1
|
Yuan X, Li W, Yan Q, Ou Y, Long Q, Zhang P. Biomarkers of mature neuronal differentiation and related diseases. Future Sci OA 2024; 10:2410146. [PMID: 39429212 PMCID: PMC11497955 DOI: 10.1080/20565623.2024.2410146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
The nervous system regulates perception, cognition and behavioral responses by serving as the body's primary communication system for receiving, regulating and transmitting information. Neurons are the fundamental structures and units of the nervous system. Their differentiation and maturation processes rely on the expression of specific biomarkers. Neuron-specific intracellular markers can be used to determine the degree of neuronal maturation. Neuronal cytoskeletal proteins dictate the shape and structure of neurons, while synaptic plasticity and signaling processes are intricately associated with neuronal synaptic markers. Furthermore, abnormal expression levels of biomarkers can serve as diagnostic indicators for nervous system diseases. This article reviews the markers of mature neuronal differentiation and their relationship with nervous system diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Wen Li
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Ya Ou
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Qingxi Long
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital Affiliated to North China University of Science & Technology, Tangshan, Hebei Province, 063000, China
- Hebei Provincial Key Laboratory of Neurobiological Function, Department of Neurology, Tangshan, Hebei Province, 063000, China
| |
Collapse
|
2
|
Ge Y, Zhang T. SNAP25 as a prognostic marker in transcriptome analysis of meningioma. Lab Med 2024:lmae085. [PMID: 39514545 DOI: 10.1093/labmed/lmae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Meningiomas are the most common intracranial tumors and their diagnosis relies mostly on neuroimaging and histology. However, the histology grades cannot predict the outcome exactly and some meningiomas tend to recur after resection of even benign tumors. Therefore, it is necessary to explore prognostic and diagnostic molecular targets. METHODS Differential expression analysis between meningiomas and meninges was performed based on the merged data of GSE43290 and GSE84263. Next, we performed gene set enrichment analysis (GSEA), immune cell infiltration analysis, protein-protein interaction analysis, and survival analysis using public data. The expression level of Synaptosome-associated-protein-25kDa (SNAP25) was verified by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) and Western blotting in meningioma tissues. RESULTS There were 263 upregulated and 592 downregulated genes identified in meningiomas by differential expression analysis. GSEA results revealed that meningiomas were negatively related to the pathway of soluble N-ethylmaleimide sensitive factor attachment protein receptor interactions in vascular transport and chemokine signaling. SNAP25 was characterized as a hub gene and downregulated in meningiomas. The Kaplan-Meier plot indicated that high expression of SNAP25 is a favorable factor. CONCLUSION SNAP25 was downregulated and identified as a potential prognostic marker in meningioma.
Collapse
Affiliation(s)
- Yu Ge
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Tao Zhang
- Department of Laboratory Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200011, China
| |
Collapse
|
3
|
Huang Z, Yao W, He W, Pan J, Chai W, Wang B, Jia Z, Fan X, Wang W, Zhang W. Moniezia benedeni drives the SNAP-25 expression of the enteric nerves in sheep's small intestine. BMC Vet Res 2024; 20:283. [PMID: 38956647 PMCID: PMC11218246 DOI: 10.1186/s12917-024-04140-6] [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: 11/28/2023] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The neuroimmune network plays a crucial role in regulating mucosal immune homeostasis within the digestive tract. Synaptosome-associated protein 25 (SNAP-25) is a presynaptic membrane-binding protein that activates ILC2s, initiating the host's anti-parasitic immune response. METHODS To investigate the effect of Moniezia benedeni (M. benedeni) infection on the distribution of SNAP-25 in the sheep's small intestine, the recombinant plasmid pET-28a-SNAP-25 was constructed and expressed in BL21, yielding the recombinant protein. Then, the rabbit anti-sheep SNAP-25 polyclonal antibody was prepared and immunofluorescence staining was performed with it. The expression levels of SNAP-25 in the intestines of normal and M. benedeni-infected sheep were detected by ELISA. RESULTS The results showed that the SNAP-25 recombinant protein was 29.3 KDa, the titer of the prepared immune serum reached 1:128,000. It was demonstrated that the rabbit anti-sheep SNAP-25 polyclonal antibody could bind to the natural protein of sheep SNAP-25 specifically. The expression levels of SNAP-25 in the sheep's small intestine revealed its primary presence in the muscular layer and lamina propria, particularly around nerve fibers surrounding the intestinal glands. Average expression levels in the duodenum, jejunum, and ileum were 130.32 pg/mg, 185.71 pg/mg, and 172.68 pg/mg, respectively. Under conditions of M. benedeni infection, the spatial distribution of SNAP-25-expressing nerve fibers remained consistent, but its expression level in each intestine segment was increased significantly (P < 0.05), up to 262.02 pg/mg, 276.84 pg/mg, and 326.65 pg/mg in the duodenum, jejunum, and ileum, and it was increased by 101.06%, 49.07%, and 89.16% respectively. CONCLUSIONS These findings suggest that M. benedeni could induce the SNAP-25 expression levels in sheep's intestinal nerves significantly. The results lay a foundation for further exploration of the molecular mechanism by which the gastrointestinal nerve-mucosal immune network perceives parasites in sheep.
Collapse
Affiliation(s)
- Zhen Huang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wanling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wanhong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jing Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wenzhu Chai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Baoshan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhitao Jia
- People's Government of Heisongyi Township, Wuwei, 733000, China
| | - Xiping Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wenhui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wangdong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
4
|
Cordano C, Werneburg S, Abdelhak A, Bennett DJ, Beaudry-Richard A, Duncan GJ, Oertel FC, Boscardin WJ, Yiu HH, Jabassini N, Merritt L, Nocera S, Sin JH, Samana IP, Condor Montes SY, Ananth K, Bischof A, Nourbakhsh B, Hauser SL, Cree BAC, Emery B, Schafer DP, Chan JR, Green AJ. Synaptic injury in the inner plexiform layer of the retina is associated with progression in multiple sclerosis. Cell Rep Med 2024; 5:101490. [PMID: 38574736 PMCID: PMC11031420 DOI: 10.1016/j.xcrm.2024.101490] [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: 07/24/2023] [Revised: 02/01/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
While neurodegeneration underlies the pathological basis for permanent disability in multiple sclerosis (MS), predictive biomarkers for progression are lacking. Using an animal model of chronic MS, we find that synaptic injury precedes neuronal loss and identify thinning of the inner plexiform layer (IPL) as an early feature of inflammatory demyelination-prior to symptom onset. As neuronal domains are anatomically segregated in the retina and can be monitored longitudinally, we hypothesize that thinning of the IPL could represent a biomarker for progression in MS. Leveraging our dataset with over 800 participants enrolled for more than 12 years, we find that IPL atrophy directly precedes progression and propose that synaptic loss is predictive of functional decline. Using a blood proteome-wide analysis, we demonstrate a strong correlation between demyelination, glial activation, and synapse loss independent of neuroaxonal injury. In summary, monitoring synaptic injury is a biologically relevant approach that reflects a potential driver of progression.
Collapse
Affiliation(s)
- Christian Cordano
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sebastian Werneburg
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Ophthalmology & Visual Sciences, Michigan Neuroscience Institute, University of Michigan - Michigan Medicine, Ann Arbor, MI, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel J Bennett
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Beaudry-Richard
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Greg J Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Frederike C Oertel
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - W John Boscardin
- Department of Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Hao H Yiu
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Merritt
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sonia Nocera
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jung H Sin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Isaac P Samana
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shivany Y Condor Montes
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Antje Bischof
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen L Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonah R Chan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ari J Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Ceddia RP, Zurawski Z, Thompson Gray A, Adegboye F, McDonald-Boyer A, Shi F, Liu D, Maldonado J, Feng J, Li Y, Alford S, Ayala JE, McGuinness OP, Collins S, Hamm HE. Gβγ-SNAP25 exocytotic brake removal enhances insulin action, promotes adipocyte browning, and protects against diet-induced obesity. J Clin Invest 2023; 133:e160617. [PMID: 37561580 PMCID: PMC10541194 DOI: 10.1172/jci160617] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Negative regulation of exocytosis from secretory cells is accomplished through inhibitory signals from Gi/o GPCRs by Gβγ subunit inhibition of 2 mechanisms: decreased calcium entry and direct interaction of Gβγ with soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) plasma membrane fusion machinery. Previously, we disabled the second mechanism with a SNAP25 truncation (SNAP25Δ3) that decreased Gβγ affinity for the SNARE complex, leaving exocytotic fusion and modulation of calcium entry intact and removing GPCR-Gβγ inhibition of SNARE-mediated exocytosis. Here, we report substantial metabolic benefit in mice carrying this mutation. Snap25Δ3/Δ3 mice exhibited enhanced insulin sensitivity and beiging of white fat. Metabolic protection was amplified in Snap25Δ3/Δ3 mice challenged with a high-fat diet. Glucose homeostasis, whole-body insulin action, and insulin-mediated glucose uptake into white adipose tissue were improved along with resistance to diet-induced obesity. Metabolic protection in Snap25Δ3/Δ3 mice occurred without compromising the physiological response to fasting or cold. All metabolic phenotypes were reversed at thermoneutrality, suggesting that basal autonomic activity was required. Direct electrode stimulation of sympathetic neuron exocytosis from Snap25Δ3/Δ3 inguinal adipose depots resulted in enhanced and prolonged norepinephrine release. Thus, the Gβγ-SNARE interaction represents a cellular mechanism that deserves further exploration as an additional avenue for combating metabolic disease.
Collapse
Affiliation(s)
- Ryan P. Ceddia
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zack Zurawski
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Feyisayo Adegboye
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Fubiao Shi
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dianxin Liu
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jose Maldonado
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Julio E. Ayala
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Sheila Collins
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Heidi E. Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
6
|
Stojanovic M, Schindler SE, Morris JC, Head D. Effect of exercise engagement and cardiovascular risk on neuronal injury. Alzheimers Dement 2023; 19:4454-4462. [PMID: 37534906 PMCID: PMC10592382 DOI: 10.1002/alz.13400] [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: 03/28/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Neuronal health as a potential underlying mechanism of the beneficial effects of exercise has been understudied in humans. Furthermore, there has been limited consideration of potential moderators (e.g., cardiovascular health) on the effects of exercise. METHODS Clinically normal middle-aged and older adults completed a validated questionnaire about exercise engagement over a 10-year period (n = 75; age 63 ± 8 years). A composite estimate of neuronal injury was formulated that included cerebrospinal fluid-based measures of visinin-like protein-1, neurogranin, synaptosomal-associated protein 25, and neurofilament light chain. Cardiovascular risk was estimated using the Framingham Risk Score. RESULTS Cross-sectional analyses showed that greater exercise engagement was associated with less neuronal injury in the group with lower cardiovascular risk (p = 0.008), but not the group with higher cardiovascular risk (p = 0.209). DISCUSSION Cardiovascular risk is an important moderator to consider when examining the effects of exercise on cognitive and neural health, and may be relevant to personalized exercise recommendations. HIGHLIGHTS We examined the association between exercise engagement and neuronal injury. Vascular risk moderated the association between exercise and neuronal injury. Cardiovascular risk may be relevant to personalized exercise recommendations.
