lamaze A, Krätschmer P, Jepson J. Neurocalcin regulates nighttime sleep and arousal in Drosophila. eLife 2019; 8:e38114. [PMID: 30865587 PMCID: PMC6415939 DOI: 10.7554/elife.38114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/29/2019] [Indexed: 01/28/2023] Open
Abstract
Sleep-like states in diverse organisms can be separated into distinct stages, each with a characteristic arousal threshold. However, the molecular pathways underlying different sleep stages remain unclear. The fruit fly, Drosophila melanogaster, exhibits consolidated sleep during both day and night, with night sleep associated with higher arousal thresholds compared to day sleep. Here we identify a role for the neuronal calcium sensor protein Neurocalcin (NCA) in promoting sleep during the night but not the day by suppressing nocturnal arousal and hyperactivity. We show that both circadian and light-sensing pathways define the temporal window in which NCA promotes sleep. Furthermore, we find that NCA promotes sleep by suppressing synaptic release from a dispersed wake-promoting neural network and demonstrate that the mushroom bodies, a sleep-regulatory center, are a module within this network. Our results advance the understanding of how sleep stages are genetically defined.
Collapse
Affiliation(s)
- Ko-Fan Chen
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Simon Lowe
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Angélique Lamaze
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Patrick Krätschmer
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - James Jepson
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| |
Collapse
|
25
|
Maklad A, Sharma A, Azimi I. Calcium Signaling in Brain Cancers: Roles and Therapeutic Targeting. Cancers (Basel) 2019; 11:cancers11020145. [PMID: 30691160 PMCID: PMC6406375 DOI: 10.3390/cancers11020145] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Calcium signaling, in addition to its numerous physiological roles, is also implicated in several pathological conditions including cancer. An increasing body of evidence suggest critical roles of calcium signaling in the promotion of different aspects of cancer, including cell proliferation, therapy resistance and metastatic-related processes. In many cases, this is associated with altered expression and/or activity of some calcium channels and pumps. Brain cancers have also been the subject of many of these studies. In addition to diverse roles of calcium signals in normal brain function, a number of proteins involved in calcium transport are implicated to have specific roles in some brain cancers including gliomas, medulloblastoma, neuroblastoma and meningioma. This review discusses research that has been conducted so far to understand diverse roles of Ca2+-transporting proteins in the progression of brain cancers, as well as any attempts to target these proteins towards a therapeutic approach for the control of brain cancers. Finally, some knowledge gaps in the field that may need to be further considered are also discussed.
Collapse
Affiliation(s)
- Ahmed Maklad
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| | - Anjana Sharma
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| | - Iman Azimi
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| |
Collapse
|
26
|
Choudhary D, Kragelund BB, Heidarsson PO, Cecconi C. The Complex Conformational Dynamics of Neuronal Calcium Sensor-1: A Single Molecule Perspective. Front Mol Neurosci 2018; 11:468. [PMID: 30618617 PMCID: PMC6304440 DOI: 10.3389/fnmol.2018.00468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/03/2018] [Indexed: 01/16/2023] Open
Abstract
The human neuronal calcium sensor-1 (NCS-1) is a multispecific two-domain EF-hand protein expressed predominantly in neurons and is a member of the NCS protein family. Structure-function relationships of NCS-1 have been extensively studied showing that conformational dynamics linked to diverse ion-binding is important to its function. NCS-1 transduces Ca2+ changes in neurons and is linked to a wide range of neuronal functions such as regulation of neurotransmitter release, voltage-gated Ca2+ channels and neuronal outgrowth. Defective NCS-1 can be deleterious to cells and has been linked to serious neuronal disorders like autism. Here, we review recent studies describing at the single molecule level the structural and mechanistic details of the folding and misfolding processes of the non-myristoylated NCS-1. By manipulating one molecule at a time with optical tweezers, the conformational equilibria of the Ca2+-bound, Mg2+-bound and apo states of NCS-1 were investigated revealing a complex folding mechanism underlain by a rugged and multidimensional energy landscape. The molecular rearrangements that NCS-1 undergoes to transit from one conformation to another and the energetics of these reactions are tightly regulated by the binding of divalent ions (Ca2+ and Mg2+) to its EF-hands. At pathologically high Ca2+ concentrations the protein sometimes follows non-productive misfolding pathways leading to kinetically trapped and potentially harmful misfolded conformations. We discuss the significance of these misfolding events as well as the role of inter-domain interactions in shaping the energy landscape and ultimately the biological function of NCS-1. The conformational equilibria of NCS-1 are also compared to those of calmodulin (CaM) and differences and similarities in the behavior of these proteins are rationalized in terms of structural properties.
Collapse
Affiliation(s)
- Dhawal Choudhary
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy.,Center S3, CNR Institute Nanoscience, Modena, Italy
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Ciro Cecconi
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy.,Center S3, CNR Institute Nanoscience, Modena, Italy
| |
Collapse
|
27
|
Tsvetkov PO, Roman AY, Baksheeva VE, Nazipova AA, Shevelyova MP, Vladimirov VI, Buyanova MF, Zinchenko DV, Zamyatnin AA, Devred F, Golovin AV, Permyakov SE, Zernii EY. Functional Status of Neuronal Calcium Sensor-1 Is Modulated by Zinc Binding. Front Mol Neurosci 2018; 11:459. [PMID: 30618610 PMCID: PMC6302015 DOI: 10.3389/fnmol.2018.00459] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 11/28/2018] [Indexed: 11/29/2022] Open
Abstract
Neuronal calcium sensor-1 (NCS-1) protein is abundantly expressed in the central nervous system and retinal neurons, where it regulates many vital processes such as synaptic transmission. It coordinates three calcium ions by EF-hands 2-4, thereby transducing Ca2+ signals to a wide range of protein targets, including G protein-coupled receptors and their kinases. Here, we demonstrate that NCS-1 also has Zn2+-binding sites, which affect its structural and functional properties upon filling. Fluorescence and circular dichroism experiments reveal the impact of Zn2+ binding on NCS-1 secondary and tertiary structure. According to atomic absorption spectroscopy and isothermal titration calorimetry studies, apo-NCS-1 has two high-affinity (4 × 106 M-1) and one low-affinity (2 × 105 M-1) Zn2+-binding sites, whereas Mg2+-loaded and Ca2+-loaded forms (which dominate under physiological conditions) bind two zinc ions with submicromolar affinity. Metal competition analysis and circular dichroism studies suggest that Zn2+-binding sites of apo- and Mg2+-loaded NCS-1 overlap with functional EF-hands of the protein. Consistently, high Zn2+ concentrations displace Mg2+ from the EF-hands and decrease the stoichiometry of Ca2+ binding. Meanwhile, one of the EF-hands of Zn2+-saturated NCS-1 exhibits a 14-fold higher calcium affinity, which increases the overall calcium sensitivity of the protein. Based on QM/MM molecular dynamics simulations, Zn2+ binding to Ca2+-loaded NCS-1 could occur at EF-hands 2 and 4. The high-affinity zinc binding increases the thermal stability of Ca2+-free NCS-1 and favours the interaction of its Ca2+-loaded form with target proteins, such as dopamine receptor D2R and GRK1. In contrast, low-affinity zinc binding promotes NCS-1 aggregation accompanied by the formation of twisted rope-like structures. Altogether, our findings suggest a complex interplay between magnesium, calcium and zinc binding to NCS-1, leading to the appearance of multiple conformations of the protein, in turn modulating its functional status.
