1
|
Fang S, Clayton PT, Garg D, Yoganathan S, Zaki MS, Helgadottir EA, Palmadottir VK, Landry M, Gospe SM, Mankad K, Bonifati V, Sharma S, Tuschl K. Consensus of Expert Opinion for the Diagnosis and Management of Hypermanganesaemia With Dystonia 1 and 2. J Inherit Metab Dis 2025; 48:e70031. [PMID: 40320765 PMCID: PMC12050909 DOI: 10.1002/jimd.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
Hypermanganesaemia with Dystonia 1 and 2 (HMNDYT1 and 2) are inherited, autosomal recessive disorders caused by pathogenic variants in the genes encoding the manganese transporters SLC30A10 and SLC39A14, respectively. Impaired hepatic and enterocytic manganese uptake (SLC39A14) and excretion (SLC30A10) lead to deposition of manganese in the basal ganglia resulting in childhood-onset dystonia-parkinsonism. HMNDYT1 is characterized by additional features due to manganese accumulation in the liver causing cirrhosis, polycythaemia, and depleted iron stores. High blood manganese levels and pathognomonic MRI brain appearances of manganese deposition resulting in T1 hyperintensity of the basal ganglia are diagnostic clues. Treatment is limited to chelation therapy and iron supplementation that can prevent disease progression. Due to their rarity, the awareness of the inherited manganese transporter defects is limited. Here, we provide consensus expert recommendations for the diagnosis and treatment of patients with HMNDYT1 and 2 in order to facilitate early diagnosis and optimize clinical outcome. These recommendations were developed through an evidence and consensus-based process led by a group of 13 international experts across the disciplines of metabolic medicine, neurology, hematology, genetics, and radiology, and address the clinical presentation, diagnostic investigations, principles of treatment, and monitoring of patients with HMNDYT1 and 2.
Collapse
Affiliation(s)
- Sherry Fang
- Department of Metabolic MedicineGreat Ormond Street Hospital for ChildrenLondonUK
| | - Peter T. Clayton
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| | - Divyani Garg
- Department of NeurologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Sangeetha Yoganathan
- Paediatric Neurology Unit, Department of Neurological SciencesChristian Medical CollegeVelloreIndia
| | - Maha S. Zaki
- Department of Clinical GeneticsHuman Genetics and Genome Research Institute, National Research CentreCairoEgypt
| | | | | | - Maude Landry
- The Moncton Hospital, Horizon Health NetworkMonctonCanada
| | - Sidney M. Gospe
- Department of Neurology and PediatricsUniversity of WashingtonSeattleWashingtonUSA
- Department of PediatricsDuke UniversityDurhamNorth CarolinaUSA
| | - Kshitij Mankad
- Department of RadiologyGreat Ormond Street Hospital for ChildrenLondonUK
| | - Vincenzo Bonifati
- Erasmus MC, University Medical Center RotterdamRotterdamthe Netherlands
| | - Suvasini Sharma
- Department of PediatricsLady Hardinge Medical College and Associated Kalawati Saran Children's HospitalDelhiIndia
| | - Karin Tuschl
- Department of Metabolic MedicineGreat Ormond Street Hospital for ChildrenLondonUK
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
2
|
Zhang Y, Hu R, Su M, Hu J. Probing the substrate binding-induced conformational change of a ZIP metal transporter using a sandwich ELISA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642161. [PMID: 40161815 PMCID: PMC11952358 DOI: 10.1101/2025.03.09.642161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Zrt-/Irt-like proteins (ZIPs), a family of divalent metal transporters, are crucial for maintaining the homeostasis of zinc, an essential trace element involved in numerous biological processes. While extensive research on the prototypical ZIP from Bordetella bronchiseptica (BbZIP) have suggested an elevator transport mechanism, the dynamic conformational changes during the transport cycle have not been thoroughly studied. In this work, we developed a sandwich ELISA using a custom anti-BbZIP monoclonal antibody to investigate the conformational change induced by the metal binding to the transport site. This was achieved by determining the accessibility of a cysteine residue introduced at a position exposed to the solvent only when the transporter adopts an outward-facing conformation. This assay allowed us to report the dissociation constants of BbZIP for Zn2+ and Cd2+ at low and sub-micromolar levels, respectively. Notably, the installation of a positive charge at the M2 site drastically reduced metal binding at the M1 site, consistent with an auxiliary role for the M2 site in metal transport. We also demonstrated that this assay can be used to rapidly screen variants for subsequent structural study. We anticipate that other transporters where substrate binding induces large conformational changes can also be studied using this method.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Ryan Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Min Su
- Electron Microscopy Core, University of Missouri, MO 65211
- Department of Biochemistry, University of Missouri, MO 65211
| | - Jian Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
- Department of Chemistry, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
3
|
Xu J, Gu H, Zhou K, Wu L, Zhang Y, Bian C, Huang Z, Chen G, Cheng X, Yin X. ZIP8 Regulates Inflammation and Macrophage Polarisation in Intervertebral Disc Degeneration via the Wnt/β-Catenin Pathway. J Cell Mol Med 2025; 29:e70431. [PMID: 39993958 PMCID: PMC11850097 DOI: 10.1111/jcmm.70431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
One main cause of persistent back discomfort is intervertebral disc degeneration (IDD), with inflammation and extracellular matrix (ECM) degradation playing critical roles. This study investigates the role of ZIP8, a zinc transporter, in IDD pathogenesis, focusing on its effects on inflammatory responses, ECM degradation and Wnt/β-catenin signalling pathway. ZIP8 was identified as a hub gene from the GSE27494 dataset through bioinformatics analysis. The role of ZIP8 was investigated in nucleus pulposus (NP) cells and RAW 264.7 macrophages. An in vivo IDD rat model was used to assess the consequences of ZIP8 overexpression. The involvement of the Wnt/β-catenin pathway was examined, and the effect on macrophage polarisation was analysed. ZIP8 overexpression in NP cells led to increased inflammatory cytokine production and enhanced NF-κB pathway activation, while ZIP8 knockdown alleviated these effects. In vitro, ZIP8 knockdown reduced IL-1β-induced apoptosis and ECM degradation, promoting cell viability. In vivo, ZIP8 overexpression exacerbated disc degeneration, as evidenced by magnetic resonance imaging (MRI) and histological assessments. Additionally, modulation of ZIP8, in conjunction with the Wnt/β-catenin signalling pathway, revealed its involvement in regulating apoptosis and proliferation in NP cells. In RAW 264.7 macrophages, ZIP8 knockdown inhibited M1 macrophage polarisation and reduced proinflammatory cytokine expression, while promoting anti-inflammatory responses. ZIP8 is a key regulator in IDD, affecting inflammation, ECM integrity and Wnt/β-catenin signalling pathways. Targeting ZIP8 by knockdown may offer therapeutic potential in IDD by modulating inflammatory responses and protecting ECM structure, offering a novel approach to IDD treatment.
