1
|
Cuevas-Rios G, Assale TA, Wissfeld J, Bungartz A, Hofmann J, Langmann T, Neumann H. Decreased sialylation elicits complement-related microglia response and bipolar cell loss in the mouse retina. Glia 2024; 72:2295-2312. [PMID: 39228105 DOI: 10.1002/glia.24613] [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: 04/08/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
Sialylation plays an important role in self-recognition, as well as keeping the complement and innate immune systems in check. It is unclear whether the reduced sialylation seen during aging and in mice heterozygous for the null mutant of UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (Gne+/-), an essential enzyme for sialic acid biosynthesis, contributes to retinal inflammation and degeneration. We found a reduction of polysialic acid and trisialic acid expression in several retinal layers in Gne+/- mice at 9 months of age compared to Gne+/+ wildtype (WT) mice, which was associated with a higher microglial expression of the lysosomal marker CD68. Furthermore, the total number of rod bipolar cells was reduced in 12 months old Gne+/- mice in comparison to WT mice, demonstrating loss of these retinal interneurons. Transcriptome analysis showed up-regulation of complement, inflammation, and apoptosis-related pathways in the retinas of Gne+/- mice. Particularly, increased gene transcript levels of the complement factors C3 and C4 and the pro-inflammatory cytokine Il-1β were observed by semi-quantitative real-time polymerase chain reaction (sqRT-PCR) in 9 months old Gne+/- mice compared to WT mice. The increased expression of CD68, loss of rod bipolar cells, and increased gene transcription of complement factor C4, were all prevented after crossing Gne+/- mice with complement factor C3-deficient animals. In conclusion, our data show that retinal hyposialylation in 9 and 12 months old Gne+/- mice was associated with complement-related inflammation and lysosomal microglia response, as well as rod bipolar cells loss, which was absent after genetic deletion of complement factor C3.
Collapse
Affiliation(s)
- German Cuevas-Rios
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Tawfik Abou Assale
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jannis Wissfeld
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Annemarie Bungartz
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Julia Hofmann
- Experimental Immunology of the Eye, Department of Ophthalmology, University Hospital Cologne, Cologne, Germany
| | - Thomas Langmann
- Experimental Immunology of the Eye, Department of Ophthalmology, University Hospital Cologne, Cologne, Germany
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
2
|
Jiao K, Zhang J, Li Q, Lv X, Yu Y, Zhu B, Zhong H, Yu X, Song J, Ke Q, Qian F, Luan X, Zhang X, Chang X, Wang L, Liu M, Dong J, Zou Z, Bu B, Jiang H, Liu L, Li Y, Yue D, Chang X, Zheng Y, Wang N, Gao M, Xia X, Cheng N, Wang T, Luo SS, Xi J, Lin J, Lu J, Zhao C, Yang H, Lin P, Hong D, Zhao Z, Wang Z, Zhu W. Novel variants and genotype-phenotype correlation in a multicentre cohort of GNE myopathy in China. J Med Genet 2024; 61:1053-1061. [PMID: 39332896 DOI: 10.1136/jmg-2024-110149] [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: 05/30/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND GlcNAc2-epimerase (GNE) myopathy is a rare autosomal recessive disorder caused by pathogenic variants in the GNE gene, which is essential for the sialic acid biosynthesis pathway. OBJECTIVE This multi-centre study aimed to delineate the clinical phenotype and GNE variant spectrum in Chinese patients, enhancing our understanding of the genetic diversity and clinical manifestation across different populations. METHODS We retrospectively analysed GNE variants from 113 patients, integrating these data with external GNE variants from online databases for a global perspective, examining their consequences, distribution, ethnicity and severity. RESULTS This study revealed 97 distinct GNE variants, including 35 (36.08%) novel variants. Two more patients with deep intronic variant c.862+870C>T were identified, while whole genome sequencing (WGS) uncovered another two novel intronic variants: c.52-8924G>T and c.1505-12G>A. Nanopore long reads sequencing (LRS) and further PCR analysis verified a 639 bp insertion at chr9:36249241. Missense variants predominantly located in the epimerase/kinase domain coding region, indicating the impairment of catalytic function as a key pathogenic consequence. Comparative studies with Japanese, Korean and Jewish, our cohorts showed later onset ages by 2 years. The high allele frequency of the non-catalytic GNE variant, c.620A>T, might underlie the milder phenotype of Chinese patients. CONCLUSIONS Comprehensive techniques such as WGS and Nanopore LRS warrants the identifying of GNE variants. Patients with the non-catalytic GNE variant, c.620A>T, had a milder disease progression and later wheelchair use.
Collapse
Affiliation(s)
- Kexin Jiao
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Jialong Zhang
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Qiuxiang Li
- Department of Neurology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Xiaoqing Lv
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yanyan Yu
- Department of Neurology and Department of Medical Genetics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bochen Zhu
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Huahua Zhong
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Xu'en Yu
- Department of Neurology, The Affiliated Hospital of Institute of Neurology, Anhui University of Chinese Medicine, Hefei, China
| | - Jia Song
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Qing Ke
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fangyuan Qian
- Department of Neurology, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Xinghua Luan
- Department of Neurology, Shanghai Sixth People's Hospital, Shanghai, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Sixth People's Hospital, Shanghai, China
| | - Xueli Chang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liang Wang
- Department of Neurology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Meirong Liu
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jihong Dong
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Zhangyu Zou
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haishan Jiang
- Department of Neurology, Southern Medical University Nanfang Hospital, Guangzhou, China, China
| | - LingChun Liu
- Department of Neurology, First People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Yue Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyue Yue
- Department of Neurology, Jing'an District Centre Hospital of Shanghai, Shanghai, Shanghai, China
| | - Xuechun Chang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Yongsheng Zheng
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Ningning Wang
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Mingshi Gao
- Department of Pathology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Xingyu Xia
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Nachuan Cheng
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Tao Wang
- Department of Anesthesiology, Zhongshan hospital, Shanghai, China
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science,Fudan University, Shanghai, China
| | - Su-Shan Luo
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Jianying Xi
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Jie Lin
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Huan Yang
- Department of Neurology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Pengfei Lin
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Daojun Hong
- Department of Neurology and Department of Medical Genetics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhe Zhao
- Department of Neuromuscular Disease, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhiqiang Wang
- The Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Xiamen, Fujian, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| |
Collapse
|
3
|
Kotioumbé M, Maiga AB, Bamba S, Cissé L, Diarra S, Diallo S, Yalcouyé A, Kané F, Diallo SH, Coulibaly D, Coulibaly T, Dembélé K, Maiga B, Guinto CO, Landouré G. A novel variant in the GNE gene in a Malian patient presenting with distal myopathy. Neurogenetics 2024; 25:487-492. [PMID: 39088149 DOI: 10.1007/s10048-024-00761-z] [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: 04/06/2024] [Accepted: 04/25/2024] [Indexed: 08/02/2024]
Abstract
GNE-myopathy (GNE-M) is a rare autosomal recessive disorder caused by variants in the GNE gene. We report a novel variant in GNE causing GNE-M in a Malian family. A 19-year-old male patient from consanguineous marriage was seen for progressive walking difficulty. Neurological examination found predominant distal muscle weakness and atrophy, decreased tendon reflexes, predominating in lower limbs. Electroneuromyography showed an axonal neuropathy pattern. However, whole exome sequencing (WES) revealed a novel biallelic variant in GNE c.1838G > A:p.Gly613Glu, segregating with the phenotype within the family. This study highlights its diagnosis challenges in sub-Saharan Africa and broadens the genetic spectrum of this rare disease.
Collapse
Affiliation(s)
- Mahamadou Kotioumbé
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Alassane B Maiga
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Salia Bamba
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Lassana Cissé
- Service de Neurologie, Centre Hospitalier Universitaire Point "G", Bamako, Mali
| | - Salimata Diarra
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
- Neurogenetics Branch, NINDS, NIH, Bethesda, MD, USA
| | - Salimata Diallo
- Service de Neurologie, Centre Hospitalier Universitaire "Gabriel Touré", Bamako, Mali
| | - Abdoulaye Yalcouyé
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Fousseyni Kané
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Seybou H Diallo
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
- Service de Neurologie, Centre Hospitalier Universitaire "Gabriel Touré", Bamako, Mali
| | - Dramane Coulibaly
- Service de Médecine, Centre Hospitalier Universitaire "Le Luxembourg", Bamako, Mali
| | - Thomas Coulibaly
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
- Service de Neurologie, Centre Hospitalier Universitaire Point "G", Bamako, Mali
| | - Kékouta Dembélé
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
| | - Boubacar Maiga
- Service de Neurologie, Centre Hospitalier Universitaire Point "G", Bamako, Mali
| | - Cheick O Guinto
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali
- Service de Neurologie, Centre Hospitalier Universitaire Point "G", Bamako, Mali
| | - Guida Landouré
- Faculté de Médecine et d'Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, BP: 1805, Mali.
- Service de Neurologie, Centre Hospitalier Universitaire Point "G", Bamako, Mali.
| |
Collapse
|
4
|
Yang S, Ma Y, Song Y, Wang X, Cong P, Meng N, Xu J, Xue C. Establishment of a targeted analysis method for gangliosides in mouse tissues by HILIC-ESI-MS/MS. Anal Bioanal Chem 2024; 416:5457-5471. [PMID: 38305860 DOI: 10.1007/s00216-024-05169-0] [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: 10/07/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Gangliosides play an imperative role in cell signaling, neuronal recovery, apoptosis, and other physiological processes. For example, GM3 can regulate hypothalamic leptin resistance and control energy homeostasis, GD3 can mediate cell proliferation and differentiation and induce apoptosis, and GQ1b can stimulate neurogenesis. Therefore, the present study sought to establish and optimize the targeted analysis method for ganglioside subclasses and their molecular species using hydrophilic interaction liquid chromatography-triple quadrupole-MS/MS (HILIC-QQQ-MS/MS). Additionally, the fragmentation pattern of different ganglioside subclasses and their retention time patterns were analyzed, providing more accurate qualitative results. The limit of quantitation (LOQ) was as low as 10-4 ng. Moreover, the molecular species of gangliosides in the liver, cortex, and hypothalamus of C57BL/6 mice were analyzed using the established method. A total of 23 ganglioside subclasses with 164 molecular species, including 40 O-acetylated ganglioside molecular species and 28 NeuGc ganglioside molecular species, were identified using the semi-quantitative analysis method of an external standard curve corrected by an internal standard. In addition to NeuGc gangliosides, the contents of ganglioside subclasses were more abundant in the mouse brain than those in the mouse liver; especially, the contents of unsaturated gangliosides in the hypothalamus were much higher than those in the liver. Among them, O-acetylated gangliosides were detected only in the cortex and hypothalamus at a concentration of up to 100 μg/mg protein (40 molecular species). Overall, the proposed method expanded the detectable number of ganglioside subclasses and molecular species in biological samples and provided more opportunities for further study of the biological functions of gangliosides.
Collapse
Affiliation(s)
- Shuo Yang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
| | - Yingxu Ma
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
| | - Yu Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
| | - Xiaoxu Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
| | - Peixu Cong
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China.
| | - Nan Meng
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
| | - Jie Xu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, 266404, Shandong, China
- Qingdao Marine Science and Technology Center, Qingdao, 266235, China
| |
Collapse
|
5
|
Jiao K, Cheng N, Huan X, Zhang J, Ding Y, Luan X, Liu L, Wang X, Zhu B, Du K, Fan J, Gao M, Xia X, Wang N, Wang T, Xi J, Luo S, Lu J, Zhao C, Yue D, Zhu W. Pseudoexon activation by deep intronic variation in GNE myopathy with thrombocytopenia. Muscle Nerve 2024; 69:708-718. [PMID: 38558464 DOI: 10.1002/mus.28092] [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: 07/25/2023] [Revised: 03/07/2024] [Accepted: 03/16/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION/AIMS GNE myopathy is a rare autosomal recessive disorder caused by pathogenic variants in the GNE gene, which is essential for the sialic acid biosynthesis pathway. Although over 300 GNE variants have been reported, some patients remain undiagnosed with monoallelic pathogenic variants. This study aims to analyze the entire GNE genomic region to identify novel pathogenic variants. METHODS Patients with clinically compatible GNE myopathy and monoallelic pathogenic variants in the GNE gene were enrolled. The other GNE pathogenic variant was verified using comprehensive methods including exon 2 quantitative polymerase chain reaction and nanopore long-read single-molecule sequencing (LRS). RESULTS A deep intronic GNE variant, c.862+870C>T, was identified in nine patients from eight unrelated families. This variant generates a cryptic splice site, resulting in the activation of a novel pseudoexon between exons 5 and 6. It results in the insertion of an extra 146 nucleotides into the messengerRNA (mRNA), which is predicted to result in a truncated humanGNE1(hGNE1) protein. Peanut agglutinin(PNA) lectin staining of muscle tissues showed reduced sialylation of mucin O-glycans on sarcolemmal glycoproteins. Notably, a third of patients with the c.862+870C>T variant exhibited thrombocytopenia. A common core haplotype harboring the deep intronic GNE variant was found in all these patients. DISCUSSION The transcript with pseudoexon activation potentially affects sialic acid biosynthesis via nonsense-mediated mRNA decay, or resulting in a truncated hGNE1 protein, which interferes with normal enzyme function. LRS is expected to be more frequently incorporated in genetic analysis given its efficacy in detecting hard-to-find pathogenic variants.
