1
|
Li Y, Ma R, Qi R, Li H, Li J, Liu W, Wan Y, Li S, Sun Z, Xu J, Zhan K. Novel insight into the feed conversion ratio in laying hens and construction of its prediction model. Poult Sci 2024; 103:104013. [PMID: 39098296 PMCID: PMC11345651 DOI: 10.1016/j.psj.2024.104013] [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: 03/04/2024] [Revised: 04/16/2024] [Accepted: 06/19/2024] [Indexed: 08/06/2024] Open
Abstract
Feed efficiency (FE) is an important economic factor in poultry production, and feed conversion ratio (FCR) is one of the most widely used measures of FE. Factors associated with FCR include genetics, the environment, and other factors. However, the mechanisms responsible for FCR in chickens are still less well appreciated. In this study, we examined the pattern changes of FCR, then delved into understanding the mechanisms behind these variations from both genetic and environmental perspectives. Most interestingly, the FCR at the front section of henhouse exhibited the lowest value. Further investigation revealed that laying rate in the high FCR (HFCR) group was lower than that in the low FCR (LFCR) group (P < 0.05). Cortisol, total antioxidant capacity (TAOC), and IgG levels in the LFCR group were significantly lower than those in the HFCR group (P < 0.05), while BUN level was significantly higher than that in the HFCR group (P < 0.05). We identified a total of 67 and 10 differentially expressed genes (DEGs) associated with FCR in ovarian and small intestine tissues, respectively. Functional enrichment analysis of DEGs revealed that they might affect FCR by modulating genes associated with salivary secretion, ferroptosis, and mineral absorption. Moreover, values for relative humidity (RH), air velocity (AV), PM2.5, ammonia (NH3), and carbon dioxide (CO2) in the LFCR group were significantly lower than those in the HFCR group (P < 0.05). Conversely, value for light intensity (LI) in the LFCR group was significantly higher than that in the HFCR group (P < 0.05). Correlation analysis revealed a positive correlation between FCR and RH, AV, PM2.5, NH3, and CO2, and a negative correlation with LI. Finally, the FCR prediction model was successfully constructed based on multiple environmental variables using the random forest algorithm, providing a valuable tool for predicting FCR in chickens.
Collapse
Affiliation(s)
- Yan Li
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Ruiyu Ma
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Renrong Qi
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Hualong Li
- Advanced Manufacturing Technology Research Center of Institute of Intelligent Machines, Hefei Institute of Physical Science, CAS, Hefei 230001 China
| | - Junying Li
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Wei Liu
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Yi Wan
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Sanjun Li
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Zhen Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036 Anhui, China
| | - Jiechi Xu
- Qianshan Tiansheng Agricultural Ecological Technology Development Co. Ltd, Qianshan, Anhui 246300, China
| | - Kai Zhan
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China.
| |
Collapse
|
2
|
Yang JC, Zhao M, Chernikova D, Arias-Jayo N, Zhou Y, Situ J, Gutta A, Chang C, Liang F, Lagishetty V, Jacobs JP. ZIP8 A391T Crohn's Disease-Linked Risk Variant Induces Colonic Metal Ion Dyshomeostasis, Microbiome Compositional Shifts, and Inflammation. Dig Dis Sci 2024; 69:3760-3772. [PMID: 39322808 PMCID: PMC11489278 DOI: 10.1007/s10620-024-08647-8] [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: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND The pathogenesis of Crohn's disease involves genetic and environmental factors, with the gut microbiome playing a crucial role. The Crohn's disease-associated variant rs13107325 in the SLC39A8 gene results in an A391T substitution in the ZIP8 metal ion transporter and has previously been linked to alterations in the colonic microbiome in variant carriers. We hypothesized that the A391T substitution alters metal ion homeostasis in the colonic mucosal-luminal interface, thereby inducing dysbiosis which may promote intestinal inflammation. METHODS To evaluate this hypothesis, we generated a SLC39A8 A393T mouse model (matching human A391T). We first examined trace element abundance in the colonic mucosal epithelium and lumen of homozygous A393T and wild-type (WT) mice to determine if the variant affected metal distribution. We also performed 16S rRNA gene sequencing on colon samples at 2 months, 3-4 months, and 12 months of age, and conducted histological scoring of colon tissue collected from 5-month and 10-month old mice. RESULTS Consistent with an effect of the variant on ZIP8 function, homozygous A393T mice exhibited increased cobalt in the colonic mucosa, but reduced iron, zinc, manganese, cobalt, copper, and cadmium in the colonic lumen. 16S rRNA gene sequencing of colon samples revealed variant-linked effects on microbiome beta diversity in 2-month-, 3-4-month-, and 12-month-old mice. Histological scoring showed spontaneous intestinal inflammation in 10-month but not in 5-month-old mice. Lastly, predicted pathway analysis of the microbiome samples revealed differential enrichment of iron-, zinc-, and cobalt-dependent pathways in A393T mice compared to wild-type controls. CONCLUSION These results suggest that the variant in SLC39A8 primarily restricts metal availability to the microbiota, resulting in compositions that can adapt to the environment and that A393T-linked dysbiosis occurs prior to the onset of inflammation. This study paves the way for future studies investigating risk variants as microbiome-disease modifiers.
Collapse
Affiliation(s)
- Julianne C Yang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Matthew Zhao
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Diana Chernikova
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90073, USA
| | - Nerea Arias-Jayo
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Yi Zhou
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- West China Hospital, Sichuan University, Chengdu, China
| | - Jamilla Situ
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Arjun Gutta
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Candace Chang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Fengting Liang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
- Goodman-Luskin Microbiome Center, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
3
|
Xie Y, Zhao Y, Zhou Y, Jiang Y, Zhang Y, Du J, Cai M, Fu J, Liu H. Shared Genetic Architecture Among Gastrointestinal Diseases, Schizophrenia, and Brain Subcortical Volumes. Schizophr Bull 2024; 50:1243-1254. [PMID: 38973257 PMCID: PMC11349026 DOI: 10.1093/schbul/sbae099] [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] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND HYPOTHESIS The gut-brain axis plays important roles in both gastrointestinal diseases (GI diseases) and schizophrenia (SCZ). Moreover, both GI diseases and SCZ exhibit notable abnormalities in brain subcortical volumes. However, the genetic mechanisms underlying the comorbidity of these diseases and the shared alterations in brain subcortical volumes remain unclear. STUDY DESIGN Using the genome-wide association studies data of SCZ, 14 brain subcortical volumes, and 8 GI diseases, the global polygenic overlap and local genetic correlations were identified, as well as the shared genetic variants among those phenotypes. Furthermore, we conducted multi-trait colocalization analyses to bolster our findings. Functional annotations, cell-type enrichment, and protein-protein interaction (PPI) analyses were carried out to reveal the critical etiology and pathology mechanisms. STUDY RESULTS The global polygenic overlap and local genetic correlations informed the close relationships between SCZ and both GI diseases and brain subcortical volumes. Moreover, 84 unique lead-shared variants were identified. The associated genes were linked to vital biological processes within the immune system. Additionally, significant correlations were observed with key immune cells and the PPI analysis identified several histone-associated hub genes. These findings highlighted the pivotal roles played by the immune system for both SCZ and GI diseases, along with the shared alterations in brain subcortical volumes. CONCLUSIONS These findings revealed the shared genetic architecture contributing to SCZ and GI diseases, as well as their shared alterations in brain subcortical volumes. These insights have substantial implications for the concurrent development of intervention and therapy targets for these diseases.
Collapse
Affiliation(s)
- Yingying Xie
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yao Zhao
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujing Zhou
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yurong Jiang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujie Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaojiao Du
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengjing Cai
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huaigui Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Taskozhina G, Batyrova G, Umarova G, Issanguzhina Z, Kereyeva N. The Manganese-Bone Connection: Investigating the Role of Manganese in Bone Health. J Clin Med 2024; 13:4679. [PMID: 39200820 PMCID: PMC11355939 DOI: 10.3390/jcm13164679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The complex relationship between trace elements and skeletal health has received increasing attention in the scientific community. Among these minerals, manganese (Mn) has emerged as a key element affecting bone metabolism and integrity. This review examines the multifaceted role of Mn in bone health, including its effects on bone regeneration, mineralization, and overall skeletal strength. This review article is based on a synthesis of experimental models, epidemiologic studies, and clinical trials of the mechanisms of the effect of Mn on bone metabolism. Current research data show that Mn is actively involved in the processes of bone remodeling by modulating the activity of osteoblasts and osteoclasts, as well as the main cells that regulate bone formation and resorption. Mn ions have a profound effect on bone mineralization and density by intricately regulating signaling pathways and enzymatic reactions in these cells. Additionally, Mn superoxide dismutase (MnSOD), located in bone mitochondria, plays a crucial role in osteoclast differentiation and function, protecting osteoclasts from oxidative damage. Understanding the nuances of Mn's interaction with bone is essential for optimizing bone strategies, potentially preventing and managing skeletal diseases. Key findings include the stimulation of osteoblast proliferation and differentiation, the inhibition of osteoclastogenesis, and the preservation of bone mass through the RANK/RANKL/OPG pathway. These results underscore the importance of Mn in maintaining bone health and highlight the need for further research into its therapeutic potential.
Collapse
Affiliation(s)
- Gulaim Taskozhina
- Department of Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan
| | - Gulnara Batyrova
- Department of Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan
| | - Gulmira Umarova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| | - Zhamilya Issanguzhina
- Department of Children Disease No. 2, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| | - Nurgul Kereyeva
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| |
Collapse
|
5
|
Manipur I, Reales G, Sul JH, Shin MK, Longerich S, Cortes A, Wallace C. CoPheScan: phenome-wide association studies accounting for linkage disequilibrium. Nat Commun 2024; 15:5862. [PMID: 38997278 PMCID: PMC11245513 DOI: 10.1038/s41467-024-49990-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Phenome-wide association studies (PheWAS) facilitate the discovery of associations between a single genetic variant with multiple phenotypes. For variants which impact a specific protein, this can help identify additional therapeutic indications or on-target side effects of intervening on that protein. However, PheWAS is restricted by an inability to distinguish confounding due to linkage disequilibrium (LD) from true pleiotropy. Here we describe CoPheScan (Coloc adapted Phenome-wide Scan), a Bayesian approach that enables an intuitive and systematic exploration of causal associations while simultaneously addressing LD confounding. We demonstrate its performance through simulation, showing considerably better control of false positive rates than a conventional approach not accounting for LD. We used CoPheScan to perform PheWAS of protein-truncating variants and fine-mapped variants from disease and pQTL studies, in 2275 disease phenotypes from the UK Biobank. Our results identify the complexity of known pleiotropic genes such as APOE, and suggest a new causal role for TGM3 in skin cancer.
Collapse
Affiliation(s)
- Ichcha Manipur
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK.
| | - Guillermo Reales
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK
| | | | | | | | - Adrian Cortes
- Human Genetics and Genomics, GSK, Heidelberg, 69117, Germany
| | - Chris Wallace
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
6
|
Gong W, Fu Y, Wu BS, Du J, Yang L, Zhang YR, Chen SD, Kang J, Mao Y, Dong Q, Tan L, Feng J, Cheng W, Yu JT. Whole-exome sequencing identifies protein-coding variants associated with brain iron in 29,828 individuals. Nat Commun 2024; 15:5540. [PMID: 38956042 PMCID: PMC11219919 DOI: 10.1038/s41467-024-49702-2] [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: 04/12/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
Iron plays a fundamental role in multiple brain disorders. However, the genetic underpinnings of brain iron and its implications for these disorders are still lacking. Here, we conduct an exome-wide association analysis of brain iron, measured by quantitative susceptibility mapping technique, across 26 brain regions among 26,789 UK Biobank participants. We find 36 genes linked to brain iron, with 29 not being previously reported, and 16 of them can be replicated in an independent dataset with 3,039 subjects. Many of these genes are involved in iron transport and homeostasis, such as FTH1 and MLX. Several genes, while not previously connected to brain iron, are associated with iron-related brain disorders like Parkinson's (STAB1, KCNA10), Alzheimer's (SHANK1), and depression (GFAP). Mendelian randomization analysis reveals six causal relationships from regional brain iron to brain disorders, such as from the hippocampus to depression and from the substantia nigra to Parkinson's. These insights advance our understanding of the genetic architecture of brain iron and offer potential therapeutic targets for brain disorders.