Collapse
Affiliation(s)
- Jun Xu
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Huijie Gu
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Kaifeng Zhou
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Liang Wu
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Yiming Zhang
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Chong Bian
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Zhongyue Huang
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Guangnan Chen
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Xiangyang Cheng
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| | - Xiaofan Yin
- Department of Orthopaedics, Minhang HospitalFudan UniversityShanghaiChina
| |
Collapse
|
4
|
Alandanoosi A, George F, Liuzzi J. Investigating the relationship among zinc status, blood manganese levels, and enzymatic markers of tissue damage: an epidemiological study using NHANES 2013-2016 data. Toxicol Lett 2025; 403:76-83. [PMID: 39631448 DOI: 10.1016/j.toxlet.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/28/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Manganese is an essential trace element required for various physiological processes. However, excessive exposure to this metal can lead to health issues. This study aims to evaluate whether adequate zinc intake can influence the relationship between blood manganese levels and markers indicating damage to the liver and other organs in populations using epidemiological data. We conducted a comprehensive analysis utilizing 2013-2016 data from the National Health and Nutrition Examination Survey (NHANES). The findings indicated that blood manganese exhibits a significant positive association with the serum levels of enzymatic markers of liver damage alkaline phosphatase and aspartate aminotransferase. However, when investigating the interaction between blood manganese and zinc intake at the second quartile, a significant negative association was found with alkaline phosphatase in three different linear regression models. A similar association was found between the fourth quartile of zinc intake and lactate dehydrogenase activity in all three models of the study. The findings suggest that unhealthy high levels of manganese in populations may lead to tissue injury and disease. Nevertheless, having an adequate zinc intake could help mitigate manganese toxicity.
Collapse
Affiliation(s)
- Afnan Alandanoosi
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, USA
| | - Florence George
- Department of Mathematics and Statistics, College of Arts, Sciences & Education, Florida International University, USA
| | - Juan Liuzzi
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, USA.
| |
Collapse
|
5
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
6
|
Vungutur V, McCabe SM, Zhao N. ZIP8 Is Upregulated in the Testis of Zip14-/- Mice. Nutrients 2024; 16:3575. [PMID: 39519408 PMCID: PMC11547875 DOI: 10.3390/nu16213575] [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: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Manganese is an essential nutrient involved in various biological processes, including reproductive health, yet the mechanisms regulating its homeostasis in the testis remain poorly understood. METHODS AND RESULTS In this study, we investigated the expression and regulation of key manganese transporters-ZIP8, ZIP14, and ZnT10-in mouse testes. Immunoblotting analyses revealed that ZIP8 is expressed in the testes, while ZIP14 and ZnT10 were undetectable. Using Zip14 knockout (Zip14-/-) mice, which exhibit systemic manganese overload, we discovered a significant increase in manganese levels in the testis, accompanied by an upregulation of ZIP8. Importantly, the levels of other essential metals, such as iron, zinc, and copper, remained unchanged. CONCLUSIONS Our findings suggest that ZIP8 plays a critical role in manganese transport in the testis, and its increased expression may contribute to manganese accumulation in the absence of ZIP14. This study advances our understanding of manganese homeostasis in the testis and its potential impact on male reproductive health.
Collapse
Affiliation(s)
| | | | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
7
|
Mori H, Goji A, Hara M. Upregulation of Intracellular Zinc Ion Level after Differentiation of the Neural Stem/Progenitor Cells In Vitro with the Changes in Gene Expression of Zinc Transporters. Biol Trace Elem Res 2024; 202:4699-4714. [PMID: 38180597 DOI: 10.1007/s12011-023-04033-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: 08/16/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
We measured the intracellular zinc ion concentration of murine fetal neural stem/progenitor cells (NSPCs) and that in the differentiated cells. The NSPCs cultured with 1.5 μM Zn2+ proliferated slightly faster than that in the zinc-deficient medium and the intracellular zinc concentration of the NSPCs and that of their differentiated cells (DCs) cultured with 1.5 μM Zn2+ was 1.34-fold and 2.00-fold higher than those in the zinc-deficient medium, respectively. The zinc transporter genes upregulated over the 3.5-fold change were Zip1, Zip4, Zip12, Zip13, ZnT1, ZnT8, and ZnT10 whereas the only downregulated one was Zip8 during the differentiation of NSPCs to DCs. The cell morphologies of both NSPCs and DCs in the low oxygen culture condition consisting of 2%O2 and 5%CO2, the high carbon dioxide condition consisting of 21%O2 and 10%CO2, and the normal condition consisting of 21%O2 and 5%CO2 were essentially the same each other. The expression of Zip4, Zip8, Zip12, and Zip14 was not drastically changed depending on the O2 and CO2 concentrations.
Collapse
Affiliation(s)
- Hideki Mori
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan
| | - Akari Goji
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan
| | - Masayuki Hara
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan.
| |
Collapse
|
8
|
Yang Z, Zhou Z, Meng Q, Chen Z, Yun L, Jiang J, He Y, Dian M, Zhang R, Ge H, Yan T, Men B, Li Z, Wu X, He J, Rao S. Dihydroartemisinin Sensitizes Lung Cancer Cells to Cisplatin Treatment by Upregulating ZIP14 Expression and Inducing Ferroptosis. Cancer Med 2024; 13:e70271. [PMID: 39394878 PMCID: PMC11470233 DOI: 10.1002/cam4.70271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Despite significant advances in lung cancer treatment, cisplatin (DDP)-based chemotherapy remains a cornerstone for managing the disease. However, the prevalence of chemoresistance presents a major challenge, limiting its effectiveness and contributing to poor outcomes. This underscores the urgent need for novel therapeutic strategies to overcome chemoresistance and improve chemotherapy efficacy in lung cancer patients. Exploring approaches to sensitize tumors to cisplatin could enhance treatment responses and overall survival rates. METHODS AND RESULTS Our study utilized a variety of lung cancer models, including cell lines, mouse models, and patient-derived organoids, to validate the synergistic cytotoxic effects of dihydroartemisinin (DHA) and cisplatin (DDP). When combined with DDP, we demonstrate that DHA is a promising therapeutic agent that effectively triggers ferroptosis in lung cancer cells, offering a potential strategy for overcoming chemoresistance. Mechanistically, the combination of DHA and DDP synergistically enhances ZIP14 expression, modulating iron homeostasis and upregulating oxidative stress, leading to both in vitro and in vivo ferroptosis. Notably, our findings revealed that the sequential administration of DDP followed by DHA significantly increases ZIP14 expression and induces superior therapeutic outcomes compared to the simultaneous administration or DHA followed by DDP. This observation underscores the importance of the drug administration order in optimizing treatment efficacy, providing new insights into enhancing chemotherapy response in lung cancer. CONCLUSION Our findings suggest that combining dihydroartemisinin (DHA) with cisplatin (DDP) presents a promising strategy to overcome chemoresistance in lung cancer patients. Importantly, administering DHA during chemotherapy intervals could further optimize treatment outcomes, enhancing the overall efficacy of lung cancer chemotherapy.
Collapse
Affiliation(s)
- Zhuoying Yang
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zehao Zhou
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Qingyu Meng
- Department of Radiation Oncology, Peking Union Medical College HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Zhijie Chen
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Liang Yun
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jianjun Jiang
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yujing He
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Meijuan Dian
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ruihao Zhang
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Haotian Ge
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Tianbao Yan
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Biying Men
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zichao Li
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xu Wu
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Junming He
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
9
|
McCabe SM, Zhao N. Expression of Manganese Transporters ZIP8, ZIP14, and ZnT10 in Brain Barrier Tissues. Int J Mol Sci 2024; 25:10342. [PMID: 39408669 PMCID: PMC11476488 DOI: 10.3390/ijms251910342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Manganese (Mn) is an essential trace mineral for brain function, but excessive accumulation can cause irreversible nervous system damage, highlighting the need for proper Mn balance. ZIP14, ZnT10, and ZIP8 are key transporters involved in maintaining Mn homeostasis, particularly in the absorption and excretion of Mn in the intestine and liver. However, their roles in the brain are less understood. The blood-cerebrospinal fluid barrier and the blood-brain barrier, formed by the choroid plexus and brain blood vessels, respectively, are critical for brain protection and brain metal homeostasis. This study identified ZIP14 on the choroid plexus epithelium, and ZIP8 and ZnT10 in brain microvascular tissue. We show that despite significant Mn accumulation in the CSF of Znt10 knockout mice, ZIP14 expression levels in the blood-cerebrospinal fluid barrier remain unchanged, indicating that ZIP14 does not have a compensatory mechanism for regulating Mn uptake in the brain in vivo. Additionally, Mn still enters the CSF without ZIP14 when systemic levels rise. This indicates that alternative transport mechanisms or compensatory pathways ensure Mn balance in the CSF, shedding light on potential strategies for managing Mn-related disorders.