Collapse
Affiliation(s)
- Kexin Jiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Nachuan Cheng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Xiao Huan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Jialong Zhang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xinghua Luan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - LingChun Liu
- The First People's Hospital of Yunnan Province, Kunming, China
| | - Xilu Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Bochen Zhu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Kunzhao Du
- Jinshan Hospital Center for Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jiale Fan
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, The Institutes of Brain Science, Shanghai, China
| | - Mingshi Gao
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xingyu Xia
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Ningning Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Tao Wang
- Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Dongyue Yue
- Department of Neurology, Jing'an District Center Hospital of Shanghai, Shanghai, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| |
Collapse
|
6
|
Filipsky F, Läubli H. Regulation of sialic acid metabolism in cancer. Carbohydr Res 2024; 539:109123. [PMID: 38669826 DOI: 10.1016/j.carres.2024.109123] [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: 03/10/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024]
Abstract
Sialic acid, the terminal structure of cell surface glycans, has essential functions in regulating immune response, cell-to-cell communication, and cell adhesion. More importantly, an increased level of sialic acid, termed hypersialylation, has emerged as a commonly observed phenotype in cancer. Therefore, targeting sialic acid ligands (sialoglycans) and their receptors (Siglecs) may provide a new therapeutic approach for cancer immunotherapy. We highlight the complexity of the sialic acid metabolism and its involvement in malignant transformation within individual cancer subtypes. In this review, we focus on the dysregulation of sialylation, the intricate nature of sialic acid synthesis, and clinical perspective. We aim to provide a brief insight into the mechanism of hypersialylation and how our understanding of these processes can be leveraged for the development of novel therapeutics.
Collapse
Affiliation(s)
- Filip Filipsky
- Department of Biomedicine, University Hospital and University of Basel, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, University Hospital and University of Basel, Switzerland; Division of Oncology, University Hospital Basel, Switzerland.
| |
Collapse
|
7
|
Mashangva F, Oswalia J, Singh S, Arya R. Potential small effector molecules restoring cellular defects due to sialic acid biosynthetic enzyme deficiency: Pathological relevance to GNE myopathy. Biochem Pharmacol 2024; 223:116199. [PMID: 38604256 DOI: 10.1016/j.bcp.2024.116199] [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: 10/29/2023] [Revised: 02/21/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
GNEM (GNE Myopathy) is a rare neuromuscular disease caused due to biallelic mutations in sialic acid biosynthetic GNE enzyme (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine Kinase). Recently direct or indirect role of GNE in other cellular functions have been elucidated. Hyposialylation of IGF-1R leads to apoptosis due to mitochondrial dysfunction while hyposialylation of β1 integrin receptor leads to altered F-actin assembly, disrupted cytoskeletal organization and slow cell migration. Other cellular defects in presence of GNE mutation include altered ER redox state and chaperone expression such as HSP70 or PrdxIV. Currently, there is no cure to treat GNEM. Possible therapeutic trials focus on supplementation with sialic acid, ManNAc, sialyllactose and gene therapy that slows the disease progression. In the present study, we analyzed the effect of small molecules like BGP-15 (HSP70 modulator), IGF-1 (IGF-1R ligand) and CGA (cofilin activator) on cellular phenotypes of GNE heterozygous knock out L6 rat skeletal muscle cell line (SKM‑GNEHz). Treatment with BGP-15 improved GNE epimerase activity by 40 % and reduced ER stress by 45 % for SKM‑GNEHz. Treatment with IGF-1 improved epimerase activity by 37.5 %, F-actin assembly by 100 %, cell migration upto 36 % (36 h) and atrophy by 0.44-fold for SKM‑GNEHz. Treatment with CGA recovered epimerase activity by 49 %, F-actin assembly by 132 % and cell migration upto 41 % (24 h) in SKM‑GNEHz. Our study shows that treatment with these small effector molecules reduces the detrimental phenotype observed in SKM‑GNEHz, thereby, providing insights into potential therapeutic targets for GNEM.
Collapse
Affiliation(s)
| | - Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shagun Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
8
|
Gueho A, Żarski D, Rime H, Guével B, Com E, Lavigne R, Nguyen T, Montfort J, Pineau C, Bobe J. Evolutionarily conserved ovarian fluid proteins are responsible for extending egg viability in salmonid fish. Sci Rep 2024; 14:9651. [PMID: 38671194 PMCID: PMC11053066 DOI: 10.1038/s41598-024-60118-2] [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: 09/05/2023] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
In contrast to most fishes, salmonids exhibit the unique ability to hold their eggs for several days after ovulation without significant loss of viability. During this period, eggs are held in the body cavity in a biological fluid, the coelomic fluid (CF) that is responsible for preserving egg viability. To identify CF proteins responsible for preserving egg viability, a proteomic comparison was performed using 3 salmonid species and 3 non-salmonid species to identify salmonid-specific highly abundant proteins. In parallel, rainbow trout CF fractions were purified and used in a biological test to estimate their egg viability preservation potential. The most biologically active CF fractions were then subjected to mass spectrometry analysis. We identified 50 proteins overabundant in salmonids and present in analytical fractions with high egg viability preservation potential. The identity of these proteins illuminates the biological processes participating in egg viability preservation. Among identified proteins of interest, the ovarian-specific expression and abundance in CF at ovulation of N-acetylneuraminic acid synthase a (Nansa) suggest a previously unsuspected role. We show that salmonid CF is a complex biological fluid containing a diversity of proteins related to immunity, calcium binding, lipid metabolism, proteolysis, extracellular matrix and sialic acid metabolic pathway that are collectively responsible for preserving egg viability.
Collapse
Affiliation(s)
- Aurélie Gueho
- INRAE UR1037, Fish Physiology and Genomics, 35000, Rennes, France
| | - Daniel Żarski
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str. 10, 10-748, Olsztyn, Poland
| | - Hélène Rime
- INRAE UR1037, Fish Physiology and Genomics, 35000, Rennes, France
| | - Blandine Guével
- Inserm, EHESP, Irset, UMR_S 1085, Univ Rennes, 35000, Rennes, France
- CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, Univ Rennes, 35000, Rennes, France
| | - Emmanuelle Com
- Inserm, EHESP, Irset, UMR_S 1085, Univ Rennes, 35000, Rennes, France
- CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, Univ Rennes, 35000, Rennes, France
| | - Régis Lavigne
- Inserm, EHESP, Irset, UMR_S 1085, Univ Rennes, 35000, Rennes, France
- CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, Univ Rennes, 35000, Rennes, France
| | - Thaovi Nguyen
- INRAE UR1037, Fish Physiology and Genomics, 35000, Rennes, France
| | - Jérôme Montfort
- INRAE UR1037, Fish Physiology and Genomics, 35000, Rennes, France
| | - Charles Pineau
- Inserm, EHESP, Irset, UMR_S 1085, Univ Rennes, 35000, Rennes, France
- CNRS, Inserm, Biosit UAR 3480 US_S 018, Protim Core Facility, Univ Rennes, 35000, Rennes, France
| | - Julien Bobe
- INRAE UR1037, Fish Physiology and Genomics, 35000, Rennes, France.
| |
Collapse
|
9
|
Neu CT, Weilepp L, Bork K, Gesper A, Horstkorte R. GNE deficiency impairs Myogenesis in C2C12 cells and cannot be rescued by ManNAc supplementation. Glycobiology 2024; 34:cwae004. [PMID: 38224318 PMCID: PMC10987290 DOI: 10.1093/glycob/cwae004] [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: 08/08/2023] [Revised: 12/19/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024] Open
Abstract
GNE myopathy (GNEM) is a late-onset muscle atrophy, caused by mutations in the gene for the key enzyme of sialic acid biosynthesis, UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE). With an incidence of one to nine cases per million it is an ultra-rare, so far untreatable, autosomal recessive disease. Several attempts have been made to treat GNEM patients by oral supplementation with sialic acid precursors (e.g. N-acetylmannosamine, ManNAc) to restore sarcolemmal sialylation and muscle strength. In most studies, however, no significant improvement was observed. The lack of a suitable mouse model makes it difficult to understand the exact pathomechanism of GNEM and many years of research have failed to identify the role of GNE in skeletal muscle due to the lack of appropriate tools. We established a CRISPR/Cas9-mediated Gne-knockout cell line using murine C2C12 cells to gain insight into the actual role of the GNE enzyme and sialylation in a muscular context. The main aspect of this study was to evaluate the therapeutic potential of ManNAc and N-acetylneuraminic acid (Neu5Ac). Treatment of Gne-deficient C2C12 cells with Neu5Ac, but not with ManNAc, showed a restoration of the sialylation level back to wild type levels-albeit only with long-term treatment, which could explain the rather low therapeutic potential. We furthermore highlight the importance of sialic acids on myogenesis, for C2C12 Gne-knockout myoblasts lack the ability to differentiate into mature myotubes.
Collapse
Affiliation(s)
- Carolin T Neu
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Linus Weilepp
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Kaya Bork
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Astrid Gesper
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Rüdiger Horstkorte
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| |
Collapse
|
10
|
Abdel-Naim AB, Kumar P, Bazuhair MA, Rizg WY, Niyazi HA, Alkuwaity K, Niyazi HA, Alharthy SA, Harakeh S, Haque S, Prakash A, Kumar V. Computational insights into dynamics and conformational stability of N-acetylmannosamine kinase mutations. J Biomol Struct Dyn 2024:1-11. [PMID: 38502682 DOI: 10.1080/07391102.2024.2323702] [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: 05/02/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024]
Abstract
The activity of UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE) is essential for the biosynthesis of sialic acid, which is involved in cellular processes in health and diseases. GNE contains an N-terminal epimerase domain and a C-terminal kinase domain (N-acetylmannosamine kinase, MNK). Mutations of the GNE protein led to hypoactivity of the enzyme and cause sialurea or autosomal recessive inclusion body myopathy/Nonaka myopathy. Here, we used all-atom molecular dynamics (MD) simulations to comprehend the folding, dynamics and conformational stability of MNK variants, including the wild type (WT) and three mutants (H677R, V696M and H677R/V696M). The deleterious and destabilizing nature of MNK mutants were predicted using different prediction tools. Results predicted that mutations modulate the stability, flexibility and function of MNK. The effect of mutations on the conformational stability and dynamics of MNK was next studied through the free-energy landscape (FEL), hydrogen-bonds and secondary structure changes. The FEL results show that the mutations interfere with various conformational transitions in both WT and mutants, exposing the structural underpinnings of protein destabilization and unfolding brought on by mutation. We discover that, when compared to the other two mutations, V696M and H677R/V696M, H677R has the most harmful effects. These findings have a strong correlation with published experimental studies that demonstrate how these mutations disrupt MNK activity. Hence, this computational study describes the structural details to unravel the mutant effects at the atomistic resolution and has implications for understanding the GNE's physiological and pathological role.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Pawan Kumar
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mohammed A Bazuhair
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Waleed Y Rizg
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hatoon A Niyazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalil Alkuwaity
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hanouf A Niyazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saif A Alharthy
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Toxicology and Forensic Sciences Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| |
Collapse
|
11
|
Wißfeld J, Abou Assale T, Cuevas-Rios G, Liao H, Neumann H. Therapeutic potential to target sialylation and SIGLECs in neurodegenerative and psychiatric diseases. Front Neurol 2024; 15:1330874. [PMID: 38529039 PMCID: PMC10961342 DOI: 10.3389/fneur.2024.1330874] [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: 10/31/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Sialic acids, commonly found as the terminal carbohydrate on the glycocalyx of mammalian cells, are pivotal checkpoint inhibitors of the innate immune system, particularly within the central nervous system (CNS). Sialic acid-binding immunoglobulin-like lectins (SIGLECs) expressed on microglia are key players in maintaining microglial homeostasis by recognizing intact sialylation. The finely balanced sialic acid-SIGLEC system ensures the prevention of excessive and detrimental immune responses in the CNS. However, loss of sialylation and SIGLEC receptor dysfunctions contribute to several chronic CNS diseases. Genetic variants of SIGLEC3/CD33, SIGLEC11, and SIGLEC14 have been associated with neurodegenerative diseases such as Alzheimer's disease, while sialyltransferase ST8SIA2 and SIGLEC4/MAG have been linked to psychiatric diseases such as schizophrenia, bipolar disorders, and autism spectrum disorders. Consequently, immune-modulatory functions of polysialic acids and SIGLEC binding antibodies have been exploited experimentally in animal models of Alzheimer's disease and inflammation-induced CNS tissue damage, including retinal damage. While the potential of these therapeutic approaches is evident, only a few therapies to target either sialylation or SIGLEC receptors have been tested in patient clinical trials. Here, we provide an overview of the critical role played by the sialic acid-SIGLEC axis in shaping microglial activation and function within the context of neurodegeneration and synaptopathies and discuss the current landscape of therapies that target sialylation or SIGLECs.