Collapse
Affiliation(s)
- Weikang Gong
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, UK.
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, China
| | - Bang-Sheng Wu
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jingnan Du
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, 02138, USA
| | - Liu Yang
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - JuJiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China
| | - Ying Mao
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Wei Cheng
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, 200433, Shanghai, China.
| | - Jin-Tai Yu
- School of Data Science, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Geng J, Wang Y, Lv F, Yu X, Gong M, Zhang J, Zhao Z, Zhu X, Zhang X, Yang J, Yang XA. Coumestrol facilitates apoptosis in colorectal cancer cells by interacting with ZIP8 protein via the ferroptosis pathway. J Cancer 2024; 15:4656-4667. [PMID: 39006076 PMCID: PMC11242349 DOI: 10.7150/jca.94628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/09/2024] [Indexed: 07/16/2024] Open
Abstract
Objective: So far, there have been no reports of coumestrol inhibiting colorectal cancer (CRC) through the ferroptosis pathway. This study is to investigate the mechanism of the traditional Chinese medicine monomer coumestrol in the treatment of CRC. Methods: Data on CRC transcriptome sequencing was obtained from the GEO database and TCGA database. Bioinformatics analyses were conducted to screen for CRC prognostic-related key genes and their potential binding monomers in traditional Chinese medicine. The inhibitory effect of coumestrol on CRC cell lines (COLO 205 & HCT 116) was determined using the CCK-8 assay, and cell apoptosis was assessed by flow cytometry. The content of ferrous ions was measured using the Ferrous Ion Content Assay Kit. The expression of ferroptosis pathway-related genes SLC39A8, NCOA4, VDAC2, and NOX2 before and after small interference RNA (siRNA) was examined through real-time PCR and Western blotting. Results: SLC39A8 was found to be associated with CRC clinical progression staging, and its encoded protein ZIP8 may bind to coumestrol. KEGG enrichment analysis suggested that ZIP8 plays a role in iron transmembrane transport and may affect the expression of ferroptosis pathway-related genes NCOA4, VDAC2, and NOX2. Coumestrol was found to induce apoptosis in CRC cell lines by upregulating the expression of ferroptosis pathway-related genes SLC39A8, NCOA4, VDAC2, and NOX2. However, coumestrol was unable to upregulate the expression of ferroptosis pathway-related genes in CRC cell lines after SLC39A8 interference. Conclusion: Coumestrol facilitates apoptosis in CRC cells by interacting with ZIP8 protein via the ferroptosis pathway.
Collapse
Affiliation(s)
- Jing Geng
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
| | - Yingying Wang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
| | - Fengchun Lv
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
| | - Xiaomin Yu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
| | - Mingyu Gong
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
| | - Jie Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Graduate School of Chengde Medical University, 067000 Chengde, China
| | - Zicheng Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Graduate School of Chengde Medical University, 067000 Chengde, China
| | - Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Graduate School of Chengde Medical University, 067000 Chengde, China
- Department of Biomedical Engineering, Chengde Medical University, 067000 Chengde, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Graduate School of Chengde Medical University, 067000 Chengde, China
| | - Jian Yang
- Institute of Basic Medical Sciences, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, 067000 Chengde, China
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, 067000 Chengde, China
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, 067000 Chengde, China
| |
Collapse
|
8
|
Tomar V, Kang J, Lin R, Brant SR, Lazarev M, Tressler C, Glunde K, Zachara N, Melia J. Aberrant N-glycosylation is a therapeutic target in carriers of a common and highly pleiotropic mutation in the manganese transporter ZIP8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601207. [PMID: 39005453 PMCID: PMC11244875 DOI: 10.1101/2024.06.28.601207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The treatment of defective glycosylation in clinical practice has been limited to patients with rare and severe phenotypes associated with congenital disorders of glycosylation (CDG). Carried by approximately 5% of the human population, the discovery of the highly pleiotropic, missense mutation in a manganese transporter ZIP8 has exposed under-appreciated roles for Mn homeostasis and aberrant Mn-dependent glycosyltransferases activity leading to defective N-glycosylation in complex human diseases. Here, we test the hypothesis that aberrant N-glycosylation contributes to disease pathogenesis of ZIP8 A391T-associated Crohn's disease. Analysis of N-glycan branching in intestinal biopsies demonstrates perturbation in active Crohn's disease and a genotype-dependent effect characterized by increased truncated N-glycans. A mouse model of ZIP8 391-Thr recapitulates the intestinal glycophenotype of patients carrying mutations in ZIP8. Borrowing from therapeutic strategies employed in the treatment of patients with CDGs, oral monosaccharide therapy with N-acetylglucosamine ameliorates the epithelial N-glycan defect, bile acid dyshomeostasis, intestinal permeability, and susceptibility to chemical-induced colitis in a mouse model of ZIP8 391-Thr. Together, these data support ZIP8 391-Thr alters N-glycosylation to contribute to disease pathogenesis, challenging the clinical paradigm that CDGs are limited to patients with rare diseases. Critically, the defect in glycosylation can be targeted with monosaccharide supplementation, providing an opportunity for genotype-driven, personalized medicine.
Collapse
|
9
|
Nauffal V, Klarqvist MDR, Hill MC, Pace DF, Di Achille P, Choi SH, Rämö JT, Pirruccello JP, Singh P, Kany S, Hou C, Ng K, Philippakis AA, Batra P, Lubitz SA, Ellinor PT. Noninvasive assessment of organ-specific and shared pathways in multi-organ fibrosis using T1 mapping. Nat Med 2024; 30:1749-1760. [PMID: 38806679 DOI: 10.1038/s41591-024-03010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fibrotic diseases affect multiple organs and are associated with morbidity and mortality. To examine organ-specific and shared biologic mechanisms that underlie fibrosis in different organs, we developed machine learning models to quantify T1 time, a marker of interstitial fibrosis, in the liver, pancreas, heart and kidney among 43,881 UK Biobank participants who underwent magnetic resonance imaging. In phenome-wide association analyses, we demonstrate the association of increased organ-specific T1 time, reflecting increased interstitial fibrosis, with prevalent diseases across multiple organ systems. In genome-wide association analyses, we identified 27, 18, 11 and 10 independent genetic loci associated with liver, pancreas, myocardial and renal cortex T1 time, respectively. There was a modest genetic correlation between the examined organs. Several loci overlapped across the examined organs implicating genes involved in a myriad of biologic pathways including metal ion transport (SLC39A8, HFE and TMPRSS6), glucose metabolism (PCK2), blood group antigens (ABO and FUT2), immune function (BANK1 and PPP3CA), inflammation (NFKB1) and mitosis (CENPE). Finally, we found that an increasing number of organs with T1 time falling in the top quintile was associated with increased mortality in the population. Individuals with a high burden of fibrosis in ≥3 organs had a 3-fold increase in mortality compared to those with a low burden of fibrosis across all examined organs in multivariable-adjusted analysis (hazard ratio = 3.31, 95% confidence interval 1.77-6.19; P = 1.78 × 10-4). By leveraging machine learning to quantify T1 time across multiple organs at scale, we uncovered new organ-specific and shared biologic pathways underlying fibrosis that may provide therapeutic targets.
Collapse
Affiliation(s)
- Victor Nauffal
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Danielle F Pace
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cody Hou
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | - Anthony A Philippakis
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Lubitz
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
10
|
Aierken Y, He H, Li R, Lin Z, Xu T, Zhang L, Wu Y, Liu Y. Inhibition of Slc39a14/Slc39a8 reduce vascular calcification via alleviating iron overload induced ferroptosis in vascular smooth muscle cells. Cardiovasc Diabetol 2024; 23:186. [PMID: 38812011 PMCID: PMC11138056 DOI: 10.1186/s12933-024-02224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/03/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Vascular calcification (VC) is an independent risk factor for cardiovascular diseases. Recently, ferroptosis has been recognised as a novel therapeutic target for cardiovascular diseases. Although an association between ferroptosis and vascular calcification has been reported, the role and mechanism of iron overload in vascular calcification are still poorly understood. Specifically, further in-depth research is required on whether metalloproteins SLC39a14 and SLC39a8 are involved in ferroptosis induced by iron overload. METHODS R language was employed for the differential analysis of the dataset, revealing the correlation between ferroptosis and calcification. The experimental approaches encompassed both in vitro and in vivo studies, incorporating the use of iron chelators and models of iron overload. Additionally, gain- and loss-of-function experiments were conducted to investigate iron's effects on vascular calcification comprehensively. Electron microscopy, immunofluorescence, western blotting, and real-time polymerase chain reaction were used to elucidate how Slc39a14 and Slc39a8 mediate iron overload and promote calcification. RESULTS Ferroptosis was observed in conjunction with vascular calcification (VC); the association was consistently confirmed by in vitro and in vivo studies. Our results showed a positive correlation between iron overload in VSMCs and calcification. Iron chelators are effective in reversing VC and iron overload exacerbates this process. The expression levels of the metal transport proteins Slc39a14 and Slc39a8 were significantly upregulated during calcification; the inhibition of their expression alleviated VC. Conversely, Slc39a14 overexpression exacerbates calcification and promotes intracellular iron accumulation in VSMCs. CONCLUSIONS Our research demonstrates that iron overload occurs during VC, and that inhibition of Slc39a14 and Slc39a8 significantly relieves VC by intercepting iron overload-induced ferroptosis in VSMCs, providing new insights into the VC treatment.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Animals
- Cation Transport Proteins/metabolism
- Cation Transport Proteins/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Iron Chelating Agents/pharmacology
- Iron Chelating Agents/therapeutic use
- Signal Transduction
- Male
- Humans
- Iron/metabolism
- Iron Overload/metabolism
- Iron Overload/pathology
Collapse
Affiliation(s)
- Yierpani Aierken
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Huqiang He
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Runwen Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Zipeng Lin
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Tongjie Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Li Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, No. 25, Taiping Street, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
11
|
Malone SG, Davis CN, Piserchia Z, Setzer MR, Toikumo S, Zhou H, Winterlind EL, Gelernter J, Justice A, Leggio L, Rentsch CT, Kranzler HR, Gray JC. Alcohol use disorder and body mass index show genetic pleiotropy and shared neural associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.03.24306773. [PMID: 38746260 PMCID: PMC11092735 DOI: 10.1101/2024.05.03.24306773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background The prevalence of co-occurring heavy alcohol consumption and obesity is increasing in the United States. Despite neurobiological overlap in the regulation of alcohol consumption and eating behavior, alcohol- and body mass index (BMI)-related phenotypes show no or minimal genetic correlation. We hypothesized that the lack of genetic correlation is due to mixed effect directions of variants shared by AUD and BMI. Methods We applied MiXeR, to investigate shared genetic architecture between AUD and BMI in individuals of European ancestry. We used conjunctional false discovery rate (conjFDR) analysis to detect loci associated with both phenotypes and their directional effect, Functional Mapping and Annotation (FUMA) to identify lead single nucleotide polymorphisms (SNPs), Genotype-Tissue Expression (GTEx) samples to examine gene expression enrichment across tissue types, and BrainXcan to evaluate the shared associations of AUD and BMI with brain image-derived phenotypes. Results MiXeR analysis indicated polygenic overlap of 80.9% between AUD and BMI, despite a genetic correlation (r g ) of -.03. ConjFDR analysis yielded 56 lead SNPs with the same effect direction and 76 with the opposite direction. Of the 132 shared lead SNPs, 53 were novel for both AUD and BMI. GTEx analyses identified significant overexpression in the frontal cortex (BA9), hypothalamus, cortex, anterior cingulate cortex (BA24), hippocampus, and amygdala. Amygdala and caudate nucleus gray matter volumes were significantly associated with both AUD and BMI in BrainXcan analyses. Conclusions More than half of variants significantly associated with AUD and BMI had opposite directions of effect for the traits, supporting our hypothesis that this is the basis for their lack of genetic correlation. Follow-up analyses identified brain regions implicated in executive functioning, reward, homeostasis, and food intake regulation. Together, these findings clarify the extensive polygenic overlap between AUD and BMI and elucidate several overlapping neurobiological mechanisms.