Collapse
Affiliation(s)
- Marta Stojanovic
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63105
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, 63110
| | - Suzanne E. Schindler
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, 63110
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, 63110
| | - John C. Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, 63110
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, 63110
| | - Denise Head
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, 63105
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, 63110
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, 63110
| |
Collapse
|
7
|
Chen S, Li F, Liu G, Li Y, Li Z, Liu Y, Nakanishi H. Construction of a Yeast Cell-Based Assay System to Analyze SNAP25-Targeting Botulinum Neurotoxins. Microorganisms 2023; 11:1125. [PMID: 37317099 DOI: 10.3390/microorganisms11051125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 06/16/2023] Open
Abstract
Herein, we describe a yeast cell-based assay system to analyze SNAP25-targeting botulinum neurotoxins (BoNTs). BoNTs are protein toxins, and, upon incorporation into neuronal cells, their light chains (BoNT-LCs) target specific synaptosomal N-ethylmaleimide-sensitive attachment protein receptor (SNARE) proteins, including synaptosomal-associated protein 25 (SNAP25). BoNT-LCs are metalloproteases, and each BoNT-LC recognizes and cleaves conserved domains in SNAREs termed the SNARE domain. In the budding yeast Saccharomyces cerevisiae, the SNAP25 ortholog Spo20 is required for production of the spore plasma membrane; thus, defects in Spo20 cause sporulation deficiencies. We found that chimeric SNAREs in which SNARE domains in Spo20 are replaced with those of SNAP25 are functional in yeast cells. The Spo20/SNAP25 chimeras, but not Spo20, are sensitive to digestion by BoNT-LCs. We demonstrate that spo20∆ yeasts harboring the chimeras exhibit sporulation defects when various SNAP25-targeting BoNT-LCs are expressed. Thus, the activities of BoNT-LCs can be assessed by colorimetric measurement of sporulation efficiencies. Although BoNTs are notorious toxins, they are also used as therapeutic and cosmetic agents. Our assay system will be useful for analyzing novel BoNTs and BoNT-like genes, as well as their manipulation.
Collapse
Affiliation(s)
- Shilin Chen
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Feng Li
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Guoyu Liu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Yuqing Li
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Zijie Li
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Yishi Liu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Hideki Nakanishi
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| |
Collapse
|
8
|
Wang B, Xu Y, Xu S, Wu H, Qu P, Tong Z, Lü P, Cheng C. Characterization of Banana SNARE Genes and Their Expression Analysis under Temperature Stress and Mutualistic and Pathogenic Fungal Colonization. PLANTS (BASEL, SWITZERLAND) 2023; 12:1599. [PMID: 37111823 PMCID: PMC10142651 DOI: 10.3390/plants12081599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 06/19/2023]
Abstract
SNAREs (soluble N-ethylmaleimide-sensitive-factor attachment protein receptors) are engines for almost all of the membrane fusion and exocytosis events in organism cells. In this study, we identified 84 SNARE genes from banana (Musa acuminata). Gene expression analysis revealed that the expression of MaSNAREs varied a lot in different banana organs. By analyzing their expression patterns under low temperature (4 °C), high temperature (45 °C), mutualistic fungus (Serendipita indica, Si) and fungal pathogen (Fusarium oxysporum f. sp. Cubense Tropical Race 4, FocTR4) treatments, many MaSNAREs were found to be stress responsive. For example, MaBET1d was up-regulate by both low and high temperature stresses; MaNPSN11a was up-regulated by low temperature but down-regulated by high temperature; and FocTR4 treatment up-regulated the expression of MaSYP121 but down-regulated MaVAMP72a and MaSNAP33a. Notably, the upregulation or downregulation effects of FocTR4 on the expression of some MaSNAREs could be alleviated by priorly colonized Si, suggesting that they play roles in the Si-enhanced banana wilt resistance. Foc resistance assays were performed in tobacco leaves transiently overexpressing MaSYP121, MaVAMP72a and MaSNAP33a. Results showed that transient overexpression of MaSYP121 and MaSNPA33a suppressed the penetration and spread of both Foc1 (Foc Race 1) and FocTR4 in tobacco leaves, suggesting that they play positive roles in resisting Foc infection. However, the transient overexpression of MaVAMP72a facilitated Foc infection. Our study can provide a basis for understanding the roles of MaSNAREs in the banana responses to temperature stress and mutualistic and pathogenic fungal colonization.
Collapse
Affiliation(s)
- Bin Wang
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yanbing Xu
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shiyao Xu
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Huan Wu
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Pengyan Qu
- College of Horticulture, Shanxi Agricultural University, Jinzhong 030801, China
| | - Zheng Tong
- Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Peitao Lü
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chunzhen Cheng
- College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- College of Horticulture, Shanxi Agricultural University, Jinzhong 030801, China
| |
Collapse
|
9
|
Smirnova EV, Rakitina TV, Ziganshin RH, Saratov GA, Arapidi GP, Belogurov AA, Kudriaeva AA. Identification of Myelin Basic Protein Proximity Interactome Using TurboID Labeling Proteomics. Cells 2023; 12:cells12060944. [PMID: 36980286 PMCID: PMC10047773 DOI: 10.3390/cells12060944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Myelin basic protein (MBP) is one of the key structural elements of the myelin sheath and has autoantigenic properties in multiple sclerosis (MS). Its intracellular interaction network is still partially deconvoluted due to the unfolded structure, abnormally basic charge, and specific cellular localization. Here we used the fusion protein of MBP with TurboID, an engineered biotin ligase that uses ATP to convert biotin to reactive biotin-AMP that covalently attaches to nearby proteins, to determine MBP interactome. Despite evident benefits, the proximity labeling proteomics technique generates high background noise, especially in the case of proteins tending to semi-specific interactions. In order to recognize unique MBP partners, we additionally mapped protein interaction networks for deaminated MBP variant and cyclin-dependent kinase inhibitor 1 (p21), mimicking MBP in terms of natively unfolded state, size and basic amino acid clusters. We found that in the plasma membrane region, MBP is colocalized with adhesion proteins occludin and myelin protein zero-like protein 1, solute carrier family transporters ZIP6 and SNAT1, Eph receptors ligand Ephrin-B1, and structural components of the vesicle transport machinery-synaptosomal-associated protein 23 (SNAP23), vesicle-associated membrane protein 3 (VAMP3), protein transport protein hSec23B and cytoplasmic dynein 1 heavy chain 1. We also detected that MBP potentially interacts with proteins involved in Fe2+ and lipid metabolism, namely, ganglioside GM2 activator protein, long-chain-fatty-acid-CoA ligase 4 (ACSL4), NADH-cytochrome b5 reductase 1 (CYB5R1) and metalloreductase STEAP3. Assuming the emerging role of ferroptosis and vesicle cargo docking in the development of autoimmune neurodegeneration, MBP may recruit and regulate the activity of these processes, thus, having a more inclusive role in the integrity of the myelin sheath.
Collapse
Affiliation(s)
- Evgeniya V Smirnova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Tatiana V Rakitina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Rustam H Ziganshin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - George A Saratov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), 141701 Dolgoprudny, Russia
| | - Georgij P Arapidi
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), 141701 Dolgoprudny, Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Alexey A Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russian Federation, 127473 Moscow, Russia
| | - Anna A Kudriaeva
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
10
|
Palfreyman MT, West SE, Jorgensen EM. SNARE Proteins in Synaptic Vesicle Fusion. ADVANCES IN NEUROBIOLOGY 2023; 33:63-118. [PMID: 37615864 DOI: 10.1007/978-3-031-34229-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Neurotransmitters are stored in small membrane-bound vesicles at synapses; a subset of synaptic vesicles is docked at release sites. Fusion of docked vesicles with the plasma membrane releases neurotransmitters. Membrane fusion at synapses, as well as all trafficking steps of the secretory pathway, is mediated by SNARE proteins. The SNAREs are the minimal fusion machinery. They zipper from N-termini to membrane-anchored C-termini to form a 4-helix bundle that forces the apposed membranes to fuse. At synapses, the SNAREs comprise a single helix from syntaxin and synaptobrevin; SNAP-25 contributes the other two helices to complete the bundle. Unc13 mediates synaptic vesicle docking and converts syntaxin into the permissive "open" configuration. The SM protein, Unc18, is required to initiate and proofread SNARE assembly. The SNAREs are then held in a half-zippered state by synaptotagmin and complexin. Calcium removes the synaptotagmin and complexin block, and the SNAREs drive vesicle fusion. After fusion, NSF and alpha-SNAP unwind the SNAREs and thereby recharge the system for further rounds of fusion. In this chapter, we will describe the discovery of the SNAREs, their relevant structural features, models for their function, and the central role of Unc18. In addition, we will touch upon the regulation of SNARE complex formation by Unc13, complexin, and synaptotagmin.
Collapse
Affiliation(s)
- Mark T Palfreyman
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - Sam E West
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - Erik M Jorgensen
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
11
|
Reynolds HM, Wen T, Farrell A, Mao R, Moore B, Boyden SE, Bayrak-Toydemir P, Nicholas TJ, Rynearson S, Holt C, Miller C, Noble K, Bentley D, Palmquist R, Ostrander B, Manberg S, Bonkowsky JL, Shayota BJ, Jenkins SM. Rapid genome sequencing identifies a novel de novo SNAP25 variant for neonatal congenital myasthenic syndrome. Cold Spring Harb Mol Case Stud 2022; 8:a006242. [PMID: 36379720 PMCID: PMC9808558 DOI: 10.1101/mcs.a006242] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital myasthenic syndrome (CMS) is a group of 32 disorders involving genetic dysfunction at the neuromuscular junction resulting in skeletal muscle weakness that worsens with physical activity. Precise diagnosis and molecular subtype identification are critical for treatment as medication for one subtype may exacerbate disease in another (Engel et al., Lancet Neurol 14: 420 [2015]; Finsterer, Orphanet J Rare Dis 14: 57 [2019]; Prior and Ghosh, J Child Neurol 36: 610 [2021]). The SNAP25-related CMS subtype (congenital myasthenic syndrome 18, CMS18; MIM #616330) is a rare disorder characterized by muscle fatigability, delayed psychomotor development, and ataxia. Herein, we performed rapid whole-genome sequencing (rWGS) on a critically ill newborn leading to the discovery of an unreported pathogenic de novo SNAP25 c.529C > T; p.Gln177Ter variant. In this report, we present a novel case of CMS18 with complex neonatal consequence. This discovery offers unique insight into the extent of phenotypic severity in CMS18, expands the reported SNAP25 variant phenotype, and paves a foundation for personalized management for CMS18.
Collapse
Affiliation(s)
- Hayley M Reynolds
- University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | - Ting Wen
- University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
- ARUP Laboratories, Salt Lake City, Utah 84108, USA
| | - Andrew Farrell
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | - Rong Mao
- University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
- ARUP Laboratories, Salt Lake City, Utah 84108, USA
| | - Barry Moore
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | - Steven E Boyden
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | - Pinar Bayrak-Toydemir
- University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
- ARUP Laboratories, Salt Lake City, Utah 84108, USA
| | - Thomas J Nicholas
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | - Shawn Rynearson
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | - Carson Holt
- Department of Human Genetics, Utah Center for Genetic Discovery, Salt Lake City, Utah 84112, USA
| | | | | | - Dawn Bentley
- Division of Neonatology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | - Rachel Palmquist
- Division of Pediatric Neurology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84113, USA
| | - Betsy Ostrander
- Division of Pediatric Neurology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84113, USA
| | - Stephanie Manberg
- Division of Pediatric Neurology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84113, USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84113, USA
- Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, Utah 84108, USA
| | - Brian J Shayota
- Division of Medical Genetics, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | - Sabrina Malone Jenkins
- Division of Neonatology, Department of Pediatrics University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| |
Collapse
|
12
|
Fang D, Yang B, Wang P, Mo T, Gan Y, Liang G, Huang R, Zeng H. Role of SNAP-25 MnlI variant in impaired working memory and brain functions in attention deficit/hyperactivity disorder. Brain Behav 2022; 12:e2758. [PMID: 36068994 PMCID: PMC9575616 DOI: 10.1002/brb3.2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Attention deficit/hyperactivity disorder (ADHD) is a hereditary neurodevelopmental disorder characterized by working memory (WM) deficits. The MnlI variant (rs3746544) of the synaptosomal-associated protein 25 (SNAP-25) gene is associated with ADHD. In this study, we investigated the role and underlying mechanism of SNAP-25 MnlI variant in cognitive impairment and brain functions in boys with ADHD. METHOD We performed WM capacity tests using the fourth version of the Wechsler Intelligence Scale for Children (WISC-IV) and regional homogeneity (ReHo) analysis for the resting-state functional magnetic resonance imaging data of 56 boys with ADHD divided into two genotypic groups (TT homozygotes and G-allele carriers). Next, Spearman's rank correlation analysis between the obtained ReHo values and the WM index (WMI) calculated for each participant. RESULTS Compared with G-allele carrier group, there were higher ReHo values for the left medial prefrontal cortex (mPFC) and higher WM capacity in TT homozygote group. Contrary to TT homozygote group, the WM capacity was negatively correlated with the peak ReHo value for the left mPFC in G-allele carrier group. CONCLUSION These findings suggest that SNAP-25 MnlI variant may underlie cognitive and brain function impairments in boys with ADHD, thus suggesting its potential as a new target for ADHD treatment.