Collapse
Affiliation(s)
| | - Ningning Zhao
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
10
|
Wang S, Gu J, Bian J, He Y, Xu X, Wang C, Li G, Zhang H, Ni B, Chen S, Shao Y, Jiang Y. Nesfatin-1 mitigates calcific aortic valve disease via suppressing ferroptosis mediated by GSH/GPX4 and ZIP8/SOD2 axes. Free Radic Biol Med 2024; 222:149-164. [PMID: 38851518 DOI: 10.1016/j.freeradbiomed.2024.06.004] [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] [Received: 01/29/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Calcific aortic valve disease (CAVD) predominantly affects the elderly and currently lacks effective medical treatments. Nesfatin-1, a peptide derived from the cleavage of Nucleobindin 2, has been implicated in various calcification processes, both physiological and pathological. This study explores the impact of Nesfatin-1 on the transformation of aortic valve interstitial cells (AVICs) in CAVD. METHODS AND RESULTS In vitro experiments showed that Nesfatin-1 treatment mitigated the osteogenic differentiation of AVICs. Corresponding in vivo studies demonstrated a deceleration in the progression of CAVD. RNA-sequencing of AVICs treated with and without Nesfatin-1 highlighted an enrichment of the Ferroptosis pathway among the top pathways identified by the Kyoto Encyclopedia of Genes and Genomes analysis. Further examination confirmed increased ferroptosis in both calcified valves and osteoblast-like AVICs, with a reduction in ferroptosis following Nesfatin-1 treatment. Within the Ferroptosis pathway, ZIP8 showed the most notable modulation by Nesfatin-1. Silencing ZIP8 in AVICs increased ferroptosis and osteogenic differentiation, decreased intracellular Mn2+ concentration, and reduced the expression and activity of superoxide dismutase (SOD2). Furthermore, the silencing of SOD2 exacerbated ferroptosis and osteogenic differentiation. Nesfatin-1 treatment was found to elevate the expression of glutathione peroxidase 4 (GPX4) and levels of glutathione (GSH), as confirmed by Western blotting and GSH concentration assays. CONCLUSION In summary, Nesfatin-1 effectively inhibits the osteogenic differentiation of AVICs by attenuating ferroptosis, primarily through the GSH/GPX4 and ZIP8/SOD2 pathways.
Collapse
Affiliation(s)
- Song Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Jiaxi Gu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Jinhui Bian
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Yuqiu He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Xiufan Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Chen Wang
- Department of Cardiovascular Surgery and Heart Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Geng Li
- Department of Cardiovascular Surgery and Heart Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Hui Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Buqing Ni
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China
| | - Si Chen
- Department of Cardiovascular Surgery and Heart Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China.
| | - Yefan Jiang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, China.
| |
Collapse
|
11
|
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
|
12
|
Fontes A, Jauch AT, Sailer J, Engler J, Azul AM, Zischka H. Metabolic Derangement of Essential Transition Metals and Potential Antioxidant Therapies. Int J Mol Sci 2024; 25:7880. [PMID: 39063122 PMCID: PMC11277342 DOI: 10.3390/ijms25147880] [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: 05/19/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Essential transition metals have key roles in oxygen transport, neurotransmitter synthesis, nucleic acid repair, cellular structure maintenance and stability, oxidative phosphorylation, and metabolism. The balance between metal deficiency and excess is typically ensured by several extracellular and intracellular mechanisms involved in uptake, distribution, and excretion. However, provoked by either intrinsic or extrinsic factors, excess iron, zinc, copper, or manganese can lead to cellular damage upon chronic or acute exposure, frequently attributed to oxidative stress. Intracellularly, mitochondria are the organelles that require the tightest control concerning reactive oxygen species production, which inevitably leaves them to be one of the most vulnerable targets of metal toxicity. Current therapies to counteract metal overload are focused on chelators, which often cause secondary effects decreasing patients' quality of life. New therapeutic options based on synthetic or natural antioxidants have proven positive effects against metal intoxication. In this review, we briefly address the cellular metabolism of transition metals, consequences of their overload, and current therapies, followed by their potential role in inducing oxidative stress and remedies thereof.
Collapse
Affiliation(s)
- Adriana Fontes
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Adrian T. Jauch
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Judith Sailer
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Jonas Engler
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| | - Anabela Marisa Azul
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, D-85764 Neuherberg, Germany;
- School of Medicine and Health, Institute of Toxicology and Environmental Hygiene, Technical University Munich, D-80802 Munich, Germany
| |
Collapse
|
13
|
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
|
14
|
Einhorn V, Haase H, Maares M. Interaction and competition for intestinal absorption by zinc, iron, copper, and manganese at the intestinal mucus layer. J Trace Elem Med Biol 2024; 84:127459. [PMID: 38640745 DOI: 10.1016/j.jtemb.2024.127459] [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] [Received: 02/09/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Trace elements such as zinc, manganese, copper, or iron are essential for a wide range of physiological functions. It is therefore crucial to ensure an adequate supply of these elements to the body. Many previous investigations have dealt with the role of transport proteins, in particular their selectivity for, and competition between, different ions. Another so far less well investigated major factor influencing the absorption of trace elements seems to be the intestinal mucus layer. This gel-like substance covers the entire gastrointestinal tract and its physiochemical properties can be mainly assigned to the glycoproteins it contains, so-called mucins. Interaction with mucins has already been demonstrated for some metals. However, knowledge about the impact on the respective bioavailability and competition between those metals is still sketchy. This review therefore aims to summarize the findings and knowledge gaps about potential effects regarding the interaction between gastrointestinal mucins and the trace elements iron, zinc, manganese, and copper. Mucins play an indispensable role in the absorption of these trace elements in the neutral to slightly alkaline environment of the intestine, by keeping them in a soluble form that can be absorbed by enterocytes. Furthermore, the studies so far indicate that the competition between these trace elements for uptake already starts at the intestinal mucus layer, yet further research is required to completely understand this interaction.
Collapse
Affiliation(s)
- Vincent Einhorn
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Hajo Haase
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Maria Maares
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany; Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal 14558, Germany.
| |
Collapse
|
15
|
Yuasa H, Morino N, Wagatsuma T, Munekane M, Ueda S, Matsunaga M, Uchida Y, Katayama T, Katoh T, Kambe T. ZNT5-6 and ZNT7 play an integral role in protein N-glycosylation by supplying Zn 2+ to Golgi α-mannosidase II. J Biol Chem 2024; 300:107378. [PMID: 38762179 PMCID: PMC11209640 DOI: 10.1016/j.jbc.2024.107378] [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: 02/19/2024] [Revised: 04/20/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024] Open
Abstract
The stepwise addition of monosaccharides to N-glycans attached to client proteins to generate a repertoire of mature proteins involves a concerted action of many glycosidases and glycosyltransferases. Here, we report that Golgi α-mannosidase II (GMII), a pivotal enzyme catalyzing the first step in the conversion of hybrid- to complex-type N-glycans, is activated by Zn2+ supplied by the early secretory compartment-resident ZNT5-ZNT6 heterodimers (ZNT5-6) and ZNT7 homodimers (ZNT7). Loss of ZNT5-6 and ZNT7 function results in marked accumulation of hybrid-type and complex/hybrid glycans with concomitant reduction of complex- and high-mannose-type glycans. In cells lacking the ZNT5-6 and ZNT7 functions, the GMII activity is substantially decreased. In contrast, the activity of its homolog, lysosomal mannosidase (LAMAN), is not decreased. Moreover, we show that the growth of pancreatic cancer MIA PaCa-2 cells lacking ZNT5-6 and ZNT7 is significantly decreased in a nude mouse xenograft model. Our results indicate the integral roles of ZNT5-6 and ZNT7 in N-glycosylation and highlight their potential as novel target proteins for cancer therapy.
