1
|
Cabana VC, Lussier MP. RNF13 variants L311S and L312P associated with developmental epileptic encephalopathy alter dendritic organization in hippocampal neurons. IBRO Neurosci Rep 2025; 18:559-573. [PMID: 40276023 PMCID: PMC12018061 DOI: 10.1016/j.ibneur.2025.04.004] [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: 12/23/2024] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Developmental and epileptic encephalopathy (DEE) is a group of rare and serious neurological disorders where seizures exacerbate developmental impairment. Recently, genetic mutations in the RNF13 gene were reported to cause DEE73. Specifically, two leucines from the ubiquitin E3 ligase RNF13 are converted to serine or proline (L311S and L312P). These mutations are located within a dileucine motif, which impairs RNF13's capacity to interact with AP-3. A second motif allows RNF13 to interact with AP-1 when the dileucine sorting motif is altered. The present study demonstrates that RNF13 variants L311S and L312P are trafficked through an AP-1-dependent pathway in HeLa cells. In cultures of primary rat hippocampal neurons, the protein level of the variants is significantly higher in dendrites than for wild-type protein. L311S and L312P variants alter dendritic components similarly to an RNF13 AP-3-defective binding variant or a dominant negative for RNF13's ubiquitin ligase activity. Compared to non-transfected neurons, the variants change the distribution of EEA1-positive early endosomes throughout the dendrites. While the WT alters the distribution of lysosomes (Lamp1-positive) in dendrites, the variants only decrease their presence in proximal dendrites. Unlike the variants, RNF13 WT increases the abundance of PSD-95 in distal dendrites. Interestingly, only the variants with altered dileucine motifs decrease the total number of postsynaptic inhibitory protein Gephyrin puncta. This study reports that genetic variants L311S and L312P mainly act as a dominant negative protein. This research provides valuable insights into the dendritic defects that occur when DEE73-associated genetic variants of RNF13 are present.
Collapse
Affiliation(s)
- Valérie C. Cabana
- Department of Chemistry, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- CERMO-FC - The Center of Excellence in Research on Ophan Diseases - Fondation Courtois, Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- PROTEO - The Quebec Network for Research on Protein Function, Engineering and Applications, Montréal, QC H3C 3P8, Canada
| | - Marc P. Lussier
- Department of Chemistry, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- CERMO-FC - The Center of Excellence in Research on Ophan Diseases - Fondation Courtois, Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- PROTEO - The Quebec Network for Research on Protein Function, Engineering and Applications, Montréal, QC H3C 3P8, Canada
| |
Collapse
|
2
|
Wang Q, Li S, Wu W, Zhou W, Yan K, Liu Z, Yan L, Zheng B, Zhang F, Jiang X, Ye Y, Wang H. RNF13 protects neurons against ischemia-reperfusion injury via stabilizing p62-mediated Nrf2/HO-1 signaling pathway. Cell Commun Signal 2024; 22:535. [PMID: 39511649 PMCID: PMC11542339 DOI: 10.1186/s12964-024-01905-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Cerebral ischemia/reperfusion injury (CIRI), a common, universal clinical problem that costs a large proportion of the economic and disease burden. Identifying the key regulators of cerebral I/R injury could provide potential strategies for clinically improving the prognosis of stroke. Ring finger protein 13 (RNF13) has been proven to be involved in the inflammatory response. Here, we aimed to identify the role of RNF13 in cerebral I/R injury and further reveal its immanent mechanisms. METHODS The CRISPR/Cas9 based knockout mice, RNA sequencing, mass spectrometry, co-immunoprecipitation, GST-pull down, immunofluorescent staining, western blot, RT-PCR were used to investigate biodistribution, function and mechanism of RNF13 during cerebral I/R injury. RESULTS In the present study, we found that RNF13 was significantly up-regulated in patients, mice and primary neurons after I/R injury. Deficiency of RNF13 aggravated I/R-induced neurological impairment, inflammatory response and apoptosis while overexpression of RNF13 inhibited I/R injury. Mechanistically, this inhibitory effect of RNF13 during I/R injury was confirmed to be dependent on the blocking of TRIM21-mediated autophagy-dependent degradation of p62 and the stabilization of the p62-mediated Nrf2/HO-1 signaling pathway. CONCLUSION RNF13 is a crucial regulator of cerebral I/R injury that plays its role in inhibiting cell apoptosis and inflammatory response by preventing the autophagy-medicated degradation of the p62/Nrf2/HO-1 signaling pathway via blocking the interaction of TRIM21-p62 complex. Therefore, RNF13 represents a potential pharmacological target in acute ischemia stroke therapy.