Collapse
Affiliation(s)
- Diangang Fang
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Binrang Yang
- Development and Behavior Specialty, Shenzhen Children's Hospital, Shenzhen, China
| | - Peng Wang
- Cardiac Rehabilitation Center, Fuwai Hospital CAMS&PUMC, Beijing, China
| | - Tong Mo
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Yungen Gan
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Guohua Liang
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Rong Huang
- Department of Radiology, Peking University Shenzhen hospital, Shenzhen, China
| | - Hongwu Zeng
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
13
|
Kha QH, Ho QT, Le NQK. Identifying SNARE Proteins Using an Alignment-Free Method Based on Multiscan Convolutional Neural Network and PSSM Profiles. J Chem Inf Model 2022; 62:4820-4826. [PMID: 36166351 PMCID: PMC9554904 DOI: 10.1021/acs.jcim.2c01034] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
![]()
Background: SNARE proteins play a vital
role in
membrane fusion and cellular physiology and pathological processes.
Many potential therapeutics for mental diseases or even cancer based
on SNAREs are also developed. Therefore, there is a dire need to predict
the SNAREs for further manipulation of these essential proteins, which
demands new and efficient approaches. Methods: Some
computational frameworks were proposed to tackle the hurdles of biological
methods, which take plenty of time and budget to conduct the identification
of SNAREs. However, the performances of existing frameworks were insufficiently
satisfied, as they failed to retain the SNARE sequence order and capture
the mass hidden features from SNAREs. This paper proposed a novel
model constructed on the multiscan convolutional neural network (CNN)
and position-specific scoring matrix (PSSM) profiles to address these
limitations. We employed and trained our model on the benchmark dataset
with fivefold cross-validation and two different independent datasets. Results: Overall, the multiscan CNN was cross-validated
on the training set and excelled in the SNARE classification reaching
0.963 in AUC and 0.955 in AUPRC. On top of that, with the sensitivity,
specificity, accuracy, and MCC of 0.842, 0.968, 0.955, and 0.767,
respectively, our proposed framework outperformed previous models
in the SNARE recognition task. Conclusions: It is
truly believed that our model can contribute to the discrimination
of SNARE proteins and general proteins.
Collapse
Affiliation(s)
- Quang-Hien Kha
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Quang-Thai Ho
- College of Information & Communication Technology, Can Tho University, Can Tho 90000, Viet Nam.,Department of Computer Science and Engineering, Yuan Ze University, Chung-Li 32003, Taiwan
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan.,Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 106, Taiwan.,Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| |
Collapse
|
14
|
Cui L, Li H, Xi Y, Hu Q, Liu H, Fan J, Xiang Y, Zhang X, Shui W, Lai Y. Vesicle trafficking and vesicle fusion: mechanisms, biological functions, and their implications for potential disease therapy. MOLECULAR BIOMEDICINE 2022; 3:29. [PMID: 36129576 PMCID: PMC9492833 DOI: 10.1186/s43556-022-00090-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
Intracellular vesicle trafficking is the fundamental process to maintain the homeostasis of membrane-enclosed organelles in eukaryotic cells. These organelles transport cargo from the donor membrane to the target membrane through the cargo containing vesicles. Vesicle trafficking pathway includes vesicle formation from the donor membrane, vesicle transport, and vesicle fusion with the target membrane. Coat protein mediated vesicle formation is a delicate membrane budding process for cargo molecules selection and package into vesicle carriers. Vesicle transport is a dynamic and specific process for the cargo containing vesicles translocation from the donor membrane to the target membrane. This process requires a group of conserved proteins such as Rab GTPases, motor adaptors, and motor proteins to ensure vesicle transport along cytoskeletal track. Soluble N-ethyl-maleimide-sensitive factor (NSF) attachment protein receptors (SNARE)-mediated vesicle fusion is the final process for vesicle unloading the cargo molecules at the target membrane. To ensure vesicle fusion occurring at a defined position and time pattern in eukaryotic cell, multiple fusogenic proteins, such as synaptotagmin (Syt), complexin (Cpx), Munc13, Munc18 and other tethering factors, cooperate together to precisely regulate the process of vesicle fusion. Dysfunctions of the fusogenic proteins in SNARE-mediated vesicle fusion are closely related to many diseases. Recent studies have suggested that stimulated membrane fusion can be manipulated pharmacologically via disruption the interface between the SNARE complex and Ca2+ sensor protein. Here, we summarize recent insights into the molecular mechanisms of vesicle trafficking, and implications for the development of new therapeutics based on the manipulation of vesicle fusion.
Collapse
|
15
|
Natalwala A, Behbehani R, Yapom R, Kunath T. An Isogenic Collection of Pluripotent Stem Cell Lines With Elevated α-Synuclein Expression Validated for Neural Induction and Cortical Neuron Differentiation. Front Cell Dev Biol 2022; 10:898560. [PMID: 35712660 PMCID: PMC9196909 DOI: 10.3389/fcell.2022.898560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
α-Synuclein (αSyn) is a small, disordered protein that becomes aggregated in Lewy body diseases, such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Human induced pluripotent stem cells (hiPSCs) potentially provide a tractable disease model to monitor early molecular changes associated with PD/DLB. We and others have previously derived hiPSC lines from patients with duplication and triplication of the SNCA gene, encoding for αSyn. It is now recognised that to perform meaningful disease modelling with these hiPSC lines, it is critical to generate isogenic control cell lines that lack the disease causing mutations. In order to complement the existing and emerging hiPSC models for PD/DLB, we have generated an allelic series of αSyn over-expressing hESC lines on the same isogenic background. An unresolved question is whether pluripotent stem cell lines, with elevated levels of αSyn, can undergo efficient differentiation into dopaminergic and cortical neurons to model PD and DLB, respectively. We took advantage of our isogenic collection of hESC lines to determine if increased expression of αSyn affects neural induction and neuronal differentiation. Clonal hESC lines with significantly different levels of αSyn expression proliferated normally and maintained expression of pluripotent markers, such as OCT4. All cell lines efficiently produced PAX6+ neuroectoderm and there was no correlation between αSyn expression and neural induction efficiency. Finally, global transcriptomic analysis of cortical differentiation of hESC lines with low or high levels of αSyn expression demonstrated robust and similar induction of cortical neuronal expression profiles. Gene expression differences observed were unrelated to neural induction and neuronal differentiation. We conclude that elevated expression of αSyn in human pluripotent stem cells does not adversely affect their neuronal differentiation potential and that collections of isogenic cell lines with differing levels of αSyn expression are valid and suitable models to investigate synucleinopathies.
Collapse
Affiliation(s)
- Ammar Natalwala
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square House, London, United Kingdom,Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, United Kingdom,Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom,*Correspondence: Ammar Natalwala, ; Tilo Kunath,
| | - Ranya Behbehani
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ratsuda Yapom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom,*Correspondence: Ammar Natalwala, ; Tilo Kunath,
| |
Collapse
|
16
|
Lei F, Tian Y, Miao J, Pan L, Tong R, Zhou Y. Immunotoxicity pathway and mechanism of benzo[a]pyrene on hemocytes of Chlamys farreri in vitro. FISH & SHELLFISH IMMUNOLOGY 2022; 124:208-218. [PMID: 35413479 DOI: 10.1016/j.fsi.2022.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 06/14/2023]
Abstract
Benzo[a]pyrene (B[a]P), a typical PAHs widely existing in the marine environment, has been extensively studied for its immunotoxicity due to its persistence and high toxicity. Nevertheless, the immunotoxicity mechanism remain incompletely understood. In this study, isolated hemocytes of Chlamys farreri were exposed at three concentrations of B[a]P (5, 10 and 15 μg/mL), and the effects of B[a]P on detoxification metabolism, signal transduction, humoral immune factors, exocytosis and phagocytosis relevant proteins and immune function at 0, 6, 12, 24 h were studied. Results illustrated the AhR, ARNT and CYP1A1 were significantly induced by B[a]P at 12 h. Additionally, the content of B[a]P metabolite BPDE increased in a dose-dependent manner with pollutants. Under B[a]P stimulation, the expressions of PTK (Src, Fyn) and PLC-Ca2+-PKC pathway gene increased significantly, while the transcription level of AC-cAMP-PKA pathway gene decreased remarkably. Additionally, the expressions of nuclear transcription factors (CREB, NF-κB), complement system genes and C-type lectin genes up-regulated obviously. The gene expressions of phagocytosis and exocytosis related proteins were also notably affected. 5 μg/mL B[a]P could promote phagocytosis in a transitory time, but with the increase of exposure time and concentration of B[a]P, the phagocytosis, antibacterial and bacteriolytic activities gradually decreased. These results indicated that similar to vertebrates, BPDE, the metabolite of B[a]P, mediated downstream signal transduction via PTK in bivalves. The declined of the immune defense ability of hemocytes might be closely related to the inhibition of AC-cAMP-PKA pathway and the imbalance of intracellular Ca2+ pathway. In addition, the results manifested that complement and lectin systems play a significant role in regulating immune response. In this study, the direct relationship between detoxification metabolism and immune signal transduction in bivalves under B[a]P stress was demonstrated for the first time, which provided important information for the potential molecular mechanism of B[a]P-induced immune system disorder in bivalves.
Collapse
Affiliation(s)
- Fengjun Lei
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Yimeng Tian
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Jingjing Miao
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China.
| | - Ruixue Tong
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Yueyao Zhou
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| |
Collapse
|
17
|
Hao X, Zhu B, Yang P, Dong D, Sahbaie P, Oliver PL, Shen WJ, Azhar S, Kraemer FB. SNAP25 mutation disrupts metabolic homeostasis, steroid hormone production and central neurobehavior. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166304. [PMID: 34826585 PMCID: PMC8759409 DOI: 10.1016/j.bbadis.2021.166304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/29/2021] [Accepted: 11/11/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE SNAP-25 is one of the key proteins involved in formation of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes that are at the core of hormonal secretion and synaptic transmission. Altered expression or function of SNAP-25 can contribute to the development of neuropsychiatric and metabolic disease. A dominant negative (DN) I67T missense mutation in the b-isoform of SNAP-25 (DN-SNAP25mut) mice leads to abnormal interactions within the SNARE complex and impaired exocytotic vesicle recycling, yet the significance of this mutation to any association between the central nervous system and metabolic homeostasis is unknown. METHODS Here we explored aspects of metabolism, steroid hormone production and neurobehavior of DN-SNAP25mut mice. RESULTS DN-SNAP25mut mice displayed enhanced insulin function through increased Akt phosphorylation, alongside increased adrenal and gonadal hormone production. In addition, increased anxiety behavior and beigeing of white adipose tissue with increased energy expenditure were observed in mutants. CONCLUSIONS Our results show that SNAP25 plays an important role in bridging central neurological systems with peripheral metabolic homeostasis, and provide potential insights between metabolic disease and neuropsychiatric disorders in humans.