Collapse
|
12
|
Moksnes MR, Hansen AF, Wolford BN, Thomas LF, Rasheed H, Simić A, Bhatta L, Brantsæter AL, Surakka I, Zhou W, Magnus P, Njølstad PR, Andreassen OA, Syversen T, Zheng J, Fritsche LG, Evans DM, Warrington NM, Nøst TH, Åsvold BO, Flaten TP, Willer CJ, Hveem K, Brumpton BM. A genome-wide association study provides insights into the genetic etiology of 57 essential and non-essential trace elements in humans. Commun Biol 2024; 7:432. [PMID: 38594418 PMCID: PMC11004147 DOI: 10.1038/s42003-024-06101-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/22/2024] [Indexed: 04/11/2024] Open
Abstract
Trace elements are important for human health but may exert toxic or adverse effects. Mechanisms of uptake, distribution, metabolism, and excretion are partly under genetic control but have not yet been extensively mapped. Here we report a comprehensive multi-element genome-wide association study of 57 essential and non-essential trace elements. We perform genome-wide association meta-analyses of 14 trace elements in up to 6564 Scandinavian whole blood samples, and genome-wide association studies of 43 trace elements in up to 2819 samples measured only in the Trøndelag Health Study (HUNT). We identify 11 novel genetic loci associated with blood concentrations of arsenic, cadmium, manganese, selenium, and zinc in genome-wide association meta-analyses. In HUNT, several genome-wide significant loci are also indicated for other trace elements. Using two-sample Mendelian randomization, we find several indications of weak to moderate effects on health outcomes, the most precise being a weak harmful effect of increased zinc on prostate cancer. However, independent validation is needed. Our current understanding of trace element-associated genetic variants may help establish consequences of trace elements on human health.
Collapse
Affiliation(s)
- Marta R Moksnes
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.
| | - Ailin F Hansen
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Laurent F Thomas
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- BioCore-Bioinformatics Core Facility, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Humaira Rasheed
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Medicine and Laboratory Sciences, University of Oslo, Oslo, Norway
| | - Anica Simić
- Department of Chemistry, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Laxmi Bhatta
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Lise Brantsæter
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | - Ole A Andreassen
- NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tore Syversen
- Department of Neuroscience, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Bristol, UK
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - David M Evans
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Nicole M Warrington
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Therese H Nøst
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bjørn Olav Åsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Trond Peder Flaten
- Department of Chemistry, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Cristen J Willer
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway
| | - Ben M Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.
- HUNT Research Centre, Department of Public Health and Nursing, NTNU-Norwegian University of Science and Technology, Levanger, Norway.
- Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
13
|
Fanelli G, Franke B, Fabbri C, Werme J, Erdogan I, De Witte W, Poelmans G, Ruisch IH, Reus LM, van Gils V, Jansen WJ, Vos SJ, Alam KA, Martinez A, Haavik J, Wimberley T, Dalsgaard S, Fóthi Á, Barta C, Fernandez-Aranda F, Jimenez-Murcia S, Berkel S, Matura S, Salas-Salvadó J, Arenella M, Serretti A, Mota NR, Bralten J. Local patterns of genetic sharing challenge the boundaries between neuropsychiatric and insulin resistance-related conditions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.07.24303921. [PMID: 38496672 PMCID: PMC10942494 DOI: 10.1101/2024.03.07.24303921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The co-occurrence of insulin resistance (IR)-related metabolic conditions with neuropsychiatric disorders is a complex public health challenge. Evidence of the genetic links between these phenotypes is emerging, but little is currently known about the genomic regions and biological functions that are involved. To address this, we performed Local Analysis of [co]Variant Association (LAVA) using large-scale (N=9,725-933,970) genome-wide association studies (GWASs) results for three IR-related conditions (type 2 diabetes mellitus, obesity, and metabolic syndrome) and nine neuropsychiatric disorders. Subsequently, positional and expression quantitative trait locus (eQTL)-based gene mapping and downstream functional genomic analyses were performed on the significant loci. Patterns of negative and positive local genetic correlations (|rg|=0.21-1, pFDR<0.05) were identified at 109 unique genomic regions across all phenotype pairs. Local correlations emerged even in the absence of global genetic correlations between IR-related conditions and Alzheimer's disease, bipolar disorder, and Tourette's syndrome. Genes mapped to the correlated regions showed enrichment in biological pathways integral to immune-inflammatory function, vesicle trafficking, insulin signalling, oxygen transport, and lipid metabolism. Colocalisation analyses further prioritised 10 genetically correlated regions for likely harbouring shared causal variants, displaying high deleterious or regulatory potential. These variants were found within or in close proximity to genes, such as SLC39A8 and HLA-DRB1, that can be targeted by supplements and already known drugs, including omega-3/6 fatty acids, immunomodulatory, antihypertensive, and cholesterol-lowering drugs. Overall, our findings underscore the complex genetic landscape of IR-neuropsychiatric multimorbidity, advocating for an integrated disease model and offering novel insights for research and treatment strategies in this domain.
Collapse
Affiliation(s)
- Giuseppe Fanelli
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Josefin Werme
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Izel Erdogan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ward De Witte
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert Poelmans
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I. Hyun Ruisch
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lianne Maria Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, California, United States
| | - Veerle van Gils
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Willemijn J. Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Stephanie J.B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | | | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson’s Disease, University of Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Norway
| | - Theresa Wimberley
- National Centre for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
- iPSYCH - The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark
| | - Søren Dalsgaard
- National Centre for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Child and Adolescent Psychiatry Glostrup, Mental Health Services of the Capital Region, Hellerup, Denmark
| | - Ábel Fóthi
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Fernando Fernandez-Aranda
- Clinical Psychology Department, University Hospital of Bellvitge, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Susana Jimenez-Murcia
- Clinical Psychology Department, University Hospital of Bellvitge, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Psychological Services, University of Barcelona, Spain
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Silke Matura
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Biochemistry and biotechnology Department, Grup Alimentació, Nutrició, Desenvolupament i Salut Mental, Unitat de Nutrició Humana, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Martina Arenella
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK
| | | | - Nina Roth Mota
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janita Bralten
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Liu Z, Huang J, Li D, Zhang C, Wan H, Zeng B, Tan Y, Zhong F, Liao H, Liu M, Chen ZS, Zou C, Liu D, Qin B. Targeting ZIP8 mediated ferroptosis as a novel strategy to protect against the retinal pigment epithelial degeneration. Free Radic Biol Med 2024; 214:42-53. [PMID: 38309537 DOI: 10.1016/j.freeradbiomed.2024.01.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
The degeneration of retinal pigment epithelium (RPE) plays an important role in the development of age-related macular degeneration (AMD). However, the underlying mechanism remains elusive. In this study, we identified that ZIP8, a metal-ion transporter, plays a crucial role in the degeneration of RPE cells mediated by ferroptosis. ZIP8 was found to be upregulated in patients with AMD through transcriptome analysis. Upregulated ZIP8 was also observed in both oxidative-stressed RPE cells and AMD mouse model. Importantly, knockdown of ZIP8 significantly inhibited ferroptosis in RPE cells induced by sodium iodate-induced oxidative stress. Blocking ZIP8 with specific antibodies reversed RPE degeneration and restored retinal function, improving visual loss in a mouse model of NaIO3-induced. Interestingly, the modification of the N-glycosylation sites N40, N72 and N88, but not N273, was essential for the intracellular iron accumulation mediated by ZIP8, which further led to increased lipid peroxidation and RPE death. These findings highlight the critical role of ZIP8 in RPE ferroptosis and provide a potential target for the treatment of diseases associated with retinal degeneration, including AMD.
Collapse
Affiliation(s)
- Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Institute of Biopharmaceutics and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Chuanhe Zhang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Huan Wan
- The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Bing Zeng
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Yao Tan
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - Fuhua Zhong
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
| | - MuYun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen Kenuo Medical Laboratory, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, USA
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China.
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China; Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China.
| |
Collapse
|
15
|
Nebert DW. Gene-Environment Interactions: My Unique Journey. Annu Rev Pharmacol Toxicol 2024; 64:1-26. [PMID: 37788491 DOI: 10.1146/annurev-pharmtox-022323-082311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
I am deeply honored to be invited to write this scientific autobiography. As a physician-scientist, pediatrician, molecular biologist, and geneticist, I have authored/coauthored more than 600 publications in the fields of clinical medicine, biochemistry, biophysics, pharmacology, drug metabolism, toxicology, molecular biology, cancer, standardized gene nomenclature, developmental toxicology and teratogenesis, mouse genetics, human genetics, and evolutionary genomics. Looking back, I think my career can be divided into four distinct research areas, which I summarize mostly chronologically in this article: (a) discovery and characterization of the AHR/CYP1 axis, (b) pharmacogenomics and genetic prediction of response to drugs and other environmental toxicants, (c) standardized drug-metabolizing gene nomenclature based on evolutionary divergence, and (d) discovery and characterization of the SLC39A8 gene encoding the ZIP8 metal cation influx transporter. Collectively, all four topics embrace gene-environment interactions, hence the title of my autobiography.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics and Molecular Developmental Biology, Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA;
| |
Collapse
|
16
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Zhang Y, Jiang C, Meng N. Targeting Ferroptosis: A Novel Strategy for the Treatment of Atherosclerosis. Mini Rev Med Chem 2024; 24:1262-1276. [PMID: 38284727 DOI: 10.2174/0113895575273164231130070920] [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: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 01/30/2024]
Abstract
Since ferroptosis was reported in 2012, its application prospects in various diseases have been widely considered, initially as a treatment direction for tumors. Recent studies have shown that ferroptosis is closely related to the occurrence and development of atherosclerosis. The primary mechanism is to affect the occurrence and development of atherosclerosis through intracellular iron homeostasis, ROS and lipid peroxide production and metabolism, and a variety of intracellular signaling pathways. Inhibition of ferroptosis is effective in inhibiting the development of atherosclerosis, and it can bring a new direction for treating atherosclerosis. In this review, we discuss the mechanism of ferroptosis and focus on the relationship between ferroptosis and atherosclerosis, summarize the different types of ferroptosis inhibitors that have been widely studied, and discuss some issues worthy of attention in the treatment of atherosclerosis by targeting ferroptosis.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|
18
|
Zhou S, Luo H, Tian Y, Li H, Zeng Y, Wang X, Shan S, Xiong J, Cheng G. Investigating the shared genetic architecture of post-traumatic stress disorder and gastrointestinal tract disorders: a genome-wide cross-trait analysis. Psychol Med 2023; 53:7627-7635. [PMID: 37218628 DOI: 10.1017/s0033291723001423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
BACKGROUND Observational studies suggest a correlation between post-traumatic stress disorder (PTSD) and gastrointestinal tract (GIT) disorders. However, the genetic overlap, causal relationships, and underlining mechanisms between PTSD and GIT disorders were absent. METHODS We obtained genome-wide association study statistics for PTSD (23 212 cases, 151 447 controls), peptic ulcer disease (PUD; 16 666 cases, 439 661 controls), gastroesophageal reflux disease (GORD; 54 854 cases, 401 473 controls), PUD and/or GORD and/or medications (PGM; 90 175 cases, 366 152 controls), irritable bowel syndrome (IBS; 28 518 cases, 426 803 controls), and inflammatory bowel disease (IBD; 7045 cases, 449 282 controls). We quantified genetic correlations, identified pleiotropic loci, and performed multi-marker analysis of genomic annotation, fast gene-based association analysis, transcriptome-wide association study analysis, and bidirectional Mendelian randomization analysis. RESULTS PTSD globally correlates with PUD (rg = 0.526, p = 9.355 × 10-7), GORD (rg = 0.398, p = 5.223 × 10-9), PGM (rg = 0.524, p = 1.251 × 10-15), and IBS (rg = 0.419, p = 8.825 × 10-6). Cross-trait meta-analyses identify seven genome-wide significant loci between PTSD and PGM (rs13107325, rs1632855, rs1800628, rs2188100, rs3129953, rs6973700, and rs73154693); three between PTSD and GORD (rs13107325, rs1632855, and rs3132450); one between PTSD and IBS/IBD (rs4937872 and rs114969413, respectively). Proximal pleiotropic genes are mainly enriched in immune response regulatory pathways, and in brain, digestive, and immune systems. Gene-level analyses identify five candidates: ABT1, BTN3A2, HIST1H3J, ZKSCAN4, and ZKSCAN8. We found significant causal effects of GORD, PGM, IBS, and IBD on PTSD. We observed no reverse causality of PTSD with GIT disorders, except for GORD. CONCLUSIONS PTSD and GIT disorders share common genetic architectures. Our work offers insights into the biological mechanisms, and provides genetic basis for translational research studies.