Collapse
Affiliation(s)
- Philipp O Tsvetkov
- Aix-Marseille University, CNRS, INP, Institute of Neurophysiopathology, Faculty of Pharmacy, Marseille, France
| | - Andrei Yu Roman
- Institute of Physiologically Active Compounds (RAS), Chernogolovka, Russia
| | - Viktoriia E Baksheeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Aliya A Nazipova
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Russia
| | - Marina P Shevelyova
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Russia
| | - Vasiliy I Vladimirov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia
| | - Michelle F Buyanova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry V Zinchenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia
| | - Andrey A Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - François Devred
- Aix-Marseille University, CNRS, INP, Institute of Neurophysiopathology, Faculty of Pharmacy, Marseille, France
| | - Andrey V Golovin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- Faculty of Computer Science, Higher School of Economics, Moscow, Russia
| | - Sergei E Permyakov
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Russia
| | - Evgeni Yu Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
28
|
Naranjo R, González P, Lopez-Hurtado A, Dopazo XM, Mellström B, Naranjo JR. Inhibition of the Neuronal Calcium Sensor DREAM Modulates Presenilin-2 Endoproteolysis. Front Mol Neurosci 2018; 11:449. [PMID: 30559648 PMCID: PMC6287014 DOI: 10.3389/fnmol.2018.00449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/21/2018] [Indexed: 11/14/2022] Open
Abstract
Deregulated intracellular Ca2+ and protein homeostasis underlie synaptic dysfunction and are common features in neurodegenerative diseases. DREAM, also known as calsenilin or KChIP-3, is a multifunctional Ca2+ binding protein of the neuronal calcium sensor superfamily with specific functions through protein-DNA and protein-protein interactions. Small-molecules able to bind DREAM, like the anti-diabetic drug repaglinide, disrupt some of the interactions with other proteins and modulate DREAM activity on Kv4 channels or on the processing of activating transcription factor 6 (ATF6). Here, we show the interaction of endogenous DREAM and presenilin-2 (PS2) in mouse brain and, using DREAM deficient mice or transgenic mice overexpressing a dominant active DREAM (daDREAM) mutant in the brain, we provide genetic evidence of the role of DREAM in the endoproteolysis of endogenous PS2. We show that repaglinide disrupts the interaction between DREAM and the C-terminal PS2 fragment (Ct-PS2) by coimmunoprecipitation assays. Exposure to sub-micromolar concentrations of repaglinide reduces the levels of Ct-PS2 fragment in N2a neuroblastoma cells. These results suggest that the interaction between DREAM and PS2 may represent a new target for modulation of PS2 processing, which could have therapeutic potential in Alzheimer’s disease (AD) treatment.
Collapse
Affiliation(s)
- Rocío Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| | - Paz González
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| | - Alejandro Lopez-Hurtado
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| | - Xosé M Dopazo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| | - Britt Mellström
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| | - José R Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,National Biotechnology Center (CNB), CSIC, Madrid, Spain
| |
Collapse
|
29
|
Franco R, Aguinaga D, Reyes I, Canela EI, Lillo J, Tarutani A, Hasegawa M, Del Ser-Badia A, Del Rio JA, Kreutz MR, Saura CA, Navarro G. N-Methyl-D-Aspartate Receptor Link to the MAP Kinase Pathway in Cortical and Hippocampal Neurons and Microglia Is Dependent on Calcium Sensors and Is Blocked by α-Synuclein, Tau, and Phospho-Tau in Non-transgenic and Transgenic APP Sw,Ind Mice. Front Mol Neurosci 2018; 11:273. [PMID: 30233307 PMCID: PMC6127644 DOI: 10.3389/fnmol.2018.00273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/18/2018] [Indexed: 11/14/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) respond to glutamate to allow the influx of calcium ions and the signaling to the mitogen-activated protein kinase (MAPK) cascade. Both MAPK- and Ca2+-mediated events are important for both neurotransmission and neural cell function and fate. Using a heterologous expression system, we demonstrate that NMDAR may interact with the EF-hand calcium-binding proteins calmodulin, calneuron-1, and NCS1 but not with caldendrin. NMDARs were present in primary cultures of both neurons and microglia from cortex and hippocampus. Calmodulin in microglia, and calmodulin and NCS1 in neurons, are necessary for NMDA-induced MAP kinase pathway activation. Remarkably, signaling to the MAP kinase pathway was blunted in primary cultures of cortical and hippocampal neurons and microglia from wild-type animals by proteins involved in neurodegenerative diseases: α-synuclein, Tau, and p-Tau. A similar blockade by pathogenic proteins was found using samples from the APPSw,Ind transgenic Alzheimer’s disease model. Interestingly, a very marked increase in NMDAR–NCS1 complexes was identified in neurons and a marked increase of both NMDAR–NCS1 and NMDAR–CaM complexes was identified in microglia from the transgenic mice. The results show that α-synuclein, Tau, and p-Tau disrupt the signaling of NMDAR to the MAPK pathway and that calcium sensors are important for NMDAR function both in neurons and microglia. Finally, it should be noted that the expression of receptor–calcium sensor complexes, specially those involving NCS1, is altered in neural cells from APPSw,Ind mouse embryos/pups.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David Aguinaga
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Reyes
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Enric I Canela
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Lillo
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | - Airi Tarutani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Anna Del Ser-Badia
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - José A Del Rio
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group Dendritic Organelles and Synaptic Function, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos A Saura
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
30
|
Angelats E, Requesens M, Aguinaga D, Kreutz MR, Franco R, Navarro G. Neuronal Calcium and cAMP Cross-Talk Mediated by Cannabinoid CB 1 Receptor and EF-Hand Calcium Sensor Interactions. Front Cell Dev Biol 2018; 6:67. [PMID: 30073165 PMCID: PMC6060245 DOI: 10.3389/fcell.2018.00067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/12/2018] [Indexed: 11/19/2022] Open
Abstract
Endocannabinoids are important players in neural development and function. They act via receptors, whose activation inhibits cAMP production. The aim of the paper was to look for calcium- and cAMP-signaling cross-talk mediated by cannabinoid CB1 receptors (CB1R) and to assess the relevance of EF-hand CaM-like calcium sensors in this regard. Using a heterologous expression system, we demonstrated that CB1R interacts with calneuron-1 and NCS1 but not with caldendrin. Furthermore, interaction motives were identified in both calcium binding proteins and the receptor, and we showed that the first two sensors competed for binding to the receptor in a Ca2+-dependent manner. Assays in neuronal primary cultures showed that, CB1R-NCS1 complexes predominate at basal Ca2+ levels, whereas in the presence of ionomycin, a calcium ionophore, CB1R-calneuron-1 complexes were more abundant. Signaling assays following forskolin-induced intracellular cAMP levels showed in mouse striatal neurons that binding of CB1R to NCS1 is required for CB1R-mediated signaling, while the binding of CB1R to calneuron-1 completely blocked Gi-mediated signaling in response to a selective receptor agonist, arachidonyl-2-chloroethylamide. Calcium levels and interaction with calcium sensors may even lead to apparent Gs coupling after CB1R agonist challenge.