Collapse
Affiliation(s)
- Jannis Wißfeld
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Tawfik Abou Assale
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - German Cuevas-Rios
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Huan Liao
- Florey Institute of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Harazi A, Yakovlev L, Ilouz N, Selke P, Horstkorte R, Fellig Y, Lahat O, Lifschytz T, Abudi N, Abramovitch R, Argov Z, Mitrani-Rosenbaum S. Induced Muscle and Liver Absence of Gne in Postnatal Mice Does Not Result in Structural or Functional Muscle Impairment. J Neuromuscul Dis 2024; 11:905-917. [PMID: 38875046 PMCID: PMC11380236 DOI: 10.3233/jnd-240056] [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] [Accepted: 05/20/2024] [Indexed: 06/16/2024]
Abstract
Background GNE Myopathy is a unique recessive neuromuscular disorder characterized by adult-onset, slowly progressive distal and proximal muscle weakness, caused by mutations in the GNE gene which is a key enzyme in the biosynthesis of sialic acid. To date, the precise pathophysiology of the disease is not well understood and no reliable animal model is available. Gne KO is embryonically lethal in mice. Objective To gain insights into GNE function in muscle, we have generated an inducible muscle Gne KO mouse. To minimize the contribution of the liver to the availability of sialic acid to muscle via the serum, we have also induced combined Gne KO in liver and muscle. Methods A mouse carrying loxp sequences flanking Gne exon3 was generated by Crispr/Cas9 and bred with a human skeletal actin (HSA) promoter driven CreERT mouse. Gne muscle knock out was induced by tamoxifen injection of the resulting homozygote GneloxpEx3loxp/HSA Cre mouse. Liver Gne KO was induced by systemic injection of AAV8 vectors carrying the Cre gene driven by the hepatic specific promoter of the thyroxine binding globulin gene. Results Characterization of these mice for a 12 months period showed no significant changes in their general behaviour, motor performance, muscle mass and structure in spite of a dramatic reduction in sialic acid content in both muscle and liver. Conclusions We conclude that post weaning lack of Gne and sialic acid in muscle and liver have no pathologic effect in adult mice. These findings could reflect a strong interspecies versatility, but also raise questions about the loss of function hypothesis in Gne Myopathy. If these findings apply to humans they have a major impact on therapeutic strategies.
Collapse
Affiliation(s)
- Avi Harazi
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lena Yakovlev
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nili Ilouz
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Philipp Selke
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Rudiger Horstkorte
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Yakov Fellig
- Department of Pathology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Olga Lahat
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tzuri Lifschytz
- Biological Psychiatry Laboratory and Hadassah BrainLabs Center for Psychedelic Research, Hadassah Medical Center, Hebrew University, Jerusalem, Israel
| | - Nathalie Abudi
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Rinat Abramovitch
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Zohar Argov
- Department of Neurology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stella Mitrani-Rosenbaum
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
13
|
Peng J, Yu L, Huang L, Paschoal VA, Chu H, de Souza CO, Varre JV, Oh DY, Kohler JJ, Xiao X, Xu L, Holland WL, Shaul PW, Mineo C. Hepatic sialic acid synthesis modulates glucose homeostasis in both liver and skeletal muscle. Mol Metab 2023; 78:101812. [PMID: 37777009 PMCID: PMC10583174 DOI: 10.1016/j.molmet.2023.101812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
OBJECTIVE Sialic acid is a terminal monosaccharide of glycans in glycoproteins and glycolipids, and its derivation from glucose is regulated by the rate-limiting enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE). Although the glycans on key endogenous hepatic proteins governing glucose metabolism are sialylated, how sialic acid synthesis and sialylation in the liver influence glucose homeostasis is unknown. Studies were designed to fill this knowledge gap. METHODS To decrease the production of sialic acid and sialylation in hepatocytes, a hepatocyte-specific GNE knockdown mouse model was generated, and systemic glucose metabolism, hepatic insulin signaling and glucagon signaling were evaluated in vivo or in primary hepatocytes. Peripheral insulin sensitivity was also assessed. Furthermore, the mechanisms by which sialylation in the liver influences hepatic insulin signaling and glucagon signaling and peripheral insulin sensitivity were identified. RESULTS Liver GNE deletion in mice caused an impairment of insulin suppression of hepatic glucose production. This was due to a decrease in the sialylation of hepatic insulin receptors (IR) and a decline in IR abundance due to exaggerated degradation through the Eph receptor B4. Hepatic GNE deficiency also caused a blunting of hepatic glucagon receptor (GCGR) function which was related to a decline in its sialylation and affinity for glucagon. An accompanying upregulation of hepatic FGF21 production caused an enhancement of skeletal muscle glucose disposal that led to an overall increase in glucose tolerance and insulin sensitivity. CONCLUSION These collective observations reveal that hepatic sialic acid synthesis and sialylation modulate glucose homeostasis in both the liver and skeletal muscle. By interrogating how hepatic sialic acid synthesis influences glucose control mechanisms in the liver, a new metabolic cycle has been identified in which a key constituent of glycans generated from glucose modulates the systemic control of its precursor.
Collapse
Affiliation(s)
- Jun Peng
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Liming Yu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Vivian A Paschoal
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Camila O de Souza
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Joseph V Varre
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Da Young Oh
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jennifer J Kohler
- Dept. of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xue Xiao
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Lin Xu
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - William L Holland
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA; Dept. of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
14
|
Wang Z, Wang X, Ma Y, Cong P, Wang X, Song Y, Xu J, Xue C. Astaxanthin alleviates ganglioside metabolism disorder in the cortex of Alzheimer's disease mice. Food Funct 2023; 14:10362-10374. [PMID: 37929718 DOI: 10.1039/d3fo03223j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The present study analyzed the amelioration effect and mechanism of two kinds of astaxanthin (AST), including free-AST (F-AST) and docosahexaenoic acid-acylated AST monoester (AST-DHA), on ganglioside (GLS) metabolism in the cortex of APP/PS1 mice using the LC-MS strategy in combination with molecular biology. Water maze and immunohistochemical experiments demonstrated that AST significantly improved the cognitive level of APP/PS1 mice and reduced Aβ deposition in the cortex. After the dietary intake of AST, the composition and level of 84 GLS molecular species in the mouse cortex were determined using the LC-MS strategy. The results showed that the total GLS was reduced, most complex GLS was decreased, and simple GLS (GM3 and GM1a) was increased in the APP/PS1 mouse cortex. Notably, F-AST mainly regulated complex GLS (p < 0.001), whereas AST-DHA primarily reacted with simple GLS (p < 0.001). OAc-GQ1a(38:1), OAc-GQ1a(36:1), GD1a(36:1), and GM3(38:1) decreased 3.73, 2.31, and 2.29-fold and increased 3.54-fold, respectively, and were identified as potential AD biomarkers in the cortices of APP/PS1 mice. Additionally, the AST diet significantly upregulated the mRNA expression of GLS synthesizing genes (st3gal5, st8sia1, b3galt4, st3fal2, and soat) and siae (p < 0.05) and down-regulated that of the GLS catabolizing gene hexa (p < 0.01). In conclusion, improving GLS homeostasis in the AD mouse cortex might be a critical pathway to explain the AD-preventing effect of AST.
Collapse
Affiliation(s)
- Zhigao Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Xiaoxu Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Yingxu Ma
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Peixu Cong
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Xincen Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Yu Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Jie Xu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266003, China.
- Qingdao Marine Science and Technology Center, Qingdao, Shandong Province 266235, China
| |
Collapse
|
15
|
Bu Q, Dai Y, Zhang H, Li M, Liu H, Huang Y, Zeng A, Qin F, Jiang L, Wang L, Chen Y, Li H, Wang X, Zhao Y, Qin M, Zhao Y, Zhang N, Kuang W, Zhao Y, Cen X. Neurodevelopmental defects in human cortical organoids with N-acetylneuraminic acid synthase mutation. SCIENCE ADVANCES 2023; 9:eadf2772. [PMID: 38000033 PMCID: PMC10672180 DOI: 10.1126/sciadv.adf2772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/26/2023] [Indexed: 11/26/2023]
Abstract
Biallelic genetic variants in N-acetylneuraminic acid synthase (NANS), a critical enzyme in endogenous sialic acid biosynthesis, are clinically associated with neurodevelopmental disorders. However, the mechanism underlying the neuropathological consequences has remained elusive. Here, we found that NANS mutation resulted in the absence of both sialic acid and protein polysialylation in the cortical organoids and notably reduced the proliferation and expansion of neural progenitors. NANS mutation dysregulated neural migration and differentiation, disturbed synapse formation, and weakened neuronal activity. Single-cell RNA sequencing revealed that NANS loss of function markedly altered transcriptional programs involved in neuronal differentiation and ribosomal biogenesis in various neuronal cell types. Similarly, Nans heterozygous mice exhibited impaired cortical neurogenesis and neurobehavioral deficits. Collectively, our findings reveal a crucial role of NANS-mediated endogenous sialic acid biosynthesis in regulating multiple features of human cortical development, thus linking NANS mutation with its clinically relevant neurodevelopmental disorders.
Collapse
Affiliation(s)
- Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huaqin Zhang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haxiaoyu Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Huang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ailing Zeng
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaojie Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ying Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ni Zhang
- Mental Health Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weihong Kuang
- Mental Health Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
16
|
Scott H, Novikov B, Ugur B, Allen B, Mertsalov I, Monagas-Valentin P, Koff M, Baas Robinson S, Aoki K, Veizaj R, Lefeber DJ, Tiemeyer M, Bellen H, Panin V. Glia-neuron coupling via a bipartite sialylation pathway promotes neural transmission and stress tolerance in Drosophila. eLife 2023; 12:e78280. [PMID: 36946697 PMCID: PMC10110239 DOI: 10.7554/elife.78280] [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: 03/01/2022] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Modification by sialylated glycans can affect protein functions, underlying mechanisms that control animal development and physiology. Sialylation relies on a dedicated pathway involving evolutionarily conserved enzymes, including CMP-sialic acid synthetase (CSAS) and sialyltransferase (SiaT) that mediate the activation of sialic acid and its transfer onto glycan termini, respectively. In Drosophila, CSAS and DSiaT genes function in the nervous system, affecting neural transmission and excitability. We found that these genes function in different cells: the function of CSAS is restricted to glia, while DSiaT functions in neurons. This partition of the sialylation pathway allows for regulation of neural functions via a glia-mediated control of neural sialylation. The sialylation genes were shown to be required for tolerance to heat and oxidative stress and for maintenance of the normal level of voltage-gated sodium channels. Our results uncovered a unique bipartite sialylation pathway that mediates glia-neuron coupling and regulates neural excitability and stress tolerance.