Collapse
Affiliation(s)
- Hana Yuasa
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Naho Morino
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takumi Wagatsuma
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Sachiko Ueda
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Mayu Matsunaga
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yasuo Uchida
- Department of Molecular Systems Pharmaceutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima City, Japan
| | - Takane Katayama
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Toshihiko Katoh
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
16
|
Zahoor SM, Ishaq S, Ahmed T. Neurotoxic effects of metals on blood brain barrier impairment and possible therapeutic approaches. VITAMINS AND HORMONES 2024; 126:1-24. [PMID: 39029969 DOI: 10.1016/bs.vh.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Exposure to neurotoxic and heavy metals (Pb2+, As3+, Mn2+, Cd2+, etc) has increased over time and has shown to negatively affect brain health. Heavy metals can cross the blood brain barrier (BBB) in various ways including receptor or carrier-mediated transport, passive diffusion, or transport via gaps in the endothelial cells of the brain. In high concentrations, these metals have been shown to cause structural and functional impairment to the BBB, by inducing oxidative stress, ion dyshomeostasis, tight junction (TJ) loss, astrocyte/pericyte damage and interference of gap junctions. The structural and functional impairment of the BBB results in increased BBB permeability, which ultimately leads to accumulation of these heavy metals in the brain and their subsequent toxicity. As a result of these effects, heavy metals are correlated with various neurological disorders. The pathological effects of these heavy metals can be effectively mitigated via chelation. In addition, it is possible to treat the associated disorders by counteracting the molecular mechanisms associated with the brain and BBB impairment.
Collapse
Affiliation(s)
- Saba Mehak Zahoor
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sara Ishaq
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
17
|
Alba-González A, Dragomir EI, Haghdousti G, Yáñez J, Dadswell C, González-Méndez R, Wilson SW, Tuschl K, Folgueira M. Manganese Overexposure Alters Neurogranin Expression and Causes Behavioral Deficits in Larval Zebrafish. Int J Mol Sci 2024; 25:4933. [PMID: 38732149 PMCID: PMC11084468 DOI: 10.3390/ijms25094933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Manganese (Mn), a cofactor for various enzyme classes, is an essential trace metal for all organisms. However, overexposure to Mn causes neurotoxicity. Here, we evaluated the effects of exposure to Mn chloride (MnCl2) on viability, morphology, synapse function (based on neurogranin expression) and behavior of zebrafish larvae. MnCl2 exposure from 2.5 h post fertilization led to reduced survival (60%) at 5 days post fertilization. Phenotypical changes affected body length, eye and olfactory organ size, and visual background adaptation. This was accompanied by a decrease in both the fluorescence intensity of neurogranin immunostaining and expression levels of the neurogranin-encoding genes nrgna and nrgnb, suggesting the presence of synaptic alterations. Furthermore, overexposure to MnCl2 resulted in larvae exhibiting postural defects, reduction in motor activity and impaired preference for light environments. Following the removal of MnCl2 from the fish water, zebrafish larvae recovered their pigmentation pattern and normalized their locomotor behavior, indicating that some aspects of Mn neurotoxicity are reversible. In summary, our results demonstrate that Mn overexposure leads to pronounced morphological alterations, changes in neurogranin expression and behavioral impairments in zebrafish larvae.
Collapse
Affiliation(s)
- Anabel Alba-González
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Elena I. Dragomir
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Golsana Haghdousti
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Julián Yáñez
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Ramón González-Méndez
- School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK; (C.D.); (R.G.-M.)
| | - Stephen W. Wilson
- Department of Cell and Developmental, University College London, London, WC1E 6BT, UK; (E.I.D.); (G.H.); (S.W.W.)
| | - Karin Tuschl
- UCL GOSH Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Mónica Folgueira
- Department of Biology, Faculty of Sciences, University of A Coruña, 15008 A Coruña, Spain; (A.A.-G.); (J.Y.)
- Centro Interdisciplinar de Química y Biología, (CICA), University of A Coruña, 15071 A Coruña, Spain
| |
Collapse
|
18
|
Nishito Y, Kamimura Y, Nagamatsu S, Yamamoto N, Yasui H, Kambe T. Zinc and manganese homeostasis closely interact in mammalian cells. FASEB J 2024; 38:e23605. [PMID: 38597508 DOI: 10.1096/fj.202400181r] [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: 01/27/2024] [Revised: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Understanding the homeostatic interactions among essential trace metals is important for explaining their roles in cellular systems. Recent studies in vertebrates suggest that cellular Mn metabolism is related to Zn metabolism in multifarious cellular processes. However, the underlying mechanism remains unclear. In this study, we examined the changes in the expression of proteins involved in cellular Zn and/or Mn homeostatic control and measured the Mn as well as Zn contents and Zn enzyme activities to elucidate the effects of Mn and Zn homeostasis on each other. Mn treatment decreased the expression of the Zn homeostatic proteins metallothionein (MT) and ZNT1 and reduced Zn enzyme activities, which were attributed to the decreased Zn content. Moreover, loss of Mn efflux transport protein decreased MT and ZNT1 expression and Zn enzyme activity without changing extracellular Mn content. This reduction was not observed when supplementing with the same Cu concentrations and in cells lacking Cu efflux proteins. Furthermore, cellular Zn homeostasis was oppositely regulated in cells expressing Zn and Mn importer ZIP8, depending on whether Zn or Mn concentration was elevated in the extracellular milieu. Our results provide novel insights into the intricate interactions between Mn and Zn homeostasis in mammalian cells and facilitate our understanding of the physiopathology of Mn, which may lead to the development of treatment strategies for Mn-related diseases in the future.
Collapse
Affiliation(s)
- Yukina Nishito
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yoshiki Kamimura
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Shino Nagamatsu
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Nao Yamamoto
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiroyuki Yasui
- Department of Analytical and Bioinorganic Chemistry, Division of Analytical and Physical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Prajapati M, Quenneville CB, Zhang JZ, Chong GS, Chiu L, Ma B, Ward LD, Tu HC, Bartnikas TB. AAV-mediated hepatic expression of SLC30A10 and the Thr95Ile variant attenuates manganese excess and other phenotypes in Slc30a10-deficient mice. J Biol Chem 2024; 300:105732. [PMID: 38336290 PMCID: PMC10933546 DOI: 10.1016/j.jbc.2024.105732] [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: 08/10/2023] [Revised: 01/17/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.