Collapse
Affiliation(s)
- Edgar Angelats
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Requesens
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - David Aguinaga
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Novel approaches to probe the binding of recoverin to membranes. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2018; 47:679-691. [DOI: 10.1007/s00249-018-1304-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/05/2018] [Accepted: 04/13/2018] [Indexed: 02/08/2023]
|
32
|
Yang T, Britt JK, Cintrón-Pérez CJ, Vázquez-Rosa E, Tobin KV, Stalker G, Hardie J, Taugher RJ, Wemmie J, Pieper AA, Lee A. Ca 2+-Binding Protein 1 Regulates Hippocampal-dependent Memory and Synaptic Plasticity. Neuroscience 2018; 380:90-102. [PMID: 29660444 DOI: 10.1016/j.neuroscience.2018.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/16/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022]
Abstract
Ca2+-binding protein 1 (CaBP1) is a Ca2+-sensing protein similar to calmodulin that potently regulates voltage-gated Ca2+ channels. Unlike calmodulin, however, CaBP1 is mainly expressed in neuronal cell-types and enriched in the hippocampus, where its function is unknown. Here, we investigated the role of CaBP1 in hippocampal-dependent behaviors using mice lacking expression of CaBP1 (C-KO). By western blot, the largest CaBP1 splice variant, caldendrin, was detected in hippocampal lysates from wild-type (WT) but not C-KO mice. Compared to WT mice, C-KO mice exhibited mild deficits in spatial learning and memory in both the Barnes maze and in Morris water maze reversal learning. In contextual but not cued fear-conditioning assays, C-KO mice showed greater freezing responses than WT mice. In addition, the number of adult-born neurons in the hippocampus of C-KO mice was ∼40% of that in WT mice, as measured by bromodeoxyuridine labeling. Moreover, hippocampal long-term potentiation was significantly reduced in C-KO mice. We conclude that CaBP1 is required for cellular mechanisms underlying optimal encoding of hippocampal-dependent spatial and fear-related memories.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jeremiah K Britt
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Coral J Cintrón-Pérez
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Edwin Vázquez-Rosa
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Grant Stalker
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jason Hardie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Rebecca J Taugher
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - John Wemmie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew A Pieper
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Free Radical, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Oncology Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Veterans Affairs, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Pappajohn Biomedical Institute and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
33
|
López-Hurtado A, Burgos DF, González P, Dopazo XM, González V, Rábano A, Mellström B, Naranjo JR. Inhibition of DREAM-ATF6 interaction delays onset of cognition deficit in a mouse model of Huntington's disease. Mol Brain 2018. [PMID: 29523177 PMCID: PMC5845147 DOI: 10.1186/s13041-018-0359-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The transcriptional repressor DREAM (downstream regulatory element antagonist modulator) is a multifunctional neuronal calcium sensor (NCS) that controls Ca2+ and protein homeostasis through gene regulation and protein-protein interactions. Downregulation of DREAM is part of an endogenous neuroprotective mechanism that improves ATF6 (activating transcription factor 6) processing, neuronal survival in the striatum, and motor coordination in R6/2 mice, a model of Huntington’s disease (HD). Whether modulation of DREAM activity can also ameliorate cognition deficits in HD mice has not been studied. Moreover, it is not known whether DREAM downregulation in HD is unique, or also occurs for other NCS family members. Using the novel object recognition test, we show that chronic administration of the DREAM-binding molecule repaglinide, or induced DREAM haplodeficiency delays onset of cognitive impairment in R6/1 mice, another HD model. The mechanism involves a notable rise in the levels of transcriptionally active ATF6 protein in the hippocampus after repaglinide administration. In addition, we show that reduction in DREAM protein in the hippocampus of HD patients was not accompanied by downregulation of other NCS family members. Our results indicate that DREAM inhibition markedly improves ATF6 processing in the hippocampus and that it might contribute to a delay in memory decline in HD mice. The mechanism of neuroprotection through DREAM silencing in HD does not apply to other NCS family members.
Collapse
Affiliation(s)
- Alejandro López-Hurtado
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain
| | - Daniel F Burgos
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain
| | - Paz González
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain
| | - Xose M Dopazo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain
| | - Valentina González
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Fundación CIEN, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Rábano
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Fundación CIEN, Instituto de Salud Carlos III, Madrid, Spain
| | - Britt Mellström
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain
| | - Jose R Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain. .,Centro Nacional de Biotecnología, CNB-CSIC, Darwin 3, E-28049, Madrid, Spain.
| |
Collapse
|
34
|
Zhou K, Cherra SJ, Goncharov A, Jin Y. Asynchronous Cholinergic Drive Correlates with Excitation-Inhibition Imbalance via a Neuronal Ca 2+ Sensor Protein. Cell Rep 2018; 19:1117-1129. [PMID: 28494862 DOI: 10.1016/j.celrep.2017.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/13/2017] [Accepted: 04/15/2017] [Indexed: 10/19/2022] Open
Abstract
Excitation-inhibition imbalance in neural networks is widely linked to neurological and neuropsychiatric disorders. However, how genetic factors alter neuronal activity, leading to excitation-inhibition imbalance, remains unclear. Here, using the C. elegans locomotor circuit, we examine how altering neuronal activity for varying time periods affects synaptic release pattern and animal behavior. We show that while short-duration activation of excitatory cholinergic neurons elicits a reversible enhancement of presynaptic strength, persistent activation results to asynchronous and reduced cholinergic drive, inducing imbalance between endogenous excitation and inhibition. We find that the neuronal calcium sensor protein NCS-2 is required for asynchronous cholinergic release in an activity-dependent manner and dampens excitability of inhibitory neurons non-cell autonomously. The function of NCS-2 requires its Ca2+ binding and membrane association domains. These results reveal a synaptic mechanism implicating asynchronous release in regulation of excitation-inhibition balance.