Collapse
Affiliation(s)
- Hilary Scott
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Boris Novikov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Berrak Ugur
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Brooke Allen
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Ilya Mertsalov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Pedro Monagas-Valentin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Melissa Koff
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Sarah Baas Robinson
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Raisa Veizaj
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Dirk J Lefeber
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Hugo Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Vladislav Panin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| |
Collapse
|
17
|
Qin M, Chen W, Hua L, Meng Y, Wang J, Li H, Yang R, Yan L, Qiao J. DNA methylation abnormalities induced by advanced maternal age in villi prime a high-risk state for spontaneous abortion. Clin Epigenetics 2023; 15:44. [PMID: 36945044 PMCID: PMC10029192 DOI: 10.1186/s13148-023-01432-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/20/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Advanced maternal age (AMA) has increased in many high-income countries in recent decades. AMA is generally associated with a higher risk of various pregnancy complications, and the underlying molecular mechanisms are largely unknown. In the current study, we profiled the DNA methylome of 24 human chorionic villi samples (CVSs) from early pregnancies in AMA and young maternal age (YMA), 11 CVSs from early spontaneous abortion (SA) cases using reduced representation bisulfite sequencing (RRBS), and the transcriptome of 10 CVSs from AMA and YMA pregnancies with mRNA sequencing(mRNA-seq). Single-cell villous transcriptional atlas presented expression patterns of targeted AMA-/SA-related genes. Trophoblast cellular impairment was investigated through the knockdown of GNE expression in HTR8-S/Vneo cells. RESULTS AMA-induced local DNA methylation changes, defined as AMA-related differentially methylated regions (DMRs), may be derived from the abnormal expression of genes involved in DNA demethylation, such as GADD45B. These DNA methylation changes were significantly enriched in the processes involved in NOTCH signaling and extracellular matrix organization and were reflected in the transcriptional alterations in the corresponding biological processes and specific genes. Furthermore, the DNA methylation level of special AMA-related DMRs not only significantly changed in AMA but also showed more excessive defects in CVS from spontaneous abortion (SA), including four AMA-related DMRs whose nearby genes overlapped with AMA-related differentially expressed genes (DEGs) (CDK11A, C19orf71, COL5A1, and GNE). The decreased DNA methylation level of DMR near GNE was positively correlated with the downregulated expression of GNE in AMA. Single-cell atlas further revealed comparatively high expression of GNE in the trophoblast lineage, and knockdown of GNE in HTR8-S/Vneo cells significantly impaired cellular proliferation and migration. CONCLUSION Our study provides valuable resources for investigating AMA-induced epigenetic abnormalities and provides new insights for explaining the increased risks of pregnancy complications in AMA pregnancies.
Collapse
Affiliation(s)
- Meng Qin
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Wei Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Lingyue Hua
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Yan Meng
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Beijing, 100096 China
| | - Jing Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Hanna Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Rui Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
| | - Liying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, 100191 China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191 China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191 China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China
- Beijing Advanced Innovation Center for Genomics, Beijing, 100871 China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871 China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing Jishuitan Hospital, Beijing, 100191 China
| |
Collapse
|
18
|
Omoto T, Wu D, Maruyama E, Tajima K, Hane M, Sato C, Kitajima K. Forced expression of α2,3-sialyltransferase IV rescues impaired heart development in α2,6-sialyltransferase I-deficient medaka. Biochem Biophys Res Commun 2023; 649:62-70. [PMID: 36745971 DOI: 10.1016/j.bbrc.2023.01.010] [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: 12/24/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Sialic acids (Sias) are often linked to galactose (Gal) residues by α2,6- and α2,3-linkages in glycans of glycoproteins. Sias are indispensable for vertebrate development, because organisms deficient in some enzymes in the Sia synthetic pathway are lethal during the development. However, it remains unknown if the difference of Siaα2,6Gal or α2,3Gal linkage has a critical meaning. To find a clue to understand significance of the linkage difference at the organism level, medaka was used as a vertebrate model. In embryos, Siaα2,6Gal epitopes recognized by Sambucus nigra lectin (SNA) and Siaα2,3Gal epitopes recognized by Maackia amurensis lectin (MAA) were enriched in the blastodisc and the yolk sphere, respectively. When these lectins were injected in the perivitelline space, SNA, but not MAA, impaired embryo body formation at 1 day post-fertilization (dpf). Most Siaα2,6Gal epitopes occurred on N-glycans owing to their sensitivity to peptide:N-glycanase. Of knockout-medaka (KO) for either of two β-galactoside:α2,6-sialyltransferase genes, ST6Gal I and ST6Gal II, only ST6Gal I-KO showed severe cardiac abnormalities at 7-16 dpf, leading to lethality at 14-18 dpf. Interestingly, however, these cardiac abnormalities of ST6Gal I-KO were rescued not only by forced expression of ST6Gal I, but also by that of ST6Gal II and the β-galactoside:α2,3-sialyltransferase IV gene (ST3Gal IV). Taken together, the Siaα2,6Gal linkage synthesized by ST6Gal I are critical in heart development; however, it can be replaced by the linkages synthesized by ST6Gal II and ST3Gal IV. These data suggest that sialylation itself is more important than its particular linkage for the heart development.
Collapse
Affiliation(s)
- Takayuki Omoto
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Di Wu
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Institute for Glyco-core Research, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Emi Maruyama
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Katsue Tajima
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Institute for Glyco-core Research, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Institute for Glyco-core Research, Nagoya University, Chikusa, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, 464-8601, Japan; Institute for Glyco-core Research, Nagoya University, Chikusa, Nagoya, 464-8601, Japan.
| |
Collapse
|
19
|
Genetic and Clinical Spectrum of GNE Myopathy in Russia. Genes (Basel) 2022; 13:genes13111991. [PMID: 36360228 PMCID: PMC9690815 DOI: 10.3390/genes13111991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/04/2022] Open
Abstract
GNE myopathy (GNEM) is a rare hereditary disease, but at the same time, it is the most common distal myopathy in several countries due to a founder effect of some pathogenic variants in the GNE gene. We collected the largest cohort of patients with GNEM from Russia and analyzed their mutational spectrum and clinical data. In our cohort, 10 novel variants were found, including 2 frameshift variants and 2 large deletions. One novel missense variant c.169_170delGCinsTT (p.(Ala57Phe)) was detected in 4 families in a homozygous state and in 3 unrelated patients in a compound heterozygous state. It was the second most frequent variant in our cohort. All families with this novel frequent variant were non-consanguineous and originated from the 3 neighboring areas in the European part of Russia. The clinical picture of the patients carrying this novel variant was typical, but the severity of clinical manifestation differed significantly. In our study, we reported two atypical cases expanding the phenotypic spectrum of GNEM. One female patient had severe quadriceps atrophy, hand joint contractures, keloid scars, and non-classical pattern on leg muscle magnetic resonance imaging, which was more similar to atypical collagenopathy rather than GNEM. Another patient initially had been observed with spinal muscular atrophy due to asymmetric atrophy of hand muscles and results of electromyography. The peculiar pattern of muscle involvement on magnetic resonance imaging consisted of pronounced changes in the posterior thigh muscle group with relatively spared muscles of the lower legs, apart from the soleus muscles. Different variants in the GNE gene were found in both atypical cases. Thus, our data expand the mutational and clinical spectrum of GNEM.
Collapse
|
20
|
Livne H, Avital T, Ruppo S, Harazi A, Mitrani-Rosenbaum S, Daya A. Generation and characterization of a novel gne Knockout Model in Zebrafish. Front Cell Dev Biol 2022; 10:976111. [PMID: 36353515 PMCID: PMC9637792 DOI: 10.3389/fcell.2022.976111] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/14/2022] [Indexed: 12/04/2022] Open
Abstract
GNE Myopathy is a rare, recessively inherited neuromuscular worldwide disorder, caused by a spectrum of bi-allelic mutations in the human GNE gene. GNE encodes a bi-functional enzyme responsible for the rate-limiting step of sialic acid biosynthesis pathway. However, the process in which GNE mutations lead to the development of a muscle pathology is not clear yet. Cellular and mouse models for GNE Myopathy established to date have not been informative. Further, additional GNE functions in muscle have been hypothesized. In these studies, we aimed to investigate gne functions using zebrafish genetic and transgenic models, and characterized them using macroscopic, microscopic, and molecular approaches. We first established transgenic zebrafish lineages expressing the human GNE cDNA carrying the M743T mutation, driven by the zebrafish gne promoter. These fish developed entirely normally. Then, we generated a gne knocked-out (KO) fish using the CRISPR/Cas9 methodology. These fish died 8–10 days post-fertilization (dpf), but a phenotype appeared less than 24 h before death and included progressive body axis curving, deflation of the swim bladder and decreasing movement and heart rate. However, muscle histology uncovered severe defects, already at 5 dpf, with compromised fiber organization. Sialic acid supplementation did not rescue the larvae from this phenotype nor prolonged their lifespan. To have deeper insights into the potential functions of gne in zebrafish, RNA sequencing was performed at 3 time points (3, 5, and 7 dpf). Genotype clustering was progressive, with only 5 genes differentially expressed in gne KO compared to gne WT siblings at 3 dpf. Enrichment analyses of the primary processes affected by the lack of gne also at 5 and 7 dpf point to the involvement of cell cycle and DNA damage/repair processes in the gne KO zebrafish. Thus, we have established a gne KO zebrafish lineage and obtained new insights into gne functions. This is the only model where GNE can be related to clear muscle defects, thus the only animal model relevant to GNE Myopathy to date. Further elucidation of gne precise mechanism-of-action in these processes could be relevant to GNE Myopathy and allow the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Hagay Livne
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tom Avital
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
| | - Shmuel Ruppo
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avi Harazi
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stella Mitrani-Rosenbaum
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- *Correspondence: Alon Daya,
| |
Collapse
|
21
|
Mullen J, Alrasheed K, Mozaffar T. GNE myopathy: History, etiology, and treatment trials. Front Neurol 2022; 13:1002310. [PMID: 36330422 PMCID: PMC9623016 DOI: 10.3389/fneur.2022.1002310] [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: 07/25/2022] [Accepted: 10/03/2022] [Indexed: 12/04/2022] Open
Abstract
GNE myopathy is an ultrarare muscle disease characterized by slowly progressive muscle weakness. Symptoms typically start in early adulthood, with weakness and atrophy in the tibialis anterior muscles and with slow progression over time, which largely spares the quadriceps muscles. Muscle biopsy shows atrophic fibers and rimmed vacuoles without inflammation. Inherited in an autosomal recessive manner, patients with GNE myopathy carry mutations in the GNE gene which affect the sialic acid synthesis pathway. Here, we look at the history and clinical aspects of GNE myopathy, as well as focus on prior treatment trials and challenges and unmet needs related to this disorder.
Collapse
Affiliation(s)
- Jeffrey Mullen
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Khalid Alrasheed
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Tahseen Mozaffar
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
- The Institute for Immunology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
22
|
Masunaga Y, Nishimura G, Takahashi K, Hishiyama T, Imamura M, Kashimada K, Kadoya M, Wada Y, Okamoto N, Oba D, Ohashi H, Ikeno M, Sakamoto Y, Fukami M, Saitsu H, Ogata T. Clinical and molecular findings in three Japanese patients with N-acetylneuraminic acid synthetase-congenital disorder of glycosylation (NANS-CDG). Sci Rep 2022; 12:17079. [PMID: 36224347 PMCID: PMC9556533 DOI: 10.1038/s41598-022-21751-x] [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: 05/02/2022] [Accepted: 09/30/2022] [Indexed: 01/04/2023] Open
Abstract
We report clinical and molecular findings in three Japanese patients with N-acetylneuraminic acid synthetase-congenital disorder of glycosylation (NANS-CDG). Patient 1 exhibited a unique constellation of clinical features including marked hydrocephalus, spondyloepimetaphyseal dysplasia (SEMD), and thrombocytopenia which is comparable to that of an infant reported by Faye-Peterson et al., whereas patients 2 and 3 showed Camera-Genevieve type SMED with intellectual/developmental disability which is currently known as the sole disease name for NANS-CDG. Molecular studies revealed a maternally inherited likely pathogenic c.207del:p.(Arg69Serfs*57) variant and a paternally derived likely pathogenic c.979_981dup:p.(Ile327dup) variant in patient 1, a homozygous likely pathogenic c.979_981dup:p.(Ile327dup) variant caused by maternal segmental isodisomy involving NANS in patient 2, and a paternally inherited pathogenic c.133-12T>A variant leading to aberrant splicing and a maternally inherited likely pathogenic c.607T>C:p.(Tyr203His) variant in patient 3 (reference mRNA: NM_018946.4). The results, together with previously reported data, imply that (1) NANS plays an important role in postnatal growth and fetal brain development; (2) SMED is recognizable at birth and shows remarkable postnatal evolution; (3) NANS-CDG is associated with low-normal serum sialic acid, obviously elevated urine N-acetylmannosamine, and normal N- and O-glycosylation of serum proteins; and (4) NANS-CDG is divided into Camera-Genevieve type and more severe Faye-Peterson type.