Collapse
Affiliation(s)
- Siquan Zhou
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hang Luo
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Ye Tian
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Haoqi Li
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Yaxian Zeng
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Xiaoyu Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shufang Shan
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jingyuan Xiong
- West China School of Public Health and West China Fourth Hospital, Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Guo Cheng
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Ahuja S, Lazar IM. Proteomic Insights into Metastatic Breast Cancer Response to Brain Cell-Secreted Factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.22.563488. [PMID: 37961261 PMCID: PMC10634729 DOI: 10.1101/2023.10.22.563488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2+ and triple negative breast cancers frequently metastasize to the brain and stay potentially dormant for years, clinging to the microvasculature, until favorable environmental conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection, diagnosis, and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain tissues and biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development, leading to slow progress in understanding the drivers of disease progression and response to therapy. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the growth and initial response of HER2+ breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment conditions were simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., human endothelial cells (HBEC5i), human astrocytes (NHA) and human microglia (HMC3) cells. Cytokine microarrays were used to investigate the cell secretomes and explore the mediators responsible for cell-cell communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The results of the study suggest that the exposure of SKBR3 cells to the brain secretomes altered their growth potential and drove them towards a state of quiescence. The cytokines, growth factors and enzymes detected in the brain cell-conditioned medium were supportive of mostly inflammatory conditions, indicating a collective functional contribution to cell activation, defense, inflammatory responses, chemotaxis, adhesion, angiogenesis, and ECM organization. The SKBR3 cells, on the other hand, secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell culture media, suggesting that upon exposure the SKBR3 cells were deprived of favorable environmental conditions required for optimal growth. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, providing insights into the cancer-host cell cross-talk that contributes to driving metastasized cancer cells into dormancy and into the opportunities that exist for developing novel therapeutic strategies that target the brain metastases of breast cancer.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
| | - Iulia M. Lazar
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Fralin Life Sciences Institute, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Carilion School of Medicine, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Division of Systems Biology/AIS, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
| |
Collapse
|
20
|
Baj J, Flieger W, Barbachowska A, Kowalska B, Flieger M, Forma A, Teresiński G, Portincasa P, Buszewicz G, Radzikowska-Büchner E, Flieger J. Consequences of Disturbing Manganese Homeostasis. Int J Mol Sci 2023; 24:14959. [PMID: 37834407 PMCID: PMC10573482 DOI: 10.3390/ijms241914959] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Manganese (Mn) is an essential trace element with unique functions in the body; it acts as a cofactor for many enzymes involved in energy metabolism, the endogenous antioxidant enzyme systems, neurotransmitter production, and the regulation of reproductive hormones. However, overexposure to Mn is toxic, particularly to the central nervous system (CNS) due to it causing the progressive destruction of nerve cells. Exposure to manganese is widespread and occurs by inhalation, ingestion, or dermal contact. Associations have been observed between Mn accumulation and neurodegenerative diseases such as manganism, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. People with genetic diseases associated with a mutation in the gene associated with impaired Mn excretion, kidney disease, iron deficiency, or a vegetarian diet are at particular risk of excessive exposure to Mn. This review has collected data on the current knowledge of the source of Mn exposure, the experimental data supporting the dispersive accumulation of Mn in the brain, the controversies surrounding the reference values of biomarkers related to Mn status in different matrices, and the competitiveness of Mn with other metals, such as iron (Fe), magnesium (Mg), zinc (Zn), copper (Cu), lead (Pb), calcium (Ca). The disturbed homeostasis of Mn in the body has been connected with susceptibility to neurodegenerative diseases, fertility, and infectious diseases. The current evidence on the involvement of Mn in metabolic diseases, such as type 2 diabetes mellitus/insulin resistance, osteoporosis, obesity, atherosclerosis, and non-alcoholic fatty liver disease, was collected and discussed.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Wojciech Flieger
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Aleksandra Barbachowska
- Department of Plastic, Reconstructive and Burn Surgery, Medical University of Lublin, 21-010 Łęczna, Poland;
| | - Beata Kowalska
- Department of Water Supply and Wastewater Disposal, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Michał Flieger
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (W.F.); (A.F.)
| | - Grzegorz Teresiński
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | - Piero Portincasa
- Clinica Medica A. Murri, Department of Biomedical Sciences & Human Oncology, Medical School, University of Bari, 70124 Bari, Italy;
| | - Grzegorz Buszewicz
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland; (M.F.); (G.T.); (G.B.)
| | | | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
21
|
Sunuwar L, Tomar V, Wildeman A, Culotta V, Melia J. Hepatobiliary manganese homeostasis is dynamic in the setting of inflammation or infection in mice. FASEB J 2023; 37:e23123. [PMID: 37561548 DOI: 10.1096/fj.202300539r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/19/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
Manganese is a diet-derived micronutrient that is essential for critical cellular processes like redox homeostasis, protein glycosylation, and lipid and carbohydrate metabolism. Control of Mn availability, especially at the local site of infection, is a key component of the innate immune response. Less has been elucidated about Mn homeostasis at the systemic level. In this work, we demonstrate that systemic Mn homeostasis is dynamic in response to inflammation and infection in mice. This phenomenon is evidenced in male and female mice, mice of two genetic backgrounds (C57BL/6 and BALB/c), in multiple models of acute (dextran sodium sulfate-induced) and chronic (enterotoxigenic Bacteroides fragilis) colitis, and systemic infection with Candida albicans. When mice were fed a standard corn-based chow with excess Mn (100 ppm), liver Mn decreased and biliary Mn increased threefold in response to infection or colitis. Liver iron, copper, and zinc were unchanged. When dietary Mn was restricted to minimally adequate amounts (10 ppm), baseline hepatic Mn levels decreased by approximately 60% in the liver, and upon induction of colitis, liver Mn did not decrease further, however, biliary Mn still increased 20-fold. In response to acute colitis, hepatic Slc39a8 mRNA (gene encoding the Mn importer, Zip8) and Slc30a10 mRNA (gene encoding the Mn exporter, Znt10) are decreased. Zip8 protein is decreased. Inflammation/infection-associated dynamic Mn homeostasis may represent a novel host immune/inflammatory response that reorganizes systemic Mn availability through differential expression of key Mn transporters with down-regulation of Zip8.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vartika Tomar
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Asia Wildeman
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Valeria Culotta
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joanna Melia
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Vicogne D, Beauval N, Durin Z, Allorge D, Kondratska K, Haustrate A, Prevarskaya N, Lupashin V, Legrand D, Foulquier F. Insights into the regulation of cellular Mn 2+ homeostasis via TMEM165. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166717. [PMID: 37062452 PMCID: PMC10639120 DOI: 10.1016/j.bbadis.2023.166717] [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: 12/08/2022] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/18/2023]
Abstract
Golgi cation homeostasis is known to be crucial for many cellular processes including vesicular fusion events, protein secretion, as well as for the activity of Golgi glycosyltransferases and glycosidases. TMEM165 was identified in 2012 as the first cation transporter related to human glycosylation diseases, namely the Congenital Disorders of Glycosylation (CDG). Interestingly, divalent manganese (Mn) supplementation has been shown to suppress the observed glycosylation defects in TMEM165-deficient cell lines, thus suggesting that TMEM165 is involved in cellular Mn homeostasis. This paper demonstrates that the origin of the Golgi glycosylation defects arises from impaired Golgi Mn homeostasis in TMEM165-depleted cells. We show that Mn supplementation fully rescues the Mn content in the secretory pathway/organelles of TMEM165-depleted cells and hence the glycosylation process. Strong cytosolic and organellar Mn accumulations can also be observed in TMEM165- and SPCA1-depleted cells upon incubation with increasing Mn concentrations, thus demonstrating the crucial involvement of these two proteins in cellular Mn homeostasis. Interestingly, our results show that the cellular Mn homeostasis maintenance in control cells is correlated with the presence of TMEM165 and that the Mn-detoxifying capacities of cells, through the activity of SPCA1, rely on the Mn-induced degradation mechanism of TMEM165. Finally, this paper highlights that TMEM165 is essential in secretory pathway/organelles Mn homeostasis maintenance to ensure both Golgi glycosylation enzyme activities and cytosolic Mn detoxification.
Collapse
Affiliation(s)
- Dorothée Vicogne
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Nicolas Beauval
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé humaine, F-59000 Lille, France
| | - Zoé Durin
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - Delphine Allorge
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé humaine, F-59000 Lille, France
| | - Kateryna Kondratska
- Univ. Lille, INSERM U1003-PHYCEL-Physiology Cellulaire, F-59000 Lille, France; Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Aurélien Haustrate
- Univ. Lille, INSERM U1003-PHYCEL-Physiology Cellulaire, F-59000 Lille, France; Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Natasha Prevarskaya
- Univ. Lille, INSERM U1003-PHYCEL-Physiology Cellulaire, F-59000 Lille, France; Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Biomed 261-2, slot 505, 200 South Cedar St., Little Rock, AR 72205, USA
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000 Lille, France.
| |
Collapse
|
23
|
Real MVF, Colvin MS, Sheehan MJ, Moeller AH. Major urinary protein ( Mup) gene family deletion drives sex-specific alterations on the house mouse gut microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551491. [PMID: 37577672 PMCID: PMC10418228 DOI: 10.1101/2023.08.01.551491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The gut microbiota is shaped by host metabolism. In house mice (Mus musculus), major urinary protein (MUP) pheromone production represents a considerable energy investment, particularly in sexually mature males. Deletion of the Mup gene family shifts mouse metabolism towards an anabolic state, marked by lipogenesis, lipid accumulation, and body mass increases. Given the metabolic implications of MUPs, they may also influence the gut microbiota. Here, we investigated the effect of deletion of the Mup gene family on the gut microbiota of sexually mature mice. Shotgun metagenomics revealed distinct taxonomic and functional profiles between wildtype and knockout males, but not females. Deletion of the Mup gene cluster significantly reduced diversity in microbial families and functions in male mice. Additionally, specific taxa of the Ruminococcaceae family, which is associated with gut health and reduced risk of developing metabolic syndrome, and several microbial functions, such as transporters involved in vitamin B5 acquisition, were significantly depleted in the microbiota of Mup-knockout males. Altogether these results show that major urinary proteins significantly affect the gut microbiota of house mouse in a sex-specific manner.
