1
|
Usher S, Toulmé E, Florea R, Yatskevich S, Jao CC, Dijkhof LRH, Colding JM, Joshi P, Zilberleyb I, Trimbuch T, Brokowski B, Hauser AS, Leitner A, Rosenmund C, Kschonsak M, Pless SA. The sodium leak channel NALCN is regulated by neuronal SNARE complex proteins. SCIENCE ADVANCES 2025; 11:eads6004. [PMID: 40085699 DOI: 10.1126/sciadv.ads6004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
NALCN (sodium leak channel, nonselective) is vital for regulating electrical activity in neurons and other excitable cells, and mutations in the channel or its auxiliary proteins lead to severe neurodevelopmental disorders. Here, we show that the neuronal SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) complex proteins syntaxin and SNAP25 (synaptosome-associated protein 25), which enable synaptic transmission in the nervous system, inhibit the activity of the NALCN channel complex in both heterologous systems and primary neurons. The existence of this interaction suggests that the neurotransmitter release machinery can regulate electrical signaling directly and therefore modulate the threshold for its own activity. We further find that reduction of NALCN currents is sufficient to promote cell survival in syntaxin-depleted cells. This suggests that disinhibited NALCN may cause the puzzling phenomenon of rapid neuronal cell death in the absence of syntaxin. This interaction could offer opportunities for future drug development against genetic diseases linked to both NALCN- and SNARE protein-containing complexes.
Collapse
Affiliation(s)
- Samuel Usher
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Estelle Toulmé
- Institut für Neurophysiologie, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Roberta Florea
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, Zürich 8093, Switzerland
| | - Stanislau Yatskevich
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christine C Jao
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Luuk R H Dijkhof
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Janne M Colding
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Prajakta Joshi
- Department of Biomolecular Resources, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Inna Zilberleyb
- Department of Biomolecular Resources, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Thorsten Trimbuch
- Institut für Neurophysiologie, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Bettina Brokowski
- Institut für Neurophysiologie, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Alexander Leitner
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, Zürich 8093, Switzerland
| | - Christian Rosenmund
- Institut für Neurophysiologie, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Marc Kschonsak
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stephan A Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
2
|
Niu Y, Liu C, Jia L, Zhao F, Wang Y, Wang L, Chen W, Gan Y, Wen Y. STX1A regulates ferroptosis and chemoresistance in gastric cancer through mitochondrial function modulation. Hum Cell 2025; 38:66. [PMID: 40056239 DOI: 10.1007/s13577-025-01195-x] [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: 05/18/2024] [Accepted: 02/27/2025] [Indexed: 03/10/2025]
Abstract
Gastric cancer is one of the leading causes of cancer-related deaths worldwide, and chemoresistance remains a major obstacle to effective treatment. Ferroptosis, a novel form of regulated cell death, has emerged as a potential therapeutic strategy to treat cancer. However, the molecular mechanisms regulating ferroptosis in gastric cancer remain largely unknown. In this study, we identified syntaxin 1A (STX1A) as a novel regulator of mitochondrial function and ferroptosis in gastric cancer. We found that STX1A is overexpressed in gastric cancer cell lines and tissues and that its knockdown inhibits cell proliferation and induces ferroptosis. Notably, we made the novel discovery that STX1A is localized to the mitochondria, providing a direct link between STX1A and mitochondrial function. Mechanistically, we demonstrated that STX1A depletion impairs mitochondrial respiration, leading to increased oxidative stress and ferroptosis. Furthermore, we showed that targeting STX1A or directly inhibiting mitochondrial function can reverse acquired resistance to 5-fluorouracil and cisplatin in gastric cancer cells by inducing ferroptosis. Our findings provide new insights into the regulation of ferroptosis in gastric cancer and suggest that the STX1A-mitochondria-ferroptosis axis may be a promising therapeutic target for overcoming chemoresistance and improving patient outcomes.
Collapse
Affiliation(s)
- Yan Niu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, People's Republic of China
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Chunyu Liu
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Lizhou Jia
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Fangxin Zhao
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Yixiao Wang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Lu Wang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Weiyi Chen
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Yanzi Gan
- Rehabilitation Department, The Third Hospital of Hohhot, Hohhot, People's Republic of China.
| | - Yongjun Wen
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, People's Republic of China.
| |
Collapse
|
3
|
Wu C, Wang Q, Li W, Han M, Zhao H, Xu Z. Research progress on pathogenesis and treatment of febrile seizures. Life Sci 2025; 362:123360. [PMID: 39746603 DOI: 10.1016/j.lfs.2024.123360] [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: 08/17/2024] [Revised: 11/26/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Febrile seizures (FSs) are the most common pediatric neurological disorder, affecting approximately 5 % of children aged 6 months to 5 years. While most FSs are self-limiting and benign, about 20-30 % present as complex FSs (CFSs), which pose a risk of acute brain injury and the development of temporal lobe epilepsy. Various factors, including age, geographical distribution, and type of infection influence the occurrence of FS. Infection is the primary external trigger for FS, while the underlying intrinsic factors are linked to the immature and incomplete myelination of the brain during specific developmental stages. Although the precise pathogenesis of FS is not yet fully understood, it is likely caused by the interaction of immature brain development, fever, neuroinflammation, and genetic susceptibility. This review discussed the pathogenesis of febrile seizures, focusing on factors such as age, fever, neuroinflammation, genetics, and intestinal microbiota, and summarized existing therapeutic approaches. Our review may facilitate the identification of new targets for mechanistic studies and clinical treatment of febrile seizures.
Collapse
Affiliation(s)
- Chang Wu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Qingmei Wang
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Wenmi Li
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Mingxuan Han
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Huawei Zhao
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Zhenghao Xu
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
4
|
Yuan X, Li W, Yuan Y, Zhu X, Meng Y, Wu Q, Yan Q, Zhang P. Characterization of neuronal differentiation in human adipose-derived stromal cells: morphological, molecular, and ultrastructural insights. J Neurosci Methods 2024; 412:110296. [PMID: 39357604 DOI: 10.1016/j.jneumeth.2024.110296] [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: 04/26/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Adipose-derived stromal cells (ADSCs) have shown promise as a potential source of neural differentiation. In this study, we investigated the morphological, molecular and ultrastructural features of ADSCs during neuronal differentiation. METHODS ADSCs were induced in vitro and their differentiation was examined at different time points. Immunocytochemical staining was performed to detect the expression of neuron-specific markers NSE and MAP-2. Immunofluorescence double labeling and Western blot detected the co-expression of presynaptic markers (CaMKII, SynCAM1, SYN) and postsynaptic markers (PSD-95, Synapsin I). Scanning electron microscopy (SEM) was performed to detect the synaptic structural features of differentiated neurons. RESULTS ADSCs showed diverse morphological features during differentiation, gradually acquiring a neuron-like spindle shape and organized arrangement. The expression of neuron-specific markers and synaptic markers peaked at 5 h of induction. Scanning electron microscopy showed polygonal protrusions of ADSC-derived neurons, and transmission electron microscopy showed characteristic ultrastructures such as nidus, synaptic vesicle-like structures, and tight junctions. CONCLUSION Our findings suggest that ADSCs differentiated for 5 h have neuronal features, including morphological, molecular, and ultrastructural resemblance to neurons, as well as the formation of synaptic structures. These insights contribute to a better understanding of ADSC-based neuronal differentiation and pave the way for future applications in regenerative medicine and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yi Yuan
- Children's hospital of Capital institute of pediatrics, department of pediatric othopedic, Beijing 100000, China
| | - Xuhong Zhu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yan Meng
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Wu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| |
Collapse
|
5
|
Toulmé E, Salazar Lázaro A, Trimbuch T, Rizo J, Rosenmund C. Neurotransmitter release is triggered by a calcium-induced rearrangement in the Synaptotagmin-1/SNARE complex primary interface. Proc Natl Acad Sci U S A 2024; 121:e2409636121. [PMID: 39374398 PMCID: PMC11494337 DOI: 10.1073/pnas.2409636121] [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: 05/14/2024] [Accepted: 08/14/2024] [Indexed: 10/09/2024] Open
Abstract
The Ca2+ sensor synaptotagmin-1 (Syt1) triggers neurotransmitter release together with the neuronal sensitive factor attachment protein receptor (SNARE) complex formed by syntaxin-1, SNAP25, and synaptobrevin. Moreover, Syt1 increases synaptic vesicle (SV) priming and impairs spontaneous vesicle release. The Syt1 C2B domain binds to the SNARE complex through a primary interface via two regions (I and II), but how exactly this interface mediates distinct functions of Syt1 and the mechanism underlying Ca2+ triggering of release are unknown. Using mutagenesis and electrophysiological experiments, we show that region II is functionally and spatially subdivided: Binding of C2B domain arginines to SNAP-25 acidic residues at one face of region II is crucial for Ca2+-evoked release but not for vesicle priming or clamping of spontaneous release, whereas other SNAP-25 and syntaxin-1 acidic residues at the other face mediate priming and clamping of spontaneous release but not evoked release. Mutations that disrupt region I impair the priming and clamping functions of Syt1 while, strikingly, mutations that enhance binding through this region increase vesicle priming and clamping of spontaneous release, but strongly inhibit evoked release and vesicle fusogenicity. These results support previous findings that the primary interface mediates the functions of Syt1 in vesicle priming and clamping of spontaneous release and, importantly, show that Ca2+ triggering of release requires a rearrangement of the primary interface involving dissociation of region I, while region II remains bound. Together with biophysical studies presented in [K. Jaczynska et al., bioRxiv [Preprint] (2024). https://doi.org/10.1101/2024.06.17.599417 (Accessed 18 June 2024)], our data suggest a model whereby this rearrangement pulls the SNARE complex to facilitate fast SV fusion.
Collapse
Affiliation(s)
- Estelle Toulmé
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Andrea Salazar Lázaro
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| |
Collapse
|
6
|
Dey H, Perez-Hurtado M, Heidelberger R. Syntaxin 3B: A SNARE Protein Required for Vision. Int J Mol Sci 2024; 25:10665. [PMID: 39408994 PMCID: PMC11476516 DOI: 10.3390/ijms251910665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Syntaxin 3 is a member of a large protein family of syntaxin proteins that mediate fusion between vesicles and their target membranes. Mutations in the ubiquitously expressed syntaxin 3A splice form give rise to a serious gastrointestinal disorder in humans called microvillus inclusion disorder, while mutations that additionally involve syntaxin 3B, a splice form that is expressed primarily in retinal photoreceptors and bipolar cells, additionally give rise to an early onset severe retinal dystrophy. In this review, we discuss recent studies elucidating the roles of syntaxin 3B and the regulation of syntaxin 3B functionality in membrane fusion and neurotransmitter release in the vertebrate retina.