Collapse
Affiliation(s)
- Xiao Hao
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States; Department of Endocrinology, First Affiliated Hospital of the Medical College of Zhengzhou University, Zhengzhou, China
| | - Bing Zhu
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States; Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Pinglin Yang
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States; Department of Orthopedics, Second Affiliated Hospital of Xi'an, Jiaotong University, Xi'an, Shaanxi, China
| | - Dachuan Dong
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States
| | - Peyman Sahbaie
- Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States
| | - Peter L Oliver
- Medical Research Council Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| | - Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States.
| | - Salman Azhar
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, CA, United States; Geriatric Research, Education, and Clinical Center, Veterans Administration Palo Alto Health Care System, Palo Alto, CA, United States.
| |
Collapse
|
18
|
Regan SL, Williams MT, Vorhees CV. Review of rodent models of attention deficit hyperactivity disorder. Neurosci Biobehav Rev 2022; 132:621-637. [PMID: 34848247 PMCID: PMC8816876 DOI: 10.1016/j.neubiorev.2021.11.041] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a polygenic neurodevelopmental disorder that affects 8-12 % of children and >4 % of adults. Environmental factors are believed to interact with genetic predispositions to increase susceptibility to ADHD. No existing rodent model captures all aspects of ADHD, but several show promise. The main genetic models are the spontaneous hypertensive rat, dopamine transporter knock-out (KO) mice, dopamine receptor subtype KO mice, Snap-25 KO mice, guanylyl cyclase-c KO mice, and latrophilin-3 KO mice and rats. Environmental factors thought to contribute to ADHD include ethanol, nicotine, PCBs, lead (Pb), ionizing irradiation, 6-hydroxydopamine, neonatal hypoxia, some pesticides, and organic pollutants. Model validation criteria are outlined, and current genetic models evaluated against these criteria. Future research should explore induced multiple gene KOs given that ADHD is polygenic and epigenetic contributions. Furthermore, genetic models should be combined with environmental agents to test for interactions.
Collapse
Affiliation(s)
- Samantha L. Regan
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229
| | - Michael T. Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Charles V. Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229,Corresponding author: Charles V. Vorhees, Ph.D., Div. of Neurology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA:
| |
Collapse
|
19
|
Luo C, Shi Y, Xiang Y. SNAREs Regulate Vesicle Trafficking During Root Growth and Development. FRONTIERS IN PLANT SCIENCE 2022; 13:853251. [PMID: 35360325 PMCID: PMC8964185 DOI: 10.3389/fpls.2022.853251] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 01/27/2022] [Indexed: 05/13/2023]
Abstract
SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins assemble to drive the final membrane fusion step of membrane trafficking. Thus, SNAREs are essential for membrane fusion and vesicular trafficking, which are fundamental mechanisms for maintaining cellular homeostasis. In plants, SNAREs have been demonstrated to be located in different subcellular compartments and involved in a variety of fundamental processes, such as cytokinesis, cytoskeleton organization, symbiosis, and biotic and abiotic stress responses. In addition, SNAREs can also contribute to the normal growth and development of Arabidopsis. Here, we review recent progress in understanding the biological functions and signaling network of SNAREs in vesicle trafficking and the regulation of root growth and development in Arabidopsis.
Collapse
|
20
|
Huang H, Ouyang Q, Zhu M, Yu H, Mei K, Liu R. mTOR-mediated phosphorylation of VAMP8 and SCFD1 regulates autophagosome maturation. Nat Commun 2021; 12:6622. [PMID: 34785650 PMCID: PMC8595342 DOI: 10.1038/s41467-021-26824-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTORC1) has been shown to regulate autophagy at different steps. However, how mTORC1 regulates the N-ethylmaleimide-sensitive protein receptor (SNARE) complex remains elusive. Here we show that mTORC1 inhibits formation of the SNARE complex (STX17-SNAP29-VAMP8) by phosphorylating VAMP8, thereby blocking autophagosome-lysosome fusion. A VAMP8 phosphorylation mimic mutant is unable to promote autophagosome-lysosome fusion in vitro. Furthermore, we identify SCFD1, a Sec1/Munc18-like protein, that localizes to the autolysosome and is required for SNARE complex formation and autophagosome-lysosome fusion. VAMP8 promotes SCFD1 recruitment to autolysosomes when dephosphorylated. Consistently, phosphorylated VAMP8 or SCFD1 depletion inhibits autophagosome-lysosome fusion, and expression of phosphomimic VAMP8 leads to increased lipid droplet accumulation when expressed in mouse liver. Thus, our study supports that mTORC1-mediated phosphorylation of VAMP8 blocks SCFD1 recruitment, thereby inhibiting STX17-SNAP29-VAMP8 complex formation and autophagosome-lysosome fusion. Autophagy relies on coordinated fusion of organelle membranes, although the interplay between the regulatory machinery is not well studied. Here, the authors show that SNARE complex formation is inhibited by mTORC1 phosphorylation of VAMP8, which prevents autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Hong Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China.,School of Life Sciences, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China
| | - Qinqin Ouyang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China.,School of Life Sciences, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China
| | - Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjing, China
| | - Rong Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China. .,School of Life Sciences, Nanjing Agricultural University, Nanjing, China. .,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China. .,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
21
|
Sauvola CW, Littleton JT. SNARE Regulatory Proteins in Synaptic Vesicle Fusion and Recycling. Front Mol Neurosci 2021; 14:733138. [PMID: 34421538 PMCID: PMC8377282 DOI: 10.3389/fnmol.2021.733138] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Membrane fusion is a universal feature of eukaryotic protein trafficking and is mediated by the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) family. SNARE proteins embedded in opposing membranes spontaneously assemble to drive membrane fusion and cargo exchange in vitro. Evolution has generated a diverse complement of SNARE regulatory proteins (SRPs) that ensure membrane fusion occurs at the right time and place in vivo. While a core set of SNAREs and SRPs are common to all eukaryotic cells, a specialized set of SRPs within neurons confer additional regulation to synaptic vesicle (SV) fusion. Neuronal communication is characterized by precise spatial and temporal control of SNARE dynamics within presynaptic subdomains specialized for neurotransmitter release. Action potential-elicited Ca2+ influx at these release sites triggers zippering of SNAREs embedded in the SV and plasma membrane to drive bilayer fusion and release of neurotransmitters that activate downstream targets. Here we discuss current models for how SRPs regulate SNARE dynamics and presynaptic output, emphasizing invertebrate genetic findings that advanced our understanding of SRP regulation of SV cycling.
Collapse
Affiliation(s)
- Chad W Sauvola
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
22
|
Differential Expression Levels of Sox9 in Early Neocortical Radial Glial Cells Regulate the Decision between Stem Cell Maintenance and Differentiation. J Neurosci 2021; 41:6969-6986. [PMID: 34266896 PMCID: PMC8372026 DOI: 10.1523/jneurosci.2905-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Radial glial progenitor cells (RGCs) in the dorsal telencephalon directly or indirectly produce excitatory projection neurons and macroglia of the neocortex. Recent evidence shows that the pool of RGCs is more heterogeneous than originally thought and that progenitor subpopulations can generate particular neuronal cell types. Using single-cell RNA sequencing, we have studied gene expression patterns of RGCs with different neurogenic behavior at early stages of cortical development. At this early age, some RGCs rapidly produce postmitotic neurons, whereas others self-renew and undergo neurogenic divisions at a later age. We have identified candidate genes that are differentially expressed among these early RGC subpopulations, including the transcription factor Sox9. Using in utero electroporation in embryonic mice of either sex, we demonstrate that elevated Sox9 expression in progenitors affects RGC cell cycle duration and leads to the generation of upper layer cortical neurons. Our data thus reveal molecular differences between progenitor cells with different neurogenic behavior at early stages of corticogenesis and indicates that Sox9 is critical for the maintenance of RGCs to regulate the generation of upper layer neurons. SIGNIFICANCE STATEMENT The existence of heterogeneity in the pool of RGCs and its relationship with the generation of cellular diversity in the cerebral cortex has been an interesting topic of debate for many years. Here we describe the existence of RGCs with reduced neurogenic behavior at early embryonic ages presenting a particular molecular signature. This molecular signature consists of differential expression of some genes including the transcription factor Sox9, which has been found to be a specific regulator of this subpopulation of progenitor cells. Functional experiments perturbing expression levels of Sox9 reveal its instructive role in the regulation of the neurogenic behavior of RGCs and its relationship with the generation of upper layer projection neurons at later ages.
Collapse
|
23
|
de Paola M, Garrido F, Zanetti MN, Michaut MA. VAMPs sensitive to tetanus toxin are required for cortical granule exocytosis in mouse oocytes. Exp Cell Res 2021; 405:112629. [PMID: 34023392 DOI: 10.1016/j.yexcr.2021.112629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/16/2023]
Abstract
Fusion of cortical granules with oocyte plasma membrane is one of the most significant secretory events to prevent polyspermy during oocyte activation. Cortical granule exocytosis (CGE) is distinct from most other exocytosis because cortical granules are not renewed after secretion. However, it is thought to be mediated by SNARE complex, which mediates membrane fusion in other exocytoses. SNAREs proteins are divided into Q (glutamine)- and R (arginine)-SNAREs. Q-SNAREs include Syntaxins and SNAP25 family, and R-SNAREs include VAMPs family. In mouse oocytes, Syntaxin4 and SNAP23 have been involved in CGE; nevertheless, it is unknown if VAMP is required. Here, we demonstrated by RT-PCR and immunoblotting that VAMP1 and VAMP3 are expressed in mouse oocyte, and they localized in the cortical region of this cell. Using a functional assay to quantify CGE, we showed that tetanus toxin -which specifically cleavages VAMP1, VAMP2 or VAMP3- inhibited CGE suggesting that at least one VAMP was necessary. Function blocking assays demonstrated that only the microinjection of anti-VAMP1 or anti-VAMP3 antibodies abolished CGE in activated oocytes. These findings demonstrate that R-SNAREs sensitive to tetanus toxin, VAMP1 and VAMP3 -but not VAMP2-, are required for CGE and demonstrate that CGE is mediated by the SNARE complex.
Collapse
Affiliation(s)
- Matilde de Paola
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Av. Libertador 80, 5500, Mendoza, Argentina
| | - Facundo Garrido
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina
| | - María N Zanetti
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina
| | - Marcela Alejandra Michaut
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Padre Jorge Contreras, 1300, Mendoza, Argentina.
| |
Collapse
|
24
|
Wang Z, Li J, Zhang T, Lu T, Wang H, Jia M, Liu J, Xiong J, Zhang D, Wang L. Family-based association study identifies SNAP25 as a susceptibility gene for autism in the Han Chinese population. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:109985. [PMID: 32479779 DOI: 10.1016/j.pnpbp.2020.109985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/09/2020] [Accepted: 05/26/2020] [Indexed: 11/18/2022]
Abstract
Autism is a neurodevelopmental disorder with high heritability. Synaptosome associated protein 25 (SNAP25) encodes a presynaptic membrane-binding protein. It plays a crucial role in neurotransmission and may be involved in the pathogenesis of autism. However, the association between SNAP25 and autism in the Han Chinese population remains unclear. To investigate whether single nucleotide polymorphisms (SNPs) in SNAP25 contribute to the risk of autism, we performed a family-based association study of 14 tagSNPs in SNAP25 in 640 Han Chinese autism trios. Our results demonstrated that rs363018 in SNAP25 was significantly associated with autism under both additive (A > G, Z = 3.144, P = .0017) and recessive models (A > G, Z = 3.055, P = .0023) after Bonferroni correction (P < .0036). An additional SNP, rs8636, was nominally associated with autism under the recessive model (C > T, Z = 1.972, P = .0487). Haplotype-based association test revealed that haplotypes A-T (Z = 2.038, P = .0415) and G-T (Z = -3.114, P = .0018) of rs363018-rs362582 were significantly associated with autism after the permutation test (P = .0158). These findings suggest that SNAP25 may represent a susceptibility gene for autism in the Han Chinese population.