Collapse
Affiliation(s)
- Keming Zhou
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Salvatore J Cherra
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexandr Goncharov
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
35
|
Thapliyal S, Babu K. C. elegans Locomotion: Finding Balance in Imbalance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:185-196. [PMID: 30637699 DOI: 10.1007/978-981-13-3065-0_14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The excitation-inhibition (E-I) imbalance in neural circuits represents a hallmark of several neuropsychiatric disorders. The tiny nematode Caenorhabditis elegans has emerged as an excellent system to study the molecular mechanisms underlying this imbalance in neuronal circuits. The C. elegans body wall muscles receive inputs from both excitatory cholinergic and inhibitory GABAergic motor neurons at neuromuscular junctions (NMJ), making it an excellent model for studying the genetic and molecular mechanisms required for maintaining E-I balance at the NMJ. The cholinergic neurons form dyadic synapses wherein they synapse onto ipsilateral body wall muscles allowing for muscle contraction as well as onto GABAergic motor neurons that in turn synapse on the contralateral body wall muscles causing muscle relaxation. An alternating wave of contraction and relaxation mediated by excitatory and inhibitory signals maintains locomotion in C. elegans. This locomotory behavior requires an intricate balance between the excitatory cholinergic signaling and the inhibitory GABAergic signaling mechanisms.Studies on the C. elegans NMJ have provided insights into several molecular mechanisms that could regulate this balance in neural circuits. This review provides a discussion on multiple genetic factors including neuropeptides and their receptors, cell adhesion molecules, and other molecular pathways that have been associated with maintaining E-I balance in C. elegans motor circuits. Further, it also discusses the implications of these studies that could help us in understanding the role of E-I balance in mammalian neural circuits and how changes in this balance could give rise to brain disorders.
Collapse
Affiliation(s)
- Shruti Thapliyal
- Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India.
| | - Kavita Babu
- Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India.
| |
Collapse
|
36
|
Dudczig S, Currie PD, Jusuf PR. Developmental and adult characterization of secretagogin expressing amacrine cells in zebrafish retina. PLoS One 2017; 12:e0185107. [PMID: 28949993 PMCID: PMC5614429 DOI: 10.1371/journal.pone.0185107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
Calcium binding proteins show stereotypical expression patterns within diverse neuron types across the central nervous system. Here, we provide a characterization of developmental and adult secretagogin-immunolabelled neurons in the zebrafish retina with an emphasis on co-expression of multiple calcium binding proteins. Secretagogin is a recently identified and cloned member of the F-hand family of calcium binding proteins, which labels distinct neuron populations in the retinas of mammalian vertebrates. Both the adult distribution of secretagogin labeled retinal neurons as well as the developmental expression indicative of the stage of neurogenesis during which this calcium binding protein is expressed was quantified. Secretagogin expression was confined to an amacrine interneuron population in the inner nuclear layer, with monostratified neurites in the center of the inner plexiform layer and a relatively regular soma distribution (regularity index > 2.5 across central–peripheral areas). However, only a subpopulation (~60%) co-labeled with gamma-aminobutyric acid as their neurotransmitter, suggesting that possibly two amacrine subtypes are secretagogin immunoreactive. Quantitative co-labeling analysis with other known amacrine subtype markers including the three main calcium binding proteins parvalbumin, calbindin and calretinin identifies secretagogin immunoreactive neurons as a distinct neuron population. The highest density of secretagogin cells of ~1800 cells / mm2 remained relatively evenly along the horizontal meridian, whilst the density dropped of to 125 cells / mm2 towards the dorsal and ventral periphery. Thus, secretagogin represents a new amacrine label within the zebrafish retina. The developmental expression suggests a possible role in late stage differentiation. This characterization forms the basis of functional studies assessing how the expression of distinct calcium binding proteins might be regulated to compensate for the loss of one of the others.
Collapse
Affiliation(s)
- Stefanie Dudczig
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
| | - Peter David Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Patricia Regina Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
- * E-mail:
| |
Collapse
|
37
|
ARX polyalanine expansion mutations lead to migration impediment in the rostral cortex coupled with a developmental deficit of calbindin-positive cortical GABAergic interneurons. Neuroscience 2017. [PMID: 28627419 DOI: 10.1016/j.neuroscience.2017.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Aristaless-related homeobox gene (ARX) is indispensable for interneuron development. Patients with ARX polyalanine expansion mutations of the first two tracts (namely PA1 and PA2) suffer from intellectual disability of varying severity, with seizures a frequent comorbidity. The impact of PA1 and PA2 mutations on the brain development is unknown, hindering the search for therapeutic interventions. Here, we characterized the disturbances to cortical interneuron development in mice modeling the two most common ARX polyalanine expansion mutations in human. We found a consistent ∼40-50% reduction of calbindin-positive interneurons, but not Stt+ or Cr+ interneurons, within the cortex of newborn hemizygous mice (p=0.024) for both mutant strains compared to wildtype (p=0.011). We demonstrate that this was a consequence of calbindin precursor cells being arrested or delayed at the ventral subpallium en route of tangential migration. Ex-vivo assay validated this migration deficit in PA1 cells (p=0.0002) suggesting that the defect is contributed by intrinsic loss of Arx function within migrating cells. Both humans and mice with PA1 mutations present with severe clinical features, including intellectual disability and infantile spasms. Our data further demonstrated the pathogenic mechanism was robustly shared between PA1 and PA2 mutations, as previously reported including Arx protein reduction and overlapping transcriptome profiles within the developing mouse brains. Data from our study demonstrated that cortical calbindin interneuron development and migration is negatively affected by ARX polyalanine expansion mutations. Understanding the cellular pathogenesis contributing to disease manifestation is necessary to screen efficacy of potential therapeutic interventions.