Collapse
Affiliation(s)
| | | | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (H.D.)
| |
Collapse
|
7
|
Toulmé E, Lázaro AS, Trimbuch T, Rizo J, Rosenmund C. Neurotransmitter release is triggered by a calcium-induced rearrangement in the Synaptotagmin-1/SNARE complex primary interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599435. [PMID: 38948868 PMCID: PMC11213007 DOI: 10.1101/2024.06.17.599435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Ca2+ sensor synaptotagmin-1 triggers neurotransmitter release together with the neuronal SNARE complex formed by syntaxin-1, SNAP25 and synaptobrevin. Moreover, synaptotagmin-1 increases synaptic vesicle priming and impairs spontaneous vesicle release. The synaptotagmin-1 C2B domain binds to the SNARE complex through a primary interface via two regions (I and II), but how exactly this interface mediates distinct functions of synaptotagmin-1, and the mechanism underlying Ca2+-triggering of release is unknown. Using mutagenesis and electrophysiological experiments, we show that region II is functionally and spatially subdivided: binding of C2B domain arginines to SNAP-25 acidic residues at one face of region II is crucial for Ca2+-evoked release but not for vesicle priming or clamping of spontaneous release, whereas other SNAP-25 and syntaxin-1 acidic residues at the other face mediate priming and clamping of spontaneous release but not evoked release. Mutations that disrupt region I impair the priming and clamping functions of synaptotagmin-1 while, strikingly, mutations that enhance binding through this region increase vesicle priming and clamping of spontaneous release, but strongly inhibit evoked release and vesicle fusogenicity. These results support previous findings that the primary interface mediates the functions of synaptotagmin-1 in vesicle priming and clamping of spontaneous release, and, importantly, show that Ca2+-triggering of release requires a rearrangement of the primary interface involving dissociation of region I, while region II remains bound. Together with modeling and biophysical studies presented in the accompanying paper, our data suggest a model whereby this rearrangement pulls the SNARE complex to facilitate fast synaptic vesicle fusion.
Collapse
Affiliation(s)
- Estelle Toulmé
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Andrea Salazar Lázaro
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Thorsten Trimbuch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Christian Rosenmund
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
8
|
Guiberson NGL, Black LS, Haller JE, Brukner A, Abramov D, Ahmad S, Xie YX, Sharma M, Burré J. Disease-linked mutations in Munc18-1 deplete synaptic Doc2. Brain 2024; 147:2185-2202. [PMID: 38242640 PMCID: PMC11146428 DOI: 10.1093/brain/awae019] [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: 05/31/2022] [Revised: 11/20/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Heterozygous de novo mutations in the neuronal protein Munc18-1/STXBP1 cause syndromic neurological symptoms, including severe epilepsy, intellectual disability, developmental delay, ataxia and tremor, summarized as STXBP1 encephalopathies. Although haploinsufficiency is the prevailing disease mechanism, it remains unclear how the reduction in Munc18-1 levels causes synaptic dysfunction in disease as well as how haploinsufficiency alone can account for the significant heterogeneity among patients in terms of the presence, onset and severity of different symptoms. Using biochemical and cell biological readouts on mouse brains, cultured mouse neurons and heterologous cells, we found that the synaptic Munc18-1 interactors Doc2A and Doc2B are unstable in the absence of Munc18-1 and aggregate in the presence of disease-causing Munc18-1 mutants. In haploinsufficiency-mimicking heterozygous knockout neurons, we found a reduction in Doc2A/B levels that is further aggravated by the presence of the disease-causing Munc18-1 mutation G544D as well as an impairment in Doc2A/B synaptic targeting in both genotypes. We also demonstrated that overexpression of Doc2A/B partially rescues synaptic dysfunction in heterozygous knockout neurons but not heterozygous knockout neurons expressing G544D Munc18-1. Our data demonstrate that STXBP1 encephalopathies are not only characterized by the dysfunction of Munc18-1 but also by the dysfunction of the Munc18-1 binding partners Doc2A and Doc2B, and that this dysfunction is exacerbated by the presence of a Munc18-1 missense mutant. These findings may offer a novel explanation for the significant heterogeneity in symptoms observed among STXBP1 encephalopathy patients.
Collapse
Affiliation(s)
- Noah Guy Lewis Guiberson
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Luca S Black
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jillian E Haller
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Aniv Brukner
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Debra Abramov
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Saad Ahmad
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yan Xin Xie
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Burré
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
9
|
Salazar Lázaro A, Trimbuch T, Vardar G, Rosenmund C. The stability of the primed pool of synaptic vesicles and the clamping of spontaneous neurotransmitter release rely on the integrity of the C-terminal half of the SNARE domain of syntaxin-1A. eLife 2024; 12:RP90775. [PMID: 38512129 PMCID: PMC10957171 DOI: 10.7554/elife.90775] [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] [Indexed: 03/22/2024] Open
Abstract
The SNARE proteins are central in membrane fusion and, at the synapse, neurotransmitter release. However, their involvement in the dual regulation of the synchronous release while maintaining a pool of readily releasable vesicles remains unclear. Using a chimeric approach, we performed a systematic analysis of the SNARE domain of STX1A by exchanging the whole SNARE domain or its N- or C-terminus subdomains with those of STX2. We expressed these chimeric constructs in STX1-null hippocampal mouse neurons. Exchanging the C-terminal half of STX1's SNARE domain with that of STX2 resulted in a reduced RRP accompanied by an increased release rate, while inserting the C-terminal half of STX1's SNARE domain into STX2 leads to an enhanced priming and decreased release rate. Additionally, we found that the mechanisms for clamping spontaneous, but not for Ca2+-evoked release, are particularly susceptible to changes in specific residues on the outer surface of the C-terminus of the SNARE domain of STX1A. Particularly, mutations of D231 and R232 affected the fusogenicity of the vesicles. We propose that the C-terminal half of the SNARE domain of STX1A plays a crucial role in the stabilization of the RRP as well as in the clamping of spontaneous synaptic vesicle fusion through the regulation of the energetic landscape for fusion, while it also plays a covert role in the speed and efficacy of Ca2+-evoked release.
Collapse
Affiliation(s)
- Andrea Salazar Lázaro
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Thorsten Trimbuch
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Gülçin Vardar
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Christian Rosenmund
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
- NeuroCure Excellence ClusterBerlinGermany
| |
Collapse
|
10
|
Dai Y, Peng Y, Hu W, Liu Y, Wang H. Prenatal amoxicillin exposure induces developmental toxicity in fetal mice and its characteristics. J Environ Sci (China) 2024; 137:287-301. [PMID: 37980015 DOI: 10.1016/j.jes.2023.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 11/20/2023]
Abstract
Amoxicillin, a widely used antibiotic in human and veterinary pharmaceuticals, is now considered as an "emerging contaminant" because it exists widespreadly in the environment and brings a series of adverse outcomes. Currently, systematic studies about the developmental toxicity of amoxicillin are still lacking. We explored the potential effects of amoxicillin exposure on pregnancy outcomes, maternal/fetal serum phenotypes, and fetal multiple organ development in mice, at different doses (75, 150, 300 mg/(kg·day)) during late-pregnancy, or at a dose of 300 mg/(kg·day) during different stages (mid-/late-pregnancy) and courses (single-/multi-course). Results showed that prenatal amoxicillin exposure (PAmE) had no significant influence on the body weights of dams, but it could inhibit the physical development and reduce the survival rate of fetuses, especially during the mid-pregnancy. Meanwhile, PAmE altered multiple maternal/fetal serum phenotypes, especially in fetuses. Fetal multi-organ function results showed that PAmE inhibited testicular/adrenal steroid synthesis, long bone/cartilage and hippocampal development, and enhanced ovarian steroid synthesis and hepatic glycogenesis/lipogenesis, and the order of severity might be gonad (testis, ovary) > liver > others. Further analysis found that PAmE-induced multi-organ developmental and functional alterations had differences in stages, courses and fetal gender, and the most obvious changes might be in high-dose, late-pregnancy and multi-course, but there was no typical rule of a dose-response relationship. In conclusion, this study confirmed that PAmE could cause abnormal development and multi-organ function alterations, which deepens our understanding of the risk of PAmE and provides an experimental basis for further exploration of the long-term harm.
Collapse
Affiliation(s)
- Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yu Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yi Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
11
|
Li P, Yan Z. An epigenetic mechanism of social isolation stress in adolescent female mice. Neurobiol Stress 2024; 29:100601. [PMID: 38213831 PMCID: PMC10776430 DOI: 10.1016/j.ynstr.2023.100601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Social isolation during adolescence can increase the risk of mental disorders. Epigenetic changes induced by chronic social isolation may serve as a mechanism underlying emotional disturbances. To test this, we exposed female mice to a post-weaning 6-week social isolation (SI) stress. We found the significantly increased methylation of histone H3 at lysine 9 (H3K9), a histone mark linked to gene repression, as well as the increased H3K9 methyltransferases SUV39H1 and SETDB1, in prefrontal cortex (PFC) of SI females. To find out potential downstream genes affected by this epigenetic alteration, we examined genes linked to neuronal and synaptic functions. Activity-dependent genes, including Arc, c-Fos and Npas4, were significantly reduced in PFC of SI females, correlated with the increased H3K9me2 occupancy around Arc enhancer. Treatment of SI females with UNC0642, a selective inhibitor of H3K9 methylation, significantly attenuated the anxiety-like behavior and elevated Arc expression. These results have revealed an epigenetic mechanism and intervention avenue for anxiety induced by chronic social isolation.
Collapse
Affiliation(s)
- Pei Li
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| |
Collapse
|
12
|
Peall KJ, Owen MJ, Hall J. Rare genetic brain disorders with overlapping neurological and psychiatric phenotypes. Nat Rev Neurol 2024; 20:7-21. [PMID: 38001363 DOI: 10.1038/s41582-023-00896-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
Understanding rare genetic brain disorders with overlapping neurological and psychiatric phenotypes is of increasing importance given the potential for developing disease models that could help to understand more common, polygenic disorders. However, the traditional clinical boundaries between neurology and psychiatry result in frequent segregation of these disorders into distinct silos, limiting cross-specialty understanding that could facilitate clinical and biological advances. In this Review, we highlight multiple genetic brain disorders in which neurological and psychiatric phenotypes are observed, but for which in-depth, cross-spectrum clinical phenotyping is rarely undertaken. We describe the combined phenotypes observed in association with genetic variants linked to epilepsy, dystonia, autism spectrum disorder and schizophrenia. We also consider common underlying mechanisms that centre on synaptic plasticity, including changes to synaptic and neuronal structure, calcium handling and the balance of excitatory and inhibitory neuronal activity. Further investigation is needed to better define and replicate these phenotypes in larger cohorts, which would help to gain greater understanding of the pathophysiological mechanisms and identify common therapeutic targets.