Collapse
Affiliation(s)
- Ziqi Wang
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Jun Li
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Tian Zhang
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Tianlan Lu
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Han Wang
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Meixiang Jia
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Jing Liu
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Jun Xiong
- Haidian Maternal & Child Health Hospital, Beijing 100080, China.
| | - Dai Zhang
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lifang Wang
- Peking University Sixth Hospital, Beijing 100191, China; Peking University Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), Beijing 100191, China; National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
25
|
Mühlenbrock P, Sari M, Steinem C. In vitro single vesicle fusion assays based on pore-spanning membranes: merits and drawbacks. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:239-252. [PMID: 33320298 PMCID: PMC8071798 DOI: 10.1007/s00249-020-01479-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022]
Abstract
Neuronal fusion mediated by soluble N-ethylmaleimide-sensitive-factor attachment protein receptors (SNAREs) is a fundamental cellular process by which two initially distinct membranes merge resulting in one interconnected structure to release neurotransmitters into the presynaptic cleft. To get access to the different stages of the fusion process, several in vitro assays have been developed. In this review, we provide a short overview of the current in vitro single vesicle fusion assays. Among those assays, we developed a single vesicle assay based on pore-spanning membranes (PSMs) on micrometre-sized pores in silicon, which might overcome some of the drawbacks associated with the other membrane architectures used for investigating fusion processes. Prepared by spreading of giant unilamellar vesicles with reconstituted t-SNAREs, PSMs provide an alternative tool to supported lipid bilayers to measure single vesicle fusion events by means of fluorescence microscopy. Here, we discuss the diffusive behaviour of the reconstituted membrane components as well as that of the fusing synthetic vesicles with reconstituted synaptobrevin 2 (v-SNARE). We compare our results with those obtained if the synthetic vesicles are replaced by natural chromaffin granules under otherwise identical conditions. The fusion efficiency as well as the different fusion states observable in this assay by means of both lipid mixing and content release are illuminated.
Collapse
Affiliation(s)
- Peter Mühlenbrock
- Georg-August-Universität Göttingen, Institute of Organic and Biomolecular Chemistry, Tammannstr. 2, 37077, Göttingen, Germany
| | - Merve Sari
- Georg-August-Universität Göttingen, Institute of Organic and Biomolecular Chemistry, Tammannstr. 2, 37077, Göttingen, Germany
| | - Claudia Steinem
- Georg-August-Universität Göttingen, Institute of Organic and Biomolecular Chemistry, Tammannstr. 2, 37077, Göttingen, Germany.
- Max-Planck-Institute for Dynamics and Self Organization, Am Faßberg 17, 37077, Göttingen, Germany.
| |
Collapse
|
26
|
Jin Y, Zhang C, Fang X, Fang C, Chen J, Du R, Hu Q, Dong L, Zhu Z, Wang T. SNAP25 protects primary cortical neurons from hypoxic-ischemic injury associated with CREB signal. IBRAIN 2021; 7:1-11. [PMID: 37786874 PMCID: PMC10528992 DOI: 10.1002/j.2769-2795.2021.tb00058.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 10/04/2023]
Abstract
Background Hypoxic-ischemic encephalopathy (HIE) could induce exacerbated changes and unpredictable effects in brain cells, and the mechanism remains unclear. Methods HIE model was established in neonatal rats, Zea-Longa score and TTC staining were used to observe the neurobehavior and brain infarct volume in rats subjected to cerebral hypoxia-ischemia (HI). Primary cortical neurons were then cultured in vitro to establish an oxygen and glucose deprivation model. To determine the role of synaptosomal-associated protein-25 (SNAP25) in HIE, PC12 cells were cultured and effective siRNA fragments were screened, and SNAP25 was transfected into primary neurons. Then, quantitative real-time polymerase chain reaction was used to detect the mRNA expression level and immunofluorescence staining was used to observe the morphological changes of neurons before and after the injury. Finally, the abundance values of SNAP25 and its associated genes were filtered using the NCBI and GeneMANIA, respectively. Results HI leads to a decrease in neuronal number and an increase in SNAP25 expression. Whereas, the interference of SNAP25 caused marked decrease in neuronal number and the length of neurite. Moreover, the expression levels of CREB and SYP were significantly decreased after interference of SNAP25. Conclusion SNAP25 exhibited several neuroprotective effects to neuronal protection in neonatal cerebral HI by regulating CREB and SYP.
Collapse
Affiliation(s)
- Yuan Jin
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Chao Zhang
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xu Fang
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Chang‐Le Fang
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research CenterDepartment of AnesthesiologyDepartment of Cardiovascular DiseaseAffiliated Traditional Chinese Medicine Hospital, Southwest Medical UniversityLuzhouSichuanChina
| | - Jie Chen
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research CenterDepartment of AnesthesiologyDepartment of Cardiovascular DiseaseAffiliated Traditional Chinese Medicine Hospital, Southwest Medical UniversityLuzhouSichuanChina
| | - Ruo‐Lan Du
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Qiao Hu
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Liang Dong
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Zhao‐Qiong Zhu
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Ting‐Hua Wang
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| |
Collapse
|
27
|
Hubrich BE, Menzel PM, Kugler B, Diederichsen U. Synthesis of PNA-Peptide Conjugates as Functional SNARE Protein Mimetics. Methods Mol Biol 2021; 2105:61-74. [PMID: 32088864 DOI: 10.1007/978-1-0716-0243-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PNA-peptide conjugates are versatile tools in chemical biology, which are employed in a variety of applications. Here, we present the synthesis of PNA-peptide conjugates that serve as SNARE protein-mimicking biooligomers. They resemble the structure of native SNARE proteins but exhibit a much simpler architecture. Incorporated into liposomes, they induce lipid mixing, so that they can be used to study the SNARE-mediated membrane fusion in a simplified setting in vitro. They consist of artificial SNARE recognition units made out of PNA oligomers, which are attached to the native linker and transmembrane domains of two neuronal SNAREs. The PNA-peptide conjugates are synthesized via solid-phase peptide synthesis in a continuous fashion starting with the peptide part, followed by assembly of the PNA recognition unit. On top, we describe a strategy to synthesize PNA-peptide conjugates in a fully automated fashion by using a peptide synthesizer.
Collapse
Affiliation(s)
- Barbara E Hubrich
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Patrick M Menzel
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Benedikt Kugler
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Ulf Diederichsen
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany.
| |
Collapse
|
28
|
Rubio C, Luna R, Rosiles A, Rubio-Osornio M. Caloric Restriction and Ketogenic Diet Therapy for Epilepsy: A Molecular Approach Involving Wnt Pathway and K ATP Channels. Front Neurol 2020; 11:584298. [PMID: 33250850 PMCID: PMC7676225 DOI: 10.3389/fneur.2020.584298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Epilepsy is a neurological disorder in which, in many cases, there is poor pharmacological control of seizures. Nevertheless, it may respond beneficially to alternative treatments such as dietary therapy, like the ketogenic diet or caloric restriction. One of the mechanisms of these diets is to produce a hyperpolarization mediated by the adenosine triphosphate (ATP)-sensitive potassium (KATP) channels (KATP channels). An extracellular increase of K+ prevents the release of Ca2+ by inhibiting the signaling of the Wnt pathway and the translocation of β-catenin to the cell nucleus. Wnt ligands hyperpolarize the cells by activating K+ current by Ca2+. Each of the diets described in this paper has in common a lower use of carbohydrates, which leads to biochemical, genetic processes presumed to be involved in the reduction of epileptic seizures. Currently, there is not much information about the genetic processes implicated as well as the possible beneficial effects of diet therapy on epilepsy. In this review, we aim to describe some of the possible genes involved in Wnt pathways, their regulation through the KATP channels which are implicated in each one of the diets, and how they can reduce epileptic seizures at the molecular level.
Collapse
Affiliation(s)
- Carmen Rubio
- Neurophysiology Department, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Rudy Luna
- Neurophysiology Department, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Artemio Rosiles
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Moisés Rubio-Osornio
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| |
Collapse
|
29
|
Mehlmann LM, Uliasz TF, Lowther KM. SNAP23 is required for constitutive and regulated exocytosis in mouse oocytes†. Biol Reprod 2020; 101:338-346. [PMID: 31201423 DOI: 10.1093/biolre/ioz106] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/17/2019] [Accepted: 06/10/2019] [Indexed: 12/25/2022] Open
Abstract
Mammalian oocytes are stored in the ovary for prolonged periods, and arrested in meiotic prophase. During this period, their plasma membranes are constantly being recycled by endocytosis and exocytosis. However, the function of this membrane turnover is unknown. Here, we investigated the requirement for exocytosis in the maintenance of meiotic arrest. Using Trim-away, a newly developed method for rapidly and specifically depleting proteins in oocytes, we have identified the SNARE protein, SNAP23, to be required for meiotic arrest. Degradation of SNAP23 causes premature meiotic resumption in follicle-enclosed oocytes. The reduction in SNAP23 is associated with loss of gap junction communication between the oocyte and surrounding follicle cells. Reduction of SNAP23 protein also inhibits regulated exocytosis in response to a Ca2+ stimulus (cortical granule exocytosis), as measured by lectin staining and cleavage of ZP2. Our results show an essential role for SNAP23 in two key processes that occur in mouse oocytes and eggs.
Collapse
Affiliation(s)
- Lisa M Mehlmann
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| | - Tracy F Uliasz
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| | - Katie M Lowther
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
30
|
MARTINIÈRE A, MOREAU P. Complex roles of Rabs and SNAREs in the secretory pathway and plant development: a never‐ending story. J Microsc 2020; 280:140-157. [DOI: 10.1111/jmi.12952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/22/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Affiliation(s)
- A. MARTINIÈRE
- Univ Montpellier, CNRS, INRAE, Montpellier SupAgro BPMP Montpellier France
| | - P. MOREAU
- UMR 5200 Membrane Biogenesis Laboratory CNRS and University of Bordeaux, INRAE Bordeaux Villenave d'Ornon France
| |
Collapse
|
31
|
Lakhssassi N, Piya S, Bekal S, Liu S, Zhou Z, Bergounioux C, Miao L, Meksem J, Lakhssassi A, Jones K, Kassem MA, Benhamed M, Bendahmane A, Lambert K, Boualem A, Hewezi T, Meksem K. A pathogenesis-related protein GmPR08-Bet VI promotes a molecular interaction between the GmSHMT08 and GmSNAP18 in resistance to Heterodera glycines. PLANT BIOTECHNOLOGY JOURNAL 2020; 18:1810-1829. [PMID: 31960590 PMCID: PMC7336373 DOI: 10.1111/pbi.13343] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/19/2019] [Accepted: 01/03/2020] [Indexed: 05/19/2023]
Abstract
Soybean cyst nematode (SCN, Heterodera glycines) is the most devastating pest affecting soybean production worldwide. SCN resistance requires both the GmSHMT08 and the GmSNAP18 in 'Peking'-type resistance. Here, we describe the molecular interaction between GmSHMT08 and GmSNAP18, which is potentiated by a pathogenesis-related protein GmPR08-Bet VI. Like GmSNAP18 and GmSHMT08, GmPR08-Bet VI expression was induced in response to SCN and its overexpression decreased SCN cysts by 65% in infected transgenic soybean roots. Overexpression of GmPR08-Bet VI did not have an effect on SCN resistance when the two cytokinin-binding sites in GmPR08-Bet VI were mutated, indicating a new role of GmPR08-Bet VI in SCN resistance. GmPR08-Bet VI was mapped to a QTL for resistance to SCN using different mapping populations. GmSHMT08, GmSNAP18 and GmPR08-Bet VI localize to the cytosol and plasma membrane. GmSNAP18 expression and localization hyper-accumulated at the plasma membrane and was specific to the root cells surrounding the nematode in SCN-resistant soybeans. Genes encoding key components of the salicylic acid signalling pathway were induced under SCN infection. GmSNAP18 and GmPR08-Bet VI were also induced under salicylic acid and cytokinin exogenous treatments, while GmSHMT08 was induced only when the resistant GmSNAP18 was present, pointing to the presence of a molecular crosstalk between SCN-resistant genes and defence genes. Expression analysis of GmSHMT08 and GmSNAP18 identified the need of a minimum expression requirement to trigger the SCN resistance reaction. These results provide insight into a new response mechanism towards plant nematode resistance involving haplotype compatibility, gene dosage and hormone signalling.