Collapse
Affiliation(s)
- Madalena V. F. Real
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Melanie S. Colvin
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Michael J. Sheehan
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Andrew H. Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
24
|
Jiang Y, Li Z, Sui D, Sharma G, Wang T, MacRenaris K, Takahashi H, Merz K, Hu J. Rational engineering of an elevator-type metal transporter ZIP8 reveals a conditional selectivity filter critically involved in determining substrate specificity. Commun Biol 2023; 6:778. [PMID: 37495662 PMCID: PMC10372143 DOI: 10.1038/s42003-023-05146-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023] Open
Abstract
Engineering of transporters to alter substrate specificity as desired holds great potential for applications, including metabolic engineering. However, the lack of knowledge on molecular mechanisms of substrate specificity hinders designing effective strategies for transporter engineering. Here, we applied an integrated approach to rationally alter the substrate preference of ZIP8, a Zrt-/Irt-like protein (ZIP) metal transporter with multiple natural substrates, and uncovered the determinants of substrate specificity. By systematically replacing the differentially conserved residues with the counterparts in the zinc transporter ZIP4, we created a zinc-preferring quadruple variant (Q180H/E343H/C310A/N357H), which exhibited largely reduced transport activities towards Cd2+, Fe2+, and Mn2+ whereas increased activity toward Zn2+. Combined mutagenesis, modeling, covariance analysis, and computational studies revealed a conditional selectivity filter which functions only when the transporter adopts the outward-facing conformation. The demonstrated approach for transporter engineering and the gained knowledge about substrate specificity will facilitate engineering and mechanistic studies of other transporters.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
| | - Zhen Li
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Gaurav Sharma
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
| | - Tianqi Wang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Keith MacRenaris
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Hideki Takahashi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Kenneth Merz
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Jian Hu
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
25
|
Kim S, Seo YA. In vitro studies of manganese transport and homeostasis. Methods Enzymol 2023; 687:185-206. [PMID: 37666632 DOI: 10.1016/bs.mie.2023.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Manganese (Mn) is an essential micronutrient required for fundamental cell functions and vital physiological processes. More than a dozen putative Mn transporters have been described over the last two decades, but few have been thoroughly evaluated. Recent genetic studies have revealed vital roles for solute carrier family 39, member 8 (SLC39A8) in Mn homeostasis. SLC39A8 can mediate the cellular uptake of the essential metals zinc, iron, and Mn, as well as the non-essential metal cadmium. However, loss-of-function mutations in SLC39A8 have been found in patients with severe Mn deficiency in the blood without affecting other metals. An in vitro study from our laboratory showed that SLC39A8 is a cell-surface transporter that strongly stimulates 54Mn incorporation into cells (Choi, Nguyen, Gupta, Iwase, & Seo, 2018). By contrast, the disease-associated mutations completely abrogated the cellular uptake of 54Mn (Choi et al., 2018), thereby providing a causal link between SLC39A8 deficiency and Mn deficiency. The importance of SLC39A8 is now increasingly recognized in multiple disease processes, and SLC39A8 has emerged as a critical regulator of Mn homeostasis. Thus, exploring the function of SLC39A8 in cellular Mn homeostasis is of significant research interest. This chapter describes the advanced methods used in our laboratory to examine Mn homeostasis and transport. Specifically, genetic and molecular approaches are described in HeLa cells overexpressing SLC39A8 and disease-associated SLC39A8 mutants. These methods are useful for characterizing the roles of Mn in diverse cellular events.
Collapse
Affiliation(s)
- Sangnam Kim
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States.
| |
Collapse
|
26
|
Jiang Y, Sui D, Hu J. Cell-based transport assay to study kinetics and substrate specificity of human ZIPs. Methods Enzymol 2023; 687:139-155. [PMID: 37666630 PMCID: PMC10999280 DOI: 10.1016/bs.mie.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Kinetic study of human ZIPs is crucial for understanding the transport mechanism and the molecular basis of substrate specificity. In this chapter, we describe the detailed experimental procedures for functional studies of two human ZIPs, including the zinc-preferring ZIP4 and the multi-metal transporter ZIP8, by using the cell-based transport assays. Kinetic study of ZIP4 is elaborated in the first section; in the second section, comparison of ZIP4 and ZIP8 in terms of the zinc/cadmium selectivity is performed by using an internal competition assay adapted from the established cell-based approach. The protocols provided in this chapter will facilitate mechanistic and engineering studies of the ZIPs.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Jian Hu
- Department of Chemistry, Michigan State University, East Lansing, MI, United States; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
27
|
Vungutur V, Yu S, McCabe S, Fung C, Zhao N. A simple and highly reproducible method for the detection of erythrocyte membrane ZIP metal transporters by immunoblotting. Methods Enzymol 2023; 687:87-102. [PMID: 37666640 PMCID: PMC10755855 DOI: 10.1016/bs.mie.2023.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Manganese is one of the essential trace elements found in erythrocytes. Metal transporters situated on the plasma membrane generally facilitate the movement of manganese into and out of cells. This study aims at determining whether two recently discovered manganese importers, ZIP8 and ZIP14, are located in the erythrocyte membrane. We outline a simple, effective and repeatable method for the isolation of erythrocyte membrane from a minimum of 50 µL mouse blood, followed by the identification of ZIP metal transporters using immunoblotting. Our results revealed that ZIP8 is expressed within the erythrocyte membrane, in contrast to ZIP14 which is not identified using immunoblotting approach. A direct measurement of the ZIP8 protein expression in erythrocyte membranes could provide valuable information for further analyzing its biological function.
Collapse
Affiliation(s)
- Varalakshmi Vungutur
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Suetmui Yu
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Shannon McCabe
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Caitlin Fung
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
28
|
Sofer T, Kurniansyah N, Murray M, Ho YL, Abner E, Esko T, Huffman JE, Cho K, Wilson PWF, Gottlieb DJ. Genome-wide association study of obstructive sleep apnoea in the Million Veteran Program uncovers genetic heterogeneity by sex. EBioMedicine 2023; 90:104536. [PMID: 36989840 PMCID: PMC10065974 DOI: 10.1016/j.ebiom.2023.104536] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) for obstructive sleep apnoea (OSA) are limited due to the underdiagnosis of OSA, leading to misclassification of OSA, which consequently reduces statistical power. We performed a GWAS of OSA in the Million Veteran Program (MVP) of the U.S. Department of Veterans Affairs (VA) healthcare system, where OSA prevalence is close to its true population prevalence. METHODS We performed GWAS of 568,576 MVP participants, stratified by biological sex and by harmonized race/ethnicity and genetic ancestry (HARE) groups of White, Black, Hispanic, and Asian individuals. We considered both BMI adjusted (BMI-adj) and unadjusted (BMI-unadj) models. We replicated associations in independent datasets, and analysed the heterogeneity of OSA genetic associations across HARE and sex groups. We finally performed a larger meta-analysis GWAS of MVP, FinnGen, and the MGB Biobank, totalling 916,696 individuals. FINDINGS MVP participants are 91% male. OSA prevalence is 21%. In MVP there were 18 and 6 genome-wide significant loci in BMI-unadj and BMI-adj analyses, respectively, corresponding to 21 association regions. Of these, 17 were not previously reported in association with OSA, and 13 replicated in FinnGen (False Discovery Rate p-value < 0.05). There were widespread significant differences in genetic effects between men and women, but less so across HARE groups. Meta-analysis of MVP, FinnGen, and MGB biobank revealed 17 additional, previously unreported, genome-wide significant regions. INTERPRETATION Sex differences in genetic associations with OSA are widespread, likely associated with multiple OSA risk factors. OSA shares genetic underpinnings with several sleep phenotypes, suggesting shared aetiology and causal pathways. FUNDING Described in acknowledgements.
Collapse
Affiliation(s)
- Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Murray
- Massachusetts Veterans Epidemiology Research and Information Center, VA Healthcare System, Boston, MA, USA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Healthcare System, Boston, MA, USA
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center, VA Healthcare System, Boston, MA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Healthcare System, Boston, MA, USA; Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Massachusetts Veterans Epidemiology Research and Information Center, VA Healthcare System, Boston, MA, USA
| |
Collapse
|
29
|
Sunuwar L, Tomar V, Wildeman A, Culotta V, Melia J. Hepatobiliary manganese homeostasis is dynamic in the setting of illness in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533688. [PMID: 36993204 PMCID: PMC10055399 DOI: 10.1101/2023.03.22.533688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Manganese is a diet-derived micronutrient that is essential for critical cellular processes like redox homeostasis, protein glycosylation, and lipid and carbohydrate metabolism. Control of Mn availability, especially at the local site of infection, is a key component of the innate immune response. Less has been elucidated about Mn homeostasis at the systemic level. In this work, we demonstrate that systemic Mn homeostasis is dynamic in response to illness in mice. This phenomenon is evidenced in male and female mice, mice of two genetic backgrounds (C57/BL6 and BALB/c), in multiple models of acute (dextran-sodium sulfate-induced) and chronic ( enterotoxigenic Bacteriodes fragilis ) colitis, and systemic infection with Candida albicans . When mice were fed a standard corn-based chow with excess Mn (100 ppm), liver Mn decreased and biliary Mn increased 3-fold in response to infection or colitis. Liver iron, copper, and zinc were unchanged. When dietary Mn was restricted to minimally adequate amounts (10ppm), baseline hepatic Mn levels decreased by approximately 60% in the liver, and upon induction of colitis, liver Mn did not decrease further, however biliary Mn still increased 20-fold. In response to acute colitis, hepatic Slc39a8 mRNA (gene encoding the Mn importer, Zip8) and Slc30a10 mRNA (gene encoding the Mn exporter, Znt10) are decreased. Zip8 protein is decreased. Illness- associated dynamic Mn homeostasis may represent a novel host immune/inflammatory response that reorganizes systemic Mn availability through differential expression of key Mn transporters with down-regulation of Zip8.
Collapse
|
30
|
Yu S, Zhao N. The Regulation of ZIP8 by Dietary Manganese in Mice. Int J Mol Sci 2023; 24:ijms24065962. [PMID: 36983036 PMCID: PMC10056016 DOI: 10.3390/ijms24065962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
ZIP8 is a newly identified manganese transporter. A lack of functional ZIP8 results in severe manganese deficiency in both humans and mice, indicating that ZIP8 plays a crucial role in maintaining body manganese homeostasis. Despite a well-acknowledged connection between ZIP8 and manganese metabolism, how ZIP8 is regulated under high-manganese conditions remains unclear. The primary goal of this study was to examine the regulation of ZIP8 by high-manganese intake. We used both neonatal and adult mouse models in which mice were supplied with dietary sources containing either a normal or a high level of manganese. We discovered that high-manganese intake caused a reduction in liver ZIP8 protein in young mice. Since a decrease in hepatic ZIP8 leads to reduced manganese reabsorption from the bile, our study identified a novel mechanism for the regulation of manganese homeostasis under high-manganese conditions: high dietary manganese intake results in a decrease in ZIP8 in the liver, which in turn decreases the reabsorption of manganese from the bile to prevent manganese overload in the liver. Interestingly, we found that a high-manganese diet did not cause a decrease in hepatic ZIP8 in adult animals. To determine the potential reason for this age-dependent variation, we compared the expressions of liver ZIP8 in 3-week-old and 12-week-old mice. We found that liver ZIP8 protein content in 12-week-old mice decreases when compared with that of 3-week-old mice under normal conditions. Overall, results from this study provide novel insights to facilitate the understanding of ZIP8's function in regulating manganese metabolism.
Collapse
Affiliation(s)
- Suetmui Yu
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
31
|
Vourdoumpa A, Paltoglou G, Charmandari E. The Genetic Basis of Childhood Obesity: A Systematic Review. Nutrients 2023; 15:1416. [PMID: 36986146 PMCID: PMC10058966 DOI: 10.3390/nu15061416] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Overweight and obesity in childhood and adolescence represents one of the most challenging public health problems of our century owing to its epidemic proportions and the associated significant morbidity, mortality, and increase in public health costs. The pathogenesis of polygenic obesity is multifactorial and is due to the interaction among genetic, epigenetic, and environmental factors. More than 1100 independent genetic loci associated with obesity traits have been currently identified, and there is great interest in the decoding of their biological functions and the gene-environment interaction. The present study aimed to systematically review the scientific evidence and to explore the relation of single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs) with changes in body mass index (BMI) and other measures of body composition in children and adolescents with obesity, as well as their response to lifestyle interventions. Twenty-seven studies were included in the qualitative synthesis, which consisted of 7928 overweight/obese children and adolescents at different stages of pubertal development who underwent multidisciplinary management. The effect of polymorphisms in 92 different genes was assessed and revealed SNPs in 24 genetic loci significantly associated with BMI and/or body composition change, which contribute to the complex metabolic imbalance of obesity, including the regulation of appetite and energy balance, the homeostasis of glucose, lipid, and adipose tissue, as well as their interactions. The decoding of the genetic and molecular/cellular pathophysiology of obesity and the gene-environment interactions, alongside with the individual genotype, will enable us to design targeted and personalized preventive and management interventions for obesity early in life.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
32
|
Ma S, Wang C, Khan A, Liu L, Dalgleish J, Kiryluk K, He Z, Ionita-Laza I. BIGKnock: fine-mapping gene-based associations via knockoff analysis of biobank-scale data. Genome Biol 2023; 24:24. [PMID: 36782330 PMCID: PMC9926792 DOI: 10.1186/s13059-023-02864-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
We propose BIGKnock (BIobank-scale Gene-based association test via Knockoffs), a computationally efficient gene-based testing approach for biobank-scale data, that leverages long-range chromatin interaction data, and performs conditional genome-wide testing via knockoffs. BIGKnock can prioritize causal genes over proxy associations at a locus. We apply BIGKnock to the UK Biobank data with 405,296 participants for multiple binary and quantitative traits, and show that relative to conventional gene-based tests, BIGKnock produces smaller sets of significant genes that contain the causal gene(s) with high probability. We further illustrate its ability to pinpoint potential causal genes at [Formula: see text] of the associated loci.