Collapse
Affiliation(s)
- Kathryn J Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Michael J Owen
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
13
|
Gheinani AH, Sack BS, Bigger-Allen A, Thaker H, Atta H, Lambrinos G, Costa K, Doyle C, Gharaee-Kermani M, Patalano S, Piper M, Cotellessa JF, Vitko D, Li H, Prabhakaran MK, Cristofaro V, Froehlich J, Lee RS, Yang W, Sullivan MP, Macoska JA, Adam RM. Integrated omics analysis unveils a DNA damage response to neurogenic injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.10.571015. [PMID: 38106029 PMCID: PMC10723451 DOI: 10.1101/2023.12.10.571015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Spinal cord injury (SCI) evokes profound bladder dysfunction. Current treatments are limited by a lack of molecular data to inform novel therapeutic avenues. Previously, we showed systemic inosine treatment improved bladder function following SCI in rats. Here, we applied multi-omics analysis to explore molecular alterations in the bladder and their sensitivity to inosine following SCI. Canonical pathways regulated by SCI included those associated with protein synthesis, neuroplasticity, wound healing, and neurotransmitter degradation. Upstream regulator analysis identified MYC as a key regulator, whereas causal network analysis predicted multiple regulators of DNA damage response signaling following injury, including PARP-1. Staining for both DNA damage (γH2AX) and PARP activity (poly-ADP-ribose) markers in the bladder was increased following SCI, and attenuated in inosine-treated tissues. Proteomics analysis suggested that SCI induced changes in protein synthesis-, neuroplasticity-, and oxidative stress-associated pathways, a subset of which were shown in transcriptomics data to be inosine-sensitive. These findings provide novel insights into the molecular landscape of the bladder following SCI, and highlight a potential role for PARP inhibition to treat neurogenic bladder dysfunction.
Collapse
Affiliation(s)
- Ali Hashemi Gheinani
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Functional Urology Research Group, Department for BioMedical Research DBMR, University of Bern, Switzerland
- Department of Urology, Inselspital University Hospital, 3010 Bern, Switzerland
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bryan S Sack
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Functional Urology Research Group, Department for BioMedical Research DBMR, University of Bern, Switzerland
| | - Alex Bigger-Allen
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Biological & Biomedical Sciences Graduate Program, Division of Medical Sciences, Harvard Medical School, Boston, MA
| | - Hatim Thaker
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Hussein Atta
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - George Lambrinos
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Kyle Costa
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
| | - Claire Doyle
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | | | - Mary Piper
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Justin F Cotellessa
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Dijana Vitko
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Haiying Li
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Manubhai Kadayil Prabhakaran
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Vivian Cristofaro
- Division of Urology, VA Boston Healthcare System, Boston, MA, USA
- University of Massachusetts, Boston, MA, USA
| | - John Froehlich
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Richard S Lee
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Wei Yang
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Maryrose P Sullivan
- Division of Urology, VA Boston Healthcare System, Boston, MA, USA
- University of Massachusetts, Boston, MA, USA
| | | | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Urology, Inselspital University Hospital, 3010 Bern, Switzerland
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
14
|
Willemse SW, Harley P, van Eijk RPA, Demaegd KC, Zelina P, Pasterkamp RJ, van Damme P, Ingre C, van Rheenen W, Veldink JH, Kiernan MC, Al-Chalabi A, van den Berg LH, Fratta P, van Es MA. UNC13A in amyotrophic lateral sclerosis: from genetic association to therapeutic target. J Neurol Neurosurg Psychiatry 2023; 94:649-656. [PMID: 36737245 PMCID: PMC10359588 DOI: 10.1136/jnnp-2022-330504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with limited treatment options and an incompletely understood pathophysiology. Although genomewide association studies (GWAS) have advanced our understanding of the disease, the precise manner in which risk polymorphisms contribute to disease pathogenesis remains unclear. Of relevance, GWAS have shown that a polymorphism (rs12608932) in the UNC13A gene is associated with risk for both ALS and frontotemporal dementia (FTD). Homozygosity for the C-allele at rs12608932 modifies the ALS phenotype, as these patients are more likely to have bulbar-onset disease, cognitive impairment and FTD at baseline as well as shorter survival. UNC13A is expressed in neuronal tissue and is involved in maintaining synaptic active zones, by enabling the priming and docking of synaptic vesicles. In the absence of functional TDP-43, risk variants in UNC13A lead to the inclusion of a cryptic exon in UNC13A messenger RNA, subsequently leading to nonsense mediated decay, with loss of functional protein. Depletion of UNC13A leads to impaired neurotransmission. Recent discoveries have identified UNC13A as a potential target for therapy development in ALS, with a confirmatory trial with lithium carbonate in UNC13A cases now underway and future approaches with antisense oligonucleotides currently under consideration. Considering UNC13A is a potent phenotypic modifier, it may also impact clinical trial outcomes. This present review describes the path from the initial discovery of UNC13A as a risk gene in ALS to the current therapeutic options being explored and how knowledge of its distinct phenotype needs to be taken into account in future trials.
Collapse
Affiliation(s)
- Sean W Willemse
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Peter Harley
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Koen C Demaegd
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Philip van Damme
- Department of Neurology, KU Leuven Hospital, Leuven, Belgium
- Laboratory of Neurobiology, VIB KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Caroline Ingre
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Matthew C Kiernan
- Bushell Chair of Neurology, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Neurology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Michael A van Es
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| |
Collapse
|
15
|
Liu Q, Hu W, Zhang Y, Ning J, Pang Y, Hu H, Chen M, Wu M, Wang M, Yang P, Bao L, Niu Y, Zhang R. Comprehensive Analysis of lncRNA-mRNA Expression Profiles in Depression-like Responses of Mice Related to Polystyrene Nanoparticle Exposure. TOXICS 2023; 11:600. [PMID: 37505566 PMCID: PMC10386552 DOI: 10.3390/toxics11070600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/29/2023]
Abstract
Plastics in the environment can break down into nanoplastics (NPs), which pose a potential threat to public health. Studies have shown that the nervous system constitutes a significant target for nanoplastics. However, the potential mechanism behind nanoplastics' neurotoxicity remains unknown. This study aimed to investigate the role of lncRNA in the depressive-like responses induced by exposure to 25 nm polystyrene nanoplastics (PS NPs). Forty mice were divided into four groups administered doses of 0, 10, 25, and 50 mg/kg via gavage for 6 months. After conducting behavioral tests, RNA sequencing was used to detect changes in mRNAs, miRNAs, and lncRNAs in the prefrontal cortex of the mice in the 0 and 50 mg/kg PS NPs groups. The results revealed that mice exposed to chronic PS NPs developed depressive-like responses in a dose-dependent manner. It was demonstrated that 987 mRNAs, 29 miRNAs, and 116 lncRNAs were significantly different between the two groups. Then, a competing endogenous RNA (ceRNA) network containing 6 lncRNAs, 18 miRNAs, and 750 mRNAs was constructed. Enrichment results suggested that PS NPs may contribute to the onset of depression-like responses through the activation of axon guidance, neurotrophin-signaling pathways, and dopaminergic synapses. This study provided evidence of the molecular relationship between PS NPs and depression-like responses.
Collapse
Affiliation(s)
- Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Wentao Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yaling Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jie Ning
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Huaifang Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Meiyu Chen
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Peihao Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Bao
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujie Niu
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
16
|
Uzay B, Kavalali ET. Genetic disorders of neurotransmitter release machinery. Front Synaptic Neurosci 2023; 15:1148957. [PMID: 37066095 PMCID: PMC10102358 DOI: 10.3389/fnsyn.2023.1148957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023] Open
Abstract
Synaptic neurotransmitter release is an evolutionarily conserved process that mediates rapid information transfer between neurons as well as several peripheral tissues. Release of neurotransmitters are ensured by successive events such as synaptic vesicle docking and priming that prepare synaptic vesicles for rapid fusion. These events are orchestrated by interaction of different presynaptic proteins and are regulated by presynaptic calcium. Recent studies have identified various mutations in different components of neurotransmitter release machinery resulting in aberrant neurotransmitter release, which underlie a wide spectrum of psychiatric and neurological symptoms. Here, we review how these genetic alterations in different components of the core neurotransmitter release machinery affect the information transfer between neurons and how aberrant synaptic release affects nervous system function.
Collapse
Affiliation(s)
- Burak Uzay
- Vanderbilt Brain Institute, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Ege T. Kavalali
- Vanderbilt Brain Institute, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
17
|
Szabó Z, Balogh M, Domonkos Á, Csányi M, Kaló P, Kiss GB. The bs5 allele of the susceptibility gene Bs5 of pepper (Capsicum annuum L.) encoding a natural deletion variant of a CYSTM protein conditions resistance to bacterial spot disease caused by Xanthomonas species. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2023; 136:64. [PMID: 36943531 PMCID: PMC10030403 DOI: 10.1007/s00122-023-04340-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/02/2023] [Indexed: 05/09/2023]
Abstract
KEY MESSAGE The bs5 resistance gene against bacterial spot was identified by map-based cloning. The recessive bs5 gene of pepper (Capsicum annuum L.) conditions a non-hypersensitive resistance trait, characterized by a slightly swollen, pale green, photosynthetically active leaf tissue, following Xanthomonas euvesicatoria infection. The isolation of the bs5 gene by map-based cloning revealed that the bs5 protein was shorter by 2 amino acids as compared to the wild type Bs5 protein. The natural 2 amino acid deletion occurred in the cysteine-rich transmembrane domain of the tail-anchored (TA) protein, Ca_CYSTM1. The protein products of the wild type Bs5 and mutant bs5 genes were shown to be located in the cell membrane, indicating an unknown function in this membrane compartment. Successful infection of the Bs5 pepper lines was abolished by the 6 bp deletion in the TM encoding domain of the Ca_CYSTM1 gene in bs5 homozygotes, suggesting, that the resulting resistance might be explained by the lack of entry of the Xanthomonas specific effector molecules into the plant cells.