Collapse
Affiliation(s)
- Naoufal Lakhssassi
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| | - Sarbottam Piya
- Department of Plant SciencesUniversity of TennesseeKnoxvilleTNUSA
| | - Sadia Bekal
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| | - Shiming Liu
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| | - Zhou Zhou
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| | - Catherine Bergounioux
- INRAInstitute of Plant Sciences Paris‐Saclay (IPS2)CNRSUniversité Paris‐SudOrsayFrance
| | - Long Miao
- Department of Plant SciencesUniversity of TennesseeKnoxvilleTNUSA
| | | | - Aicha Lakhssassi
- Faculty of Sciences and TechnologiesUniversity of LorraineNancyFrance
| | - Karen Jones
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| | | | - Moussa Benhamed
- INRAInstitute of Plant Sciences Paris‐Saclay (IPS2)CNRSUniversité Paris‐SudOrsayFrance
| | - Abdelhafid Bendahmane
- INRAInstitute of Plant Sciences Paris‐Saclay (IPS2)CNRSUniversité Paris‐SudOrsayFrance
| | - Kris Lambert
- Department of Crop SciencesUniversity of IllinoisUrbanaILUSA
| | - Adnane Boualem
- INRAInstitute of Plant Sciences Paris‐Saclay (IPS2)CNRSUniversité Paris‐SudOrsayFrance
| | - Tarek Hewezi
- Department of Plant SciencesUniversity of TennesseeKnoxvilleTNUSA
| | - Khalid Meksem
- Department of Plant, Soil and Agricultural SystemsSouthern Illinois UniversityCarbondaleILUSA
| |
Collapse
|
32
|
Shao K, Li F, Yang Y, Wang N, Gao XD, Nakanishi H. Characteristics of SNARE proteins are defined by distinctive properties of SNARE motifs. Biochim Biophys Acta Gen Subj 2020; 1864:129658. [PMID: 32512168 DOI: 10.1016/j.bbagen.2020.129658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Syntaxin-1A and Sso1 are syntaxin family SNARE proteins engaged in synaptic vesicle fusion and yeast exocytosis. The syntaxin-1A SNARE motif can form a fusogenic SNARE complex with Sso1 partners. However, a chimera in which the SNARE motif in syntaxin-1A is introduced into Sso1 was not functional in yeast because the chimera is retained in the ER. Through the analysis of the transport defect of Sso1/syntaxin-1A chimeric SNAREs, we found that their SNARE motifs have distinctive properties. METHODS Sso1, syntaxin-1A, and Sso1/syntaxin-1A chimeric SNAREs were expressed in yeast cells and their localization and interaction with other SNAREs are analyzed. RESULTS SNARE proteins containing the syntaxin-1A SNARE motif exhibit a transport defect because they form a cis-SNARE complex in the ER. Ectopic SNARE complex formation can be prevented in syntaxin-1A by binding to a Sec1/Munc-18-like (SM) protein. In contrast, the SNARE motif of Sso1 does not form an ectopic SNARE complex. Additionally, we found that the SNARE motif in syntaxin-1A, but not that in Sso1, self-interacts, even when it is in the inactive form and bound to the SM protein. CONCLUSIONS The SNARE motif in syntaxin-1A, but not in Sso1, likely forms ectopic SNARE complex. Because of this property, the SM protein is necessary for syntaxin-1A to prevent its promiscuous assembly and to promote its export from the ER. GENERAL SIGNIFICANCE Properties of SNARE motifs affect characteristics of SNARE proteins. The regulatory mechanisms of SNARE proteins are, in part, designed to handle such properties.
Collapse
Affiliation(s)
- Kankai Shao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Feng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Yan Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Ning Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| | - Hideki Nakanishi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| |
Collapse
|
33
|
PI(4,5)P 2-dependent regulation of exocytosis by amisyn, the vertebrate-specific competitor of synaptobrevin 2. Proc Natl Acad Sci U S A 2020; 117:13468-13479. [PMID: 32467162 DOI: 10.1073/pnas.1908232117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The functions of nervous and neuroendocrine systems rely on fast and tightly regulated release of neurotransmitters stored in secretory vesicles through SNARE-mediated exocytosis. Few proteins, including tomosyn (STXBP5) and amisyn (STXBP6), were proposed to negatively regulate exocytosis. Little is known about amisyn, a 24-kDa brain-enriched protein with a SNARE motif. We report here that full-length amisyn forms a stable SNARE complex with syntaxin-1 and SNAP-25 through its C-terminal SNARE motif and competes with synaptobrevin-2/VAMP2 for the SNARE-complex assembly. Furthermore, amisyn contains an N-terminal pleckstrin homology domain that mediates its transient association with the plasma membrane of neurosecretory cells by binding to phospholipid PI(4,5)P2 However, unlike synaptrobrevin-2, the SNARE motif of amisyn is not sufficient to account for the role of amisyn in exocytosis: Both the pleckstrin homology domain and the SNARE motif are needed for its inhibitory function. Mechanistically, amisyn interferes with the priming of secretory vesicles and the sizes of releasable vesicle pools, but not vesicle fusion properties. Our biochemical and functional analyses of this vertebrate-specific protein unveil key aspects of negative regulation of exocytosis.
Collapse
|
34
|
Won KH, Kim H. Functions of the Plant Qbc SNARE SNAP25 in Cytokinesis and Biotic and Abiotic Stress Responses. Mol Cells 2020; 43:313-322. [PMID: 32274918 PMCID: PMC7191049 DOI: 10.14348/molcells.2020.2245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/29/2022] Open
Abstract
Eukaryotes transport biomolecules between intracellular organelles and between cells and the environment via vesicle trafficking. Soluble N -ethylmaleimide-sensitive factor attachment protein receptors (SNARE proteins) play pivotal roles in vesicle and membrane trafficking. These proteins are categorized as Qa, Qb, Qc, and R SNAREs and form a complex that induces vesicle fusion for targeting of vesicle cargos. As the core components of the SNARE complex, the SNAP25 Qbc SNAREs perform various functions related to cellular homeostasis. The Arabidopsis thaliana SNAP25 homolog AtSNAP33 interacts with Qa and R SNAREs and plays a key role in cytokinesis and in triggering innate immune responses. However, other Arabidopsis SNAP25 homologs, such as AtSNAP29 and AtSNAP30, are not well studied; this includes their localization, interactions, structures, and functions. Here, we discuss three biological functions of plant SNAP25 orthologs in the context of AtSNAP33 and highlight recent findings on SNAP25 orthologs in various plants. We propose future directions for determining the roles of the less well-characterized AtSNAP29 and AtSNAP30 proteins.
Collapse
Affiliation(s)
- Kang-Hee Won
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Hyeran Kim
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
35
|
Hasegawa T, Ikeda HO, Gotoh N, Iida K, Iwai S, Nakano N, Kakizuka A, Tsujikawa A. Effect of VCP modulators on gene expression profiles of retinal ganglion cells in an acute injury mouse model. Sci Rep 2020; 10:4251. [PMID: 32144342 PMCID: PMC7060332 DOI: 10.1038/s41598-020-61160-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/10/2020] [Indexed: 11/09/2022] Open
Abstract
In glaucoma, retinal ganglion cells are damaged, leading to the progressive constriction of the visual field. We have previously shown that the valosin-containing protein (VCP) modulators, Kyoto University Substance (KUS)121 and KUS187, prevent the death of retinal ganglion cells in animal models of glaucoma, including the one generated by N-methyl-D-aspartate (NMDA)-induced neurotoxicity. KUSs appeared to avert endoplasmic reticulum (ER) stress by maintaining ATP levels, resulting in the protection of ganglion cells from cell death. To further elucidate the protective mechanisms of KUSs, we examined gene expression profiles in affected ganglion cells. We first injected KUS-treated mice with NMDA and then isolated the affected retinal ganglion cells using fluorescence-activated cell sorting. Gene expression in the cells was quantified using a next-generation sequencer. Resultantly, we found that KUS121 upregulated several genes involved in energy metabolism. In addition, we observed the upregulation of Zfp667, which has been reported to suppress apoptosis-related genes and prevent cell death. These results further support the suitability of KUS121 as a therapeutic drug in protecting retinal ganglion cells in ophthalmic disorders, such as glaucoma.
Collapse
Affiliation(s)
- Tomoko Hasegawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Hanako Ohashi Ikeda
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan.
| | - Norimoto Gotoh
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Kei Iida
- Medical Research Support Center, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Sachiko Iwai
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Noriko Nakano
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Akira Kakizuka
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies & Solution Oriented Research for Science and Technology, Kyoto, 606-8501, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| |
Collapse
|
36
|
Gopaul KR, Irfan M, Miry O, Vose LR, Moghadam A, Subah G, Hökfelt T, Bark C, Stanton PK. Developmental Time Course of SNAP-25 Isoforms Regulate Hippocampal Long-Term Synaptic Plasticity and Hippocampus-Dependent Learning. Int J Mol Sci 2020; 21:ijms21041448. [PMID: 32093363 PMCID: PMC7073020 DOI: 10.3390/ijms21041448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
SNAP-25 is essential to activity-dependent vesicle fusion and neurotransmitter release in the nervous system. During early development and adulthood, SNAP-25 appears to have differential influences on short- and long-term synaptic plasticity. The involvement of SNAP-25 in these processes may be different at hippocampal and neocortical synapses because of the presence of two different splice variants, which are developmentally regulated. We show here that the isoform SNAP-25a, which is expressed first developmentally in rodent brain, contributes to developmental regulation of the expression of both long-term depression (LTD) and long-term potentiation (LTP) at Schaffer collateral-CA1 synapses in the hippocampus. In one month old mice lacking the developmentally later expressed isoform SNAP-25b, Schaffer collateral-CA1 synapses showed faster release kinetics, decreased LTP and enhanced LTD. By four months of age, SNAP-25b-deficient mice appeared to have compensated for the lack of the adult SNAP-25b isoform, now exhibiting larger LTP and no differences in LTD compared to wild type mice. Interestingly, learning a hippocampus-dependent task reversed the reductions in LTP, but not LTD, seen at one month of age. In four month old adult mice, learning prevented the compensatory up-regulation of LTD that we observed prior to training. These findings support the hypothesis that SNAP-25b promotes stronger LTP and weakens LTD at Schaffer collateral-CA1 synapses in young mice, and suggest that compensatory mechanisms can reverse alterations in synaptic plasticity associated with a lack of SNAP-25b, once mice reach adulthood.