Collapse
Affiliation(s)
- Shiyang Ma
- Department of Biostatistics, Columbia University, New York, NY, USA
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linxi Liu
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Dalgleish
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zihuai He
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
33
|
Noel M, Chasman DI, Mora S, Otvos JD, Palmer CD, Parsons PJ, Smoller JW, Cummings RD, Mealer RG. The Inflammation Biomarker GlycA Reflects Plasma N-Glycan Branching. Clin Chem 2023; 69:80-87. [PMID: 36254612 PMCID: PMC10726709 DOI: 10.1093/clinchem/hvac160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/11/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND GlycA is a nuclear magnetic resonance (NMR) signal in plasma that correlates with inflammation and cardiovascular outcomes in large data sets. The signal is thought to originate from N-acetylglucosamine (GlcNAc) residues of branched plasma N-glycans, though direct experimental evidence is limited. Trace element concentrations affect plasma glycosylation patterns and may thereby also influence GlycA. METHODS NMR GlycA signal was measured in plasma samples from 87 individuals and correlated with MALDI-MS N-glycomics and trace element analysis. We further evaluated the genetic association with GlycA at rs13107325, a single nucleotide polymorphism resulting in a missense variant within SLC39A8, a manganese transporter that influences N-glycan branching, both in our samples and existing genome-wide association studies data from 22 835 participants in the Women's Health Study (WHS). RESULTS GlycA signal was correlated with both N-glycan branching (r2 ranging from 0.125-0.265; all P < 0.001) and copper concentration (r2 = 0.348, P < 0.0001). In addition, GlycA levels were associated with rs13107325 genotype in the WHS (β [standard error of the mean] = -4.66 [1.2674], P = 0.0002). CONCLUSIONS These results provide the first direct experimental evidence linking the GlycA NMR signal to N-glycan branching commonly associated with acute phase reactive proteins involved in inflammation.
Collapse
Affiliation(s)
- Maxence Noel
- Department of Surgery, National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel I Chasman
- Preventive Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Samia Mora
- Preventive Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Lipid Metabolomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - James D Otvos
- Laboratory Corporation of America Holdings, Morrisville, NC, USA
| | - Christopher D Palmer
- New York State Department of Health, Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - Patrick J Parsons
- New York State Department of Health, Laboratory of Inorganic and Nuclear Chemistry, Wadsworth Center, Albany, NY, USA
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - Jordan W Smoller
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit and Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard D Cummings
- Department of Surgery, National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert G Mealer
- Department of Surgery, National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit and Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Martínez D, Oyarzún-Salazar R, Quilapi AM, Coronado J, Enriquez R, Vargas-Lagos C, Oliver C, Santibañez N, Godoy M, Muñoz JL, Vargas-Chacoff L, Romero A. Live and inactivated Piscirickettsia salmonis activated nutritional immunity in Atlantic salmon ( Salmo salar). Front Immunol 2023; 14:1187209. [PMID: 37187753 PMCID: PMC10175622 DOI: 10.3389/fimmu.2023.1187209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Nutritional immunity regulates the homeostasis of micronutrients such as iron, manganese, and zinc at the systemic and cellular levels, preventing the invading microorganisms from gaining access and thereby limiting their growth. Therefore, the objective of this study was to evaluate the activation of nutritional immunity in specimens of Atlantic salmon (Salmo salar) that are intraperitoneally stimulated with both live and inactivated Piscirickettsia salmonis. The study used liver tissue and blood/plasma samples on days 3, 7, and 14 post-injections (dpi) for the analysis. Genetic material (DNA) of P. salmonis was detected in the liver tissue of fish stimulated with both live and inactivated P. salmonis at 14 dpi. Additionally, the hematocrit percentage decreased at 3 and 7 dpi in fish stimulated with live P. salmonis, unchanged in fish challenged with inactivated P. salmonis. On the other hand, plasma iron content decreased during the experimental course in fish stimulated with both live and inactivated P. salmonis, although this decrease was statistically significant only at 3 dpi. Regarding the immune-nutritional markers such as tfr1, dmt1, and ireg1 were modulated in the two experimental conditions, compared to zip8, ft-h, and hamp, which were down-regulated in fish stimulated with live and inactivated P. salmonis during the course experimental. Finally, the intracellular iron content in the liver increased at 7 and 14 dpi in fish stimulated with live and inactivated P. salmonis, while the zinc content decreased at 14 dpi under both experimental conditions. However, stimulation with live and inactivated P. salmonis did not alter the manganese content in the fish. The results suggest that nutritional immunity does not distinguish between live and inactivated P. salmonis and elicits a similar immune response. Probably, this immune mechanism would be self-activated with the detection of PAMPs, instead of a sequestration and/or competition of micronutrients by the living microorganism.
Collapse
Affiliation(s)
- Danixa Martínez
- Laboratorio Institucional de Investigación, Facultad de Ciencias de la Naturaleza, Universidad San Sebastián, Puerto Montt, Chile
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Danixa Martínez, ; Luis Vargas-Chacoff, ; Alex Romero,
| | - Ricardo Oyarzún-Salazar
- Laboratorio Institucional de Investigación, Facultad de Ciencias de la Naturaleza, Universidad San Sebastián, Puerto Montt, Chile
| | - Ana María Quilapi
- Escuela de Tecnología Médica, Facultad de la Salud, Universidad Santo Tomás, Osorno, Chile
| | - José Coronado
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Ricardo Enriquez
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Vargas-Lagos
- Escuela de Tecnología Médica, Facultad de la Salud, Universidad Santo Tomás, Osorno, Chile
| | - Cristian Oliver
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Natacha Santibañez
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Marcos Godoy
- Laboratorio Institucional de Investigación, Facultad de Ciencias de la Naturaleza, Universidad San Sebastián, Puerto Montt, Chile
- Centro de Investigaciones Biológicas Aplicadas (CIBA), Puerto Montt, Chile
| | - José Luis Muñoz
- Centro de Investigación y Desarrollo i~mar, Universidad de los Lagos, Puerto Montt, Chile
| | - Luis Vargas-Chacoff
- Instituto de Ciencias Marinas y Limnológicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
- Centro Fondap de Investigación de Altas Latitudes (IDEAL), Universidad Austral de Chile, Valdivia, Chile
- Millennium Institute Biodiversity of Antarctic and Subantarctic Ecosystems, Biodiversity of Antarctic and Subantarctic Ecosystems (BASE), University Austral of Chile, Valdivia, Chile
- *Correspondence: Danixa Martínez, ; Luis Vargas-Chacoff, ; Alex Romero,
| | - Alex Romero
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- Centro Fondap Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
- *Correspondence: Danixa Martínez, ; Luis Vargas-Chacoff, ; Alex Romero,
| |
Collapse
|
35
|
Tsuo K, Zhou W, Wang Y, Kanai M, Namba S, Gupta R, Majara L, Nkambule LL, Morisaki T, Okada Y, Neale BM, Daly MJ, Martin AR. Multi-ancestry meta-analysis of asthma identifies novel associations and highlights the value of increased power and diversity. CELL GENOMICS 2022; 2:100212. [PMID: 36778051 PMCID: PMC9903683 DOI: 10.1016/j.xgen.2022.100212] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
Abstract
Asthma is a complex disease that varies widely in prevalence across populations. The extent to which genetic variation contributes to these disparities is unclear, as the genetics underlying asthma have been investigated primarily in populations of European descent. As part of the Global Biobank Meta-analysis Initiative, we conducted a large-scale genome-wide association study of asthma (153,763 cases and 1,647,022 controls) via meta-analysis across 22 biobanks spanning multiple ancestries. We discovered 179 asthma-associated loci, 49 of which were not previously reported. Despite the wide range in asthma prevalence among biobanks, we found largely consistent genetic effects across biobanks and ancestries. The meta-analysis also improved polygenic risk prediction in non-European populations compared with previous studies. Additionally, we found considerable genetic overlap between age-of-onset subtypes and between asthma and comorbid diseases. Our work underscores the multi-factorial nature of asthma development and offers insight into its shared genetic architecture.
Collapse
Affiliation(s)
- Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wei Zhou
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Rahul Gupta
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Lerato Majara
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lethukuthula L. Nkambule
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Takayuki Morisaki
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Minatu-ku, Tokyo, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| | - Benjamin M. Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Global Biobank Meta-analysis Initiative
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Minatu-ku, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Mark J. Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Alicia R. Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
36
|
Tan HW, Xu YM, Liang ZL, Cai NL, Wu YY, Lau ATY. Single-gene knockout-coupled omics analysis identifies C9orf85 and CXorf38 as two uncharacterized human proteins associated with ZIP8 malfunction. Front Mol Biosci 2022; 9:991308. [PMID: 36330220 PMCID: PMC9623088 DOI: 10.3389/fmolb.2022.991308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/13/2022] [Indexed: 02/05/2023] Open
Abstract
Human transmembrane protein metal cation symporter ZIP8 (SLC39A8) is a member of the solute carrier gene family responsible for intracellular transportation of essential micronutrients, including manganese, selenium, and zinc. Previously, we established a ZIP8-knockout (KO) human cell model using the CRISPR/Cas9 system and explored how the expression of ZIP8 could possibly contribute to a wide range of human diseases. To further assess the biophysiological role of ZIP8, in the current study, we employed isobaric tags for relative and absolute quantitation (iTRAQ) and detected the changes of the proteome in ZIP8-KO cells (proteomic data are available via ProteomeXchange with identifier PXD036680). A total of 286 differentially expressed proteins (206 downregulated and 80 upregulated proteins) were detected in the ZIP8-KO cell model, and subsequent bioinformatics analyses (GO, KEGG, KOG, and PPI) were performed on these proteins. Interestingly, four "uncharacterized" proteins (proteins with unknown biological function) were identified in the differentially expressed proteins: C1orf198, C9orf85, C17orf75, and CXorf38-all of which were under-expressed in the ZIP8-KO cells. Notably, C9orf85 and CXorf38 were amongst the top-10 most downregulated proteins, and their expressions could be selectively induced by essential micronutrients. Furthermore, clinical-based bioinformatic analysis indicated that positive correlations between the gene expressions of ZIP8 and C9orf85 or CXorf38 were observed in multiple cancer types. Overall, this study reveals the proteomic landscape of cells with impaired ZIP8 and uncovers the potential relationships between essential micronutrients and uncharacterized proteins C9orf85 and CXorf38. The differentially expressed proteins identified in ZIP8-KO cells could be the potential targets for diagnosing and/or treating human ZIP8-associated diseases, including but not limited to malnutrition, viral infection, and cancers.
Collapse
Affiliation(s)
- Heng Wee Tan
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | | | | | | | | | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
37
|
Zaks N, Austin C, Arora M, Reichenberg A. Reprint of: Elemental dysregulation in psychotic spectrum disorders: A review and research synthesis. Schizophr Res 2022; 247:33-40. [PMID: 36075821 DOI: 10.1016/j.schres.2022.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 10/14/2022]
Abstract
Accumulating evidence from observational studies, genetic research, and animal models suggests a relationship between toxic and nutritive elements and psychotic spectrum disorders (PSD). This review systematically evaluates the current research evidence for two hypotheses: 1) that exposures to abnormal levels of toxic and nutritive elements early in life contributes to the subsequent development of PSD, and 2) that an imbalance of element levels is linked to psychotic illness and clinical severity. We focused on the extant literature on five elements, lead (Pb), copper (Cu), magnesium (Mg), manganese (Mn), and zinc (Zn), because of their previously documented associations with psychiatric problems and the availability of pertinent literature. The review identified 38 studies of which 11 measured Pb, 27 measured Cu, 16 measured Mg, 15 measured Mn, and 25 measured Zn concentrations in PSD patients and controls. A majority of research has been conducted on nutritive element imbalance, and findings are largely mixed. While it is biologically plausible that element dysregulation is an important modifiable risk factor for PSD, more research into exposure in early life is needed to better characterize this relationship.