Collapse
Affiliation(s)
- Yohei Masunaga
- grid.505613.40000 0000 8937 6696Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Gen Nishimura
- grid.430047.40000 0004 0640 5017Center for Intractable Diseases, Saitama Medical University Hospital, Saitama, Japan
| | - Koji Takahashi
- grid.410824.b0000 0004 1764 0813Department of Pediatrics, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Tomiyuki Hishiyama
- grid.410824.b0000 0004 1764 0813Department of Neonatology, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Masatoshi Imamura
- grid.410824.b0000 0004 1764 0813Department of Neonatology, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Kenichi Kashimada
- grid.410824.b0000 0004 1764 0813Department of Pediatrics, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan ,grid.265073.50000 0001 1014 9130Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Machiko Kadoya
- grid.416629.e0000 0004 0377 2137Department of Molecular Medicine, Osaka Women’s and Children’s Hospital, Osaka, Japan
| | - Yoshinao Wada
- grid.416629.e0000 0004 0377 2137Department of Molecular Medicine, Osaka Women’s and Children’s Hospital, Osaka, Japan
| | - Nobuhiko Okamoto
- grid.416629.e0000 0004 0377 2137Department of Molecular Medicine, Osaka Women’s and Children’s Hospital, Osaka, Japan
| | - Daiju Oba
- grid.416697.b0000 0004 0569 8102Division of Medical Genetics, Saitama Children’s Medical Center, Saitama, Japan
| | - Hirofumi Ohashi
- grid.416697.b0000 0004 0569 8102Division of Medical Genetics, Saitama Children’s Medical Center, Saitama, Japan
| | - Mitsuru Ikeno
- grid.258269.20000 0004 1762 2738Department of Pediatrics, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuko Sakamoto
- grid.482668.60000 0004 1769 1784Department of Orthopedics, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Maki Fukami
- grid.63906.3a0000 0004 0377 2305Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hirotomo Saitsu
- grid.505613.40000 0000 8937 6696Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tsutomu Ogata
- grid.505613.40000 0000 8937 6696Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan ,grid.63906.3a0000 0004 0377 2305Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan ,grid.505613.40000 0000 8937 6696Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan ,grid.413553.50000 0004 1772 534XDepartment of Pediatrics, Hamamatsu Medical Center, Hamamatsu, Japan
| |
Collapse
|
23
|
Ilouz N, Harazi A, Guttman M, Daya A, Ruppo S, Yakovlev L, Mitrani-Rosenbaum S. In vivo and in vitro genome editing to explore GNE functions. Front Genome Ed 2022; 4:930110. [PMID: 36237634 PMCID: PMC9552322 DOI: 10.3389/fgeed.2022.930110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022] Open
Abstract
GNE myopathy is an adult onset neuromuscular disorder characterized by slowly progressive distal and proximal muscle weakness, caused by missense recessive mutations in the GNE gene. Although the encoded bifunctional enzyme is well known as the limiting factor in the biosynthesis of sialic acid, no clear mechanisms have been recognized to account for the muscle atrophic pathology, and novel functions for GNE have been hypothesized. Two major issues impair studies on this protein. First, the expression of the GNE protein is minimal in human and mice muscles and there is no reliable antibody to follow up endogenous expression. Second, no reliable animal model is available for the disease and cellular models from GNE myopathy patients’ muscle cells (expressing the mutated protein) are less informative than expected. In order to broaden our knowledge on GNE functions in muscle, we have taken advantage of the CRISPR/Cas9 method for genome editing to first, add a tag to the endogenous Gne gene in mouse, allowing the determination of the spatiotemporal expression of the protein in the organism, using well established and reliable antibodies against the specific tag. In addition we have generated a Gne knock out murine muscle cell lineage to identify the events resulting from the total lack of the protein. A thorough multi-omics analysis of both cellular systems including transcriptomics, proteomics, phosphoproteomics and ubiquitination, unraveled novel pathways for Gne, in particular its involvement in cell cycle control and in the DNA damage/repair pathways. The elucidation of fundamental mechanisms of Gne in normal muscle may contribute to the identification of the disrupted functions in GNE myopathy, thus, to the definition of novel biomarkers and possible therapeutic targets for this disease.
Collapse
Affiliation(s)
- Nili Ilouz
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avi Harazi
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miriam Guttman
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
| | - Shmuel Ruppo
- Bioinformatics Unit of the I-CORE at the Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - Lena Yakovlev
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stella Mitrani-Rosenbaum
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- *Correspondence: Stella Mitrani-Rosenbaum,
| |
Collapse
|
24
|
Myogenesis defects in a patient-derived iPSC model of hereditary GNE myopathy. NPJ Regen Med 2022; 7:48. [PMID: 36085325 PMCID: PMC9463157 DOI: 10.1038/s41536-022-00238-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 08/10/2022] [Indexed: 11/08/2022] Open
Abstract
Hereditary muscle diseases are disabling disorders lacking effective treatments. UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE) myopathy (GNEM) is an autosomal recessive distal myopathy with rimmed vacuoles typically manifesting in late adolescence/early adulthood. GNE encodes the rate-limiting enzyme in sialic acid biosynthesis, which is necessary for the proper function of numerous biological processes. Outside of the causative gene, very little is known about the mechanisms contributing to the development of GNE myopathy. In the present study, we aimed to address this knowledge gap by querying the underlying mechanisms of GNE myopathy using a patient-derived induced pluripotent stem-cell (iPSC) model. Control and patient-specific iPSCs were differentiated down a skeletal muscle lineage, whereby patient-derived GNEM iPSC clones were able to recapitulate key characteristics of the human pathology and further demonstrated defects in myogenic progression. Single-cell RNA sequencing time course studies revealed clear differences between control and GNEM iPSC-derived muscle precursor cells (iMPCs), while pathway studies implicated altered stress and autophagy signaling in GNEM iMPCs. Treatment of GNEM patient-derived iMPCs with an autophagy activator improved myogenic differentiation. In summary, we report an in vitro, iPSC-based model of GNE myopathy and implicate defective myogenesis as a contributing mechanism to the etiology of GNE myopathy.
Collapse
|
25
|
Yadav R, Devi SS, Oswalia J, Ramalingam S, Arya R. Role of HSP70 chaperone in protein aggregate phenomenon of GNE mutant cells: Therapeutic lead for GNE Myopathy. Int J Biochem Cell Biol 2022; 149:106258. [PMID: 35777599 DOI: 10.1016/j.biocel.2022.106258] [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: 03/22/2022] [Revised: 06/04/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
Limited treatment options and research in understanding the pathomechanisms of rare diseases has raised concerns about their therapeutic development. One such poorly understood ultra-rare neuromuscular disorder is GNE Myopathy (GNEM) which is caused due to mutation in key sialic acid biosynthetic enzyme, GNE. Treatment with sialic acid or its derivatives/precursors slows the disease progression, but curative strategies need to be explored further. Pathologically, muscle biopsy samples of GNEM patients reveal rimmed vacuole formation due to aggregation of β-amyloid, Tau, presenilin proteins with unknown mechanism. The present study aims to understand the mechanism of protein aggregate formation in GNE mutant cells to decipher role of chaperones in disease phenotype. The pathologically relevant GNE mutations expressed as recombinant proteins in HEK cells was used as a model system for GNEM to estimate extent of protein aggregation. We identified HSP70, a chaperone, as binding partner of GNE. Downregulation of HSP70 with altered BAG3, JNK, BAX expression levels was observed in GNE mutant cells. The cell apoptosis was observed in GNE mutation specific manner. An activator of HSP70 chaperone, BGP-15, rescued the phenotypic defects due to GNE mutation, thereby, reducing protein aggregation significantly. The results were further validated in rat skeletal muscle cell lines carrying single Gne allele. Our study suggests that HSP70 activators can be a promising therapeutic target in the treatment of ultra-rare GNE Myopathy disease.
Collapse
Affiliation(s)
- Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | | | - Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | | | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; Special Center for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
26
|
Wu D, Gilormini PA, Toda S, Biot C, Lion C, Guérardel Y, Sato C, Kitajima K. A novel C-domain-dependent inhibition of the rainbow trout CMP-sialic acid synthetase activity by CMP-deaminoneuraminic acid. Biochem Biophys Res Commun 2022; 617:16-21. [DOI: 10.1016/j.bbrc.2022.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
|
27
|
Ertunc N, Phitak T, Wu D, Fujita H, Hane M, Sato C, Kitajima K. Sulfation of sialic acid is ubiquitous and essential for vertebrate development. Sci Rep 2022; 12:12496. [PMID: 35864127 PMCID: PMC9304399 DOI: 10.1038/s41598-022-15143-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/20/2022] [Indexed: 11/09/2022] Open
Abstract
Glycosylation of proteins and lipids occurs in vertebrates, usually terminating with sialylation, which regulates the physicochemical and biological properties of these glycoconjugates. Although less commonly known, sialic acid residues also undergo various modifications, such as acetylation, methylation, and sulfation. However, except for acetylation, the enzymes or functions of the other modification processes are unknown. To the best of our knowledge, this study is the first to demonstrate the ubiquitous occurrence of sulfated sialic acids and two genes encoding the sialate: O-sulfotransferases 1 and 2 in vertebrates. These two enzymes showed about 50% amino acid sequence identity, and appeared to be complementary to each other in acceptor substrate preferences. Gene targeting experiments showed that the deficiency of these genes was lethal for medaka fish during young fry development and accompanied by different phenotypes. Thus, the sulfation of sialic acids is essential for the vertebrate development.
Collapse
Affiliation(s)
- Nursah Ertunc
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Molecular Cell Biology, Faculty of Medical Technology, Graduate School of Health Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Thanyaluck Phitak
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Biochemistry Department, Faculty of Medicine, Chiangmai University, Chiangmai, 50200, Thailand
| | - Di Wu
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Hiroshi Fujita
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan. .,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan.
| |
Collapse
|
28
|
Naito-Matsui Y. Physiological Significance of Animal- and Tissue-specific Sialic Acid Composition. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.2036.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
29
|
Naito-Matsui Y. Physiological Significance of Animal- and Tissue-specific Sialic Acid Composition. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.2036.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
30
|
Liu F, Simpson AB, D'Costa E, Bunn FS, van Leeuwen SS. Sialic acid, the secret gift for the brain. Crit Rev Food Sci Nutr 2022; 63:9875-9894. [PMID: 35531941 DOI: 10.1080/10408398.2022.2072270] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human brain grows rapidly in early life which requires adequate nutrition. Human milk provides optimal nutrition for the developing brain, and breastfeeding significantly improves the cognition development of infants. These benefits have been largely attributed to human milk oligosaccharides (HMOS), associated with sialic acid (Sia). Subsequently, sialylated HMOS present a vital source of exogenous Sia to infants. Sialic acid is a key molecule essential for proper development of gangliosides, and therefore critical in brain development and function. Recent pre-clinical studies suggest dietary supplementation with Sia or sialylated oligosaccharides enhances intelligence and cognition performance in early and later life. Furthermore, emerging evidence suggests the involvement of Sia in brain homeostasis and disbalance correlates with common pathologies such as Alzheimer's disease (AD). Therefore, this review will discuss early brain health and development and the role of Sia in this process. Additionally, studies associating breastfeeding and specific HMOS to benefits in cognitive development are critically assessed. Furthermore, the review will assess studies implying the potential role of HMOS and microbiota in brain development via the gut-brain axis. Finally, the review will summarize recent advances regarding the role of Sia in neurodegenerative disease in later life and potential roles of dietary Sia sources.