Collapse
Affiliation(s)
- Zoltán Szabó
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary.
| | - Márta Balogh
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary
| | - Ágota Domonkos
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary
| | - Márta Csányi
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary
| | - Péter Kaló
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary
- Institute of Plant Biology, Biological Research Center, Eötvös Lóránd Research Network, Szeged, Hungary
| | - György B Kiss
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi A. U. 4., 2100, Gödöllő, Hungary
- AMBIS Biotechnology Research and Development Ltd., Budapest, Hungary
| |
Collapse
|
18
|
An Atypical, Staged Cell Death Pathway Induced by Depletion of SNARE-Proteins MUNC18-1 or Syntaxin-1. J Neurosci 2023; 43:347-358. [PMID: 36517239 PMCID: PMC9864589 DOI: 10.1523/jneurosci.0611-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/23/2022] Open
Abstract
The presynaptic proteins MUNC18-1, syntaxin-1, and SNAP25 drive SNARE-mediated synaptic vesicle fusion and are also required for neuronal viability. Their absence triggers rapid, cell-autonomous, neuron-specific degeneration, unrelated to synaptic vesicle deficits. The underlying cell death pathways remain poorly understood. Here, we show that hippocampi of munc18-1 null mice (unknown sex) express apoptosis hallmarks cleaved caspase 3 (CC-3) and phosphorylated p53, and have condensed nuclei. However, side-by-side in vitro comparison with classical apoptosis induced by camptothecin uncovered striking differences to syntaxin-1 and MUNC18-1 depleted neurons. First, live-cell imaging revealed consecutive neurite retraction hours before cell death in MUNC18-1 or syntaxin-1 depleted neurons, whereas all neurites retracted at once, directly before cell death in classical apoptosis. Second, CC-3 activation was observed only after loss of all neurites and cellular breakdown, whereas CC-3 is activated before any neurite loss in classical apoptosis. Third, a pan-caspase inhibitor and a p53 inhibitor both arrested classical apoptosis, as expected, but not cell death in MUNC18-1 or syntaxin-1 depleted neurons. Neuron-specific cell death, consecutive neurite retraction, and late CC-3 activation were conserved in syntaxin-1 depleted human neurons. Finally, no indications were observed for involvement of other established cell death pathways, including necroptosis, Wallerian degeneration, autophagic cell death, and pyroptosis. Together, these data show that depletion of presynaptic proteins MUNC18-1 or syntaxin-1 triggers an atypical, staged cell death pathway characterized by consecutive neurite retraction, ultimately leading to, but not driven by, apoptosis.SIGNIFICANCE STATEMENT Neuronal cell death can occur via a multitude of pathways and plays an important role in the developing nervous system as well as neurodegenerative diseases. One poorly understood pathway to neuronal cell death takes place on depletion of presynaptic SNARE proteins syntaxin-1, SNAP25, or MUNC18-1. The current study demonstrates that MUNC18-1 or syntaxin-1 depleted neurons show a new, atypical, staged cell death that does not resemble any of the established cell death pathways in neurons. Cell death on MUNC18-1 or syntaxin-1 depletion is characterized by consecutive neurite retraction, ultimately involving, but not driven by, classical apoptosis.
Collapse
|
19
|
Martinovic J, Zaric Kontic M, Dragic M, Todorovic A, Gusevac Stojanovic I, Mitrovic N, Grkovic I, Drakulic D. Chronic oral d-galactose intake provokes age-related changes in the rat prefrontal cortex. Behav Brain Res 2023; 436:114072. [DOI: 10.1016/j.bbr.2022.114072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 12/01/2022]
|
20
|
Yang X, Tu W, Gao X, Zhang Q, Guan J, Zhang J. Functional regulation of syntaxin-1: An underlying mechanism mediating exocytosis in neuroendocrine cells. Front Endocrinol (Lausanne) 2023; 14:1096365. [PMID: 36742381 PMCID: PMC9892835 DOI: 10.3389/fendo.2023.1096365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
The fusion of the secretory vesicle with the plasma membrane requires the assembly of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein complexes formed by synaptobrevin, syntaxin-1, and SNAP-25. Within the pathway leading to exocytosis, the transitions between the "open" and "closed" conformations of syntaxin-1 function as a switch for the fusion of vesicles with the plasma membranes; rapid assembly and disassembly of syntaxin-1 clusters on the plasma membrane provide docking and fusion sites for secretory vesicles in neuroendocrine cells; and the fully zippered trans-SNARE complex, which requires the orderly, rapid and accurate binding of syntaxin-1 to other SNARE proteins, play key roles in triggering fusion. All of these reactions that affect exocytosis under physiological conditions are tightly regulated by multiple factors. Here, we review the current evidence for the involvement of syntaxin-1 in the mechanism of neuroendocrine cell exocytosis, discuss the roles of multiple factors such as proteins, lipids, protein kinases, drugs, and toxins in SNARE complex-mediated membrane fusion, and present an overview of syntaxin-1 mutation-associated diseases with a view to developing novel mechanistic therapeutic targets for the treatment of neuroendocrine disorders.
Collapse
Affiliation(s)
- Xinquan Yang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Weifeng Tu
- Faculty of Anesthesioloy, Suzhou Hospital Affiliated to Medical School of Nanjing University, Suzhou, China
| | - Xuzhu Gao
- Department of Central Laboratory, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, China
| | - Qi Zhang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Jinping Guan
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Junlong Zhang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
- *Correspondence: Junlong Zhang,
| |
Collapse
|
21
|
Weber-Boyvat M, Kroll J, Trimbuch T, Olkkonen VM, Rosenmund C. The lipid transporter ORP2 regulates synaptic neurotransmitter release via two distinct mechanisms. Cell Rep 2022; 41:111882. [PMID: 36577376 DOI: 10.1016/j.celrep.2022.111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/26/2022] [Accepted: 12/02/2022] [Indexed: 12/28/2022] Open
Abstract
Cholesterol is crucial for neuronal synaptic transmission, assisting in the molecular and structural organization of lipid rafts, ion channels, and exocytic proteins. Although cholesterol absence was shown to result in impaired neurotransmission, how cholesterol locally traffics and its route of action are still under debate. Here, we characterized the lipid transfer protein ORP2 in murine hippocampal neurons. We show that ORP2 preferentially localizes to the presynapse. Loss of ORP2 reduces presynaptic cholesterol levels by 50%, coinciding with a profoundly reduced release probability, enhanced facilitation, and impaired presynaptic calcium influx. In addition, ORP2 plays a cholesterol-transport-independent role in regulating vesicle priming and spontaneous release, likely by competing with Munc18-1 in syntaxin1A binding. To conclude, we identified a dual function of ORP2 as a physiological modulator of the synaptic cholesterol content and a regulator of neuronal exocytosis.
Collapse
Affiliation(s)
- Marion Weber-Boyvat
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jana Kroll
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Thorsten Trimbuch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Christian Rosenmund
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
22
|
Li N, Wang H, Xin S, Min R, Zhang Y, Deng Y. Confinement induces oxidative damage and synaptic dysfunction in mice. Front Physiol 2022; 13:999574. [PMID: 36505063 PMCID: PMC9729776 DOI: 10.3389/fphys.2022.999574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
A confined environment is an enclosed area where entry or exit is highly restricted, which is a risk factor for a work crew's mental health. Previous studies have shown that a crew is more susceptible to developing anxiety or depression in a confined environment. However, the underlying mechanism by which negative emotion is induced by confinement is not fully understood. Hence, in this study, mice were retained in a tube to simulate short-term confinement. The mice exhibited depressive-like behavior. Additionally, the levels of H2O2 and malondialdehyde in the prefrontal cortex were significantly increased in the confinement group. Furthermore, a label-free quantitative proteomic strategy was applied to analyze the abundance of proteins in the prefrontal cortex of mice. A total of 71 proteins were considered differentially abundant proteins among 3,023 identified proteins. Two differentially abundant proteins, superoxide dismutase [Mn] and syntaxin-1A, were also validated by a parallel reaction monitoring assay. Strikingly, the differentially abundant proteins were highly enriched in the respiratory chain, oxidative phosphorylation, and the synaptic vesicle cycle, which might lead to oxidative damage and synaptic dysfunction. The results of this study provide valuable information to better understand the mechanisms of depressive-like behavior induced by confined environments.
Collapse
Affiliation(s)
- Nuomin Li
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Hao Wang
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Shuchen Xin
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Rui Min
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yongqian Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China,*Correspondence: Yongqian Zhang,
| | - Yulin Deng
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China,School of Life Science, Beijing Institute of Technology, Beijing, China,Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
23
|
Van Berkel AA, Koopmans F, Gonzalez-Lozano MA, Lammertse HCA, Feringa F, Bryois J, Sullivan PF, Smit AB, Toonen RF, Verhage M. Dysregulation of synaptic and developmental transcriptomic/proteomic profiles upon depletion of MUNC18-1. eNeuro 2022; 9:ENEURO.0186-22.2022. [PMID: 36257704 PMCID: PMC9668351 DOI: 10.1523/eneuro.0186-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Absence of presynaptic protein MUNC18-1 (gene: Stxbp1) leads to neuronal cell death at an immature stage before synapse formation. Here, we performed transcriptomic and proteomic profiling of immature Stxbp1 knockout (KO) cells to discover which cellular processes depend on MUNC18-1. Hippocampi of Stxbp1 KO mice showed cell-type specific dysregulation of 2123 transcripts primarily related to synaptic transmission and immune response. To further investigate direct, neuron-specific effects of MUNC18-1 depletion, a proteomic screen was performed on murine neuronal cultures at two developmental timepoints prior to onset of neuron degeneration. 399 proteins were differentially expressed, which were primarily involved in synaptic function (especially synaptic vesicle exocytosis) and neuron development. We further show that many of the downregulated proteins upon loss of MUNC18-1 are normally upregulated during this developmental stage. Thus, absence of MUNC18-1 extensively dysregulates the transcriptome and proteome, primarily affecting synaptic and developmental profiles. Lack of synaptic activity is unlikely to underlie these effects, as the changes were observed in immature neurons without functional synapses, and minimal overlap was found to activity-dependent proteins. We hypothesize that presence of MUNC18-1 is essential to advance neuron development, serving as a 'checkpoint' for neurons to initiate cell death in its absence.Significance StatementPresynaptic protein MUNC18-1 is essential for neuronal functioning. Pathogenic variants in its gene, STXBP1, are among the most common found in patients with developmental delay and epilepsy. To discern the pathogenesis in these patients, a thorough understanding of MUNC18-1's function in neurons is required. Here, we show that loss of MUNC18-1 results in extensive dysregulation of synaptic and developmental proteins in immature neurons before synapse formation. Many of the downregulated proteins are normally upregulated during this developmental stage. This indicates that MUNC18-1 is a critical regulator of neuronal development, which could play an important role in the pathogenesis of STXBP1 variant carriers.