Collapse
Affiliation(s)
- Milankumar Prajapati
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | | | - Jared Z Zhang
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Grace S Chong
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Lauren Chiu
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Bangyi Ma
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Lucas D Ward
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Ho-Chou Tu
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA.
| | - Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
20
|
Gu L, Zhao C, Wang Y, Wang C, Yin X, Ye Q, Liu Y, Zou X, Wang L, Zhuge Y, Wu J, Zhang F. Senescence of Hepatic Stellate Cells by Specific Delivery of Manganese for Limiting Liver Fibrosis. NANO LETTERS 2024; 24:1062-1073. [PMID: 38164915 PMCID: PMC10836362 DOI: 10.1021/acs.nanolett.3c03689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Senescence of activated hepatic stellate cells (HSCs) is crucial for the regression of liver fibrosis. However, impaired immune clearance can result in the accumulation of senescent HSCs, exacerbating liver fibrosis. The activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is essential for both senescence and the innate immune response. Additionally, the specific delivery to activated HSCs is hindered by their inaccessible anatomical location, capillarization of liver sinusoidal endothelial cells (LSECs), and loss of substance exchange. Herein, we propose an antifibrotic strategy that combines prosenescence with enhanced immune clearance through targeted delivery of manganese (a cGAS-STING stimulator) via albumin-mediated transcytosis, specifically aimed at inducing senescence and eliminating activated HSCs in liver fibrosis. Our findings demonstrate that only albumin efficiently transfers manganese to activated HSCs from LSECs via transcytosis compared to liposomes, resulting in significant antifibrotic effects in vivo while exhibiting negligible toxicity.
Collapse
Affiliation(s)
- Lihong Gu
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Wuxi
No. 2 People’s Hospital, Wuxi, Jiangsu 214002, People’s Republic of China
| | - Chenxuan Zhao
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Yixuan Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Chao Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Xiaochun Yin
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| | - Qingsong Ye
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Yan Liu
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| | - Xiaoping Zou
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| | - Lei Wang
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| | - Yuzheng Zhuge
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| | - Jinhui Wu
- State
Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine
Innovation Center, Medical School of Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
- Jiangsu
Key Laboratory for Nano Technology, Nanjing
University, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Feng Zhang
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, Jiangsu 210008, People’s
Republic of China
| |
Collapse
|
21
|
Ghosn ZA, Sparks KM, Spaulding JL, Vutukuri S, Ahmed MJJ, VanBerkum MFA. Divalent metal content in diet affects severity of manganese toxicity in Drosophila. Biol Open 2024; 13:bio060204. [PMID: 38117005 PMCID: PMC10810561 DOI: 10.1242/bio.060204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
Dysregulation of manganese (Mn) homeostasis is a contributing factor in many neuro-degenerative diseases. Adult Drosophila are sensitive to excessive levels of dietary Mn, dying relatively early, and exhibiting biochemical and mobility changes reminiscent of Parkinsonian conditions. To further study Mn homeostasis in Drosophila, we sought to test lower levels of dietary Mn (5 mM) and noted a striking difference in Canton-S adult survivorship on different food. On a cornmeal diet, Mn-treated flies live only about half as long as untreated siblings. Yet, with the same Mn concentration in a molasses diet, adults survive about 80% as long as untreated siblings, and adults raised on a sucrose-yeast diet are completely insensitive to this low dose of dietary Mn. By manipulating metal ion content in the cornmeal diet, and measuring the metal content in each diet, we traced the difference in lifespan to the levels of calcium and magnesium in the food, suggesting that these ions are involved in Mn uptake and/or use. Based on these findings, it is recommended that the total dietary load of metal ions be considered when assessing Mn toxicity.
Collapse
Affiliation(s)
- Zahraa A. Ghosn
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Kailynn M. Sparks
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Jacob L. Spaulding
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Sanjana Vutukuri
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Mirza J. J. Ahmed
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Mark F. A. VanBerkum
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
22
|
Zhang Z, Xiao Y, Long P, Yu Y, Liu Y, Liu K, Yang H, Li X, He M, Wu T, Yuan Y. Associations between plasma metal/metalloid mixtures and the risk of central obesity: A prospective cohort study of Chinese adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115838. [PMID: 38128312 DOI: 10.1016/j.ecoenv.2023.115838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Central obesity has increased rapidly over the past decade and posed a substantial disease burden worldwide. Exposure to metals/metalloids has been acknowledged to be involved in the development of central obesity through regulation of cortisol, insulin resistance, and glucocorticoid receptor reduction. Despite the importance, it is lack of prospective study which comprehensively evaluate the relations between multiple metals exposure and central obesity. We explored the prospective associations of plasma metal concentrations with central obesity in a prospective study of the Dongfeng-Tongji cohort. The present study included 2127 participants with a 6.87-year mean follow-up duration. We measured 23 plasma metal/metalloid concentrations at baseline. The associations between metals and incident central obesity were examined utilizing the Cox proportional hazard regression in single and multiple metals models. Additionally, we applied elastic net (ENET), Bayesian kernel machine regression (BKMR), plasma metal score (PMS), and quantile-based g-computation (Qgcomp) models to explore the joint associations of metal mixtures with central obesity. After adjusting potential confounders, we found significant associations of plasma manganese (Mn) and thallium (Tl) concentrations with a higher risk of central obesity, whereas plasma rubidium (Rb) concentration was associated with a lower risk of central obesity both in single and multiple metals models (all FDR <0.05). The ENET and Qqcomp models verified similar metals (Mn, Rb, and Tl) as important predictors for central obesity. The results of both BKMR model and PMS suggested cumulative exposure to metal mixtures was associated with a higher risk of central obesity. Our findings suggested that co-exposure to metals was associated with a higher risk of central obesity. This study expands our knowledge that the management of metals/metalloids exposure may be beneficial for the prevention of new-onset central obesity, which may subsequently alleviate the disease burden of late-life health outcomes.
Collapse
Affiliation(s)
- Zirui Zhang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Xiao
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinpin Long
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqiu Yu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyi Liu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Liu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Handong Yang
- Department of Cardiovascular Diseases, Dongfeng Central Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiulou Li
- Department of Cardiovascular Diseases, Dongfeng Central Hospital, Hubei University of Medicine, Shiyan, China
| | - Meian He
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tangchun Wu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yuan
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Smerchek DT, Rients EL, McLaughlin AM, Henderson JA, Ortner BM, Thornton KJ, Hansen SL. The influence of steroidal implants and manganese sulfate supplementation on growth performance, trace mineral status, hepatic gene expression, hepatic enzyme activity, and circulating metabolites in feedlot steers. J Anim Sci 2024; 102:skae062. [PMID: 38456567 PMCID: PMC10959487 DOI: 10.1093/jas/skae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/06/2024] [Indexed: 03/09/2024] Open
Abstract
Angus-cross steers (n = 144; 359 kg ± 13.4) were used to assess the effect of dietary Mn and steroidal implants on performance, trace minerals (TM) status, hepatic enzyme activity, hepatic gene expression, and serum metabolites. Steers (n = 6/pen) were stratified by BW in a 3 × 2 factorial. GrowSafe bunks recorded individual feed intake (experimental unit = steer; n = 24/treatment). Dietary treatments included (MANG; 8 pens/treatment; Mn as MnSO4): (1) no supplemental Mn (analyzed 14 mg Mn/kg DM; Mn0); (2) 20 mg supplemental Mn/kg DM (Mn20); (3) 50 mg supplemental Mn/kg DM (Mn50). Within MANG, steers received a steroidal implant treatment (IMP) on day 0: (1) no implant; NO; or (2) combination implant (Revalor-200; REV). Liver biopsies for TM analysis and qPCR, and blood for serum glucose, insulin, non-esterified fatty acids, and urea-N (SUN) analysis were collected on days 0, 20, 40, and 77. Data were analyzed as a randomized complete block with a factorial arrangement of treatments including fixed effects of Mn treatment (MANG) and implant (IMP) using PROC MIXED of SAS 9.4 using initial BW as a covariate. Liver TM, serum metabolite, enzyme activity, and gene expression data were analyzed as repeated measures. No MANG × IMP effects were noted (P ≥ 0.12) for growth performance or carcass characteristic measures. Dietary Mn did not influence final body weight, overall ADG, or overall G:F (P ≥ 0.14). Liver Mn concentration increased with supplemental Mn concentration (MANG; P = 0.01). An IMP × DAY effect was noted for liver Mn (P = 0.01) where NO and REV were similar on day 0 but NO cattle increased liver Mn from days 0 to 20 while REV liver Mn decreased. Relative expression of MnSOD in the liver was greater in REV (P = 0.02) compared to NO and within a MANG × IMP effect (P = 0.01) REV increased liver MnSOD activity. These data indicate current NASEM Mn recommendations are adequate to meet the demands of finishing beef cattle given a steroidal implant. Despite the roles of Mn in metabolic pathways and antioxidant defense, a basal diet containing 14 mg Mn/kg DM was sufficient for the normal growth of finishing steers. This study also provided novel insight into how implants and supplemental Mn influence genes related to arginine metabolism, urea synthesis, antioxidant capacity, and TM homeostasis as well as arginase and MnSOD activity in hepatic tissue of beef steers.