Collapse
|
38
|
Hu P, Luo BH. Integrin αv
β8
Adopts a High Affinity State for Soluble Ligands Under Physiological Conditions. J Cell Biochem 2017; 118:2044-2052. [DOI: 10.1002/jcb.25780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/01/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Ping Hu
- Department of Biological Sciences; Louisiana State University; Baton Rouge Louisiana
| | - Bing-Hao Luo
- Department of Biological Sciences; Louisiana State University; Baton Rouge Louisiana
| |
Collapse
|
39
|
Riessland M, Kaczmarek A, Schneider S, Swoboda KJ, Löhr H, Bradler C, Grysko V, Dimitriadi M, Hosseinibarkooie S, Torres-Benito L, Peters M, Upadhyay A, Biglari N, Kröber S, Hölker I, Garbes L, Gilissen C, Hoischen A, Nürnberg G, Nürnberg P, Walter M, Rigo F, Bennett CF, Kye MJ, Hart AC, Hammerschmidt M, Kloppenburg P, Wirth B. Neurocalcin Delta Suppression Protects against Spinal Muscular Atrophy in Humans and across Species by Restoring Impaired Endocytosis. Am J Hum Genet 2017; 100:297-315. [PMID: 28132687 DOI: 10.1016/j.ajhg.2017.01.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/05/2017] [Indexed: 01/17/2023] Open
Abstract
Homozygous SMN1 loss causes spinal muscular atrophy (SMA), the most common lethal genetic childhood motor neuron disease. SMN1 encodes SMN, a ubiquitous housekeeping protein, which makes the primarily motor neuron-specific phenotype rather unexpected. SMA-affected individuals harbor low SMN expression from one to six SMN2 copies, which is insufficient to functionally compensate for SMN1 loss. However, rarely individuals with homozygous absence of SMN1 and only three to four SMN2 copies are fully asymptomatic, suggesting protection through genetic modifier(s). Previously, we identified plastin 3 (PLS3) overexpression as an SMA protective modifier in humans and showed that SMN deficit impairs endocytosis, which is rescued by elevated PLS3 levels. Here, we identify reduction of the neuronal calcium sensor Neurocalcin delta (NCALD) as a protective SMA modifier in five asymptomatic SMN1-deleted individuals carrying only four SMN2 copies. We demonstrate that NCALD is a Ca2+-dependent negative regulator of endocytosis, as NCALD knockdown improves endocytosis in SMA models and ameliorates pharmacologically induced endocytosis defects in zebrafish. Importantly, NCALD knockdown effectively ameliorates SMA-associated pathological defects across species, including worm, zebrafish, and mouse. In conclusion, our study identifies a previously unknown protective SMA modifier in humans, demonstrates modifier impact in three different SMA animal models, and suggests a potential combinatorial therapeutic strategy to efficiently treat SMA. Since both protective modifiers restore endocytosis, our results confirm that endocytosis is a major cellular mechanism perturbed in SMA and emphasize the power of protective modifiers for understanding disease mechanism and developing therapies.
Collapse
|
40
|
Gong Y, Zhu Y, Zou Y, Ma B, Nussinov R, Zhang Q. Human Neuronal Calcium Sensor-1 Protein Avoids Histidine Residues To Decrease pH Sensitivity. J Phys Chem B 2017; 121:508-517. [PMID: 28030949 PMCID: PMC6413881 DOI: 10.1021/acs.jpcb.6b11094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
pH is highly regulated in mammalian central nervous systems. Neuronal calcium sensor-1 (NCS-1) can interact with numerous target proteins. Compared to that in the NCS-1 protein of Caenorhabditis elegans, evolution has avoided the placement of histidine residues at positions 102 and 83 in the NCS-1 protein of humans and Xenopus laevis, possibly to decrease the conformational sensitivity to pH gradients in synaptic processes. We used all-atom molecular dynamics simulations to investigate the effects of amino acid substitutions between species on human NCS-1 by substituting Arg102 and Ser83 for histidine at neutral (R102H and S83H) and acidic pHs (R102Hp and S83Hp). Our cumulative 5 μs simulations revealed that the R102H mutation slightly increases the structural flexibility of loop L2 and the R102Hp mutation decreases protein stability. Community network analysis illustrates that the R102H and S83H mutations weaken the interdomain and strengthen the intradomain communications. Secondary structure contents in the S83H and S83Hp mutants are similar to those in the wild type, whereas the global structural stabilities and salt-bridge probabilities decrease. This study highlights the conformational dynamics effects of the R102H and S83H mutations on the local structural flexibility and global stability of NCS-1, whereas protonated histidine decreases the stability of NCS-1. Thus, histidines at positions 102 and 83 may not be compatible with the function of NCS-1 whether in the neutral or protonated state.
Collapse
Affiliation(s)
- Yehong Gong
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Yuzhen Zhu
- Shanghai Normal University Physical Education College, 100 Gui Lin Road, Shanghai, 200234, China
| | - Yu Zou
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Department of Human Genetics and Molecular Medicine, Sackler Inst. of Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| |
Collapse
|
41
|
Cui SY, Li SJ, Cui XY, Zhang XQ, Yu B, Huang YL, Cao Q, Xu YP, Yang G, Ding H, Song JZ, Ye H, Sheng ZF, Wang ZJ, Zhang YH. Ca(2+) in the dorsal raphe nucleus promotes wakefulness via endogenous sleep-wake regulating pathway in the rats. Mol Brain 2016; 9:71. [PMID: 27456222 PMCID: PMC4960696 DOI: 10.1186/s13041-016-0252-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023] Open
Abstract
Serotonergic neurons in the dorsal raphe nucleus (DRN) are involved in the control of sleep-wake states. Our previous studies have indicated that calcium (Ca(2+)) modulation in the DRN plays an important role in rapid-eye-movement sleep (REMS) and non-REMS (NREMS) regulation during pentobarbital hypnosis. The present study investigated the effects of Ca(2+) in the DRN on sleep-wake regulation and the related neuronal mechanism in freely moving rats. Our results showed that microinjection of CaCl2 (25 or 50 nmol) in the DRN promoted wakefulness and suppressed NREMS including slow wave sleep and REMS in freely moving rats. Application of CaCl2 (25 or 50 nmol) in the DRN significantly increased serotonin in the DRN and hypothalamus, and noradrenaline in the locus coeruleus and hypothalamus. Immunohistochemistry study indicated that application of CaCl2 (25 or 50 nmol) in the DRN significantly increased c-Fos expression ratio in wake-promoting neurons including serotonergic neurons in the DRN, noradrenergic neurons in the locus coeruleus, and orxinergic neurons in the perifornical nucleus, but decreased c-Fos expression ratio of GABAergic sleep-promoting neurons in the ventrolateral preoptic nucleus. These results suggest that Ca(2+) in the DRN exert arousal effects via up-regulating serotonergic functions in the endogenous sleep-wake regulating pathways.