Collapse
Affiliation(s)
- Nina Zaks
- Department of Psychiatry, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine, Mount Sinai, NY, USA; Department of Environmental Medicine and Public Health, Icahn School of Medicine, Mount Sinai, NY, USA; Seaver Center of Research and Treatment, Icahn School of Medicine, Mount Sinai, NY, USA; Friedman Brain Institute, Icahn School of Medicine, Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Computational Modeling of Macrophage Iron Sequestration during Host Defense against Aspergillus. mSphere 2022; 7:e0007422. [PMID: 35862797 PMCID: PMC9429928 DOI: 10.1128/msphere.00074-22] [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] [Indexed: 11/21/2022] Open
Abstract
Iron is essential to the virulence of Aspergillus species, and restricting iron availability is a critical mechanism of antimicrobial host defense. Macrophages recruited to the site of infection are at the crux of this process, employing multiple intersecting mechanisms to orchestrate iron sequestration from pathogens. To gain an integrated understanding of how this is achieved in aspergillosis, we generated a transcriptomic time series of the response of human monocyte-derived macrophages to Aspergillus and used this and the available literature to construct a mechanistic computational model of iron handling of macrophages during this infection. We found an overwhelming macrophage response beginning 2 to 4 h after exposure to the fungus, which included upregulated transcription of iron import proteins transferrin receptor-1, divalent metal transporter-1, and ZIP family transporters, and downregulated transcription of the iron exporter ferroportin. The computational model, based on a discrete dynamical systems framework, consisted of 21 3-state nodes, and was validated with additional experimental data that were not used in model generation. The model accurately captures the steady state and the trajectories of most of the quantitatively measured nodes. In the experimental data, we surprisingly found that transferrin receptor-1 upregulation preceded the induction of inflammatory cytokines, a feature that deviated from model predictions. Model simulations suggested that direct induction of transferrin receptor-1 (TfR1) after fungal recognition, independent of the iron regulatory protein-labile iron pool (IRP-LIP) system, explains this finding. We anticipate that this model will contribute to a quantitative understanding of iron regulation as a fundamental host defense mechanism during aspergillosis. IMPORTANCE Invasive pulmonary aspergillosis is a major cause of death among immunosuppressed individuals despite the best available therapy. Depriving the pathogen of iron is an essential component of host defense in this infection, but the mechanisms by which the host achieves this are complex. To understand how recruited macrophages mediate iron deprivation during the infection, we developed and validated a mechanistic computational model that integrates the available information in the field. The insights provided by this approach can help in designing iron modulation therapies as anti-fungal treatments.
Collapse
|
39
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
40
|
Hafey MJ, Aleksunes LM, Bridges CC, Brouwer KR, Chien HC, Leslie EM, Hu S, Li Y, Shen J, Sparreboom A, Sprowl J, Tweedie D, Lai Y. Transporters and Toxicity: Insights from the International Transporter Consortium Workshop 4. Clin Pharmacol Ther 2022; 112:527-539. [PMID: 35546260 DOI: 10.1002/cpt.2638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/30/2022] [Indexed: 12/29/2022]
Abstract
Over the last decade, significant progress been made in elucidating the role of membrane transporters in altering drug disposition, with important toxicological consequences due to changes in localized concentrations of compounds. The topic of "Transporters and Toxicity" was recently highlighted as a scientific session at the International Transporter Consortium (ITC) Workshop 4 in 2021. The current white paper is not intended to be an extensive review on the topic of transporters and toxicity but an opportunity to highlight aspects of the role of transporters in various toxicities with clinically relevant implications as covered during the session. This includes a review of the role of solute carrier transporters in anticancer drug-induced organ injury, transporters as key players in organ barrier function, and the role of transporters in metal/metalloid toxicity.
Collapse
Affiliation(s)
- Michael J Hafey
- ADME and Discovery Toxicology, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey, USA
| | - Christy C Bridges
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, Georgia, USA
| | | | - Huan-Chieh Chien
- Pharmacokinetics and Drug Metabolism, Amgen, Inc., South San Francisco, California, USA
| | - Elaine M Leslie
- Departments of Physiology and Lab Med and Path, Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Shuiying Hu
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jinshan Shen
- Relay Therapeutics, Cambridge, Massachusetts, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jason Sprowl
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| |
Collapse
|
41
|
Wang W, Chen T, Liu Y, Wang S, Yang N, Luo M. Predictive value of single-nucleotide polymorphisms in curve progression of adolescent idiopathic scoliosis. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2022; 31:2311-2325. [PMID: 35434775 DOI: 10.1007/s00586-022-07213-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE Genetic diagnosis is a promising approach because several single-nucleotide polymorphisms (SNPs) associated with adolescent idiopathic scoliosis (AIS) progression have been reported. We review the predictive value of SNPs in curve progression of adolescent idiopathic scoliosis. METHODS We reviewed DNA-based prognostic testing to predict curve progression. Then, the multiple polymorphisms in loci related to AIS progression were also reviewed, and we elucidated the predictive value of SNPs from four functional perspectives, including endocrine metabolism, neuromuscular system, cartilage and extracellular matrix, enzymes, and cytokines. RESULTS The ScoliScores were less successful predictors than expected, and the weak power of predictive SNPs might account for its failure. Susceptibility loci in ESR1, ESR2, GPER, and IGF1, which related to endocrine metabolism, have been reported to predict AIS progression. Neuromuscular imbalance might be a potential mechanism of scoliosis, and SNPs in LBX1, NTF3, and SOCS3 have been reported to predict the curve progression of AIS. Susceptibility loci in SOX9, MATN1, AJAP1, MMP9, and TIMP2, which are related to cartilage and extracellular matrix, are also potentially related to AIS progression. Enzymes and cytokines play essential roles in regulating bone metabolism and embryonic development. SNPs in BNC2, SLC39A8, TGFB1, IL-6, IL-17RC, and CHD7 were suggested as predictive loci for AIS curve progression. CONCLUSIONS Many promising SNPs have been identified to predict the curve progression of AIS. However, conflicting results from replication studies and different ethnic groups hamper their reliability. Convincing SNPs from multiethnic populations and functional verification are needed.
Collapse
Affiliation(s)
- Wengang Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Tailong Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Yibin Liu
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Songsong Wang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Ningning Yang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China. .,Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China.
| | - Ming Luo
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
42
|
Martínez DP, Oliver C, Santibañez N, Coronado JL, Oyarzún-Salazar R, Enriquez R, Vargas-Chacoff L, Romero A. PAMPs of Piscirickettsia salmonis Trigger the Transcription of Genes Involved in Nutritional Immunity in a Salmon Macrophage-Like Cell Line. Front Immunol 2022; 13:849752. [PMID: 35493529 PMCID: PMC9046600 DOI: 10.3389/fimmu.2022.849752] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune system can limit the growth of invading pathogens by depleting micronutrients at a cellular and tissue level. However, it is not known whether nutrient depletion mechanisms discriminate between living pathogens (which require nutrients) and pathogen-associated molecular patterns (PAMPs) (which do not). We stimulated SHK-1 cells with different PAMPs (outer membrane vesicles of Piscirickettsia salmonis "OMVs", protein extract of P. salmonis "TP" and lipopolysaccharides of P. salmonis "LPS") isolated from P. salmonis and evaluated transcriptional changes in nutritional immunity associated genes. Our experimental treatments were: Control (SHK-1 stimulated with bacterial culture medium), OMVs (SHK-1 stimulated with 1μg of outer membrane vesicles), TP (SHK-1 stimulated with 1μg of total protein extract) and LPS (SHK-1 stimulated with 1μg of lipopolysaccharides). Cells were sampled at 15-, 30-, 60- and 120-minutes post-stimulation. We detected increased transcription of zip8, zip14, irp1, irp2 and tfr1 in all three experimental conditions and increased transcription of dmt1 in cells stimulated with OMVs and TP, but not LPS. Additionally, we observed generally increased transcription of ireg-1, il-6, hamp, irp1, ft-h and ft-m in all three experimental conditions, but we also detected decreased transcription of these markers in cells stimulated with TP and LPS at specific time points. Our results demonstrate that SHK-1 cells stimulated with P. salmonis PAMPs increase transcription of markers involved in the transport, uptake, storage and regulation of micronutrients such as iron, manganese and zinc.
Collapse
Affiliation(s)
- Danixa Pamela Martínez
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- Laboratorio de Fisiología de peces, Instituto de Ciencias Marinas y Limnológicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Cristian Oliver
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Natacha Santibañez
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - José Leonardo Coronado
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Ricardo Oyarzún-Salazar
- Laboratorio de Fisiología de peces, Instituto de Ciencias Marinas y Limnológicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Ricardo Enriquez
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Vargas-Chacoff
- Laboratorio de Fisiología de peces, Instituto de Ciencias Marinas y Limnológicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
- Centro Fondap de Investigación de Altas Latitudes (IDEAL), Universidad Austral de Chile, Valdivia, Chile
- Millennium Institute Biodiversity of Antarctic and Subantarctic Ecosystems, BASE, University Austral of Chile, Valdivia, Chile
| | - Alex Romero
- Laboratorio de Inmunología y estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- Centro Fondap Interdisciplinary Center for Aquaculture Research (INCAR), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
43
|
Mealer RG, Williams SE, Noel M, Yang B, D’Souza AK, Nakata T, Graham DB, Creasey EA, Cetinbas M, Sadreyev RI, Scolnick EM, Woo CM, Smoller JW, Xavier RJ, Cummings RD. The schizophrenia-associated variant in SLC39A8 alters protein glycosylation in the mouse brain. Mol Psychiatry 2022; 27:1405-1415. [PMID: 35260802 PMCID: PMC9106890 DOI: 10.1038/s41380-022-01490-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 01/13/2023]
Abstract
A missense mutation (A391T) in SLC39A8 is strongly associated with schizophrenia in genomic studies, though the molecular connection to the brain is unknown. Human carriers of A391T have reduced serum manganese, altered plasma glycosylation, and brain MRI changes consistent with altered metal transport. Here, using a knock-in mouse model homozygous for A391T, we show that the schizophrenia-associated variant changes protein glycosylation in the brain. Glycosylation of Asn residues in glycoproteins (N-glycosylation) was most significantly impaired, with effects differing between regions. RNAseq analysis showed negligible regional variation, consistent with changes in the activity of glycosylation enzymes rather than gene expression. Finally, nearly one-third of detected glycoproteins were differentially N-glycosylated in the cortex, including members of several pathways previously implicated in schizophrenia, such as cell adhesion molecules and neurotransmitter receptors that are expressed across all cell types. These findings provide a mechanistic link between a risk allele and potentially reversible biochemical changes in the brain, furthering our molecular understanding of the pathophysiology of schizophrenia and a novel opportunity for therapeutic development.
Collapse
Affiliation(s)
- Robert G. Mealer
- Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital. Harvard Medical School, Boston, MA.,National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.,The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA.,Corresponding Author: Robert Gene Mealer, M.D., Ph.D., Richard B. Simches Research Center, 185 Cambridge St, 6th Floor, Boston, MA 02114,
| | - Sarah E. Williams
- Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Maxence Noel
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Bo Yang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA
| | | | - Toru Nakata
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel B. Graham
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth A. Creasey
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Edward M. Scolnick
- Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA
| | - Christina M. Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital. Harvard Medical School, Boston, MA.,The Stanley Center for Psychiatric Research at Broad Institute of Harvard/MIT, Cambridge, MA
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Richard D. Cummings
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Li Y, Chen Y, Jiang L, Zhang J, Tong X, Chen D, Le W. Intestinal Inflammation and Parkinson's Disease. Aging Dis 2021; 12:2052-2068. [PMID: 34881085 PMCID: PMC8612622 DOI: 10.14336/ad.2021.0418] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease which significantly influences the life quality of patients. The protein α-synuclein plays an important driving role in PD occurrence and development. Braak's hypothesis suggests that α-synuclein is produced in intestine, and then spreads into the central nervous system through the vagus nerve. The abnormal expression of α-synuclein has been found in inflammatory bowel disease (IBD). Intestinal inflammation and intestinal dysbiosis have been involved in the occurrence and development of PD. The present review aimed to summarize recent advancements in studies focusing on intestinal inflammation and PD, especially the mechanisms through which link intestinal inflammation and PD. The intestinal dysfunctions such as constipation have been introduced as non-motor manifestations of PD. The possible linkages between IBD and PD, including genetic overlaps, inflammatory responses, intestinal permeability, and intestinal dysbiosis, are mainly discussed. Although it is not confirmed whether PD starts from intestine, intestinal dysfunction may affect intestinal microenvironment to influence central nervous system, including the α-synuclein pathologies and systematic inflammation. It is expected to develop some new strategies in the diagnosis and treatment of PD from the aspect of intestine. It may also become an exciting direction to find better ways to regulate the composition of gut microorganism to treat PD.