Collapse
Affiliation(s)
- A A Van Berkel
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - F Koopmans
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - M A Gonzalez-Lozano
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - H C A Lammertse
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - F Feringa
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - J Bryois
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Nobels vag 12A, 171 77 Stockholm, Sweden
| | - P F Sullivan
- UNC Center for Psychiatric Genomics, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599-7160, USA
- Karolinska Institutet, Department of Medical Epidemiology and Biostatistics, Nobels vag 12A, 171 77 Stockholm, Sweden
| | - A B Smit
- Dept. Molecular & Cellular Neurobiology, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - R F Toonen
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - M Verhage
- Dept. Functional Genomics, CNCR, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Functional Genomics, Department of Human Genetics, CNCR, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
24
|
Özdemir Ç, Şahin N, Edgünlü T. Vesicle trafficking with snares: a perspective for autism. Mol Biol Rep 2022; 49:12193-12202. [PMID: 36198849 DOI: 10.1007/s11033-022-07970-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022]
Abstract
Vesicle-mediated membrane traffic is the mechanism fundamental to many biological events, especially the release of neurotransmitters. The main proteins of the mechanism that mediates membrane fusion in vesicle-mediated membrane traffic are N-ethylmaleimide sensitive factor (NSF) supplemental protein (SNAP) receptor (SNAREs) proteins. SNAREs are classified into vesicle-associated SNAREs (vesicle-SNAREs/v-SNAREs) and target membrane-associated SNAREs (target-SNARE/t-SNAREs). Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by many symptoms, especially complications in social communication and stereotypical behaviours. Defects in synaptogenesis and neurotransmission, oxidative stress, and developmental defects in the early stages of development are defined in the pathogenesis of the disease. SNARE proteins are on the basis of synaptogenesis and neurotransmission. Although the formation mechanisms and underlying causes of the SNARE complex are not fully understood, expression differences, polymorphisms, abnormal expressions or dysfunctions of the proteins that make up the SNARE complex have been associated with many neurodevelopmental diseases, including autism. Further understanding of SNARE mechanisms is crucial both for understanding ASD and for developing new treatments. In this review, the formation mechanisms of the SNARE complex and the roles of various factors involved in this process are explained. In addition, a brief evaluation of clinical and basic studies on the SNARE complex in autism spectrum disorders was made.
Collapse
Affiliation(s)
- Çilem Özdemir
- Department of Medical Biology, Health Sciences Institution, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Nilfer Şahin
- Department of Child and Adolescent Mental Health Diseases School of Medicine, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Tuba Edgünlü
- Department of Medical Biology, School of Medicine, Muğla Sıtkı Koçman University, 48000, Mugla, Turkey.
| |
Collapse
|
25
|
Ejdrup AL, Lycas MD, Lorenzen N, Konomi A, Herborg F, Madsen KL, Gether U. A density-based enrichment measure for assessing colocalization in single-molecule localization microscopy data. Nat Commun 2022; 13:4388. [PMID: 35902578 PMCID: PMC9334352 DOI: 10.1038/s41467-022-32064-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/15/2022] [Indexed: 11/20/2022] Open
Abstract
Dual-color single-molecule localization microscopy (SMLM) provides unprecedented possibilities for detailed studies of colocalization of different molecular species in a cell. However, the informational richness of the data is not fully exploited by current analysis tools that often reduce colocalization to a single value. Here, we describe a tool specifically designed for determination of co-localization in both 2D and 3D from SMLM data. The approach uses a function that describes the relative enrichment of one molecular species on the density distribution of a reference species. The function reframes the question of colocalization by providing a density-context relevant to multiple biological questions. Moreover, the function visualize enrichment (i.e. colocalization) directly in the images for easy interpretation. We demonstrate the approach's functionality on both simulated data and cultured neurons, and compare it to current alternative measures. The method is available in a Python function for easy and parameter-free implementation.
Collapse
Affiliation(s)
- Aske L Ejdrup
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Matthew D Lycas
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Lorenzen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ainoa Konomi
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Freja Herborg
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth L Madsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Lazarevic V, Yang Y, Paslawski W, Svenningsson P. α-Synuclein induced cholesterol lowering increases tonic and reduces depolarization-evoked synaptic vesicle recycling and glutamate release. NPJ Parkinsons Dis 2022; 8:71. [PMID: 35672421 PMCID: PMC9174203 DOI: 10.1038/s41531-022-00334-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-syn) is a key molecule linked to Parkinson's disease pathology. Physiologically, the monomeric α-syn in the presynaptic termini is involved in regulation of neurotransmission, but the pathophysiology of extracellular monomeric α-syn is still unknown. Utilizing both in vivo and in vitro approaches, we investigated how extracellular α-syn impact presynaptic structure and function. Our data revealed that treatment with exogenous α-syn leads to increased tonic and decreased depolarization-evoked synaptic vesicle (SV) recycling and glutamate release. This was associated with mobilization of molecularly distinct SV pools and reorganization of active zone components. Our study also showed that exogenous α-syn impaired neuronal cholesterol level and that the cholesterol binding domain of α-syn was sufficient to exert the same presynaptic phenotype as the full-length protein. The present study sheds new light on physiological functions of extracellular α-syn in overall maintenance of presynaptic activity that involves the reorganization of both presynaptic compartment and cholesterol-rich plasma membrane domains.
Collapse
Affiliation(s)
- Vesna Lazarevic
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Yunting Yang
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
27
|
Vardar G, Salazar-Lázaro A, Zobel S, Trimbuch T, Rosenmund C. Syntaxin-1A modulates vesicle fusion in mammalian neurons via juxtamembrane domain dependent palmitoylation of its transmembrane domain. eLife 2022; 11:78182. [PMID: 35638903 PMCID: PMC9183232 DOI: 10.7554/elife.78182] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
SNAREs are undoubtedly one of the core elements of synaptic transmission. Contrary to the well characterized function of their SNARE domains bringing the plasma and vesicular membranes together, the level of contribution of their juxtamembrane domain (JMD) and the transmembrane domain (TMD) to the vesicle fusion is still under debate. To elucidate this issue, we analyzed three groups of STX1A mutations in cultured mouse hippocampal neurons: (1) elongation of STX1A’s JMD by three amino acid insertions in the junction of SNARE-JMD or JMD-TMD; (2) charge reversal mutations in STX1A’s JMD; and (3) palmitoylation deficiency mutations in STX1A’s TMD. We found that both JMD elongations and charge reversal mutations have position-dependent differential effects on Ca2+-evoked and spontaneous neurotransmitter release. Importantly, we show that STX1A’s JMD regulates the palmitoylation of STX1A’s TMD and loss of STX1A palmitoylation either through charge reversal mutation K260E or by loss of TMD cysteines inhibits spontaneous vesicle fusion. Interestingly, the retinal ribbon specific STX3B has a glutamate in the position corresponding to the K260E mutation in STX1A and mutating it with E259K acts as a molecular on-switch. Furthermore, palmitoylation of post-synaptic STX3A can be induced by the exchange of its JMD with STX1A’s JMD together with the incorporation of two cysteines into its TMD. Forced palmitoylation of STX3A dramatically enhances spontaneous vesicle fusion suggesting that STX1A regulates spontaneous release through two distinct mechanisms: one through the C-terminal half of its SNARE domain and the other through the palmitoylation of its TMD.
Collapse
Affiliation(s)
- Gülçin Vardar
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Salazar-Lázaro
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sina Zobel
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thorsten Trimbuch
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Rosenmund
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
28
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
29
|
S327 phosphorylation of the presynaptic protein SEPTIN5 increases in the early stages of neurofibrillary pathology and alters the functionality of SEPTIN5. Neurobiol Dis 2022; 163:105603. [DOI: 10.1016/j.nbd.2021.105603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/02/2021] [Accepted: 12/22/2021] [Indexed: 12/25/2022] Open
|
30
|
Barone E. Brain insulin resistance: an early risk factor for Alzheimer's disease development in Down syndrome. Neural Regen Res 2022; 17:333-335. [PMID: 34269205 PMCID: PMC8463986 DOI: 10.4103/1673-5374.317979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro, Roma, Italy
| |
Collapse
|
31
|
Lamb R, Dhar B, Cherra SJ. PXF-1 promotes synapse development at the neuromuscular junction in Caenorhabditis elegans. Front Mol Neurosci 2022; 15:945680. [PMID: 36311020 PMCID: PMC9606220 DOI: 10.3389/fnmol.2022.945680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/27/2022] [Indexed: 11/30/2022] Open
Abstract
Guanine nucleotide exchange factors (GEFs) are a family of proteins that modulate small G protein signaling. Mutations in a subfamily of GEFs that act on Rap, known as RapGEFs, have been associated with neurological disorders, and knockout mice display impairments in neuronal activity. However, the precise functions of RapGEFs in the nervous system remain unclear. Here, we have used the Caenorhabditis elegans neuromuscular junction, to investigate how the RapGEF homolog, PXF-1, regulates synaptic function. We found that loss of function mutations in pxf-1 reduced cholinergic activity at the neuromuscular junction. We observed that PXF-1 is expressed in the nervous system, and its expression in neurons is sufficient to promote synaptic activity. In pxf-1 mutant animals, there is a reduction in the levels of synaptic vesicles in cholinergic motor neurons but no change in the overall synapse numbers. In addition to synaptic vesicles proteins, we also found that filamentous actin, a scaffold for nascent synapses, was reduced at developing cholinergic synapses in pxf-1 mutant animals. Our studies indicate that PXF-1 regulates neuromuscular function by promoting the formation of actin filaments to support the development of motor neuron synapses.
Collapse
Affiliation(s)
- Reagan Lamb
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Bithika Dhar
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Salvatore J Cherra
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| |
Collapse
|
32
|
Vardar G, Salazar-Lázaro A, Brockmann M, Weber-Boyvat M, Zobel S, Kumbol VWA, Trimbuch T, Rosenmund C. Reexamination of N-terminal domains of syntaxin-1 in vesicle fusion from central murine synapses. eLife 2021; 10:69498. [PMID: 34427183 PMCID: PMC8416022 DOI: 10.7554/elife.69498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Syntaxin-1 (STX1) and Munc18-1 are two requisite components of synaptic vesicular release machinery, so much so synaptic transmission cannot proceed in their absence. They form a tight complex through two major binding modes: through STX1’s N-peptide and through STX1’s closed conformation driven by its Habc- domain. However, physiological roles of these two reportedly different binding modes in synapses are still controversial. Here we characterized the roles of STX1’s N-peptide, Habc-domain, and open conformation with and without N-peptide deletion using our STX1-null mouse model system and exogenous reintroduction of STX1A mutants. We show, on the contrary to the general view, that the Habc-domain is absolutely required and N-peptide is dispensable for synaptic transmission. However, STX1A’s N-peptide plays a regulatory role, particularly in the Ca2+-sensitivity and the short-term plasticity of vesicular release, whereas STX1’s open conformation governs the vesicle fusogenicity. Strikingly, we also show neurotransmitter release still proceeds when the two interaction modes between STX1A and Munc18-1 are presumably intervened, necessitating a refinement of the conceptualization of STX1A–Munc18-1 interaction.