Collapse
Affiliation(s)
- Fan Liu
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna Bella Simpson
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Esmée D'Costa
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fanny Sophia Bunn
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander S van Leeuwen
- Department of Laboratory Medicine, Sector Human Nutrition and Health, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Effect of GNE Mutations on Cytoskeletal Network Proteins: Potential Gateway to Understand Pathomechanism of GNEM. Neuromolecular Med 2022; 24:452-468. [PMID: 35503500 DOI: 10.1007/s12017-022-08711-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 04/09/2022] [Indexed: 12/27/2022]
Abstract
GNE myopathy is an inherited neuromuscular disorder caused by mutations in GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetyl mannosamine kinase) gene catalyzing the sialic acid biosynthesis pathway. The characteristic features include muscle weakness in upper and lower extremities, skeletal muscle wasting, and rimmed vacuole formation. More than 200 GNE mutations in either epimerase or kinase domain have been reported worldwide. In Indian subcontinent, several GNE mutations have been recently identified with unknown functional correlation. Alternate role of GNE in various cellular processes such as cell adhesion, migration, apoptosis, protein aggregation, and cytoskeletal organization have been proposed in recent studies. We aim to understand and compare the effect of various GNE mutations from Indian origin on regulation of the cytoskeletal network. In particular, F-actin dynamics was determined quantitatively by determining F/G-actin ratios in immunoblots for specific proteins. The extent of F-actin polymerization was visualized by immunostaining with Phalloidin using confocal microscopy. The proteins regulating F-actin dynamics such as RhoA, cofilin, Arp2, and alpha-actinin were studied in various GNE mutants. The altered level of cytoskeletal organization network proteins affected cell migration of GNE mutant proteins as measured by wound healing assay. The functional comparison of GNE mutations will help in better understanding of the genotypic severity of the disease in the Indian population. Our study offers a potential for identification of therapeutic molecules regulating actin dynamics in GNE specific mutations.
Collapse
|
32
|
Functional characterization of GNE mutations prevalent in Asian subjects with GNE myopathy, an ultra-rare neuromuscular disorder. Biochimie 2022; 199:36-45. [DOI: 10.1016/j.biochi.2022.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 12/19/2022]
|
33
|
Upregulation of sialyltransferases ST3Gal1 and ST6Gal1 promotes stabilization of erythrocyte mass and recovery of anemia in Trypanosoma brucei brucei-infected pigs. Res Vet Sci 2022; 145:102-108. [DOI: 10.1016/j.rvsc.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/14/2022] [Accepted: 02/04/2022] [Indexed: 11/23/2022]
|
34
|
Abstract
Sialic acids are present in humans and other metazoans, playing essential roles in physiological and pathological processes. Commensal and pathogenic bacteria have evolved the capacity to utilize sialic acids as nutrient and energy sources. However, in some actinobacteria, sialic acid catabolism (SAC) is associated with free-living populations. To unravel the distribution and evolutionary history of SAC in the phylum Actinobacteria, we analyzed the presence and diversity of the putative SAC gene cluster (nan) in 7,180 high-quality, nonredundant actinobacterial genomes that covered 1,969 species. The results showed that ∼13% of actinobacterial species had the potential to utilize sialic acids, with 45 species capable of anhydro-SAC, all except two of them through the canonical pathway. These species belonged to 20 orders and 81 genera, with ∼36% of them from four genera, Actinomyces, Bifidobacterium, Corynebacterium, and Streptomyces. Moreover, ∼40% of the nan-positive species are free living. Phylogenetic analysis of the key nan genes, nanA, nanK, and nanE, revealed a strong signal of horizontal gene transfer (HGT), accompanied with vertical inheritance and gene loss. This evolutionary pattern led to high diversity and differential distribution of nan among actinobacterial taxa and might cause the cluster to spread to some free-living species while losing in some host-associated species. The evolution of SAC in actinobacteria probably represents the evolution of certain kinds of noncore bacterial functions for environmental adaptation and lifestyle switch, in which HGT plays a dominant role. IMPORTANCE Sialic acids play essential roles in the physiology of humans and other metazoan animals, and microbial sialic acid catabolism (SAC) is one of the processes critical for pathogenesis. To date, microbial SAC is studied mainly in commensals and pathogens, while its distribution in free-living microbes and evolutionary pathway remain largely unexplored. Here, by examining all actinobacterial genomes available, we demonstrate that putative SAC is present in a small proportion of actinobacterial species, of which, however, ∼40% are free-living species. We also reveal remarkable difference in the distribution of SAC among actinobacterial taxa and high diversity of the putative SAC gene clusters. HGT plays a significant role in the evolution of SAC, accompanied with vertical inheritance and gene loss. Our results provide a comprehensive and systematic picture of the distribution and evolutionary history of SAC in actinobacteria, expanding the current knowledge on bacterial adaptation and diversification.
Collapse
|
35
|
From high-throughput to therapeutic: host-directed interventions against influenza viruses. Curr Opin Virol 2022; 53:101198. [PMID: 35030353 PMCID: PMC9086133 DOI: 10.1016/j.coviro.2021.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Influenza viruses are simultaneously supported and antagonized by factors within the host cell. This close relationship is the theoretical basis for future antivirals that target the host rather than the virus itself, a concept termed host-directed therapeutics. Genetic screening has led to the identification of host factors capable of modulating influenza virus infections, and these factors represent candidate targets for host-directed antiviral strategies. Despite advances in understanding host targets, however, there are currently no host-directed interventions for influenza viruses in clinical use. In this brief review, we discuss some host factors identified in knockout/knockdown and overexpression screens that could potentially be targeted as host-directed influenza intervention strategies. We further comment on the feasibility of changing gene expression in the respiratory tract with RNA delivery vectors and transient CRISPR-mediated gene targeting.
Collapse
|
36
|
Attri S, Sharma V, Kumar A, Verma C, Gahlawat SK. Dissecting role of founder mutation p.V727M in GNE in Indian HIBM cohort. Open Med (Wars) 2021; 16:1733-1744. [PMID: 34825065 PMCID: PMC8593392 DOI: 10.1515/med-2021-0391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
GNE gene-specific c.2179G>A(p.V727M) is a key alteration reported in patients with hereditary inclusion body myopathy (HIBM) and represents an ethnic founder mutation in the Indian cohort. However, the underlying role of this mutation in pathogenesis remains largely unknown. Thus, in this study, we aimed to access possible mechanisms of V727M mutation that could be leading to myopathy. We evaluated various in silico tools to predict the effect of this mutation on pathogenicity, structural or possible interactions, that could induce myopathy. Our results propose that V727M mutation could induce deleterious effects or pathogenicity and affect the stability of GNE protein. Analysis of differential genes reported in the V727 mutant case suggests that it can affect GNE protein interaction with Myc-proto-oncogene (MYC) transcription factor. Our in silico analysis also suggests a possible interaction between GNE ManNac-kinase domain with MYC protein at the C-terminal DNA-binding domain. MYC targets reported in skeletal muscles via ChIP-seq suggest that it plays a key role in regulating the expression of many genes reported differentially expressed in V727M-mutated HIBMs. We conclude that V727M mutation could alter the interaction of GNE with MYC thereby altering transcription of sialyltransferase and neuromuscular genes, thus understanding these effects could pave the way for developing effective therapies against HIBM.
Collapse
Affiliation(s)
- Shivangi Attri
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, 440002, India
| | - Vikas Sharma
- General Facility, Centralized Core Research Facility, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Amit Kumar
- ICMR-AIIMS Computational Genomics Centre, Division of Biomedical Informatics, Indian Council of Medical Research, New Delhi, 110029, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, The Ohio State University, OH-43210, Ohio, United States of America
| | - Suresh Kumar Gahlawat
- Department of Biotechnology, Chaudhary Devi Lal University, Sirsa, Haryana, 125055, India
| |
Collapse
|
37
|
Mitrani-Rosenbaum S, Yakovlev L, Becker Cohen M, Argov Z, Fellig Y, Harazi A. Pre Clinical Assessment of AAVrh74.MCK.GNE Viral Vector Therapeutic Potential: Robust Activity Despite Lack of Consistent Animal Model for GNE Myopathy. J Neuromuscul Dis 2021; 9:179-192. [PMID: 34806613 PMCID: PMC8842764 DOI: 10.3233/jnd-210755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: GNE myopathy is a unique adult onset rare neuromuscular disease caused by recessive mutations in the GNE gene. The pathophysiological mechanism of this disorder is not well understood and to date, there is no available therapy for this debilitating disease. We have previously established proof of concept that AAV based gene therapy can effectively deliver the wild type human GNE into cultured muscle cells from human patients and in mice, using a CMV promoter driven human wild type GNE plasmid delivered through an adeno associated virus (AAV8) based platform. Objective: In the present study we have generated a muscle specific GNE construct, driven by the MCK promoter and packaged with the AAVrh74 serotype for efficacy evaluation in an animal model of GNE Myopathy. Methods: The viral vector was systemically delivered at 2 doses to two age groups of a Gne–/– hGNED207V Tg mouse described as a preclinical model of GNE Myopathy, and treatment was monitored for long term efficacy. Results: In spite of the fact that the full described characteristics of the preclinical model could not be reproduced, the systemic injection of the rAAVrh74.MCK.GNE viral vector resulted in a long term presence and expression of human wt GNE in the murine muscles and in some improvements of their mild phenotype. The Gne–/– hGNED207V Tg mice are smaller from birth, but cannot be differentiated from littermates by muscle function (grip strength and Rotarod) and their muscle histology is normal, even at advanced age. Conclusions: The rAAVrh74.MCK.GNE vector is a robust tool for the development of GNE Myopathy therapies that supply the intact GNE. However, there is still no reliable animal model to fully assess its efficacy since the previously developed Gne–/– hGNED207V Tg mice do not present disease characteristics.
Collapse
Affiliation(s)
- Stella Mitrani-Rosenbaum
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lena Yakovlev
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Becker Cohen
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zohar Argov
- Department of Neurology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yakov Fellig
- Department of Pathology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avi Harazi
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
38
|
Zhang KY, Duan HQ, Li QX, Luo YB, Bi FF, Huang K, Yang H. Expanding the clinicopathological-genetic spectrum of GNE myopathy by a Chinese neuromuscular centre. J Cell Mol Med 2021; 25:10494-10503. [PMID: 34676965 PMCID: PMC8581342 DOI: 10.1111/jcmm.16978] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/19/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
GNE myopathy is a heterogeneous group of ultrarare neuromuscular disorders caused by mutations in the GNE gene. An estimated prevalence of 1~21/1,000,000 leads to a deficiency of data and a lack of availability of samples to conduct clinical research on this neuromuscular disorder. Although GNE, which is the mutated gene responsible for the disease, is well known as the key enzyme in the biosynthesis pathway of sialic acid, the clinicopathological-genetic spectrum of GNE mutant patients is still unclear and expanding. This study presents ten unrelated patients with GNE myopathy, discovering five novel missense mutations. Clinical, electrophysiological, imaging, pathological and genetic data are presented in a retrospective manner. Interestingly, several patients in the cohort were found to have peripheral neuropathy and inflammatory cell infiltration in muscle biopsies, which have seldom been reported. This study, conducted by a neuromuscular centre in China, is the first attempt to highlight these abnormal clinicopathological features and associate them with genetic mutations in GNE myopathy.