Collapse
Affiliation(s)
- Qiangping Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Shuang Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wenke Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, China
| | - Kaixuan Yan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Zhen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Lanlan Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Baoping Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Fangcheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China.
| | - Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277# Jiefang Avenue, Wuhan, Hubei, 430022, China.
| |
Collapse
|
3
|
Cabana VC, Sénécal AM, Bouchard AY, Kourrich S, Cappadocia L, Lussier MP. AP-1 contributes to endosomal targeting of the ubiquitin ligase RNF13 via a secondary and novel non-canonical binding motif. J Cell Sci 2024; 137:jcs262035. [PMID: 39206621 DOI: 10.1242/jcs.262035] [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: 02/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cellular trafficking between organelles is typically assured by short motifs that contact carrier proteins to transport them to their destination. The ubiquitin E3 ligase RING finger protein 13 (RNF13), a regulator of proliferation, apoptosis and protein trafficking, localizes to endolysosomal compartments through the binding of a dileucine motif to clathrin adaptor protein complex AP-3. Mutations within this motif reduce the ability of RNF13 to interact with AP-3. Here, our study shows the discovery of a glutamine-based motif that resembles a tyrosine-based motif within the C-terminal region of RNF13 that binds to the clathrin adaptor protein complex AP-1, notably without a functional interaction with AP-3. Using biochemical, molecular and cellular approaches in HeLa cells, our study demonstrates that a RNF13 dileucine variant uses an AP-1-dependent pathway to be exported from the Golgi towards the endosomal compartment. Overall, this study provides mechanistic insights into the alternate route used by this variant of the dileucine sorting motif of RNF13.
Collapse
Affiliation(s)
- Valérie C Cabana
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Audrey M Sénécal
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Antoine Y Bouchard
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Saïd Kourrich
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Laurent Cappadocia
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Marc P Lussier
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| |
Collapse
|
4
|
Liu W, Wang Y, Liu S, Zhang X, Cao X, Jiang M. E3 Ubiquitin Ligase RNF13 Suppresses TLR Lysosomal Degradation by Promoting LAMP-1 Proteasomal Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309560. [PMID: 39031743 PMCID: PMC11348240 DOI: 10.1002/advs.202309560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/27/2024] [Indexed: 07/22/2024]
Abstract
As a highly organized system, endo-lysosomes play a crucial role in maintaining immune homeostasis. However, the mechanisms involved in regulating endo-lysosome progression and subsequent inflammatory responses are not fully understood. By screening 103 E3 ubiquitin ligases in regulating endo-lysosomal acidification, it is discovered that lysosomal RNF13 inhibits lysosome maturation and promotes inflammatory responses mediated by endosomal Toll-like receptors (TLRs) in macrophages. Mechanistically, RNF13 mediates K48-linked polyubiquitination of LAMP-1 at residue K128 for proteasomal degradation. Upon TLRs activation, LAMP-1 promotes lysosomes maturation, which accelerates lysosomal degradation of TLRs and reduces TLR signaling in macrophages. Furthermore, peripheral blood mononuclear cells (PBMCs) from patients with rheumatoid arthritis (RA) show increased RNF13 levels and decreased LAMP-1 expression. Accordingly, the immunosuppressive agent hydroxychloroquine (HCQ) can increase the polyubiquitination of RNF13. Taken together, the study establishes a linkage between proteasomal and lysosomal degradation mechanisms for the induction of appropriate innate immune response, and offers a promising approach for the treatment of inflammatory diseases by targeting intracellular TLRs.