Collapse
Affiliation(s)
- Su-Ying Cui
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Sheng-Jie Li
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Xiang-Yu Cui
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Xue-Qiong Zhang
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Bin Yu
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Yuan-Li Huang
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Qing Cao
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Ya-Ping Xu
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Guang Yang
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Hui Ding
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Jin-Zhi Song
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Hui Ye
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Zhao-Fu Sheng
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Zi-Jun Wang
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| | - Yong-He Zhang
- Department of pharmacology, Peking University, School of Basic Medical Science, 38 Xueyuan Road, Beijing, 100191 China
| |
Collapse
|
42
|
Todd PAC, McCue HV, Haynes LP, Barclay JW, Burgoyne RD. Interaction of ARF-1.1 and neuronal calcium sensor-1 in the control of the temperature-dependency of locomotion in Caenorhabditis elegans. Sci Rep 2016; 6:30023. [PMID: 27435667 PMCID: PMC4951722 DOI: 10.1038/srep30023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/27/2016] [Indexed: 12/15/2022] Open
Abstract
Neuronal calcium sensor-1 (NCS-1) mediates changes in cellular function by regulating various target proteins. Many potential targets have been identified but the physiological significance of only a few has been established. Upon temperature elevation, Caenorhabditis elegans exhibits reversible paralysis. In the absence of NCS-1, worms show delayed onset and a shorter duration of paralysis. This phenotype can be rescued by re-expression of ncs-1 in AIY neurons. Mutants with defects in four potential NCS-1 targets (arf-1.1, pifk-1, trp-1 and trp-2) showed qualitatively similar phenotypes to ncs-1 null worms, although the effect of pifk-1 mutation on time to paralysis was considerably delayed. Inhibition of pifk-1 also resulted in a locomotion phenotype. Analysis of double mutants showed no additive effects between mutations in ncs-1 and trp-1 or trp-2. In contrast, double mutants of arf-1.1 and ncs-1 had an intermediate phenotype, consistent with NCS-1 and ARF-1.1 acting in the same pathway. Over-expression of arf-1.1 in the AIY neurons was sufficient to rescue partially the phenotype of both the arf-1.1 and the ncs-1 null worms. These findings suggest that ARF-1.1 interacts with NCS-1 in AIY neurons and potentially pifk-1 in the Ca(2+) signaling pathway that leads to inhibited locomotion at an elevated temperature.
Collapse
Affiliation(s)
- Paul A. C. Todd
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| | - Hannah V. McCue
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| | - Lee P. Haynes
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| | - Jeff W. Barclay
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| | - Robert D. Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| |
Collapse
|
43
|
Zhu Y, Ma B, Nussinov R, Zhang Q. Temperature-Dependent Conformational Properties of Human Neuronal Calcium Sensor-1 Protein Revealed by All-Atom Simulations. J Phys Chem B 2016; 120:3551-9. [PMID: 27007011 PMCID: PMC6415918 DOI: 10.1021/acs.jpcb.5b12299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuronal calcium sensor-1 (NCS-1) protein has orthologues from Saccharomyces cerevisiae to human with highly conserved amino acid sequences. NCS-1 is an important factor controlling the animal's response to temperature change. This leads us to investigate the temperature effects on the conformational dynamics of human NCS-1 at 310 and 316 K by all-atom molecular dynamics (MD) simulations and dynamic community network analysis. Four independent 500 ns MD simulations show that secondary structure content at 316 K is similar to that at 310 K, whereas the global protein structure is expanded. Loop 3 (L3) adopts an extended state occuping the hydrophobic crevice, and the number of suboptimal communication paths between residue D176 and V190 is reduced at 316 K. The dynamic community network analysis suggests that the interdomain correlation is weakened, and the intradomain coupling is strengthened at 316 K. The elevated temperature reduces the number of the salt bridges, especially in C-domain. This study suggests that the elevated temperature affects the conformational dynamics of human NCS-1 protein. Comparison of the structural dynamics of R102Q mutant and Δ176-190 truncated NCS-1 suggests that the structural and dynamical response of NCS-1 protein to elevated temperature may be one of its intrinsic functional properties.
Collapse
Affiliation(s)
- Yuzhen Zhu
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Inst. of Molecular Medicine Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| |
Collapse
|
44
|
Naqvi MM, Heidarsson PO, Otazo MR, Mossa A, Kragelund BB, Cecconi C. Single-molecule folding mechanisms of the apo- and Mg(2+)-bound states of human neuronal calcium sensor-1. Biophys J 2016; 109:113-23. [PMID: 26153708 DOI: 10.1016/j.bpj.2015.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 04/08/2015] [Accepted: 05/20/2015] [Indexed: 01/21/2023] Open
Abstract
Neuronal calcium sensor-1 (NCS-1) is the primordial member of a family of proteins responsible primarily for sensing changes in neuronal Ca(2+) concentration. NCS-1 is a multispecific protein interacting with a number of binding partners in both calcium-dependent and independent manners, and acting in a variety of cellular processes in which it has been linked to a number of disorders such as schizophrenia and autism. Despite extensive studies on the Ca(2+)-activated state of NCS proteins, little is known about the conformational dynamics of the Mg(2+)-bound and apo states, both of which are populated, at least transiently, at resting Ca(2+) conditions. Here, we used optical tweezers to study the folding behavior of individual NCS-1 molecules in the presence of Mg(2+) and in the absence of divalent ions. Under tension, the Mg(2+)-bound state of NCS-1 unfolds and refolds in a three-state process by populating one intermediate state consisting of a folded C-domain and an unfolded N-domain. The interconversion at equilibrium between the different molecular states populated by NCS-1 was monitored in real time through constant-force measurements and the energy landscapes underlying the observed transitions were reconstructed through hidden Markov model analysis. Unlike what has been observed with the Ca(2+)-bound state, the presence of Mg(2+) allows both the N- and C-domain to fold through all-or-none transitions with similar refolding rates. In the absence of divalent ions, NCS-1 unfolds and refolds reversibly in a two-state reaction involving only the C-domain, whereas the N-domain has no detectable transitions. Overall, the results allowed us to trace the progression of NCS-1 folding along its energy landscapes and provided a solid platform for understanding the conformational dynamics of similar EF-hand proteins.