Collapse
Affiliation(s)
- Yu Li
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Yuanyuan Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Lili Jiang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Jingyu Zhang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Xuhui Tong
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
- Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Pujol‐Giménez J, Poirier M, Bühlmann S, Schuppisser C, Bhardwaj R, Awale M, Visini R, Javor S, Hediger MA, Reymond J. Inhibitors of Human Divalent Metal Transporters DMT1 (SLC11A2) and ZIP8 (SLC39A8) from a GDB-17 Fragment Library. ChemMedChem 2021; 16:3306-3314. [PMID: 34309203 PMCID: PMC8596699 DOI: 10.1002/cmdc.202100467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Indexed: 11/06/2022]
Abstract
Solute carrier proteins (SLCs) are membrane proteins controlling fluxes across biological membranes and represent an emerging class of drug targets. Here we searched for inhibitors of divalent metal transporters in a library of 1,676 commercially available 3D-shaped fragment-like molecules from the generated database GDB-17, which lists all possible organic molecules up to 17 atoms of C, N, O, S and halogen following simple criteria for chemical stability and synthetic feasibility. While screening against DMT1 (SLC11A2), an iron transporter associated with hemochromatosis and for which only very few inhibitors are known, only yielded two weak inhibitors, our approach led to the discovery of the first inhibitor of ZIP8 (SLC39A8), a zinc transporter associated with manganese homeostasis and osteoarthritis but with no previously reported pharmacology, demonstrating that this target is druggable.
Collapse
Affiliation(s)
- Jonai Pujol‐Giménez
- Department of Biomedical Research and Department of Nephrology and Hypertension Membrane Transport Discovery Lab Inselspital, Bern University HospitalUniversity of BernCH-3010BernSwitzerland
| | - Marion Poirier
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Sven Bühlmann
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Céline Schuppisser
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Rajesh Bhardwaj
- Department of Biomedical Research and Department of Nephrology and Hypertension Membrane Transport Discovery Lab Inselspital, Bern University HospitalUniversity of BernCH-3010BernSwitzerland
| | - Mahendra Awale
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Ricardo Visini
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Sacha Javor
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Matthias A. Hediger
- Department of Biomedical Research and Department of Nephrology and Hypertension Membrane Transport Discovery Lab Inselspital, Bern University HospitalUniversity of BernCH-3010BernSwitzerland
| | - Jean‐Louis Reymond
- Department of Chemistry Biochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| |
Collapse
|
46
|
Budinger D, Barral S, Soo AKS, Kurian MA. The role of manganese dysregulation in neurological disease: emerging evidence. Lancet Neurol 2021; 20:956-968. [PMID: 34687639 DOI: 10.1016/s1474-4422(21)00238-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022]
Abstract
Manganese is an essential trace metal. The dysregulation of manganese seen in a broad spectrum of neurological disorders reflects its importance in brain development and key neurophysiological processes. Historically, the observation of acquired manganism in miners and people who misuse drugs provided early evidence of brain toxicity related to manganese exposure. The identification of inherited manganese transportopathies, which cause neurodevelopmental and neurodegenerative syndromes, further corroborates the neurotoxic potential of this element. Moreover, manganese dyshomoeostasis is also implicated in Parkinson's disease and other neurodegenerative conditions, such as Alzheimer's disease and Huntington's disease. Ongoing and future research will facilitate the development of better targeted therapeutical strategies than are currently available for manganese-associated neurological disorders.
Collapse
Affiliation(s)
- Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Audrey K S Soo
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK; Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK; Department of Neurology, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
47
|
Park JH, Marquardt T. Treatment Options in Congenital Disorders of Glycosylation. Front Genet 2021; 12:735348. [PMID: 34567084 PMCID: PMC8461064 DOI: 10.3389/fgene.2021.735348] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Despite advances in the identification and diagnosis of congenital disorders of glycosylation (CDG), treatment options remain limited and are often constrained to symptomatic management of disease manifestations. However, recent years have seen significant advances in treatment and novel therapies aimed both at the causative defect and secondary disease manifestations have been transferred from bench to bedside. In this review, we aim to give a detailed overview of the available therapies and rising concepts to treat these ultra-rare diseases.
Collapse
Affiliation(s)
- Julien H Park
- Department of General Pediatrics, Metabolic Diseases, University Children's Hospital Münster, Münster, Germany
| | - Thorsten Marquardt
- Department of General Pediatrics, Metabolic Diseases, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
48
|
Barowsky S, Jung JY, Nesbit N, Silberstein M, Fava M, Loggia ML, Smoller JW, Lee PH. Cross-Disorder Genomics Data Analysis Elucidates a Shared Genetic Basis Between Major Depression and Osteoarthritis Pain. Front Genet 2021; 12:687687. [PMID: 34603368 PMCID: PMC8481820 DOI: 10.3389/fgene.2021.687687] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/23/2021] [Indexed: 11/24/2022] Open
Abstract
Osteoarthritis (OA) and major depression (MD) are two debilitating disorders that frequently co-occur and affect millions of the elderly each year. Despite the greater symptom severity, poorer clinical outcomes, and increased mortality of the comorbid conditions, we have a limited understanding of their etiologic relationships. In this study, we conducted the first cross-disorder investigations of OA and MD, using genome-wide association data representing over 247K cases and 475K controls. Along with significant positive genome-wide genetic correlations (r g = 0.299 ± 0.026, p = 9.10 × 10-31), Mendelian randomization (MR) analysis identified a bidirectional causal effect between OA and MD (βOA → MD = 0.09, SE = 0.02, z-score p-value < 1.02 × 10-5; βMD → OA = 0.19, SE = 0.026, p < 2.67 × 10-13), indicating genetic variants affecting OA risk are, in part, shared with those influencing MD risk. Cross-disorder meta-analysis of OA and MD identified 56 genomic risk loci (P meta ≤ 5 × 10-8), which show heightened expression of the associated genes in the brain and pituitary. Gene-set enrichment analysis highlighted "mechanosensory behavior" genes (GO:0007638; P gene_set = 2.45 × 10-8) as potential biological mechanisms that simultaneously increase susceptibility to these mental and physical health conditions. Taken together, these findings show that OA and MD share common genetic risk mechanisms, one of which centers on the neural response to the sensation of mechanical stimulus. Further investigation is warranted to elaborate the etiologic mechanisms of the pleiotropic risk genes, as well as to develop early intervention and integrative clinical care of these serious conditions that disproportionally affect the aging population.
Collapse
Affiliation(s)
- Sophie Barowsky
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Jae-Yoon Jung
- Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Nicholas Nesbit
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Micah Silberstein
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Maurizio Fava
- Department of Psychiatry, Harvard Medical School and Massachusetts General Hospital, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, United States
| | - Marco L. Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School and Massachusetts General Hospital, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Phil H. Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School and Massachusetts General Hospital, Boston, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
49
|
Bonaventura E, Barone R, Sturiale L, Pasquariello R, Alessandrì MG, Pinto AM, Renieri A, Panteghini C, Garavaglia B, Cioni G, Battini R. Clinical, molecular and glycophenotype insights in SLC39A8-CDG. Orphanet J Rare Dis 2021; 16:307. [PMID: 34246313 PMCID: PMC8272319 DOI: 10.1186/s13023-021-01941-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/02/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND SLC39A8, a gene located on chromosome 4q24, encodes for the manganese (Mn) transporter ZIP8 and its detrimental variants cause a type 2 congenital disorder of glycosylation (CDG). The common SLC39A8 missense variant A391T is associated with increased risk for multiple neurological and systemic disorders and with decreased serum Mn. Patients with SLC39A8-CDG present with different clinical and neuroradiological features linked to variable transferrin glycosylation profile. Galactose and Mn supplementation therapy results in the biochemical and clinical amelioration of treated patients. RESULTS Here, we report clinical manifestations, neuroradiological features and glycophenotypes associated with novel SLC39A8 variants (c.1048G > A; p.Gly350Arg and c.131C > G; p.Ser44Trp) in two siblings of the same Italian family. Furthermore, we describe a third patient with overlapping clinical features harbouring the homozygous missense variant A391T. The clinical phenotype of the three patients was characterized by severe developmental disability, dystonic postural pattern and dyskinesia with a more severe progression of the disease in the two affected siblings. Neuroimaging showed a Leigh syndrome-like pattern involving the basal ganglia, thalami and white matter. In the two siblings, atrophic cerebral and cerebellum changes consistent with SLC39A8-CDG were detected as well. Serum transferrin isoelectric focusing (IEF) yielded variable results with slight increase of trisialotransferrin isoforms or even normal pattern. MALDI-MS showed the presence of hypogalactosylated transferrin N-glycans, spontaneously decreasing during the disease course, only in one affected sibling. Total serum N-glycome depicted a distinct pattern for the three patients, with increased levels of undergalactosylated and undersialylated precursors of fully sialylated biantennary glycans, including the monosialo-monogalacto-biantennary species A2G1S1. CONCLUSIONS Clinical, MRI and glycosylation features of patients are consistent with SLC39A8-CDG. We document two novel variants associated with Leigh syndrome-like disease presentation of SLC39A8-CDG. We show, for the first time, a severe neurological phenotype overlapping with that described for SLC39A8-CDG in association with the homozygous A391T missense variant. We observed a spontaneous amelioration of transferrin N-glycome, highlighting the efficacy of MS-based serum glycomics as auxiliary tool for the diagnosis and clinical management of therapy response in patients with SLC39A8-CDG. Further studies are needed to analyse more in depth the influence of SLC39A8 variants, including the common missense variant, on the expression and function of ZIP8 protein, and their impact on clinical, biochemical and neuroradiological features.
Collapse
Affiliation(s)
- Eleonora Bonaventura
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 331, Calambrone, 56128, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rita Barone
- Department of Clinical and Experimental Medicine-Child Neuropsychiatry Section, University of Catania, Catania, Italy
- CNR, Institute for Polymers, Composites and Biomaterials, IPCB, Catania, Italy
| | - Luisa Sturiale
- CNR, Institute for Polymers, Composites and Biomaterials, IPCB, Catania, Italy
| | - Rosa Pasquariello
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 331, Calambrone, 56128, Pisa, Italy
| | - Maria Grazia Alessandrì
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 331, Calambrone, 56128, Pisa, Italy
| | - Anna Maria Pinto
- Department of Medical Genetics, University of Siena, Siena, Italy
| | - Alessandra Renieri
- Department of Medical Genetics, University of Siena, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, Siena, Italy
| | - Celeste Panteghini
- Department of Molecular Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Barbara Garavaglia
- Department of Molecular Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 331, Calambrone, 56128, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Viale del Tirreno 331, Calambrone, 56128, Pisa, Italy.
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
50
|
Elemental dysregulation in psychotic spectrum disorders: A review and research synthesis. Schizophr Res 2021; 233:64-71. [PMID: 34242950 DOI: 10.1016/j.schres.2021.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
Accumulating evidence from observational studies, genetic research, and animal models suggests a relationship between toxic and nutritive elements and psychotic spectrum disorders (PSD). This review systematically evaluates the current research evidence for two hypotheses: 1) that exposures to abnormal levels of toxic and nutritive elements early in life contributes to the subsequent development of PSD, and 2) that an imbalance of element levels is linked to psychotic illness and clinical severity. We focused on the extant literature on five elements, lead (Pb), copper (Cu), magnesium (Mg), manganese (Mn), and zinc (Zn), because of their previously documented associations with psychiatric problems and the availability of pertinent literature. The review identified 38 studies of which 11 measured Pb, 27 measured Cu, 16 measured Mg, 15 measured Mn, and 25 measured Zn concentrations in PSD patients and controls. A majority of research has been conducted on nutritive element imbalance, and findings are largely mixed. While it is biologically plausible that element dysregulation is an important modifiable risk factor for PSD, more research into exposure in early life is needed to better characterize this relationship.
Collapse
|