Collapse
Affiliation(s)
- Wei Liu
- Department of ImmunologyCenter for ImmunotherapyInstitute of Basic Medical SciencesPeking Union Medical CollegeChinese Academy of Medical SciencesBeijing100005China
- Department of RheumatologyBeijing HospitalNational Center of GerontologyInstitute of Geriatric MedicineChinese Academy of Medical SciencesBeijing100730China
| | - Yuyang Wang
- Department of ImmunologyCenter for ImmunotherapyInstitute of Basic Medical SciencesPeking Union Medical CollegeChinese Academy of Medical SciencesBeijing100005China
| | - Shuo Liu
- Department of ImmunologyCenter for ImmunotherapyInstitute of Basic Medical SciencesPeking Union Medical CollegeChinese Academy of Medical SciencesBeijing100005China
| | - Xuan Zhang
- Department of RheumatologyBeijing HospitalNational Center of GerontologyInstitute of Geriatric MedicineChinese Academy of Medical SciencesBeijing100730China
| | - Xuetao Cao
- Department of ImmunologyCenter for ImmunotherapyInstitute of Basic Medical SciencesPeking Union Medical CollegeChinese Academy of Medical SciencesBeijing100005China
| | - Minghong Jiang
- Department of ImmunologyCenter for ImmunotherapyInstitute of Basic Medical SciencesPeking Union Medical CollegeChinese Academy of Medical SciencesBeijing100005China
| |
Collapse
|
5
|
Yang XA, Wang Y, Gong M, Zhao Z, Lv F, Zhang X, Li Y. RNF149 negatively regulates LPS/TLR4 signal transduction by ubiquitination-mediated CD63 degradation. Heliyon 2024; 10:e34350. [PMID: 39104473 PMCID: PMC11298846 DOI: 10.1016/j.heliyon.2024.e34350] [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: 01/10/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
This study aims to investigate the role of RNF149 and tetraspanin CD63 in lipopolysaccharide/Toll-like receptor 4 (LPS/TLR4) signal transduction. TNF-α was assessed using enzyme-linked immunosorbent assay. The distribution of TLR4 was examined through flow cytometry after CD63 knockdown. Real-time polymerase chain reaction was used to analyze the expression of the target genes RNF149 and CD63 under different conditions. Western blotting was employed to detect gene expression, while immunoprecipitation and confocal microscopy were used to evaluate protein interactions. Transcriptome array data from stimulated monocytes (GSE7547) was obtained from GEO and subjected to bioinformatic analysis. It is suggested that CD63 may serve as a substrate of RNF149, with RNF149 capable of directly interacting with CD63. RNF149 degrades CD63 through covalent modification of CD63 at lysine 29 of the ubiquitin monomer, leading to the formation of a multiubiquitin chain. Both RNF149 and CD63 interact with TLR4, with CD63 promoting LPS/TLR4 signaling and RNF149 inhibits it. CD63 does not impact the distribution of TLR4 on the cell surface and does not directly interact with TIRAP, IRAK4, or TRAF6, but does interact with Myd88.RNF149 plays a negative regulatory role in LPS/TLR4 signal transduction by mediating ubiquitination-induced CD63 degradation.
Collapse
Affiliation(s)
- Xiu-An Yang
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde 067000, China
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde 067000, China
| | - Yingying Wang
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde 067000, China
| | - Mingyu Gong
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde 067000, China
| | - Zicheng Zhao
- Department of Biomedical Engineering, Chengde Medical University, Chengde 067000, China
| | - Fengchun Lv
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde 067000, China
| | - Xiaoyu Zhang
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde 067000, China
- Graduate School of Chengde Medical University, 067000 Chengde, China
| | - Yan Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
6
|
Pollegioni L, Campanini B, Good JM, Motta Z, Murtas G, Buoli Comani V, Pavlidou DC, Mercier N, Mittaz-Crettol L, Sacchi S, Marchesani F. L-serine deficiency: on the properties of the Asn133Ser variant of human phosphoserine phosphatase. Sci Rep 2024; 14:12463. [PMID: 38816452 PMCID: PMC11139964 DOI: 10.1038/s41598-024-63164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024] Open
Abstract
The non-essential amino acid L-serine is involved in a number of metabolic pathways and in the brain its level is largely due to the biosynthesis from the glycolytic intermediate D-3-phosphoglycerate by the phosphorylated pathway (PP). This cytosolic pathway is made by three enzymes proposed to generate a reversible metabolon named the "serinosome". Phosphoserine phosphatase (PSP) catalyses the last and irreversible step, representing the driving force pushing L-serine synthesis. Genetic defects of the PP enzymes result in strong neurological phenotypes. Recently, we identified the homozygous missense variant [NM_004577.4: c.398A > G p.(Asn133Ser)] in the PSPH, the PSP encoding gene, in two siblings with a neurodevelopmental syndrome and a myelopathy. The recombinant Asn133Ser enzyme does not show significant alterations in protein conformation and dimeric oligomerization state, as well as in enzymatic activity and functionality of the reconstructed PP. However, the Asn133Ser variant is less stable than wild-type PSP, a feature also apparent at cellular level. Studies on patients' fibroblasts also highlight a strong decrease in the level of the enzymes of the PP, a partial nuclear and perinuclear localization of variant PSP and a stronger perinuclear aggregates formation. We propose that these alterations contribute to the formation of a dysfunctional serinosome and thus to the observed reduction of L-serine, glycine and D-serine levels (the latter playing a crucial role in modulating NMDA receptors). The characterization of patients harbouring the Asn133Ser PSP substitution allows to go deep into the molecular mechanisms related to L-serine deficit and to suggest treatments to cope with the observed amino acids alterations.