Collapse
Affiliation(s)
- Dathan T Smerchek
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Emma L Rients
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Amy M McLaughlin
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Jacob A Henderson
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Brock M Ortner
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Kara J Thornton
- Department of Animal, Dairy, and Veterinary Science, Utah State University, Logan, UT, 84322, USA
| | - Stephanie L Hansen
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
24
|
Zhang K, Qi C, Cai K. Manganese-Based Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205409. [PMID: 36121368 DOI: 10.1002/adma.202205409] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/26/2022] [Indexed: 05/12/2023]
Abstract
As an essential micronutrient, manganese (Mn) participates in various physiological processes and plays important roles in host immune system, hematopoiesis, endocrine function, and oxidative stress regulation. Mn-based nanoparticles are considered to be biocompatible and show versatile applications in nanomedicine, in particular utilized in tumor immunotherapy in the following ways: 1) acting as a biocompatible nanocarrier to deliver immunotherapeutic agents for tumor immunotherapy; 2) serving as an adjuvant to regulate tumor immune microenvironment and enhance immunotherapy; 3) activating host's immune system through the cGAS-STING pathway to trigger tumor immunotherapy; 4) real-time monitoring tumor immunotherapy effect by magnetic resonance imaging (MRI) since Mn2+ ions are ideal MRI contrast agent which can significantly enhance the T1 -weighted MRI signal after binding to proteins. This comprehensive review focuses on the most recent progress of Mn-based nanoplatforms in tumor immunotherapy. The characteristics of Mn are first discussed to guide the design of Mn-based multifunctional nanoplatforms. Then the biomedical applications of Mn-based nanoplatforms, including immunotherapy alone, immunotherapy-involved multimodal synergistic therapy, and imaging-guided immunotherapy are discussed in detail. Finally, the challenges and future developments of Mn-based tumor immunotherapy are highlighted.
Collapse
Affiliation(s)
- Ke Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chao Qi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
25
|
Santos-Díaz AI, Solís-López J, Díaz-Torres E, Guadarrama-Olmos JC, Osorio B, Kroll T, Webb SM, Hiriart M, Jiménez-Estrada I, Missirlis F. Metal ion content of internal organs in the calorically restricted Wistar rat. J Trace Elem Med Biol 2023; 78:127182. [PMID: 37130496 DOI: 10.1016/j.jtemb.2023.127182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/09/2023] [Accepted: 04/26/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Despite the agreed principle that access to food is a human right, undernourishment and metal ion deficiencies are public health problems worldwide, exacerbated in impoverished or war-affected areas. It is known that maternal malnutrition causes growth retardation and affects behavioral and cognitive development of the newborn. Here we ask whether severe caloric restriction leads per se to disrupted metal accumulation in different organs of the Wistar rat. METHODS Inductively coupled plasma optical emission spectroscopy was used to determine the concentration of multiple elements in the small and large intestine, heart, lung, liver, kidney, pancreas, spleen, brain, spinal cord, and three skeletal muscles from control and calorically restricted Wistar rats. The caloric restriction protocol was initiated from the mothers prior to mating and continued throughout gestation, lactation, and post-weaning up to sixty days of age. RESULTS Both sexes were analyzed but dimorphism was rare. The pancreas was the most affected organ presenting a higher concentration of all the elements analyzed. Copper concentration decreased in the kidney and increased in the liver. Each skeletal muscle responded to the treatment differentially: Extensor Digitorum Longus accumulated calcium and manganese, gastrocnemius decreased copper and manganese, whereas soleus decreased iron concentrations. Differences were also observed in the concentration of elements between organs independently of treatment: The soleus muscle presents a higher concentration of Zn compared to the other muscles and the rest of the organs. Notably, the spinal cord showed large accumulations of calcium and half the concentration of zinc compared to brain. X-ray fluorescence imaging suggests that the extra calcium is attributable to the presence of ossifications whereas the latter finding is attributable to the low abundance of zinc synapses in the spinal cord. CONCLUSION Severe caloric restriction did not lead to systemic metal deficiencies but caused instead specific metal responses in few organs.
Collapse
Affiliation(s)
- Alma I Santos-Díaz
- Department of Physiology, Biophysics and Neuroscience, Cinvestav, 07360 Mexico City, Mexico
| | | | - Elizabeth Díaz-Torres
- Department of Physiology, Biophysics and Neuroscience, Cinvestav, 07360 Mexico City, Mexico
| | | | - Beatriz Osorio
- Department of Physiology, Biophysics and Neuroscience, Cinvestav, 07360 Mexico City, Mexico
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA
| | - Samuel M Webb
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA
| | - Marcia Hiriart
- Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ismael Jiménez-Estrada
- Department of Physiology, Biophysics and Neuroscience, Cinvestav, 07360 Mexico City, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neuroscience, Cinvestav, 07360 Mexico City, Mexico.
| |
Collapse
|
26
|
Powers M, Minchella D, Gonzalez-Acevedo M, Escutia-Plaza D, Wu J, Heger C, Milne G, Aschner M, Liu Z. Loss of hepatic manganese transporter ZIP8 disrupts serum transferrin glycosylation and the glutamate-glutamine cycle. J Trace Elem Med Biol 2023; 78:127184. [PMID: 37163821 DOI: 10.1016/j.jtemb.2023.127184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/07/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND ZIP8, encoded by SLC39A8, is a membrane transporter that facilitates the cellular uptake of divalent biometals including zinc (Zn), manganese (Mn), and iron (Fe). The hepatic system has long been accepted as the central modulator for whole-body biometal distribution. Earlier investigations suggest the propensity of ZIP8 to prioritize Mn influx, as opposed to Fe or Zn, in hepatocytes. Hepatic ZIP8 Mn transport is crucial for maintaining homeostasis of various Mn-dependent metalloenzymes and their associated pathways. Herein, we hypothesize that a drastic decrease in systemic Mn, via the loss of hepatic ZIP8, disrupts two unique cellular pathways, post-translational glycosylation and the glutamate-glutamine cycle. METHODS ZIP8 liver-specific knockout (LSKO) mice were chosen in an attempt to substantially decrease whole-body Mn levels. To further elucidate the role of Mn in serum glycosylation, a Mn-deficient diet was adopted in conjunction with the LSKO mice to model a near-complete loss of systemic Mn. After the treatment course, transferrin sialylation profiles were determined using imaged capillary isoelectric focusing (icIEF). We also investigated the role of Mn in the glutamate-glutamine cycle; the conversion of glutamate to glutamine in F/F and LSKO mice was assessed by the glutamine/glutamate ratio in cerebrospinal fluid (CSF) via HPLC-MS. An open-field study was ultimately conducted to check if these mice displayed atypical behavior. RESULTS Two major biological pathways were found to be significantly altered due to the loss of hepatic ZIP8. We identified a disparity between F/F and LSKO transferrin sialylation profiles that were exacerbated under a Mn-deficient diet. Additionally, we discovered a neurotransmitter imbalance between the levels of glutamine and glutamate, exclusive to LSKO mice. This was characterized by the decreased glutamine/glutamate ratio in CSF. Secondary to the neurotransmitter alteration, LSKO mice exhibited an increase in locomotor activity in an open-field. CONCLUSION Our model successfully established a connection between the loss of hepatic ZIP8 and two Mn-dependent cellular pathways, namely, protein glycosylation and the glutamate-glutamine cycle.