Collapse
Affiliation(s)
- Gülçin Vardar
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Andrea Salazar-Lázaro
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Marisa Brockmann
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Marion Weber-Boyvat
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Sina Zobel
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | | | - Thorsten Trimbuch
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Christian Rosenmund
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
33
|
Extreme Glycemic Fluctuations Debilitate NRG1, ErbB Receptors and Olig1 Function: Association with Regeneration, Cognition and Mood Alterations During Diabetes. Mol Neurobiol 2021; 58:4727-4744. [PMID: 34165684 DOI: 10.1007/s12035-021-02455-1] [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: 04/15/2021] [Accepted: 06/16/2021] [Indexed: 12/28/2022]
Abstract
Neuronal regeneration is crucial for maintaining intact neural interactions for perpetuation of cognitive and emotional functioning. The NRG1-ErbB receptor signaling is a key pathway for regeneration in adult brain and also associated with learning and mood stabilization by modulating synaptic transmission. Extreme glycemic stress is known to affect NRG1-ErbB-mediated regeneration in brain; yet, it remains unclear how the ErbB receptor subtypes are differentially affected due to such metabolic variations. Here, we assessed the alterations in NRG1, ErbB receptor subtypes to study the regenerative potential, both in rodents as well as in neuronal and glial cell models of hyperglycemia and hypoglycemic insults during hyperglycemia. The pro-oxidant and anti-oxidant status leading to degenerative changes in brain regions were determined. The spatial memory and anxiogenic behaviour of experimental rodents were tested using 'T' maze and Elevated Plus Maze. Our data revealed that the extreme glycemic discrepancies during diabetes and recurrent hypoglycemia lead to altered expression of NRG1, ErbB receptor subtypes, Syntaxin1 and Olig1 that shows association with impaired regeneration, synaptic dysfunction, demyelination, cognitive deficits and anxiety.
Collapse
|
34
|
Tracking Single Molecule Dynamics in the Adult Drosophila Brain. eNeuro 2021; 8:ENEURO.0057-21.2021. [PMID: 33875453 PMCID: PMC8174007 DOI: 10.1523/eneuro.0057-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
Super-resolution microscopy provides valuable insight for understanding the nanoscale organization within living tissue, although this method is typically restricted to cultured or dissociated cells. Here, we develop a method to track the mobility of individual proteins in ex vivo adult Drosophila melanogaster brains, focusing on a key component of the presynaptic release machinery, syntaxin1A (Sx1a). We show that individual Sx1a dynamics can be reliably tracked within neurons in the whole fly brain, and that the mobility of Sx1a molecules increases following conditional neural stimulation. We then apply this preparation to the problem of general anesthesia, to address how different anesthetics might affect single molecule dynamics in intact brain synapses. We find that propofol, etomidate, and isoflurane significantly impair Sx1a mobility, while ketamine and sevoflurane have little effect. Resolving single molecule dynamics in intact fly brains provides a novel approach to link localized molecular effects with systems-level phenomena such as general anesthesia.
Collapse
|
35
|
Janecke AR, Liu X, Adam R, Punuru S, Viestenz A, Strauß V, Laass M, Sanchez E, Adachi R, Schatz MP, Saboo US, Mittal N, Rohrschneider K, Escher J, Ganesh A, Al Zuhaibi S, Al Murshedi F, AlSaleem B, Alfadhel M, Al Sinani S, Alkuraya FS, Huber LA, Müller T, Heidelberger R, Janz R. Pathogenic STX3 variants affecting the retinal and intestinal transcripts cause an early-onset severe retinal dystrophy in microvillus inclusion disease subjects. Hum Genet 2021; 140:1143-1156. [PMID: 33974130 PMCID: PMC8263458 DOI: 10.1007/s00439-021-02284-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/15/2021] [Indexed: 01/12/2023]
Abstract
Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic-intestinal and retinal-disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.
Collapse
Affiliation(s)
- Andreas R Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Xiaoqin Liu
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
| | - Rüdiger Adam
- University Children's Hospital, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Sumanth Punuru
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
| | - Arne Viestenz
- Department of Ophthalmology, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Valeria Strauß
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Halle, Halle, Germany
| | - Martin Laass
- Klinik und Poliklinik f. Kinder- u. Jugendmedizin, University of Dresden, Dresden, Germany
| | - Elizabeth Sanchez
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Adachi
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martha P Schatz
- Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ujwala S Saboo
- Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Naveen Mittal
- Department of Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Johanna Escher
- Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Anuradha Ganesh
- Department of Ophthalmology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sana Al Zuhaibi
- Department of Ophthalmology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Fathiya Al Murshedi
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| | - Badr AlSaleem
- King Fahad Medical City, Children's Specialized Hospital, Riyadh, Saudi Arabia
| | - Majid Alfadhel
- Genetics Division and Medical Genomic Research Lab, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Siham Al Sinani
- Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA.
| | - Roger Janz
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
- Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Moro A, van Nifterick A, Toonen RF, Verhage M. Dynamin controls neuropeptide secretion by organizing dense-core vesicle fusion sites. SCIENCE ADVANCES 2021; 7:eabf0659. [PMID: 34020952 PMCID: PMC8139595 DOI: 10.1126/sciadv.abf0659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/02/2021] [Indexed: 05/13/2023]
Abstract
Synaptic vesicles (SVs) release neurotransmitters at specialized active zones, but release sites and organizing principles for the other major secretory pathway, neuropeptide/neuromodulator release from dense-core vesicles (DCVs), remain elusive. We identify dynamins, yeast Vps1 orthologs, as DCV fusion site organizers in mammalian neurons. Genetic or pharmacological inactivation of all three dynamins strongly impaired DCV exocytosis, while SV exocytosis remained unaffected. Wild-type dynamin restored normal exocytosis but not guanosine triphosphatase-deficient or membrane-binding mutants that cause neurodevelopmental syndromes. During prolonged stimulation, repeated use of the same DCV fusion location was impaired in dynamin 1-3 triple knockout neurons. The syntaxin-1 staining efficiency, but not its expression level, was reduced. αSNAP (α-soluble N-ethylmaleimide-sensitive factor attachment protein) expression restored this. We conclude that mammalian dynamins organize DCV fusion sites, downstream of αSNAP, by regulating the equilibrium between fusogenic and non-fusogenic syntaxin-1 promoting its availability for SNARE (SNAP receptor) complex formation and DCV exocytosis.
Collapse
Affiliation(s)
- Alessandro Moro
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1087, 1081 HV Amsterdam, Netherlands
| | - Anne van Nifterick
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1087, 1081 HV Amsterdam, Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1087, 1081 HV Amsterdam, Netherlands.
| | - Matthijs Verhage
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands.
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, de Boelelaan 1087, 1081 HV Amsterdam, Netherlands
| |
Collapse
|
37
|
Liu C, Hu Q, Chen Y, Wu L, Liu X, Liang D. Behavioral and Gene Expression Analysis of Stxbp6-Knockout Mice. Brain Sci 2021; 11:brainsci11040436. [PMID: 33805317 PMCID: PMC8066043 DOI: 10.3390/brainsci11040436] [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: 02/08/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
Since the first report that Stxbp6, a brain-enriched protein, regulates the assembly of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, little has been discovered about its functions over the past two decades. To determine the effects of Stxbp6 loss on nervous-system-associated phenotypes and underlying mechanisms, we constructed a global Stxbp6-knockout mouse. We found that Stxbp6-null mice survive normally, with normal behavior, but gained less weight relative to age- and sex-matched wildtype mice. RNA-seq analysis of the cerebral cortex of Stxbp6-null mice relative to wildtype controls identified 126 differentially expressed genes. Of these, 57 were upregulated and 69 were downregulated. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the most significant enriched KEGG term was “complement and coagulation cascades”. Our results suggest some potential regulatory pathways of Stxbp6 in the central nervous system, providing a remarkable new resource for understanding Stxbp6 function at the organism level.
Collapse
|
38
|
Tawfik B, Martins JS, Houy S, Imig C, Pinheiro PS, Wojcik SM, Brose N, Cooper BH, Sørensen JB. Synaptotagmin-7 places dense-core vesicles at the cell membrane to promote Munc13-2- and Ca 2+-dependent priming. eLife 2021; 10:64527. [PMID: 33749593 PMCID: PMC8012061 DOI: 10.7554/elife.64527] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
Synaptotagmins confer calcium-dependence to the exocytosis of secretory vesicles, but how coexpressed synaptotagmins interact remains unclear. We find that synaptotagmin-1 and synaptotagmin-7 when present alone act as standalone fast and slow Ca2+-sensors for vesicle fusion in mouse chromaffin cells. When present together, synaptotagmin-1 and synaptotagmin-7 are found in largely non-overlapping clusters on dense-core vesicles. Synaptotagmin-7 stimulates Ca2+-dependent vesicle priming and inhibits depriming, and it promotes ubMunc13-2- and phorbolester-dependent priming, especially at low resting calcium concentrations. The priming effect of synaptotagmin-7 increases the number of vesicles fusing via synaptotagmin-1, while negatively affecting their fusion speed, indicating both synergistic and competitive interactions between synaptotagmins. Synaptotagmin-7 places vesicles in close membrane apposition (<6 nm); without it, vesicles accumulate out of reach of the fusion complex (20-40 nm). We suggest that a synaptotagmin-7-dependent movement toward the membrane is involved in Munc13-2/phorbolester/Ca2+-dependent priming as a prelude to fast and slow exocytosis triggering.