Collapse
Affiliation(s)
- Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, via J.H. Dunant 3, 21100, Varese, Italy.
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Jean-Marc Good
- Division of Genetic Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Zoraide Motta
- Department of Biotechnology and Life Sciences, University of Insubria, via J.H. Dunant 3, 21100, Varese, Italy
| | - Giulia Murtas
- Department of Biotechnology and Life Sciences, University of Insubria, via J.H. Dunant 3, 21100, Varese, Italy
| | | | - Despina-Christina Pavlidou
- Division of Genetic Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Noëlle Mercier
- Department of Epileptology, Institution of Lavigny, Lavigny, Switzerland
| | - Laureane Mittaz-Crettol
- Division of Genetic Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Silvia Sacchi
- Department of Biotechnology and Life Sciences, University of Insubria, via J.H. Dunant 3, 21100, Varese, Italy
| | | |
Collapse
|
7
|
Poliquin S, Nwosu G, Randhave K, Shen W, Flamm C, Kang JQ. Modulating Endoplasmic Reticulum Chaperones and Mutant Protein Degradation in GABRG2(Q390X) Associated with Genetic Epilepsy with Febrile Seizures Plus and Dravet Syndrome. Int J Mol Sci 2024; 25:4601. [PMID: 38731820 PMCID: PMC11083348 DOI: 10.3390/ijms25094601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
A significant number of patients with genetic epilepsy do not obtain seizure freedom, despite developments in new antiseizure drugs, suggesting a need for novel therapeutic approaches. Many genetic epilepsies are associated with misfolded mutant proteins, including GABRG2(Q390X)-associated Dravet syndrome, which we have previously shown to result in intracellular accumulation of mutant GABAA receptor γ2(Q390X) subunit protein. Thus, a potentially promising therapeutic approach is modulation of proteostasis, such as increasing endoplasmic reticulum (ER)-associated degradation (ERAD). To that end, we have here identified an ERAD-associated E3 ubiquitin ligase, HRD1, among other ubiquitin ligases, as a strong modulator of wildtype and mutant γ2 subunit expression. Overexpressing HRD1 or knockdown of HRD1 dose-dependently reduced the γ2(Q390X) subunit. Additionally, we show that zonisamide (ZNS)-an antiseizure drug reported to upregulate HRD1-reduces seizures in the Gabrg2+/Q390X mouse. We propose that a possible mechanism for this effect is a partial rescue of surface trafficking of GABAA receptors, which are otherwise sequestered in the ER due to the dominant-negative effect of the γ2(Q390X) subunit. Furthermore, this partial rescue was not due to changes in ER chaperones BiP and calnexin, as total expression of these chaperones was unchanged in γ2(Q390X) models. Our results here suggest that leveraging the endogenous ERAD pathway may present a potential method to degrade neurotoxic mutant proteins like the γ2(Q390X) subunit. We also demonstrate a pharmacological means of regulating proteostasis, as ZNS alters protein trafficking, providing further support for the use of proteostasis regulators for the treatment of genetic epilepsies.