Collapse
Affiliation(s)
- Michael Powers
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Dean Minchella
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | | | | | - Jiaqi Wu
- ProteinSimple, A Bio-Techne Brand, San Jose, CA, USA
| | - Chris Heger
- ProteinSimple, A Bio-Techne Brand, San Jose, CA, USA
| | - Ginger Milne
- Neurochemistry Core, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Michael Aschner
- Department of Cellular Biology and Pharmacology, Albert Einstein Medical College, New York, USA
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, USA.
| |
Collapse
|
27
|
Nakagawa Y, Yamada S. The Relationships Among Metal Homeostasis, Mitochondria, and Locus Coeruleus in Psychiatric and Neurodegenerative Disorders: Potential Pathogenetic Mechanism and Therapeutic Implications. Cell Mol Neurobiol 2023; 43:963-989. [PMID: 35635600 PMCID: PMC11414457 DOI: 10.1007/s10571-022-01234-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/15/2022] [Indexed: 11/03/2022]
Abstract
While alterations in the locus coeruleus-noradrenergic system are present during early stages of neuropsychiatric disorders, it is unclear what causes these changes and how they contribute to other pathologies in these conditions. Data suggest that the onset of major depressive disorder and schizophrenia is associated with metal dyshomeostasis that causes glial cell mitochondrial dysfunction and hyperactivation in the locus coeruleus. The effect of the overactive locus coeruleus on the hippocampus, amygdala, thalamus, and prefrontal cortex can be responsible for some of the psychiatric symptoms. Although locus coeruleus overactivation may diminish over time, neuroinflammation-induced alterations are presumably ongoing due to continued metal dyshomeostasis and mitochondrial dysfunction. In early Alzheimer's and Parkinson's diseases, metal dyshomeostasis and mitochondrial dysfunction likely induce locus coeruleus hyperactivation, pathological tau or α-synuclein formation, and neurodegeneration, while reduction of glymphatic and cerebrospinal fluid flow might be responsible for β-amyloid aggregation in the olfactory regions before the onset of dementia. It is possible that the overactive noradrenergic system stimulates the apoptosis signaling pathway and pathogenic protein formation, leading to further pathological changes which can occur in the presence or absence of locus coeruleus hypoactivation. Data are presented in this review indicating that although locus coeruleus hyperactivation is involved in pathological changes at prodromal and early stages of these neuropsychiatric disorders, metal dyshomeostasis and mitochondrial dysfunction are critical factors in maintaining ongoing neuropathology throughout the course of these conditions. The proposed mechanistic model includes multiple pharmacological sites that may be targeted for the treatment of neuropsychiatric disorders commonly.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
28
|
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
|
29
|
Taylor KM. The LIV-1 Subfamily of Zinc Transporters: From Origins to Present Day Discoveries. Int J Mol Sci 2023; 24:ijms24021255. [PMID: 36674777 PMCID: PMC9861476 DOI: 10.3390/ijms24021255] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
This review explains the origin of the LIV-1 family of zinc transporters, paying attention to how this family of nine human proteins was originally discovered. Structural and functional differences between these nine human LIV-1 family members and the five other ZIP transporters are examined. These differences are both related to aspects of the protein sequence, the conservation of important motifs and to the effect this may have on their overall function. The LIV-1 family are dependent on various post-translational modifications, such as phosphorylation and cleavage, which play an important role in their ability to transport zinc. These modifications and their implications are discussed in detail. Some of these proteins have been implicated in cancer which is examined. Furthermore, some additional areas of potential fruitful discovery are discussed and suggested as worthy of examination in the future.
Collapse
Affiliation(s)
- Kathryn M Taylor
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VIIth Avenue, Cardiff CF10 3NB, UK
| |
Collapse
|
30
|
McCabe S, Limesand K, Zhao N. Recent progress toward understanding the role of ZIP14 in regulating systemic manganese homeostasis. Comput Struct Biotechnol J 2023; 21:2332-2338. [PMID: 37020930 PMCID: PMC10070054 DOI: 10.1016/j.csbj.2023.03.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
ZIP14 is a metal transporter essential for the regulation of body manganese homeostasis. The physiological functions of ZIP14 have been uncovered mainly through two lines of in vivo studies that examined the phenotypes of ZIP14 loss, including studies of humans with ZIP14 mutations and animals with ZIP14 deficiency. This mini review aims at presenting an updated view of the important advances made towards understanding the genetic and pathological mechanisms of brain manganese overload caused by ZIP14 deficiency.
Collapse
|
31
|
Wu R, Yao F, Li X, Shi C, Zang X, Shu X, Liu H, Zhang W. Manganese Pollution and Its Remediation: A Review of Biological Removal and Promising Combination Strategies. Microorganisms 2022; 10:2411. [PMID: 36557664 PMCID: PMC9781601 DOI: 10.3390/microorganisms10122411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Manganese (Mn), as a cofactor of multiple enzymes, exhibits great significance to the human body, plants and animals. It is also a critical raw material and alloying element. However, extensive employment for industrial purposes leads to its excessive emission into the environment and turns into a significant threat to the ecosystem and public health. This review firstly introduces the essentiality, toxicity and regulation of Mn. Several traditional physicochemical methods and their problems are briefly discussed as well. Biological remediation, especially microorganism-mediated strategies, is a potential alternative for remediating Mn-polluted environments in a cost-efficient and eco-friendly manner. Among them, microbially induced carbonate precipitation (MICP), biosorption, bioaccumulation, bio-oxidation are discussed in detail, including their mechanisms, pivotal influencing factors along with strengths and limitations. In order to promote bioremediation efficiency, the combination of different techniques is preferable, and their research progress is also summarized. Finally, we propose the future directions of Mn bioremediation by microbes. Conclusively, this review provides a scientific basis for the microbial remediation performance for Mn pollution and guides the development of a comprehensive competent strategy towards practical Mn remediation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hengwei Liu
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wenchao Zhang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| |
Collapse
|
32
|
Zhou Y, Zhang J, Guan Q, Tao X, Wang J, Li W. The role of ferroptosis in the development of acute and chronic kidney diseases. J Cell Physiol 2022; 237:4412-4427. [PMID: 36260516 DOI: 10.1002/jcp.30901] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Ferroptosis, a novel form of regulated cell death, is characterized by imbalance of intracellular iron and redox systems, resulting from overgeneration of toxic lipid peroxidation products. In recent years, the verified crucial role of ferroptosis has been widely concerned in rudimentary pathogenesis and development of various acute and chronic kidney disease (CKD), comprehending the potential patterns of cell death can afford more reliable bases and principles for treatment and prevention of renal disease. In this review, the regulatory mechanisms of ferroptosis were introduced and the important roles of ferroptosis in diverse renal diseases such as acute kidney injury, CKD, and renal fibrosis were outlined to illuminate the potential of restraining ferroptosis in treatment and prevention of kidney disease.