Collapse
Affiliation(s)
- Mohsin M Naqvi
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy; CNR Institute of Nanoscience S3, Modena, Italy
| | - Pétur O Heidarsson
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Mariela R Otazo
- CNR Institute of Nanoscience S3, Modena, Italy; Department of Physics, Center of Applied Technologies and Nuclear Development (CEADEN), Miramar, La Habana, Cuba
| | - Alessandro Mossa
- Department of Physics, University of Bari and INFN, Sezione di Bari, Bari, Italy
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark.
| | - Ciro Cecconi
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy; CNR Institute of Nanoscience S3, Modena, Italy.
| |
Collapse
|
45
|
Bidlingmaier S, Ha K, Lee NK, Su Y, Liu B. Proteome-wide Identification of Novel Ceramide-binding Proteins by Yeast Surface cDNA Display and Deep Sequencing. Mol Cell Proteomics 2016; 15:1232-45. [PMID: 26729710 DOI: 10.1074/mcp.m115.055954] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 11/06/2022] Open
Abstract
Although the bioactive sphingolipid ceramide is an important cell signaling molecule, relatively few direct ceramide-interacting proteins are known. We used an approach combining yeast surface cDNA display and deep sequencing technology to identify novel proteins binding directly to ceramide. We identified 234 candidate ceramide-binding protein fragments and validated binding for 20. Most (17) bound selectively to ceramide, although a few (3) bound to other lipids as well. Several novel ceramide-binding domains were discovered, including the EF-hand calcium-binding motif, the heat shock chaperonin-binding motif STI1, the SCP2 sterol-binding domain, and the tetratricopeptide repeat region motif. Interestingly, four of the verified ceramide-binding proteins (HPCA, HPCAL1, NCS1, and VSNL1) and an additional three candidate ceramide-binding proteins (NCALD, HPCAL4, and KCNIP3) belong to the neuronal calcium sensor family of EF hand-containing proteins. We used mutagenesis to map the ceramide-binding site in HPCA and to create a mutant HPCA that does not bind to ceramide. We demonstrated selective binding to ceramide by mammalian cell-produced wild type but not mutant HPCA. Intriguingly, we also identified a fragment from prostaglandin D2synthase that binds preferentially to ceramide 1-phosphate. The wide variety of proteins and domains capable of binding to ceramide suggests that many of the signaling functions of ceramide may be regulated by direct binding to these proteins. Based on the deep sequencing data, we estimate that our yeast surface cDNA display library covers ∼60% of the human proteome and our selection/deep sequencing protocol can identify target-interacting protein fragments that are present at extremely low frequency in the starting library. Thus, the yeast surface cDNA display/deep sequencing approach is a rapid, comprehensive, and flexible method for the analysis of protein-ligand interactions, particularly for the study of non-protein ligands.
Collapse
Affiliation(s)
- Scott Bidlingmaier
- From the Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110
| | - Kevin Ha
- From the Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110
| | - Nam-Kyung Lee
- From the Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110
| | - Yang Su
- From the Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110
| | - Bin Liu
- From the Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110
| |
Collapse
|
46
|
Lemire S, Jeromin A, Boisselier É. Membrane binding of Neuronal Calcium Sensor-1 (NCS1). Colloids Surf B Biointerfaces 2015; 139:138-47. [PMID: 26705828 DOI: 10.1016/j.colsurfb.2015.11.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/29/2015] [Accepted: 11/22/2015] [Indexed: 01/10/2023]
Abstract
Neuronal Calcium Sensor-1 (NCS1) belongs to the family of Neuronal Calcium Sensor (NCS) proteins. NCS1 is composed of four EF-hand motifs and an N-terminal myristoylation. However, the presence of a calcium-myristoyl switch in NCS1 and its role in the membrane binding are controversial. The model of Langmuir lipid monolayers is thus used to mimic the cell membrane in order to characterize the membrane interactions of NCS1. Two binding parameters are calculated from monolayer measurements: the maximum insertion pressure, up to which protein binding is energetically favorable, and the synergy, reporting attractive or repulsive interactions with the lipid monolayers. Binding membrane measurements performed in the presence of myristoylated NCS1 reveal better binding interactions for phospholipids composed of phosphoethanolamine polar head groups and unsaturated fatty acyl chains. In the absence of calcium, the membrane binding measurements are drastically modified and suggest that the protein is more strongly bound to the membrane. Indeed, the binding of calcium by three EF-hand motifs of NCS1 leads to a conformation change. NCS1 arrangement at the membrane could thus be reshuffled for better interactions with its substrates. The N-terminal peptide of NCS1 is composed of two amphiphilic helices involved in the membrane interactions of NCS1. Moreover, the presence of the myristoyl group has a weak influence on the membrane binding of NCS1 suggesting the absence of a calcium-myristoyl switch mechanism in this protein. The myristoylation could thus have a structural role required in the folding/unfolding of NCS1 which is essential to its multiple biological functions.
Collapse
Affiliation(s)
- Samuel Lemire
- CUO-Recherche, Hôpital du Saint-Sacrement, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada
| | | | - Élodie Boisselier
- CUO-Recherche, Hôpital du Saint-Sacrement, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
47
|
Kumar RP, Ranaghan MJ, Ganjei AY, Oprian DD. Crystal Structure of Recoverin with Calcium Ions Bound to Both Functional EF Hands. Biochemistry 2015; 54:7222-8. [PMID: 26584024 DOI: 10.1021/acs.biochem.5b01160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recoverin (Rv), a small Ca(2+)-binding protein that inhibits rhodopsin kinase (RK), has four EF hands, two of which are functional (EF2 and EF3). Activation requires Ca(2+) in both EF hands, but crystal structures have never been observed with Ca(2+) ions in both sites; all previous structures have Ca(2+) bound to only EF3. We suspected that this was due to an intermolecular crystal contact between T80 and a surface glutamate (E153) that precluded coordination of a Ca(2+) ion in EF2. We constructed the E153A mutant, determined its X-ray crystal structure to 1.2 Å resolution, and showed that two Ca(2+) ions are bound, one in EF3 and one in EF2. Additionally, several other residues are shown to adopt conformations in the 2Ca(2+) structure not seen previously and not seen in a second structure of the E153A mutant containing Na(+) instead of Ca(2+) in the EF2 site. The side-chain rearrangements in these residues form a 28 Å allosteric cascade along the surface of the protein connecting the Ca(2+)-binding site of EF2 with the active-site pocket responsible for binding RK.