Collapse
Affiliation(s)
- Sarah Poliquin
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN 37232, USA;
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
| | - Gerald Nwosu
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Karishma Randhave
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Wangzhen Shen
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Carson Flamm
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Jing-Qiong Kang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Kennedy Center of Human Development, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
8
|
Kim P. Understanding the Unfolded Protein Response (UPR) Pathway: Insights into Neuropsychiatric Disorders and Therapeutic Potentials. Biomol Ther (Seoul) 2024; 32:183-191. [PMID: 38410073 PMCID: PMC10902702 DOI: 10.4062/biomolther.2023.181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 02/28/2024] Open
Abstract
The Unfolded Protein Response (UPR) serves as a critical cellular mechanism dedicated to maintaining protein homeostasis, primarily within the endoplasmic reticulum (ER). This pathway diligently responds to a variety of intracellular indicators of ER stress with the objective of reinstating balance by diminishing the accumulation of unfolded proteins, amplifying the ER's folding capacity, and eliminating slow-folding proteins. Prolonged ER stress and UPR irregularities have been linked to a range of neuropsychiatric disorders, including major depressive disorder, bipolar disorder, and schizophrenia. This review offers a comprehensive overview of the UPR pathway, delineating its activation mechanisms and its role in the pathophysiology of neuropsychiatric disorders. It highlights the intricate interplay within the UPR and its profound influence on brain function, synaptic perturbations, and neural developmental processes. Additionally, it explores evolving therapeutic strategies targeting the UPR within the context of these disorders, underscoring the necessity for precision and further research to effective treatments. The research findings presented in this work underscore the promising potential of UPR-focused therapeutic approaches to address the complex landscape of neuropsychiatric disorders, giving rise to optimism for improving outcomes for individuals facing these complex conditions.
Collapse
Affiliation(s)
- Pitna Kim
- Department of Cell, Developmental, and Integrative Biology (CDIB), University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
9
|
Taylor A, Kashyape PS, Jain R, El Naofal M, Tayoun AA. Heterozygous gain of function variants in a critical region of RNF13 cause congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive. Am J Med Genet A 2023; 191:2723-2727. [PMID: 37668308 DOI: 10.1002/ajmg.a.63390] [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: 01/09/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Missense variants in the RNF13 gene have been previously known to cause congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive through a gain-of-function disease mechanism. Here, we identify a nonsense variant, expected to result in protein truncation, in a similarly affected patient. We show that this nonsense variant, residing in the terminal exon, is likely to escape nonsense-mediated decay while removing a critical region for protein function, thus resulting in a gain-of-function effect. We review the literature and disease databases and identify several other affected individuals with overlapping phenotypes carrying distinct truncating variants in the terminal exon upstream of the putative critical region. Furthermore, we analyze truncating variants from the general population, namely, the Genome Aggregation Database (gnomAD), and provide additional evidence supporting our hypothesis, and ruling out haploinsufficiency as an alternative disease mechanism. In summary, our case report, literature review, and analysis of disease and population databases strongly support the hypothesis that heterozygous gain-of-function variants in a critical region of RNF13 cause congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive.
Collapse
Affiliation(s)
- Alan Taylor
- Al Jalila Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Pawan S Kashyape
- Neurology Department, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Ruchi Jain
- Al Jalila Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Maha El Naofal
- Al Jalila Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Ahmad Abou Tayoun
- Al Jalila Genomics Center of Excellence, Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
- Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
10
|
Khani M, Nafissi S, Shamshiri H, Moazzeni H, Taheri H, Elahi E. Identification of RNF13 as cause of recessively inherited ALS in a multi-case pedigree. J Med Genet 2022; 60:jmedgenet-2022-108645. [PMID: 35879052 DOI: 10.1136/jmg-2022-108645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/05/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease. The approximately 50 known ALS-associated genes do not fully explain its heritability, which suggests the existence of yet unidentified causative genes. We report results of studies aimed at identification of the genetic cause of ALS in a pedigree (three patients) without mutations in the common ALS-causative genes. METHODS Clinical investigations included thorough neurological and non-neurological examinations and testings. Genetic analysis was performed by exome sequencing. Functional studies included identification of altered splicing by PCR and sequencing, and mutated proteins by western blot analysis. Apoptosis in the presence and absence of tunicamycin was assessed in transfected HEK293T cells using an Annexin-PE-7AAD kit in conjunction with flow cytometry. RESULTS Clinical features are described in detail. Disease progression in the patients of the pedigree was relatively slow and survival was relatively long. An RNF13 mutation was identified as the cause of the recessively inherited ALS in the pedigree. The gene is highly conserved, and its encoded protein (RING finger protein 13) can potentially affect various neurodegenerative-relevant functions, including protein homeostasis. The RNF13 splice site mutation caused expression of two mis-spliced forms of RNF13 mRNA and an aberrant RNF13 protein, and affected apoptosis. CONCLUSION RNF13 was identified as a novel causative gene of recessively inherited ALS. The gene affects protein homeostasis, which is one of most important components of the pathology of neurodegeneration. The contribution of RNF13 to the aetiology of another neurodegenerative disease is discussed.