Collapse
Affiliation(s)
- Bassam Tawfik
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Joana S Martins
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Sébastien Houy
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Paulo S Pinheiro
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | | |
Collapse
|
39
|
Lanzillotta C, Tramutola A, Di Giacomo G, Marini F, Butterfield DA, Di Domenico F, Perluigi M, Barone E. Insulin resistance, oxidative stress and mitochondrial defects in Ts65dn mice brain: A harmful synergistic path in down syndrome. Free Radic Biol Med 2021; 165:152-170. [PMID: 33516914 DOI: 10.1016/j.freeradbiomed.2021.01.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of brain insulin signaling with reduced downstream neuronal survival and plasticity mechanisms are fundamental abnormalities observed in Alzheimer disease (AD). This phenomenon, known as brain insulin resistance, is associated with poor cognitive performance and is driven by the inhibition of IRS1. Since Down syndrome (DS) and AD neuropathology share many common features, we investigated metabolic aspects of neurodegeneration in DS and whether they contribute to early onset AD in DS. We evaluated levels and activation of proteins belonging to the insulin signaling pathway (IR, IRS1, BVR-A, MAPK, PTEN, Akt, GSK3β, PKCζ, AS160, GLUT4) in the frontal cortex of Ts65dn (DS model) (n = 5-6/group) and euploid mice (n = 6/group) at different ages (1, 3, 9 and 18 months). Furthermore, we analyzed whether changes of brain insulin signaling were associated with alterations of: (i) proteins regulating brain energy metabolism (mitochondrial complexes, hexokinase-II, Sirt1); (ii) oxidative stress (OS) markers (iii) APP cleavage; and (iv) proteins mediating synaptic plasticity mechanisms (PSD95, syntaxin-1 and BDNF). Ts65dn mice showed an overall impairment of the above-mentioned pathways, mainly characterized by defects of proteins activation state. Such alterations start early in life (at 1 month, during brain maturation). In particular, accumulation of inhibited IRS1, together with the uncoupling among the proteins downstream from IRS1 (brain insulin resistance), characterize Ts65dn mice. Furthermore, reduced levels of mitochondrial complexes and Sirt1, as well as increased indices of OS also were observed. These alterations precede the accumulation of APP-C99 in Ts65dn mice. Tellingly, oxidative stress levels were negatively associated with IR, IRS1 and AS160 activation as well as mitochondrial complexes levels in Ts65dn mice, suggesting a role for oxidative stress in the observed alterations. We propose that a close link exists among brain insulin resistance, mitochondrial defects and OS that contributes to brain dysfunctions observed in DS, likely favoring the development of AD in DS.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Graziella Di Giacomo
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy.
| |
Collapse
|
40
|
Abramov D, Guiberson NGL, Daab A, Na Y, Petsko GA, Sharma M, Burré J. Targeted stabilization of Munc18-1 function via pharmacological chaperones. EMBO Mol Med 2021; 13:e12354. [PMID: 33332765 PMCID: PMC7799358 DOI: 10.15252/emmm.202012354] [Citation(s) in RCA: 8] [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: 03/17/2020] [Revised: 11/01/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
Heterozygous de novo mutations in the neuronal protein Munc18-1 cause syndromic neurological symptoms, including severe epilepsy, intellectual disability, developmental delay, ataxia, and tremor. No disease-modifying therapy exists to treat these disorders, and while chemical chaperones have been shown to alleviate neuronal dysfunction caused by missense mutations in Munc18-1, their required high concentrations and potential toxicity necessitate a Munc18-1-targeted therapy. Munc18-1 is essential for neurotransmitter release, and mutations in Munc18-1 have been shown to cause neuronal dysfunction via aggregation and co-aggregation of the wild-type protein, reducing functional Munc18-1 levels well below hemizygous levels. Here, we identify two pharmacological chaperones via structure-based drug design, that bind to wild-type and mutant Munc18-1, and revert Munc18-1 aggregation and neuronal dysfunction in vitro and in vivo, providing the first targeted treatment strategy for these severe pediatric encephalopathies.
Collapse
Affiliation(s)
- Debra Abramov
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | - Noah Guy Lewis Guiberson
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | - Andrew Daab
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
- Present address:
University of BathBathUK
| | - Yoonmi Na
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | - Gregory A Petsko
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
- Present address:
Ann Romney Center for Neurologic DiseasesDepartment of NeurologyBrigham and Women’s Hospital and Harvard Medical SchoolBostonMA, USA
| | - Manu Sharma
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | - Jacqueline Burré
- Appel Institute for Alzheimer’s Disease ResearchBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
41
|
Kunii M, Noguchi Y, Yoshimura SI, Kanda S, Iwano T, Avriyanti E, Atik N, Sato T, Sato K, Ogawa M, Harada A. SNAP23 deficiency causes severe brain dysplasia through the loss of radial glial cell polarity. J Cell Biol 2021; 220:e201910080. [PMID: 33332551 PMCID: PMC7754684 DOI: 10.1083/jcb.201910080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 08/23/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
In the developing brain, the polarity of neural progenitor cells, termed radial glial cells (RGCs), is important for neurogenesis. Intercellular adhesions, termed apical junctional complexes (AJCs), at the apical surface between RGCs are necessary for cell polarization. However, the mechanism by which AJCs are established remains unclear. Here, we show that a SNARE complex composed of SNAP23, VAMP8, and Syntaxin1B has crucial roles in AJC formation and RGC polarization. Central nervous system (CNS)-specific ablation of SNAP23 (NcKO) results in mice with severe hypoplasia of the neocortex and no hippocampus or cerebellum. In the developing NcKO brain, RGCs lose their polarity following the disruption of AJCs and exhibit reduced proliferation, increased differentiation, and increased apoptosis. SNAP23 and its partner SNAREs, VAMP8 and Syntaxin1B, are important for the localization of an AJC protein, N-cadherin, to the apical plasma membrane of RGCs. Altogether, SNARE-mediated localization of N-cadherin is essential for AJC formation and RGC polarization during brain development.
Collapse
Affiliation(s)
- Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yuria Noguchi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Satoshi Kanda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomohiko Iwano
- Department of Anatomy and Cell Biology, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Erda Avriyanti
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Dermatology and Venereology, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Nur Atik
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Biomedical Sciences, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Takashi Sato
- Laboratory of Developmental Biology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | | | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| |
Collapse
|
42
|
Combined cellomics and proteomics analysis reveals shared neuronal morphology and molecular pathway phenotypes for multiple schizophrenia risk genes. Mol Psychiatry 2021; 26:784-799. [PMID: 31142819 PMCID: PMC7910218 DOI: 10.1038/s41380-019-0436-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/05/2019] [Accepted: 04/17/2019] [Indexed: 01/23/2023]
Abstract
An enigma in studies of neuropsychiatric disorders is how to translate polygenic risk into disease biology. For schizophrenia, where > 145 significant GWAS loci have been identified and only a few genes directly implicated, addressing this issue is a particular challenge. We used a combined cellomics and proteomics approach to show that polygenic risk can be disentangled by searching for shared neuronal morphology and cellular pathway phenotypes of candidate schizophrenia risk genes. We first performed an automated high-content cellular screen to characterize neuronal morphology phenotypes of 41 candidate schizophrenia risk genes. The transcription factors Tcf4 and Tbr1 and the RNA topoisomerase Top3b shared a neuronal phenotype marked by an early and progressive reduction in synapse numbers upon knockdown in mouse primary neuronal cultures. Proteomics analysis subsequently showed that these three genes converge onto the syntaxin-mediated neurotransmitter release pathway, which was previously implicated in schizophrenia, but for which genetic evidence was weak. We show that dysregulation of multiple proteins in this pathway may be due to the combined effects of schizophrenia risk genes Tcf4, Tbr1, and Top3b. Together, our data provide new biological functions for schizophrenia risk genes and support the idea that polygenic risk is the result of multiple small impacts on common neuronal signaling pathways.
Collapse
|
43
|
van Berkel AA, Santos TC, Shaweis H, van Weering JRT, Toonen RF, Verhage M. Loss of MUNC18-1 leads to retrograde transport defects in neurons. J Neurochem 2020; 157:450-466. [PMID: 33259669 PMCID: PMC8247427 DOI: 10.1111/jnc.15256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
Loss of the exocytic Sec1/MUNC18 protein MUNC18-1 or its target-SNARE partners SNAP25 and syntaxin-1 results in rapid, cell-autonomous and unexplained neurodegeneration, which is independent of their known role in synaptic vesicle exocytosis. cis-Golgi abnormalities are the earliest cellular phenotypes before degeneration occurs. Here, we investigated whether loss of MUNC18-1 causes defects in intracellular membrane transport pathways in primary murine neurons that may explain neurodegeneration. Electron, confocal and super resolution microscopy confirmed that loss of MUNC18-1 expression results in a smaller cis-Golgi. In addition, we now show that medial-Golgi and the trans-Golgi Network are also affected. However, stacking and cisternae ultrastructure of the Golgi were normal. Overall, ultrastructure of null mutant neurons was remarkably normal just hours before cell death occurred. By synchronizing protein trafficking by conditional cargo retention in the endoplasmic reticulum using selective hooks (RUSH) and immunocytochemistry, we show that anterograde Endoplasmic Reticulum-to-Golgi and Golgi exit of endogenous and exogenous proteins were normal. In contrast, loss of MUNC18-1 caused reduced retrograde Cholera Toxin B-subunit transport from the plasma membrane to the Golgi. In addition, MUNC18-1-deficiency resulted in abnormalities in retrograde TrkB trafficking in an antibody uptake assay. We conclude that MUNC18-1 deficient neurons have normal anterograde but reduced retrograde transport to the Golgi. The impairments in retrograde pathways suggest a role of MUNC18-1 in endosomal SNARE-dependent fusion and provide a plausible explanation for the observed Golgi abnormalities and cell death in MUNC18-1 deficient neurons.
Collapse
Affiliation(s)
- Annemiek A van Berkel
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center Amsterdam, Amsterdam, The Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
| | - Tatiana C Santos
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
| | - Hesho Shaweis
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
| | - Jan R T van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center Amsterdam, Amsterdam, The Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Melland H, Carr EM, Gordon SL. Disorders of synaptic vesicle fusion machinery. J Neurochem 2020; 157:130-164. [PMID: 32916768 DOI: 10.1111/jnc.15181] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022]
Abstract
The revolution in genetic technology has ushered in a new age for our understanding of the underlying causes of neurodevelopmental, neuromuscular and neurodegenerative disorders, revealing that the presynaptic machinery governing synaptic vesicle fusion is compromised in many of these neurological disorders. This builds upon decades of research showing that disturbance to neurotransmitter release via toxins can cause acute neurological dysfunction. In this review, we focus on disorders of synaptic vesicle fusion caused either by toxic insult to the presynapse or alterations to genes encoding the key proteins that control and regulate fusion: the SNARE proteins (synaptobrevin, syntaxin-1 and SNAP-25), Munc18, Munc13, synaptotagmin, complexin, CSPα, α-synuclein, PRRT2 and tomosyn. We discuss the roles of these proteins and the cellular and molecular mechanisms underpinning neurological deficits in these disorders.