Collapse
Affiliation(s)
- Ramasamy P Kumar
- Department of Biochemistry, Brandeis University , Waltham, Massachusetts 02454, United States
| | - Matthew J Ranaghan
- Department of Biochemistry, Brandeis University , Waltham, Massachusetts 02454, United States
| | - Allen Y Ganjei
- Department of Biochemistry, Brandeis University , Waltham, Massachusetts 02454, United States
| | - Daniel D Oprian
- Department of Biochemistry, Brandeis University , Waltham, Massachusetts 02454, United States
| |
Collapse
|
48
|
Viviano J, Krishnan A, Wu H, Venkataraman V. Electrophoretic mobility shift in native gels indicates calcium-dependent structural changes of neuronal calcium sensor proteins. Anal Biochem 2015; 494:93-100. [PMID: 26617128 DOI: 10.1016/j.ab.2015.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/21/2015] [Accepted: 11/13/2015] [Indexed: 10/22/2022]
Abstract
In proteins of the neuronal calcium sensor (NCS) family, changes in structure as well as function are brought about by the binding of calcium. In this article, we demonstrate that these structural changes, solely due to calcium binding, can be assessed through electrophoresis in native gels. The results demonstrate that the NCS proteins undergo ligand-dependent conformational changes that are detectable in native gels as a gradual decrease in mobility with increasing calcium but not other tested divalent cations such as magnesium, strontium, and barium. Surprisingly, such a gradual change over the entire tested range is exhibited only by the NCS proteins but not by other tested calcium-binding proteins such as calmodulin and S100B, indicating that the change in mobility may be linked to a unique NCS family feature--the calcium-myristoyl switch. Even within the NCS family, the changes in mobility are characteristic of the protein, indicating that the technique is sensitive to the individual features of the protein. Thus, electrophoretic mobility on native gels provides a simple and elegant method to investigate calcium (small ligand)-induced structural changes at least in the superfamily of NCS proteins.
Collapse
Affiliation(s)
- Jeffrey Viviano
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Anuradha Krishnan
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Hao Wu
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Venkat Venkataraman
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA; School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA.
| |
Collapse
|
49
|
Zhu Y, Yang S, Qi R, Zou Y, Ma B, Nussinov R, Zhang Q. Effects of the C-Terminal Tail on the Conformational Dynamics of Human Neuronal Calcium Sensor-1 Protein. J Phys Chem B 2015; 119:14236-44. [PMID: 26447771 PMCID: PMC6456052 DOI: 10.1021/acs.jpcb.5b07962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neuronal calcium sensor-1 (NCS-1) protein has been implicated in multiple neuronal functions by binding partners mostly through a largely exposed hydrophobic crevice (HC). In the absence of a ligand, the C-terminal tail (loop L3, residues D176 to V190) binds directly to the HC pocket as a ligand mimetic, occupying the HC and regulating its conformational stability. A recent experimental study reported that L3 deletion resulted in global structure destabilization. However, the influence of C-terminal tail on the conformations of NCS-1 protein is unclear at the atomic level. In this study, we investigated the structural properties and the conformational dynamics of wild type NCS-1 and L3 truncation variant by extensive all-atom molecular dynamics (MD) simulations. Our cumulative 2 μs MD simulations demonstrated that L3 deletion increased the structural flexibility of the C-domain and the distant N-domain. The community network analysis illustrated that C-terminal tail truncation weakened the interdomain correlation. Moreover, our data showed that the variant significantly disrupted the salt bridges network and expanded simultaneously the global structure and HC. These conformational changes caused by C-terminal tail truncation may affect the regulation of target interactions. Our study provides atomic details of the conformational dynamics effects of the C-terminal tail on human wild type NCS-1.
Collapse
Affiliation(s)
- Yuzhen Zhu
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai 200438, China
| | - Shuang Yang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai 200438, China
| | - Ruxi Qi
- Department of Physics, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Yu Zou
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai 200438, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Inst. of Molecular Medicine Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai 200438, China
| |
Collapse
|
50
|
Zernii EY, Grigoriev II, Nazipova AA, Scholten A, Kolpakova TV, Zinchenko DV, Kazakov AS, Senin II, Permyakov SE, Dell'Orco D, Philippov PP, Koch KW. Regulatory function of the C-terminal segment of guanylate cyclase-activating protein 2. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1325-37. [PMID: 26001899 DOI: 10.1016/j.bbapap.2015.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/22/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022]
Abstract
Neuronal responses to Ca2+-signals are provided by EF-hand-type neuronal Ca2+-sensor (NCS) proteins, which have similar core domains containing Ca2+-binding and target-recognizing sites. NCS proteins vary in functional specificity, probably depending on the structure and conformation of their non-conserved C-terminal segments. Here, we investigated the role of the C-terminal segment in guanylate cyclase activating protein-2, GCAP2, an NCS protein controlling the Ca2+-dependent regulation of photoreceptor guanylate cyclases. We obtained two chimeric proteins by exchanging C-terminal segments between GCAP2 and its photoreceptor homolog recoverin, a Ca2+-sensor controlling rhodopsin kinase (RK) activity. The exchange affected neither the structural integrity of GCAP2 and recoverin nor the Ca2+-sensitivity of GCAP2. Intrinsic fluorescence, circular dichroism, biochemical studies and hydrophobic dye probing revealed Ca2+-dependent conformational transition of the C-terminal segment of GCAP2 occurring in the molecular environment of both proteins. In Ca2+-GCAP2, the C-terminal segment was constrained and its replacement provided the protein with approximately two-fold inhibitory activity towards RK, suggesting that the segment contributes to specific target recognition by interfering with RK-binding. Upon Ca2+-release, it became less constrained and more available for phosphorylation by cyclic nucleotide-dependent protein kinase. The transition from the Ca2+-bound to the apo-state exposed hydrophobic sites in GCAP2, and was associated with its activating function without affecting its dimerization. The released C-terminal segment participated further in photoreceptor membrane binding making it sensitive to phosphorylation. Thus, the C-terminal segment in GCAP2 confers target selectivity, facilitates membrane binding and provides sensitivity of the membrane localization of the protein to phosphorylation by signaling kinases.
Collapse
Affiliation(s)
- Evgeni Yu Zernii
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Ilya I Grigoriev
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Aliya A Nazipova
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, 142290 Russia
| | - Alexander Scholten
- Department of Neurosciences, Biochemistry Group, University of Oldenburg, Oldenburg, 26111 Germany
| | - Tatiana V Kolpakova
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Dmitry V Zinchenko
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow region, 142290 Russia
| | - Alexey S Kazakov
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, 142290 Russia
| | - Ivan I Senin
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Sergei E Permyakov
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, 142290 Russia
| | - Daniele Dell'Orco
- Department of Life Sciences and Reproduction, Section of Biological Chemistry and Center for BioMedical Computing, University of Verona, Verona, 37134 Italy
| | - Pavel P Philippov
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Karl-W Koch
- Department of Neurosciences, Biochemistry Group, University of Oldenburg, Oldenburg, 26111 Germany.
| |
Collapse
|