Collapse
Affiliation(s)
- Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Shahriar Nafissi
- Department of Neurology, Tehran University of Medical Sciences, Tehran, The Islamic Republic of Iran
| | - Hosein Shamshiri
- Department of Neurology, Tehran University of Medical Sciences, Tehran, The Islamic Republic of Iran
| | - Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Hanieh Taheri
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| |
Collapse
|
11
|
Emerging roles of endoplasmic reticulum proteostasis in brain development. Cells Dev 2022; 170:203781. [DOI: 10.1016/j.cdev.2022.203781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/12/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022]
|
12
|
Disruption of the Ubiquitin-Proteasome System and Elevated Endoplasmic Reticulum Stress in Epilepsy. Biomedicines 2022; 10:biomedicines10030647. [PMID: 35327449 PMCID: PMC8945847 DOI: 10.3390/biomedicines10030647] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
The epilepsies are a broad group of conditions characterized by repeated seizures, and together are one of the most common neurological disorders. Additionally, epilepsy is comorbid with many neurological disorders, including lysosomal storage diseases, syndromic intellectual disability, and autism spectrum disorder. Despite the prevalence, treatments are still unsatisfactory: approximately 30% of epileptic patients do not adequately respond to existing therapeutics, which primarily target ion channels. Therefore, new therapeutic approaches are needed. Disturbed proteostasis is an emerging mechanism in epilepsy, with profound effects on neuronal health and function. Proteostasis, the dynamic balance of protein synthesis and degradation, can be directly disrupted by epilepsy-associated mutations in various components of the ubiquitin-proteasome system (UPS), or impairments can be secondary to seizure activity or misfolded proteins. Endoplasmic reticulum (ER) stress can arise from failed proteostasis and result in neuronal death. In light of this, several treatment modalities that modify components of proteostasis have shown promise in the management of neurological disorders. These include chemical chaperones to assist proper folding of proteins, inhibitors of overly active protein degradation, and enhancers of endogenous proteolytic pathways, such as the UPS. This review summarizes recent work on the pathomechanisms of abnormal protein folding and degradation in epilepsy, as well as treatment developments targeting this area.
Collapse
|
13
|
From Drosophila to Human: Biological Function of E3 Ligase Godzilla and Its Role in Disease. Cells 2022; 11:cells11030380. [PMID: 35159190 PMCID: PMC8834447 DOI: 10.3390/cells11030380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin–proteasome system is of fundamental importance in all fields of biology due to its impact on proteostasis and in regulating cellular processes. Ubiquitination, a type of protein post-translational modification, involves complex enzymatic machinery, such as E3 ubiquitin ligases. The E3 ligases regulate the covalent attachment of ubiquitin to a target protein and are involved in various cellular mechanisms, including the cell cycle, cell division, endoplasmic reticulum stress, and neurotransmission. Because the E3 ligases regulate so many physiological events, they are also associated with pathologic conditions, such as cancer, neurological disorders, and immune-related diseases. This review focuses specifically on the protease-associated transmembrane-containing the Really Interesting New Gene (RING) subset of E3 ligases. We describe the structure, partners, and physiological functions of the Drosophila Godzilla E3 ligase and its human homologues, RNF13, RNF167, and ZNRF4. Also, we summarize the information that has emerged during the last decade regarding the association of these E3 ligases with pathophysiological conditions, such as cancer, asthma, and rare genetic disorders. We conclude by highlighting the limitations of the current knowledge and pinpointing the unresolved questions relevant to RNF13, RNF167, and ZNRF4 ubiquitin ligases.