Collapse
Affiliation(s)
- Kai-Yue Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Clinic Medicine of 8-year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Qian Duan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Xiang Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yue-Bei Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Fang-Fang Bi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Kun Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Devi SS, Yadav R, Mashangva F, Chaudhary P, Sharma S, Arya R. Generation and Characterization of a Skeletal Muscle Cell-Based Model Carrying One Single Gne Allele: Implications in Actin Dynamics. Mol Neurobiol 2021; 58:6316-6334. [PMID: 34510381 DOI: 10.1007/s12035-021-02549-w] [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: 03/01/2021] [Accepted: 08/28/2021] [Indexed: 12/13/2022]
Abstract
UDP-N-Acetyl glucosamine-2 epimerase/N-acetyl mannosamine kinase (GNE) catalyzes key enzymatic reactions in the biosynthesis of sialic acid. Mutation in GNE gene causes GNE myopathy (GNEM) characterized by adult-onset muscle weakness and degeneration. However, recent studies propose alternate roles of GNE in other cellular processes beside sialic acid biosynthesis, particularly interaction of GNE with α-actinin 1 and 2. Lack of appropriate model system limits drug and treatment options for GNEM as GNE knockout was found to be embryonically lethal. In the present study, we have generated L6 rat skeletal muscle myoblast cell-based model system carrying one single Gne allele where GNE gene is knocked out at exon-3 using AAV mediated SEPT homology recombination (SKM-GNEHz). The cell line was heterozygous for GNE gene with one wild type and one truncated allele as confirmed by sequencing. The phenotype showed reduced GNE epimerase activity with little reduction in sialic acid content. In addition, the heterozygous GNE knockout cells revealed altered cytoskeletal organization with disrupted actin filament. Further, we observed increased levels of RhoA leading to reduced cofilin activity and causing reduced F-actin polymerization. The disturbed signaling cascade resulted in reduced migration of SKM-GNEHz cells. Our study indicates possible role of GNE in regulating actin dynamics and cell migration of skeletal muscle cell. The skeletal muscle cell-based system offers great potential in understanding pathomechanism and target identification for GNEM.
Collapse
Affiliation(s)
| | - Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | | | - Priyanka Chaudhary
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | - Shweta Sharma
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India. .,Special Centre for Systems Medicine (Concurrent Faculty), Jawaharlal Nehru University, New Mehrauli Road, 110067, New Delhi, India.
| |
Collapse
|
40
|
Crowe KE, Zygmunt DA, Heller K, Rodino-Klapac L, Noguchi S, Nishino I, Martin PT. Visualizing Muscle Sialic Acid Expression in the GNED207VTgGne-/- Cmah-/- Model of GNE Myopathy: A Comparison of Dietary and Gene Therapy Approaches. J Neuromuscul Dis 2021; 9:53-71. [PMID: 34511508 DOI: 10.3233/jnd-200575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND GNE myopathy (GNEM) is a rare, adult-onset, inclusion body myopathy that results from partial loss of function mutations in the GNE gene. GNE encodes UDP-GlcNAc epimerase/Mannose-6 kinase, a protein with two enzymatic activities that comprise the committed step in biosynthesis of sialic acid (SA), an essential glycan that appears on the terminal positions of many extracellular oligosaccharide chains. These GNE mutations can cause a reduction of SA in many tissues, although pathology is restricted to skeletal muscles through a poorly understood mechanism. OBJECTIVE Despite recent advances in the field, it remains unclear which therapeutic avenue is most promising for the restoration of SA level in skeletal muscle affected by GNEM. Our objective was to assess dietary and gene therapy strategies for GNEM in Cmah-deficient GNED207VTgGne-/- mice, a model that allows for the visualization of orally delivered N-glycolylneuraminic acid (Neu5Gc), one of the two predominant SA forms in muscle. METHODS Methods included in situ physiology studies of the tibialis anterior muscle, studies of ambulation and limb grip strength, and muscle staining using MAA, SNA, and anti-Neu5Gc antibody, along with qPCR, qRT-PCR, western blot, and HPLC studies to assess virally introduced DNA, GNE gene expression, GNE protein expression, and SA expression. RESULTS We found that a diet enriched in Neu5Gc-containing glycoproteins had no impact on Neu5Gc immunostaining in muscles of GNEM model mice. Delivery of a single high dose oral Neu5Gc therapy, however, did increase Neu5Gc immunostaining, though to levels below those found in wild type mice. Delivery of a single dose of GNE gene therapy using a recombinant Adeno Associated Virus (rAAV) vector with a liver-specific or a muscle-specific promoter both caused increased muscle Neu5Gc immunostaining that exceeded that seen with single dose monosaccharide therapy. CONCLUSIONS Our findings indicate that dietary loading of Neu5Gc-containing glycoproteins is not effective in increasing muscle Neu5Gc expression, while single dose oral Neu5Gc monosaccharide or GNE gene therapy are. Neu5Gc immunostaining, however, showed greater changes than did lectin staining or HPLC analysis. Taken together, these results suggest that Neu5Gc immunostaining may be more sensitive technique to follow SA expression than other more commonly used methods and that liver expression of GNE may contribute overall muscle SA content.
Collapse
Affiliation(s)
- Kelly E Crowe
- Department of Biology, Mount St. Joseph University Cincinnati, OH, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute Children's Drive Columbus, OH, USA
| | - Kristin Heller
- Center for Gene Therapy, Abigail Wexner Research Institute Children's Drive Columbus, OH, USA
| | - Louise Rodino-Klapac
- Center for Gene Therapy, Abigail Wexner Research Institute Children's Drive Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine Columbus, OH, USA
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience Tokyo, Japan
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute Children's Drive Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine Columbus, OH, USA
| |
Collapse
|
41
|
Passaponti S, Pavone V, Cresti L, Ietta F. The expression and role of glycans at the feto-maternal interface in humans. Tissue Cell 2021; 73:101630. [PMID: 34454366 DOI: 10.1016/j.tice.2021.101630] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022]
Abstract
During pregnancy, both the maternal endometrium and the blastocyst have highly glycosylated proteins with glycosylations controlled in a specific manner. Carbohydrates play a fundamental role in cell-cell and cell-matrix recognition and are involved in defining the structure and integrity of tissues. The uterus' secretions, which are rich in glycoproteins and glycogen and the presence of a functional glycocalyx on the uterine epithelium, establish a favourable milieu, which is essential for the correct implantation and subsequent development of the blastocyst. Likewise, carbohydrate residues such as fucose and sialic acid present at the placental level are determinant in creating an immuno-environment, which supports the mother's tolerance towards the fetal antigens. In this review, we explore the literature concerning the role of important glycan-epitopes at the feto-maternal interface in the human species. Moreover, we also show some unpublished interesting results on changes of glycan residues in human placenta tissues from the first trimester of pregnancy.
Collapse
Affiliation(s)
- Sofia Passaponti
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Valentina Pavone
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Laura Cresti
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| |
Collapse
|
42
|
Jahan M, Francis N, Wynn P, Wang B. The Potential for Sialic Acid and Sialylated Glycoconjugates as Feed Additives to Enhance Pig Health and Production. Animals (Basel) 2021; 11:ani11082318. [PMID: 34438776 PMCID: PMC8388453 DOI: 10.3390/ani11082318] [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: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary This review discusses the current challenges in the pig industry and the potential nutritional significance of sialic acid (Sia) and glycoconjugates (Sia-GC’s) for pig health and nutrition. Sia is a nine-carbon acidic sugar which is present in various organs and body fluids of humans and animals. Sias contribute to many beneficial biological functions including pathogen resistance, immunomodulation, gut microbiota development, gut maturation, anti-inflammation and neurodevelopment. The role of Sias in regulating the metabolism of pigs has seldom been reported. However, we have documented significant beneficial effects of specific Sia-GC’s on health and production performance of sows and piglets. These findings are reviewed in relation to other studies while noting the beneficial effects of the inclusion of Sia, Sia containing oligosaccharide or the sialo-protein lactoferrin in the diets of gilts and sows. The importance of the passive transfer of of Sia and Sia-GC’s through milk to the young and the implications for their growth and development is also reviewed. This information will assist in optimizing the composition of sow/gilt milk replacers designed to increases the survival of IUGR piglets or piglets with dams suffering from agalactia, a common problem in pig production systems worldwide. Abstract Swine are one of the most important agricultural species for human food production. Given the significant disease challenges confronting commercial pig farming systems, introduction of a new feed additive that can enhance animal performance by improving growth and immune status represents a major opportunity. One such candidate is sialic acid (Sia), a diverse family of nine-carbon acidic sugar, present in various organs and body fluid, as well as an essential structural and functional constituent of brain ganglioside of humans and animals. Sias are key monosaccharide and biomarker of sialylated milk oligosaccharide (Sia-MOS’s), sialylated glycoproteins and glycolipids in milk and all vertebrate cells. Sias accomplish many critical endogenous functions by virtue of their physiochemical properties and via recognition by intrinsic receptors. Human milk sialylated glycoconjugates (Sia-GC’s) are bioactive compounds known to act as prebiotics that promote gut microbiota development, gut maturation, pathogen resistance, immunomodulation, anti-inflammation and neurodevelopment. However, the importance of Sia in pig health, especially in the growth, development, immunity of developing piglet and in pig production remains unknown. This review aims to critically discuss the current status of knowledge of the biology and nutritional role of Sia and Sia-GC’s on health of both female sow and newborn piglets.
Collapse
Affiliation(s)
| | | | | | - Bing Wang
- Correspondence: ; Tel.: +61-2-6933-4549
| |
Collapse
|
43
|
Serum Antibodies to N-Glycolylneuraminic Acid Are Elevated in Duchenne Muscular Dystrophy and Correlate with Increased Disease Pathology in Cmah -/-mdx Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1474-1486. [PMID: 34294193 DOI: 10.1016/j.ajpath.2021.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 10/20/2022]
Abstract
Humans cannot synthesize the common mammalian sialic acid N-glycolylneuraminic acid (Neu5Gc) because of an inactivating deletion in the cytidine-5'-monophospho-(CMP)-N-acetylneuraminic acid hydroxylase (CMAH) gene responsible for its synthesis. Human Neu5Gc deficiency can lead to development of anti-Neu5Gc serum antibodies, the levels of which can be affected by Neu5Gc-containing diets and by disease. Metabolic incorporation of dietary Neu5Gc into human tissues in the face of circulating antibodies against Neu5Gc-bearing glycans is thought to exacerbate inflammation-driven diseases like cancer and atherosclerosis. Probing of sera with sialoglycan arrays indicated that patients with Duchenne muscular dystrophy (DMD) had a threefold increase in overall anti-Neu5Gc antibody titer compared with age-matched controls. These antibodies recognized a broad spectrum of Neu5Gc-containing glycans. Human-like inactivation of the Cmah gene in mice is known to modulate severity in a variety of mouse models of human disease, including the X chromosome-linked muscular dystrophy (mdx) model for DMD. Cmah-/-mdx mice can be induced to develop anti-Neu5Gc-glycan antibodies as humans do. The presence of anti-Neu5Gc antibodies, in concert with induced Neu5Gc expression, correlated with increased severity of disease pathology in Cmah-/-mdx mice, including increased muscle fibrosis, expression of inflammatory markers in the heart, and decreased survival. These studies suggest that patients with DMD who harbor anti-Neu5Gc serum antibodies might exacerbate disease severity when they ingest Neu5Gc-rich foods, like red meats.
Collapse
|
44
|
Safety and efficacy of N-acetylmannosamine (ManNAc) in patients with GNE myopathy: an open-label phase 2 study. Genet Med 2021; 23:2067-2075. [PMID: 34257421 PMCID: PMC8553608 DOI: 10.1038/s41436-021-01259-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 01/09/2023] Open
Abstract
Purpose To evaluate the safety and efficacy of N-acetylmannosamine (ManNAc) in GNE myopathy, a genetic muscle disease caused by deficiency of the rate-limiting enzyme in N-acetylneuraminic acid (Neu5Ac) biosynthesis. Methods We conducted an open-label, phase 2, single-center (NIH, USA) study to evaluate oral ManNAc in 12 patients with GNE myopathy (ClinicalTrials.gov NCT02346461). Primary endpoints were safety and biochemical efficacy as determined by change in plasma Neu5Ac and sarcolemmal sialylation. Clinical efficacy was evaluated using secondary outcome measures as part of study extensions, and a disease progression model (GNE-DPM) was tested as an efficacy analysis method. Results Most drug-related adverse events were gastrointestinal, and there were no serious adverse events. Increased plasma Neu5Ac (+2,159 nmol/L, p < 0.0001) and sarcolemmal sialylation (p = 0.0090) were observed at day 90 compared to baseline. A slower rate of decline was observed for upper extremity strength (p = 0.0139), lower extremity strength (p = 0.0006), and the Adult Myopathy Assessment Tool (p = 0.0453), compared to natural history. Decreased disease progression was estimated at 12 (γ = 0.61 [95% CI: 0.09, 1.27]) and 18 months (γ = 0.55 [95% CI: 0.12, 1.02]) using the GNE-DPM. Conclusion ManNAc showed long-term safety, biochemical efficacy consistent with the intended mechanism of action, and preliminary evidence clinical efficacy in patients with GNE myopathy.