Collapse
Affiliation(s)
- Katisha R. Gopaul
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Muhammad Irfan
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Omid Miry
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Linnea R. Vose
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Alexander Moghadam
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Galadu Subah
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Christina Bark
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
- Correspondence: (C.B.); (P.K.S.); Tel. +46-085-248-6984 (C.B.); +1-914-594-4883 (P.K.S.)
| | - Patric K. Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
- Correspondence: (C.B.); (P.K.S.); Tel. +46-085-248-6984 (C.B.); +1-914-594-4883 (P.K.S.)
| |
Collapse
|
37
|
Liu P, Song C, Wang C, Li Y, Su L, Li J, Zhao Q, Wang Z, Shen M, Wang G, Yu Y, Zhang L. Spinal SNAP-25 regulates membrane trafficking of GluA1-containing AMPA receptors in spinal injury-induced neuropathic pain in rats. Neurosci Lett 2020; 715:134616. [PMID: 31705923 DOI: 10.1016/j.neulet.2019.134616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/21/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Synaptosomal associated proteins of 25 kDa (SNAP-25), as a member of stable soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex, is critical for membrane fusion and required for the release of neurotransmitters. The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor is implicated in pathologic pain. This study aimed to investigate whether and how SNAP-25 regulated AMPA receptors in neuropathic pain. METHODS Male Sprague-Dawley rats underwent L4 spinal nerve ligation (SNL) or the sham procedure. After assessing mechanical allodynia and thermal sensitivity, the ipsilateral portion of the L4-5 spinal cord was harvested. The expression level of SNAP-25 was analyzed by Western blot analysis and real-time quantitative polymerase chain reaction. SNAP-25 phosphorylation and AMPA receptor membrane trafficking levels were evaluated with Western blot analysis. An association between SNAP-25 and AMPA membrane trafficking was confirmed by SNAP-25 expression or phosphorylation inhibition. RESULTS The SNL procedure induced and maintained mechanical allodynia and thermal hyperalgesia. SNL increased the expression and phosphorylation of SNAP-25 and the membrane trafficking of AMPA receptors in the spinal cord. SNAP-25 expression or phosphorylation inhibition alleviated neuropathic pain and downregulated membrane trafficking of AMPA receptors after SNL. GluA1-containing AMPA receptor inhibition relieved mechanical allodynia and thermal hyperalgesia after SNL. CONCLUSIONS The upregulation of SNAP-25-dependent membrane trafficking of AMPA receptors via SNAP-25 phosphorylation at Ser187 contributed to SNL-induced neuropathic pain. Thus, the inhibition of SNAP-25 expression or phosphorylation might serve as a treatment for neuropathic pain. However, the mechanism of GluA1-containing AMPA receptor membrane trafficking mediated by SNAP-25 phosphorylation in neuropathic pain deserves further exploration.
Collapse
Affiliation(s)
- Peng Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chengcheng Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Chunyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Lin Su
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Qi Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Zhen Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Mengxi Shen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| |
Collapse
|
38
|
SNAP-25 Puts SNAREs at Center Stage in Metabolic Disease. Neuroscience 2019; 420:86-96. [DOI: 10.1016/j.neuroscience.2018.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022]
|
39
|
The SNAP-25 Protein Family. Neuroscience 2019; 420:50-71. [DOI: 10.1016/j.neuroscience.2018.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
|
40
|
Raynaud F, Homburger V, Seveno M, Vigy O, Moutin E, Fagni L, Perroy J. SNAP23-Kif5 complex controls mGlu1 receptor trafficking. J Mol Cell Biol 2019; 10:423-436. [PMID: 29762713 DOI: 10.1093/jmcb/mjy031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/11/2018] [Indexed: 11/13/2022] Open
Abstract
Metabotropic glutamate receptors are expressed at excitatory synapses and control synaptic transmission in mammalian brain. These receptors are involved in numerous patho-physiological functions. However, little is known about the molecular determinants responsible for their intracellular transport and membrane targeting. Here we investigated the nature of the molecular motor and adaptor protein responsible for trafficking and membrane localization of the group I metabotropic glutamate mGlu1 postsynaptic receptor in cultured hippocampal neurons. In proteomic studies, we identified the synaptosome-associated protein 23 (SNAP23) and the molecular motor Kif5 kinesin as proteins interacting with mGlu1 receptor. We showed that SNAP23, but not Kif5, directly interacts with mGlu1 receptor carboxyl terminus. Using a recombination approach to impair or enhance the interaction between SNAP23 and Kif5, we found that the SNAP23-Kif5 complex controls the trafficking of mGlu1 receptor along microtubules. Additional fluorescence recovery after cleavage experiments allowed us to identify a role of the complex in the receptor cell surface targeting. In conclusion, our study indicates that along dendritic processes Kif5-SNAP23 complex contributes to proper mGlu1 receptor trafficking and cell surface expression.
Collapse
Affiliation(s)
| | | | - Martial Seveno
- BioCampus Montpellier, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Oana Vigy
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Enora Moutin
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Laurent Fagni
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Julie Perroy
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| |
Collapse
|
41
|
Wang M, Gu X, Huang X, Zhang Q, Chen X, Wu J. STX1A gene variations contribute to the susceptibility of children attention-deficit/hyperactivity disorder: a case-control association study. Eur Arch Psychiatry Clin Neurosci 2019; 269:689-699. [PMID: 30976917 DOI: 10.1007/s00406-019-01010-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/26/2019] [Indexed: 12/19/2022]
Abstract
It was presumed syntaxin-1A (STX1A) might relate to the pathophysiology of attention-deficit/hyperactivity disorder (ADHD), but the results were inconsistent. The present study aims to confirm whether the STX1A gene is involved in the susceptibility of children ADHD. We genotyped three single nucleotide polymorphisms (SNPs) of STX1A gene using Sequenom MassARRAY technology. A case-control study was performed among Chinese Han population including 754 cases and 772 controls from two different provinces. The Conners Parent Symptom Questionnaire and Integrated Visual and Auditory Continuous Performance Test were used to assess ADHD clinical symptoms. We found for the first time that rs3793243 GG genotype carriers had a lower risk of ADHD compared with AA genotype (OR 0.564, 95% confidence interval (CI) 0.406-0.692, P = 0.001), and rs875342 was also associated with children ADHD (OR 1.806, 95% CI 1.349-2.591, P = 0.001). In addition, the two positive SNPs were also significantly associated with the clinical characteristics of ADHD. Expression quantitative trait loci analysis indicated that rs3793243 might mediate STX1A gene expression. Using a case-control study to explore the association between STX1A gene and children ADHD in Chinese Han population, our results suggest STX1A genetic variants might contribute to the susceptibility of children ADHD.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xue Gu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xin Huang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qi Zhang
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinzhen Chen
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China.,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, 430030, People's Republic of China. .,Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
42
|
Construction of functional chimeras of syntaxin-1A and its yeast orthologue, and their application to the yeast cell-based assay for botulinum neurotoxin serotype C. Biochim Biophys Acta Gen Subj 2019; 1863:129396. [PMID: 31302181 DOI: 10.1016/j.bbagen.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/14/2019] [Accepted: 07/10/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Botulinum neurotoxins (BoNTs) prevent synaptic transmission because they hydrolyze synaptic N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). BoNT serotype C (BoNT/C) targets syntaxin-1A and SNAP-25, and is expected to be applied to cosmetic and therapeutic uses. SNAREs are evolutionally conserved proteins and in yeast a syntaxin-1A orthologue Sso1 is involved in exocytosis. The substrate specificity of BoNT/C is strict and it cannot cleave Sso1. METHODS Domain swapping and mutational screenings were performed to generate functional chimeras SNAREs of syntaxin-1A and Sso1. Such chimeras are expressed in yeast cells and assessed whether they are susceptible to BoNT/C digestion. RESULTS The Sso1 and syntaxin-1A chimera (Sso1/STX1A), in which the SNARE domain in Sso1 was replaced with that of syntaxin-1A, was not functional in yeast. The functional incompatibility of Sso1/STX1A was attributable to its accumulation in the ER. We found several mutations that could release Sso1/STX1A from the ER to make the chimera functional in yeast. Yeast cells harboring the mutant chimeras grew similarly to wild-type cells. However, unlike wild-type, yeast harboring the mutant chimeras exhibited a severe growth defect upon expression of BoNT/C. Results of further domain swapping analyses suggest that Sso1 is not digested by BoNT/C because it lacks a binding region to BoNT/C (α-exosite-binding region). CONCLUSIONS We obtained functional Sso1/STX1A chimeras, which can be applied to a yeast cell-based BoNT/C assay. BoNT/C can recognize these chimeras in a similar manner to syntaxin-1A. GENERAL SIGNIFICANCE The yeast cell-based BoNT/C assay would be useful to characterize and engineer BoNT/C.
Collapse
|
43
|
Martin CA, Radhakrishnan S, Nagarajan S, Muthukoori S, Dueñas JMM, Gómez Ribelles JL, Lakshmi BS, E A K N, Gómez-Tejedor JA, Reddy MS, Sellathamby S, Rela M, Subbaraya NK. An innovative bioresorbable gelatin based 3D scaffold that maintains the stemness of adipose tissue derived stem cells and the plasticity of differentiated neurons. RSC Adv 2019; 9:14452-14464. [PMID: 35519343 PMCID: PMC9064131 DOI: 10.1039/c8ra09688k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/05/2019] [Indexed: 12/02/2022] Open
Abstract
Neural tissue engineering aims at producing a simulated environment using a matrix that is suitable to grow specialized neurons/glial cells pertaining to CNS/PNS which replace damaged or lost tissues. The primary goal of this study is to design a compatible scaffold that supports the development of neural-lineage cells which aids in neural regeneration. The fabricated, freeze-dried scaffolds consisted of biocompatible, natural and synthetic polymers: gelatin and polyvinyl pyrrolidone. Physiochemical characterization was carried out using Fourier Transform Infrared Spectroscopy (FT-IR) and Scanning Electron Microscopy (SEM) imaging. The 3D construct retains good swelling proficiency and holds the integrated structure that supports cell adhesion and proliferation. The composite of PVP-gelatin is blended in such a way that it matches the mechanical strength of the brain tissue. The cytocompatibility analysis shows that the scaffolds are compatible and permissible for the growth of both stem cells as well as differentiated neurons. A change in the ratios of the scaffold components resulted in varied sizes of pores giving diverse surface morphology, greatly influencing the properties of the neurons. However, there is no change in stem cell properties. Different types of neurons are characterized by the type of gene associated with the neurotransmitter secreted by them. The change in the neuron properties could be attributed to neuroplasticity. The plasticity of the neurons was analyzed using quantitative gene expression studies. It has been observed that the gelatin-rich construct supports the prolonged proliferation of stem cells and multiple neurons along with their plasticity.