Collapse
|
14
|
Cabana VC, Bouchard AY, Sénécal AM, Ghilarducci K, Kourrich S, Cappadocia L, Lussier MP. RNF13 Dileucine Motif Variants L311S and L312P Interfere with Endosomal Localization and AP-3 Complex Association. Cells 2021; 10:cells10113063. [PMID: 34831286 PMCID: PMC8620429 DOI: 10.3390/cells10113063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEE) are rare and serious neurological disorders characterized by severe epilepsy with refractory seizures and a significant developmental delay. Recently, DEE73 was linked to genetic alterations of the RNF13 gene, which convert positions 311 or 312 in the RNF13 protein from leucine to serine or proline, respectively (L311S and L312P). Using a fluorescence microscopy approach to investigate the molecular and cellular mechanisms affected by RNF13 protein variants, the current study shows that wild-type RNF13 localizes extensively with endosomes and lysosomes, while L311S and L312P do not extensively colocalize with the lysosomal marker Lamp1. Our results show that RNF13 L311S and L312P proteins affect the size of endosomal vesicles along with the temporal and spatial progression of fluorescently labeled epidermal growth factor, but not transferrin, in the endolysosomal system. Furthermore, GST-pulldown and co-immunoprecipitation show that RNF13 variants disrupt association with AP-3 complex. Knockdown of AP-3 complex subunit AP3D1 alters the lysosomal localization of wild-type RNF13 and similarly affects the size of endosomal vesicles. Importantly, our study provides a first step toward understanding the cellular and molecular mechanism altered by DEE73-associated genetic variations of RNF13.
Collapse
Affiliation(s)
- Valérie C. Cabana
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Antoine Y. Bouchard
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Audrey M. Sénécal
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Kim Ghilarducci
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Saïd Kourrich
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laurent Cappadocia
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Marc P. Lussier
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
- Correspondence: ; Tel.: +1-(514)-987-3000 (ext. 5591); Fax: +1-(514)-987-4054
| |
Collapse
|
15
|
Tenorio-Castaño JA, Arias P, Fernández-Jaén A, Lay-Son G, Bueno-Lozano G, Bayat A, Faivre L, Gallego N, Ramos S, Butler KM, Morel C, Hadjiyannakis S, Lespinasse J, Tran-Mau-Them F, Santos-Simarro F, Pinson L, Martínez-Monseny AF, O'Callaghan Cord MDM, Álvarez S, Stolerman ES, Washington C, Ramos FJ, The S O G R I Consortium, Lapunzina P. Tenorio syndrome: Description of 14 novel cases and review of the clinical and molecular features. Clin Genet 2021; 100:405-411. [PMID: 34196401 DOI: 10.1111/cge.14020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 11/28/2022]
Abstract
Tenorio syndrome (TNORS) (OMIM #616260) is a relatively recent disorder with very few cases described so far. Clinical features included macrocephaly, intellectual disability, hypotonia, enlarged ventricles and autoimmune diseases. Molecular underlying mechanism demonstrated missense variants and a large deletion encompassing RNF125, a gene that encodes for an U3 ubiquitin ligase protein. Since the initial description of the disorder in six patients from four families, several new patients were diagnosed, adding more evidence to the clinical spectrum. In this article, we described 14 additional cases with deep phenotyping and make an overall review of all the cases with pathogenic variants in RNF125. Not all patients presented with overgrowth, but instead, most patients showed a common pattern of neurodevelopmental disease, macrocephaly and/or large forehead. Segregation analysis showed that, though the variant was inherited in some patients from an apparently asymptomatic parent, deep phenotyping suggested a mild form of the disease in some of them. The mechanism underlying the development of this disease is not well understood yet and the report of further cases will help to a better understanding and clinical characterization of the syndrome.