Collapse
Affiliation(s)
- Holly Melland
- The Florey Institute of Neuroscience and Mental Health, Melbourne Dementia Research Centre, The University of Melbourne, Melbourne, Vic., Australia
| | - Elysa M Carr
- The Florey Institute of Neuroscience and Mental Health, Melbourne Dementia Research Centre, The University of Melbourne, Melbourne, Vic., Australia
| | - Sarah L Gordon
- The Florey Institute of Neuroscience and Mental Health, Melbourne Dementia Research Centre, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
45
|
Tang BL. SNAREs and developmental disorders. J Cell Physiol 2020; 236:2482-2504. [PMID: 32959907 DOI: 10.1002/jcp.30067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family mediate membrane fusion processes associated with vesicular trafficking and autophagy. SNAREs mediate core membrane fusion processes essential for all cells, but some SNAREs serve cell/tissue type-specific exocytic/endocytic functions, and are therefore critical for various aspects of embryonic development. Mutations or variants of their encoding genes could give rise to developmental disorders, such as those affecting the nervous system and immune system in humans. Mutations to components in the canonical synaptic vesicle fusion SNARE complex (VAMP2, STX1A/B, and SNAP25) and a key regulator of SNARE complex formation MUNC18-1, produce variant phenotypes of autism, intellectual disability, movement disorders, and epilepsy. STX11 and MUNC18-2 mutations underlie 2 subtypes of familial hemophagocytic lymphohistiocytosis. STX3 mutations contribute to variant microvillus inclusion disease. Chromosomal microdeletions involving STX16 play a role in pseudohypoparathyroidism type IB associated with abnormal imprinting of the GNAS complex locus. In this short review, I discuss these and other SNARE gene mutations and variants that are known to be associated with a variety developmental disorders, with a focus on their underlying cellular and molecular pathological basis deciphered through disease modeling. Possible pathogenic potentials of other SNAREs whose variants could be disease predisposing are also speculated upon.
Collapse
Affiliation(s)
- Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
46
|
ORP/Osh mediate cross-talk between ER-plasma membrane contact site components and plasma membrane SNAREs. Cell Mol Life Sci 2020; 78:1689-1708. [PMID: 32734583 PMCID: PMC7904734 DOI: 10.1007/s00018-020-03604-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
OSBP-homologous proteins (ORPs, Oshp) are lipid binding/transfer proteins. Several ORP/Oshp localize to membrane contacts between the endoplasmic reticulum (ER) and the plasma membrane, where they mediate lipid transfer or regulate lipid-modifying enzymes. A common way in which they target contacts is by binding to the ER proteins, VAP/Scs2p, while the second membrane is targeted by other interactions with lipids or proteins.We have studied the cross-talk of secretory SNARE proteins and their regulators with ORP/Oshp and VAPA/Scs2p at ER-plasma membrane contact sites in yeast and murine primary neurons. We show that Oshp-Scs2p interactions depend on intact secretory SNARE proteins, especially Sec9p. SNAP-25/Sec9p directly interact with ORP/Osh proteins and their disruption destabilized the ORP/Osh proteins, associated with dysfunction of VAPA/Scs2p. Deleting OSH1-3 in yeast or knocking down ORP2 in primary neurons reduced the oligomerization of VAPA/Scs2p and affected their multiple interactions with SNAREs. These observations reveal a novel cross-talk between the machineries of ER-plasma membrane contact sites and those driving exocytosis.
Collapse
|
47
|
Hoerder-Suabedissen A, Korrell KV, Hayashi S, Jeans A, Ramirez DMO, Grant E, Christian HC, Kavalali ET, Wilson MC, Molnár Z. Cell-Specific Loss of SNAP25 from Cortical Projection Neurons Allows Normal Development but Causes Subsequent Neurodegeneration. Cereb Cortex 2020; 29:2148-2159. [PMID: 29850799 DOI: 10.1093/cercor/bhy127] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Indexed: 11/13/2022] Open
Abstract
Synaptosomal associated protein 25 kDa (SNAP25) is an essential component of the SNARE complex regulating synaptic vesicle fusion. SNAP25 deficiency has been implicated in a variety of cognitive disorders. We ablated SNAP25 from selected neuronal populations by generating a transgenic mouse (B6-Snap25tm3mcw (Snap25-flox)) with LoxP sites flanking exon5a/5b. In the presence of Cre-recombinase, Snap25-flox is recombined to a truncated transcript. Evoked synaptic vesicle release is severely reduced in Snap25 conditional knockout (cKO) neurons as shown by live cell imaging of synaptic vesicle fusion and whole cell patch clamp recordings in cultured hippocampal neurons. We studied Snap25 cKO in subsets of cortical projection neurons in vivo (L5-Rbp4-Cre; L6-Ntsr1-Cre; L6b-Drd1a-Cre). cKO neurons develop normal axonal projections, but axons are not maintained appropriately, showing signs of swelling, fragmentation and eventually complete absence. Onset and progression of degeneration are dependent on the neuron type, with L5 cells showing the earliest and most severe axonal loss. Ultrastructural examination revealed that cKO neurites contain autophagosome/lysosome-like structures. Markers of inflammation such as Iba1 and lipofuscin are increased only in adult cKO cortex. Snap25 cKO can provide a model to study genetic interactions with environmental influences in several disorders.
Collapse
Affiliation(s)
- Anna Hoerder-Suabedissen
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Kim V Korrell
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Shuichi Hayashi
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | | | - Denise M O Ramirez
- Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, USA
| | - Eleanor Grant
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Helen C Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Ege T Kavalali
- Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, USA.,Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, USA
| | - Michael C Wilson
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, USA
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| |
Collapse
|
48
|
Vardar G, Gerth F, Schmitt XJ, Rautenstrauch P, Trimbuch T, Schubert J, Lerche H, Rosenmund C, Freund C. Epilepsy-causing STX1B mutations translate altered protein functions into distinct phenotypes in mouse neurons. Brain 2020; 143:2119-2138. [DOI: 10.1093/brain/awaa151] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/05/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
AbstractSyntaxin 1B (STX1B) is a core component of the N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex that is critical for the exocytosis of synaptic vesicles in the presynapse. SNARE-mediated vesicle fusion is assisted by Munc18-1, which recruits STX1B in the auto-inhibited conformation, while Munc13 catalyses the fast and efficient pairing of helices during SNARE complex formation. Mutations within the STX1B gene are associated with epilepsy. Here we analysed three STX1B mutations by biochemical and electrophysiological means. These three paradigmatic mutations cause epilepsy syndromes of different severity, from benign fever-associated seizures in childhood to severe epileptic encephalopathies. An insertion/deletion (K45/RMCIE, L46M) mutation (STX1BInDel), causing mild epilepsy and located in the early helical Habc domain, leads to an unfolded protein unable to sustain neurotransmission. STX1BG226R, causing epileptic encephalopathies, strongly compromises the interaction with Munc18-1 and reduces expression of both proteins, the size of the readily releasable pool of vesicles, and Ca2+-triggered neurotransmitter release when expressed in STX1-null neurons. The mutation STX1BV216E, also causing epileptic encephalopathies, only slightly diminishes Munc18-1 and Munc13 interactions, but leads to enhanced fusogenicity and increased vesicular release probability, also in STX1-null neurons. Even though the synaptic output remained unchanged in excitatory hippocampal STX1B+/− neurons exogenously expressing STX1B mutants, the manifestation of clear and distinct molecular disease mechanisms by these mutants suggest that certain forms of epilepsies can be conceptualized by assigning mutations to structurally sensitive regions of the STX1B−Munc18-1 interface, translating into distinct neurophysiological phenotypes.
Collapse
Affiliation(s)
- Gülçin Vardar
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Fabian Gerth
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Xiao Jakob Schmitt
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Pia Rautenstrauch
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Julian Schubert
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | | | - Christian Freund
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| |
Collapse
|
49
|
Verhage M, Sørensen JB. SNAREopathies: Diversity in Mechanisms and Symptoms. Neuron 2020; 107:22-37. [PMID: 32559416 DOI: 10.1016/j.neuron.2020.05.036] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/29/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Neuronal SNAREs and their key regulators together drive synaptic vesicle exocytosis and synaptic transmission as a single integrated membrane fusion machine. Human pathogenic mutations have now been reported for all eight core components, but patients are diagnosed with very different neurodevelopmental syndromes. We propose to unify these syndromes, based on etiology and mechanism, as "SNAREopathies." Here, we review the strikingly diverse clinical phenomenology and disease severity and the also remarkably diverse genetic mechanisms. We argue that disease severity generally scales with functional redundancy and, conversely, that the large effect of mutations in some SNARE genes is the price paid for extensive integration and exceptional specialization. Finally, we discuss how subtle differences in components being rate limiting in different types of neurons helps to explain the main symptoms.
Collapse
Affiliation(s)
- Matthijs Verhage
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands; Department of Clinical Genetics, UMC Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands.
| | - Jakob B Sørensen
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|
50
|
Kajihara R, Numakawa T, Odaka H, Yaginuma Y, Fusaki N, Okumiya T, Furuya H, Inui S, Era T. Novel Drug Candidates Improve Ganglioside Accumulation and Neural Dysfunction in GM1 Gangliosidosis Models with Autophagy Activation. Stem Cell Reports 2020; 14:909-923. [PMID: 32302553 PMCID: PMC7220856 DOI: 10.1016/j.stemcr.2020.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022] Open
Abstract
GM1 gangliosidosis is a lysosomal storage disease caused by loss of lysosomal β-galactosidase activity and characterized by progressive neurodegeneration due to massive accumulation of GM1 ganglioside in the brain. Here, we generated induced pluripotent stem cells (iPSCs) derived from patients with GM1 gangliosidosis, and the resultant neurons showed impaired neurotransmitter release as a presynaptic function and accumulation of GM1 ganglioside. Treatment of normal neurons with GM1 ganglioside also disturbed presynaptic function. A high-content drug-screening system was then established and identified two compounds as drug candidates for GM1 gangliosidosis. Treatment of the patient-derived neurons with the candidate agents activated autophagy pathways, reducing GM1 ganglioside accumulation in vitro and in vivo, and restoring the presynaptic dysfunction. Our findings thus demonstrated the potential value of patient-derived iPSC lines as cellular models of GM1 gangliosidosis and revealed two potential therapeutic agents for future clinical application. GM1 ganglioside accumulation affects presynaptic function in normal neurons Presynaptic function is impaired in neurons from GM1 gangliosidosis iPSCs The drugs candidates are identified using GM1 gangliosidosis iPSCs The drug candidates activate both autophagy and sphingolipid degradation
Collapse
Affiliation(s)
- Ryutaro Kajihara
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Haruki Odaka
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Yuji Yaginuma
- Department of Morphological and Physiological Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Noemi Fusaki
- University Research Administration Center, Office of Research Promotion, Tohoku University, Tohoku 980-8577, Japan
| | - Toshika Okumiya
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Hirokazu Furuya
- Department of Neurology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Seiji Inui
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|