Collapse
Affiliation(s)
- Catherine Ann Martin
- Crystal Growth Centre, Anna University Chennai India
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | - Subathra Radhakrishnan
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
- Department of Biomedicine, Bharathidasan University India
| | | | | | - J M Meseguer Dueñas
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | - José Luis Gómez Ribelles
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | | | | | - José Antonio Gómez-Tejedor
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | - Mettu Srinivas Reddy
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | | | - Mohamed Rela
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | | |
Collapse
|
44
|
Simó A, Cilleros-Mañé V, Just-Borràs L, Hurtado E, Nadal L, Tomàs M, Garcia N, Lanuza MA, Tomàs J. nPKCε Mediates SNAP-25 Phosphorylation of Ser-187 in Basal Conditions and After Synaptic Activity at the Neuromuscular Junction. Mol Neurobiol 2019; 56:5346-5364. [PMID: 30607888 DOI: 10.1007/s12035-018-1462-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022]
Abstract
Protein kinase C (PKC) and substrates like SNAP-25 regulate neurotransmission. At the neuromuscular junction (NMJ), PKC promotes neurotransmitter release during synaptic activity. Thirty minutes of muscle contraction enhances presynaptic PKC isoform levels, specifically cPKCβI and nPKCε, through retrograde BDNF/TrkB signaling. This establishes a larger pool of these PKC isoforms ready to promote neuromuscular transmission. The PKC phosphorylation site in SNAP-25 has been mapped to the serine 187 (Ser-187), which is known to enhance calcium-dependent neurotransmitter release in vitro. Here, we localize SNAP-25 at the NMJ and investigate whether cPKCβI and/or nPKCε regulate SNAP-25 phosphorylation. We also investigate whether nerve and muscle cell activities regulate differently SNAP-25 phosphorylation and the involvement of BDNF/TrkB signaling. Our results demonstrate that nPKCε isoform is essential to positively regulate SNAP-25 phosphorylation on Ser-187 and that muscle contraction prevents it. TrkB and cPKCβI do not regulate SNAP-25 protein level or its phosphorylation during neuromuscular activity. The results provide evidence that nerve terminals need both pre- and postsynaptic activities to modulate SNAP-25 phosphorylation and ensure an accurate neurotransmission process.
Collapse
Affiliation(s)
- Anna Simó
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Victor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
45
|
Yu H, Crisman L, Stowell MHB, Shen J. Functional Reconstitution of Intracellular Vesicle Fusion Using Purified SNAREs and Sec1/Munc18 (SM) Proteins. Methods Mol Biol 2019; 1860:237-249. [PMID: 30317509 DOI: 10.1007/978-1-4939-8760-3_15] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fusion of intracellular vesicles with target membranes is mediated by two classes of conserved molecules-soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAP receptors or SNAREs) and Sec1/Munc18 (SM) proteins. A conserved function of SM proteins is to recognize their cognate trans-SNARE complexes and accelerate fusion kinetics. Here, we describe a physiologically relevant reconstitution system in which macromolecular crowding agents are included to recapitulate the crowded intracellular environment. Through this system, we elucidate the molecular mechanisms by which SNAREs and SM proteins drive vesicle fusion.
Collapse
Affiliation(s)
- Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China. .,Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| | - Lauren Crisman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Michael H B Stowell
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
46
|
Kandachar V, Tam BM, Moritz OL, Deretic D. An interaction network between the SNARE VAMP7 and Rab GTPases within a ciliary membrane-targeting complex. J Cell Sci 2018; 131:jcs.222034. [PMID: 30404838 DOI: 10.1242/jcs.222034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
The Arf4-rhodopsin complex (mediated by the VxPx motif in rhodopsin) initiates expansion of vertebrate rod photoreceptor cilia-derived light-sensing organelles through stepwise assembly of a conserved trafficking network. Here, we examine its role in the sorting of VAMP7 (also known as TI-VAMP) - an R-SNARE possessing a regulatory longin domain (LD) - into rhodopsin transport carriers (RTCs). During RTC formation and trafficking, VAMP7 colocalizes with the ciliary cargo rhodopsin and interacts with the Rab11-Rabin8-Rab8 trafficking module. Rab11 and Rab8 bind the VAMP7 LD, whereas Rabin8 (also known as RAB3IP) interacts with the SNARE domain. The Arf/Rab11 effector FIP3 (also known as RAB11FIP3) regulates VAMP7 access to Rab11. At the ciliary base, VAMP7 forms a complex with the cognate SNAREs syntaxin 3 and SNAP-25. When expressed in transgenic animals, a GFP-VAMP7ΔLD fusion protein and a Y45E phosphomimetic mutant colocalize with endogenous VAMP7. The GFP-VAMP7-R150E mutant displays considerable localization defects that imply an important role of the R-SNARE motif in intracellular trafficking, rather than cognate SNARE pairing. Our study defines the link between VAMP7 and the ciliary targeting nexus that is conserved across diverse cell types, and contributes to general understanding of how functional Arf and Rab networks assemble SNAREs in membrane trafficking.
Collapse
Affiliation(s)
- Vasundhara Kandachar
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Beatrice M Tam
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Dusanka Deretic
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, USA .,Cell Biology and Physiology, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
47
|
Karmakar S, Sharma LG, Roy A, Patel A, Pandey LM. Neuronal SNARE complex: A protein folding system with intricate protein-protein interactions, and its common neuropathological hallmark, SNAP25. Neurochem Int 2018; 122:196-207. [PMID: 30517887 DOI: 10.1016/j.neuint.2018.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/08/2018] [Accepted: 12/01/2018] [Indexed: 12/26/2022]
Abstract
SNARE (Soluble NSF(N-ethylmaleimide-sensitive factor) Attachment Receptor) complex is a trimeric supramolecular organization of SNAP25, syntaxin, and VAMP which mediates fusion of synaptic vesicles with the presynaptic plasma membrane. The functioning of this entire protein assembly is dependent on its tetrahelical coiled coil structure alongside its interaction with a large spectrum of regulatory proteins like synaptotagmin, complexin, intersectin, etc. Defects arising in SNARE complex assembly due to mutations or faulty post-translational modifications are associated to severe synaptopathies like Schizophrenia and also proteopathies like Alzheimer's disease. The review primarily focuses on SNAP25, which is the prime contributor in the complex assembly. It is conceptualized that the network of protein interactions of this helical protein assists as a chaperoning system for attaining functional structure. Additionally, the innate disordered nature of SNAP25 and its amyloidogenic propensities have been highlighted employing computational methods. The intrinsic nature of SNAP25 is anticipated to form higher-order aggregates due to its cysteine rich domain, which is also a target for several post-translational modifications. Furthermore, the aberrations in the structure and expression profile of the protein display common patterns in the pathogenesis of a diverse synaptopathies and proteopathies. This work of SNARE literature aims to provide a new comprehensive outlook and research directions towards SNARE complex and presents SNAP25 as a common neuropathological hallmark which can be a diagnostic or therapeutic target.
Collapse
Affiliation(s)
- Srijeeb Karmakar
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Laipubam Gayatri Sharma
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Abhishek Roy
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Anjali Patel
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| | - Lalit Mohan Pandey
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| |
Collapse
|
48
|
Pozzi D, Corradini I, Matteoli M. The Control of Neuronal Calcium Homeostasis by SNAP-25 and its Impact on Neurotransmitter Release. Neuroscience 2018; 420:72-78. [PMID: 30476527 DOI: 10.1016/j.neuroscience.2018.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 01/14/2023]
Abstract
The process of neurotransmitter release is central to the control of cell-to-cell communication in brain. SNAP-25 is a component of the SNARE complex, which, together with syntaxin-1 and synaptobrevin, mediates synaptic vesicle fusion with the plasma membrane. The genetic ablation of the protein or its proteolytic cleavage by botulinum neurotoxins results in a complete block of synaptic transmission. In the last years, several evidences have indicated that SNAP-25 also plays additional modulatory roles in neurotransmission through the control of voltage-gated calcium channels and presynaptic calcium ion concentration. Consistently, reduced levels of the protein affect presynaptic calcium homeostasis and result in pathologically enhanced glutamate exocytosis. The SNAP-25-dependent alterations of synaptic calcium dynamics may have direct impact on the development of neuropsychiatric disorders where the Snap-25 gene has been found to be involved.
Collapse
Affiliation(s)
- Davide Pozzi
- Humanitas University, Via Rita Levi Montalcini, 4, 20090 Pieve Emanuele, Milano, Italy; IRCCS Humanitas, via Manzoni 56, 20089 Rozzano, Italy.
| | - Irene Corradini
- CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milano, Italy
| | - Michela Matteoli
- Humanitas University, Via Rita Levi Montalcini, 4, 20090 Pieve Emanuele, Milano, Italy; IRCCS Humanitas, via Manzoni 56, 20089 Rozzano, Italy.
| |
Collapse
|
49
|
Shen C, Liu Y, Yu H, Gulbranson DR, Kogut I, Bilousova G, Zhang C, Stowell MHB, Shen J. The N-peptide-binding mode is critical to Munc18-1 function in synaptic exocytosis. J Biol Chem 2018; 293:18309-18317. [PMID: 30275014 DOI: 10.1074/jbc.ra118.005254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/25/2018] [Indexed: 01/09/2023] Open
Abstract
Sec1/Munc18 (SM) proteins promote intracellular vesicle fusion by binding to N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). A key SNARE-binding mode of SM proteins involves the N-terminal peptide (N-peptide) motif of syntaxin, a SNARE subunit localized to the target membrane. In in vitro membrane fusion assays, inhibition of N-peptide motif binding previously has been shown to abrogate the stimulatory function of Munc18-1, a SM protein involved in synaptic exocytosis in neurons. The physiological role of the N-peptide-binding mode, however, remains unclear. In this work, we addressed this key question using a "clogged" Munc18-1 protein, in which an ectopic copy of the syntaxin N-peptide motif was directly fused to Munc18-1. We found that the ectopic N-peptide motif blocks the N-peptide-binding pocket of Munc18-1, preventing the latter from binding to the native N-peptide motif on syntaxin-1. In a reconstituted system, we observed that clogged Munc18-1 is defective in promoting SNARE zippering. When introduced into induced neuronal cells (iN cells) derived from human pluripotent stem cells, clogged Munc18-1 failed to mediate synaptic exocytosis. As a result, both spontaneous and evoked synaptic transmission was abolished. These genetic findings provide direct evidence for the crucial role of the N-peptide-binding mode of Munc18-1 in synaptic exocytosis. We suggest that clogged SM proteins will also be instrumental in defining the physiological roles of the N-peptide-binding mode in other vesicle-fusion pathways.
Collapse
Affiliation(s)
- Chong Shen
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Yinghui Liu
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309,; the Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Haijia Yu
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309,; the Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China,.
| | - Daniel R Gulbranson
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Igor Kogut
- the Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, and
| | - Ganna Bilousova
- the Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, and
| | - Chen Zhang
- the School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Michael H B Stowell
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Jingshi Shen
- From the Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309,.
| |
Collapse
|
50
|
The impact of SNAP25 on brain functional connectivity density and working memory in ADHD. Biol Psychol 2018; 138:35-40. [PMID: 30092259 DOI: 10.1016/j.biopsycho.2018.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/15/2018] [Accepted: 08/04/2018] [Indexed: 11/20/2022]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopment disorder. The deficit in working memory is a central cognitive impairment in ADHD. The SNAP-25 is a neurotransmitter vesicular docking protein whose MnlI polymorphism (rs3746544) is located in the 3'-untranslated region (3'-UTR) and known to be linked to ADHD, but the underlying mechanism of this polymorphism remains unclear. Using a functional connectivity density (FCD) mapping method based on resting-state functional magnetic resonance imaging in a sample of male children diagnosed with ADHD, we first investigated the correlation between SNAP-25 rs3746544 and FCD hubs. Compared with rs3746544 G-allele carriers, TT homozygous, which confers a high risk for ADHD, exhibited significantly decreased local and long-range FCD in anterior cingulate cortex, and decreased local FCD in the dorsal lateral prefrontal cortex. Moreover, both higher local and long-range FCD could predict better WM capacity. The current findings provide new insights into the underlying neural mechanisms linking SNAP-25 rs3746544 with the risk for ADHD via the endophenotype of brain functional connectivity.
Collapse
|