Collapse
Affiliation(s)
- Jair Antonio Tenorio-Castaño
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Ithaca, European Reference Network, Brussels, Belgium
| | - Pedro Arias
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | | | - Guillermo Lay-Son
- Unidad de Genética, División de Pediatría, Facultad de Medicina, Pontificia Universidad Católica de, Chile
| | - Gloria Bueno-Lozano
- Unit of Clinical Genetics, Service of Paediatrics, School of Medicine, University Hospital 'Lozano Blesa, University of Zaragoza, CIBERER-GCV02 and ISS-Aragón, Zaragoza, Spain
| | - Allan Bayat
- Department of Pediatrics, Hvidovre Hospital, University of Copenhagen, Denmark
| | - Laurence Faivre
- Ithaca, European Reference Network, Brussels, Belgium.,Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France.,UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | - Natalia Gallego
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Ithaca, European Reference Network, Brussels, Belgium
| | - Sergio Ramos
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Kameryn M Butler
- Cytogenetics Laboratory, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Chantal Morel
- University Health Network, Fred A. Litwin Family Centre in Genetic Medicine, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stasia Hadjiyannakis
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - James Lespinasse
- Service de Cytogenetique, Centre Hospitalier de Chambéry, Chambéry, France
| | - Frederic Tran-Mau-Them
- UF6254 Innovation en Diagnostic Genomique des Maladies Rares Bat, Pôle de Biologie, CHU, Dijon, France
| | - Fernando Santos-Simarro
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Ithaca, European Reference Network, Brussels, Belgium.,Clinical Genetics section, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Lucile Pinson
- Départment de Génétique Médicale, Maladies Rares et Médecine Personnalisée, CHU de Montpellier, Montpellier, France
| | - Antonio Federico Martínez-Monseny
- Clinical Genetics section, Department of Genetic and Molecular Medicine and Pediatric Institute of Rare Diseases (IPER), Hospital Sant Joan de Déu, Barcelona, Spain.,Department of Pediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - María Del Mar O'Callaghan Cord
- Clinical Genetics section, Department of Genetic and Molecular Medicine and Pediatric Institute of Rare Diseases (IPER), Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sara Álvarez
- NIMGENETICS, Calle de Anabel Segura, Madrid, Spain
| | - Elliot S Stolerman
- Cytogenetics Laboratory, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Camerun Washington
- Cytogenetics Laboratory, Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Feliciano J Ramos
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Unit of Clinical Genetics, Service of Paediatrics, School of Medicine, University Hospital 'Lozano Blesa, University of Zaragoza, CIBERER-GCV02 and ISS-Aragón, Zaragoza, Spain.,Unit of Pediatric Endocrinology, Service of Paediatrics, University Hospital Lozano Blesa, Zaragoza, Spain
| | - The S O G R I Consortium
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Ithaca, European Reference Network, Brussels, Belgium
| |
Collapse
|
16
|
Abstract
Developmental and epileptic encephalopathies (DEEs) can be primarily attributed to genetic causes. The genetic landscape of DEEs has been largely shaped by the rise of high-throughput sequencing, which led to the discovery of new DEE-associated genes and helped identify de novo pathogenic variants. We discuss briefly the contribution of de novo variants to DEE and also focus on alternative inheritance models that contribute to DEE. First, autosomal recessive inheritance in outbred populations may have a larger contribution than previously appreciated, accounting for up to 13% of DEEs. A small subset of genes that typically harbor de novo variants have been associated with recessive inheritance, and often these individuals have more severe clinical presentations. Additionally, pathogenic variants in X-linked genes have been identified in both affected males and females, possibly due to a lack of X-chromosome inactivation skewing. Collectively, exome sequencing has resulted in a molecular diagnosis for many individuals with DEE, but this still leaves many cases unsolved. Multiple factors contribute to the missing etiology, including nonexonic variants, mosaicism, epigenetics, and oligogenic inheritance. Here, we focus on the first 2 factors. We discuss the promises and challenges of genome sequencing, which allows for a more comprehensive analysis of the genome, including interpretation of structural and noncoding variants and also yields a high number of de novo variants for interpretation. We also consider the contribution of genetic mosaicism, both what it means for a molecular diagnosis in mosaic individuals and the important implications for genetic counseling.
Collapse
Affiliation(s)
- Hannah C Happ
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gemma L Carvill
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|