Collapse
|
45
|
Wedekind H, Kats E, Weiss AC, Thiesler H, Klaus C, Kispert A, Horstkorte R, Neumann H, Weinhold B, Münster-Kühnel A, Abeln M. Gne deletion in mice leads to lethal intracerebral hemorrhage during embryonic development. Glycobiology 2021; 31:1478-1489. [PMID: 34224569 DOI: 10.1093/glycob/cwab069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 11/15/2022] Open
Abstract
Among the enzymes of the biosynthesis of sialoglycoconjugates, UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE), catalyzing the first essential step of the sialic acid (Sia) de novo biosynthesis, and CMP-Sia synthase (CMAS), activating Sia to CMP-Sia, are particularly important. The knockout of either of these enzymes in mice is embryonically lethal. While the lethality of Cmas-/- mice has been attributed to a maternal complement attack against asialo fetal placental cells, the cause of lethality in Gne-deficient embryos has remained elusive. Here, we advanced the significance of sialylation for embryonic development through detailed histological analyses of Gne-/- embryos and placentae. We found that Gne-/- embryonic and extraembryonic tissues are hyposialylated, rather than completely deficient of sialoglycans which holds true for Cmas-/- embryos. Residual sialylation of Gne-/- cells can be explained by scavenging free Sia from sialylated maternal serum glycoconjugates via the lysosomal salvage pathway. The placental architecture of Gne-/- mice was unaffected, but severe hemorrhages in the neuroepithelium with extensive bleeding into the cephalic ventricles were present at E12.5 in the mutants. At E13.5, the vast majority of Gne-/- embryos were asystolic. This phenotype persisted when Gne-/- mice were backcrossed to a complement component 3-deficient background, confirming distinct pathomechanisms of Cmas-/- and Gne-/- mice. We conclude that the low level of sialylation observed in Gne-/- mice is sufficient, both for immune homeostasis at the fetal-maternal interface and for embryonic development until E12.5. However, formation of the neural microvasculature is the first critical process depending on a higher degree of sialylation during development of the embryo proper.
Collapse
Affiliation(s)
- Henri Wedekind
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Elina Kats
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anna-Carina Weiss
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Christine Klaus
- Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Rüdiger Horstkorte
- Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Germany
| | - Birgit Weinhold
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anja Münster-Kühnel
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Markus Abeln
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
46
|
Khamirani HJ, Zoghi S, Faghihi F, Dastgheib SA, Hassanipour H, Bagher Tabei SM, Mohammadi S, Masoudi M, Poorang S, Ehsani E, Dianatpour M. Phenotype of ST3GAL3 deficient patients: A case and review of the literature. Eur J Med Genet 2021; 64:104250. [PMID: 34022416 DOI: 10.1016/j.ejmg.2021.104250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/19/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022]
Abstract
ST3GAL3 deficiency is an extremely rare autosomal recessive disorder caused by pathogenic mutations in the ST3GAL3 gene. Epilepsy, motor development delay, severe intellectual disability, and behavioral disorders have been reported to be associated with ST3GAL3 deficiency. In the present study, ST3GAL3 deficiency was caused by a homozygous splice-site mutation (NM_174964.4: c.936+1delG) in ST3GAL3. The patient described in this study was clinically similar to previously reported cases; nevertheless, we were able to detect repetitive behavior, previously not reported manifestations.
Collapse
Affiliation(s)
- Hossein Jafari Khamirani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran; Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Zoghi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Faghihi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | | | - Seyed Mohammad Bagher Tabei
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran; Maternal-fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Mohammadi
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Masoudi
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Poorang
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Ehsani
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran; Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
47
|
Leite-Lima F, Bastos VC, Vitório JG, Duarte-Andrade FF, Pereira TDSF, Martins-Chaves RR, Cruz AF, de Lacerda JCT, Lebron YAR, Moreira VR, Santos LVDS, Lange LC, de Macedo AN, Diniz MG, Gomes CC, de Castro WH, Canuto GAB, Gomez RS. Unveiling metabolic changes in marsupialized odontogenic keratocyst: A pilot study. Oral Dis 2021; 28:2219-2229. [PMID: 33978981 DOI: 10.1111/odi.13913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/08/2021] [Accepted: 05/05/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE We aimed to assess which metabolic pathways would be implicated in the phenotypic changes of the epithelial lining of odontogenic keratocyst after marsupialization, comparing pre- and post-marsupialized lesions with adjacent oral mucosa. MATERIALS AND METHODS Eighteen formalin-fixed and paraffin-embedded tissues from six subjects were divided into three paired groups: odontogenic keratocyst pre- (n = 6) and post-marsupialization (n = 6), and adjacent oral mucosa (n = 6). The metabolic pathways found in these groups were obtained by high-performance liquid chromatography-mass spectrometry-based untargeted metabolomics performed. RESULTS Through putative metabolite annotation followed by pathway enrichment and predictive analysis with automated algorithms (Mummichog and Gene Set Enrichment Analysis), we found differences in many cellular processes that may be involved in inflammation, oxidative stress response, keratinocyte-basal membrane attachment, differentiation, and proliferation functions, all relevant to odontogenic keratocyst pathobiology and the phenotype acquired after marsupialization. CONCLUSION Our study was able to identify several metabolic pathways potentially involved in the metaplastic changes induced by marsupialization of odontogenic keratocysts. An improved comprehension of this process could pave the way for the development of targeted therapies.
Collapse
Affiliation(s)
- Flávia Leite-Lima
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Victor Coutinho Bastos
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Jéssica Gardone Vitório
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Filipe Fideles Duarte-Andrade
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thaís Dos Santos Fontes Pereira
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Roberta Rayra Martins-Chaves
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Aline Fernanda Cruz
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Júlio César Tanos de Lacerda
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Yuri Abner Rocha Lebron
- Department of Sanitation and Environmental Engineering, School of Engineering, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Victor Rezende Moreira
- Department of Sanitation and Environmental Engineering, School of Engineering, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucilaine Valéria de Souza Santos
- Department of Sanitation and Environmental Engineering, School of Engineering, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Liséte Celina Lange
- Department of Sanitation and Environmental Engineering, School of Engineering, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Adriana Nori de Macedo
- Department of Chemistry, Exact Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marina Gonçalves Diniz
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Carolina Cavaliéri Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Wagner Henriques de Castro
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gisele André Baptista Canuto
- Department of Analytical Chemistry, Institute of Chemistry, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
48
|
Rawal P, Zhao L. Sialometabolism in Brain Health and Alzheimer's Disease. Front Neurosci 2021; 15:648617. [PMID: 33867926 PMCID: PMC8044809 DOI: 10.3389/fnins.2021.648617] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acids refer to a unique family of acidic sugars with a 9-carbon backbone that are mostly found as terminal residues in glycan structures of glycoconjugates including both glycoproteins and glycolipids. The highest levels of sialic acids are expressed in the brain where they regulate neuronal sprouting and plasticity, axon myelination and myelin stability, as well as remodeling of mature neuronal connections. Moreover, sialic acids are the sole ligands for microglial Siglecs (sialic acid-binding immunoglobulin-type lectins), and sialic acid-Siglec interactions have been indicated to play a critical role in the regulation of microglial homeostasis in a healthy brain. The recent discovery of CD33, a microglial Siglec, as a novel genetic risk factor for late-onset Alzheimer's disease (AD), highlights the potential role of sialic acids in the development of microglial dysfunction and neuroinflammation in AD. Apart from microglia, sialic acids have been found to be involved in several other major changes associated with AD. Elevated levels of serum sialic acids have been reported in AD patients. Alterations in ganglioside (major sialic acid carrier) metabolism have been demonstrated as an aggravating factor in the formation of amyloid pathology in AD. Polysialic acids are linear homopolymers of sialic acids and have been implicated to be an important regulator of neurogenesis that contributes to neuronal repair and recovery from neurodegeneration such as in AD. In summary, this article reviews current understanding of neural functions of sialic acids and alterations of sialometabolism in aging and AD brains. Furthermore, we discuss the possibility of looking at sialic acids as a promising novel therapeutic target for AD intervention.
Collapse
Affiliation(s)
- Punam Rawal
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
49
|
Devi SS, Yadav R, Arya R. Altered Actin Dynamics in Cell Migration of GNE Mutant Cells. Front Cell Dev Biol 2021; 9:603742. [PMID: 33816461 PMCID: PMC8012676 DOI: 10.3389/fcell.2021.603742] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/09/2021] [Indexed: 11/27/2022] Open
Abstract
Cell migration is an essential cellular process that requires coordination of cytoskeletal dynamics, reorganization, and signal transduction. The actin cytoskeleton is central in maintaining the cellular structure as well as regulating the mechanisms of cell motility. Glycosylation, particularly sialylation of cell surface proteins like integrins, regulates signal transduction from the extracellular matrix to the cytoskeletal network. The activation of integrin by extracellular cues leads to recruitment of different focal adhesion complex proteins (Src, FAK, paxillin, etc.) and activates the signal including Rho GTPases for the regulation of actin assembly and disassembly. During cell migration, the assembly and disassembly of actin filament provides the essential force for the cell to move. Abnormal sialylation can lead to actin signaling dysfunction leading to aberrant cell migration, one of the main characteristics of cancer and myopathies. In the present study, we have reported altered F-actin to G-actin ratios in GNE mutated cells. These cells exhibit pathologically relevant mutations of GNE (UDP N-acetylneuraminic 2-epimerase/N-acetylmannosamine kinase), a key sialic acid biosynthetic enzyme. It was found that GNE neither affects the actin polymerization nor binds directly to actin. However, mutation in GNE resulted in increased binding of α-actinin to actin filaments. Further, through confocal imaging, GNE was found to be localized in focal adhesion complex along with paxillin. We further elucidated that mutation in GNE resulted in upregulation of RhoA protein and Cofilin activity is downregulated, which could be rescued with Rhosin and chlorogenic acid, respectively. Lastly, mutant in GNE reduced cell migration as implicated from wound healing assay. Our study indicates that molecules altering Cofilin function could significantly revert the cell migration defect due to GNE mutation in sialic acid-deficient cells. We propose cytoskeletal proteins to be alternate drug targets for disorders associated with GNE such as GNE myopathy.
Collapse
Affiliation(s)
| | - Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
50
|
Spatial N-glycomics of the human aortic valve in development and pediatric endstage congenital aortic valve stenosis. J Mol Cell Cardiol 2021; 154:6-20. [PMID: 33516683 DOI: 10.1016/j.yjmcc.2021.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022]
Abstract
Congenital aortic valve stenosis (AS) progresses as an obstructive narrowing of the aortic orifice due to deregulated extracellular matrix (ECM) production by aortic valve (AV) leaflets and leads to heart failure with no effective therapies. Changes in glycoprotein and proteoglycan distribution are a hallmark of AS, yet valvular carbohydrate content remains virtually uncharacterized at the molecular level. While almost all glycoproteins clinically linked to stenotic valvular modeling contain multiple sites for N-glycosylation, there are very few reports aimed at understanding how N-glycosylation contributes to the valve structure in disease. Here, we tested for spatial localization of N-glycan structures within pediatric congenital aortic valve stenosis. The study was done on valvular tissues 0-17 years of age with de-identified clinical data reporting pre-operative valve function spanning normal development, aortic valve insufficiency (AVI), and pediatric endstage AS. High mass accuracy imaging mass spectrometry (IMS) was used to localize N-glycan profiles in the AV structure. RNA-Seq was used to identify regulation of N-glycan related enzymes. The N-glycome was found to be spatially localized in the normal aortic valve, aligning with fibrosa, spongiosa or ventricularis. In AVI diagnosed tissue, N-glycans localized to hypertrophic commissures with increases in pauci-mannose structures. In all valve types, sialic acid (N-acetylneuraminic acid) N-glycans were the most abundant N-glycan group. Three sialylated N-glycans showed common elevation in AS independent of age. On-tissue chemical methods optimized for valvular tissue determined that aortic valve tissue sialylation shows both α2,6 and α2,3 linkages. Specialized enzymatic strategies demonstrated that core fucosylation is the primary fucose configuration and localizes to the normal fibrosa with disparate patterning in AS. This study identifies that the human aortic valve structure is spatially defined by N-glycomic signaling and may generate new research directions for the treatment of human aortic valve disease.
Collapse
|