Collapse
Affiliation(s)
- Yijun Zhou
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong Province, China
| | - Junlan Zhang
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong Province, China
| | - Qingyan Guan
- School of Nursing, Weifang Medical University, Weifang, Shandong Province, China
| | - Xun Tao
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong Province, China
| | - Jinling Wang
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong Province, China
| | - Wentong Li
- Department of Pathology, Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
33
|
Zhu H, Tan J, Wang Z, Wu Z, Zhou W, Zhang Z, Li M, Zhao Y. Bioinformatics analysis constructs potential ferroptosis-related ceRNA network involved in the formation of intracranial aneurysm. Front Cell Neurosci 2022; 16:1016682. [PMCID: PMC9612944 DOI: 10.3389/fncel.2022.1016682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIntracranial aneurysm (IA) causes more than 80% of nontraumatic subarachnoid hemorrhages (SAHs). The mechanism of ferroptosis involved in IA formation remains unclear. The roles played by competitive endogenous RNA (ceRNA) regulation networks in many diseases are becoming clearer. The goal of this study was to understand more fully the ferroptosis-related ceRNA regulation network in IA.Materials and methodsTo identify differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs), and differentially expressed lncRNAs (DELs) across IA and control samples, the GEO datasets GSE122897 and GSE66239 were downloaded and analyzed with the aid of R. Ferroptosis DEGs were discovered by exploring the DEGs of ferroptosis-related genes of the ferroptosis database. Potentially interacting miRNAs and lncRNAs were predicted using miRWalk and StarBase. Enrichment analysis was also performed. We utilized the STRING database and Cytoscape software to identify protein-protein interactions and networks. DAB-enhanced Prussian blue staining was used to detect iron in IA tissues.ResultsIron deposition was evident in IA tissue. In all, 30 ferroptosis DEGs, 5 key DEMs, and 17 key DELs were screened out for constructing a triple regulatory network. According to expression regulation of DELs, DEMs, and DEGs, a hub triple regulatory network was built. As the functions of lncRNAs are determined by their cellular location, PVT1-hsa-miR-4644-SLC39A14 ceRNA and DUXAP8-hsa-miR-378e/378f-SLC2A3 ceRNA networks were constructed.ConclusionCeRNA (PVT1-hsa-miR-4644-SLC39A14 and DUXAP8-hsa-miR-378e/378f-SLC2A3) overexpression networks associated with ferroptosis in IA were established.
Collapse
|
34
|
Lv WH, Zhao T, Pantopoulos K, Chen GH, Wei XL, Zhang DG, Luo Z. Manganese-Induced Oxidative Stress Contributes to Intestinal Lipid Deposition via the Deacetylation of PPARγ at K339 by SIRT1. Antioxid Redox Signal 2022; 37:417-436. [PMID: 35293223 DOI: 10.1089/ars.2021.0190] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aims: Excessive manganese (Mn) exposure is toxic, and induces lipid deposition, but the underlying mechanisms remain elusive. Herein, we explored how dietary Mn supplementation affects lipid deposition and metabolism in the intestine of vertebrates using the yellow catfish Pelteobagrus fulvidraco as the model. Results: High-Mn (H-Mn) diet increased intestinal Mn content, promoted lipid accumulation and lipogenesis, and inhibited lipolysis. In addition, it induced oxidative stress, upregulated metal-response element-binding transcription factor-1 (MTF-1), and peroxisome proliferator-activated receptor gamma (PPARγ) protein expression in the nucleus, induced PPARγ acetylation, and the interaction between PPARγ and retinoid X receptor alpha (RXRα), while it downregulated sirtuin 1 (SIRT1) expression and activity. Mechanistically, Mn activated the MTF-1/divalent metal transporter 1 (DMT1) pathway, increased Mn accumulation in the mitochondria, and induced oxidative stress. This in turn promoted lipid deposition via deacetylation of PPARγ at K339 by SIRT1. Subsequently, PPARγ mediated Mn-induced lipid accumulation through transcriptionally activating fatty acid translocase, stearoyl-CoA desaturase 1, and perilipin 2 promoters. Innovation: These studies uncover a previously unknown mechanism by which Mn induces lipid deposition in the intestine via the oxidative stress-SIRT1-PPARγ pathway. Conclusion: High dietary Mn intake activates MTF-1/DMT1 and oxidative stress pathways. Oxidative stress-mediated PPARγ deacetylation at K339 site contributes to increased lipid accumulation. Our results provided a direct link between Mn and lipid metabolism via the oxidative stress-SIRT1-PPARγ axis. Antioxid. Redox Signal. 37, 417-436.
Collapse
Affiliation(s)
- Wu-Hong Lv
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhao
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Canada
| | - Guang-Hui Chen
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Xiao-Lei Wei
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Dian-Guang Zhang
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Zhi Luo
- Laboratory of Molecular Nutrition for Aquatic Economic Animals, Fishery College, Huazhong Agricultural University, Wuhan, China.,Hubei Hongshan Laboratory, Wuhan, China
| |
Collapse
|
35
|
Wong WPS, Wang JC, Meyers MS, Wang NJ, Sponenburg RA, Allen NB, Edwards JR, El Muayed M. A novel chronic in vivo oral cadmium exposure-washout mouse model for studying cadmium toxicity and complex diabetogenic effects. Toxicol Appl Pharmacol 2022; 447:116057. [PMID: 35550884 PMCID: PMC9854171 DOI: 10.1016/j.taap.2022.116057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 01/25/2023]
Abstract
Type II diabetes mellitus (T2DM) is characterized by insulin resistance, β-cell dysfunction and hyperglycemia. In addition to well known risk factors such as lifestyle and genetic risk score, accumulation of environmental toxicants in organs relevant to glucose metabolism is increasingly recognized as additional risk factors for T2DM. Here, we describe the development of an in vivo oral cadmium (Cd) exposure model. It was shown that oral Cd exposure in drinking water followed by washout and high fat diet (HFD) in C57BL/6N mice results in islet Cd bioaccumulation comparable to that found in native human islets while mitigating the anorexic effects of Cd to achieve the same weight gain required to induce insulin resistance as in Cd naïve control mice. Inter individual variation in plasma glucose and insulin levels as well as islet Cd bioaccumulation was observed in both female and male mice. Regression analysis showed an inverse correlation between islet Cd level and plasma insulin following a glucose challenge in males but not in females. This finding highlights the need to account for inter individual target tissue Cd concentrations when interpreting results from in vivo Cd exposure models. No effect of Cd on insulin secretion was observed in islets ex vivo, highlighting differences between in vivo and ex vivo cadmium exposure models. In summary, our oral in vivo Cd exposure-washout with HFD model resulted in islet Cd bioaccumulation that is relevant in the context of environmental cadmium exposure in humans. Here, we showed that islet Cd bioaccumulation is associated with complex cadmium-mediated changes in glucose clearance and β-cell function. The model described here will serve as a useful tool to further examine the relationship between Cd exposure, islet Cd bioaccumulation, dysglycemia and their underlying mechanisms.
Collapse
Affiliation(s)
- Winifred P S Wong
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Janice C Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Matthew S Meyers
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nathan J Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rebecca A Sponenburg
- Quantitative Bio-element Imaging Centre, Chemistry of Life Processes, Northwestern University, Evanston, IL 60208, USA
| | - Norrina B Allen
- Institute for Public Health and Medicine, Center for Epidemiology and Population Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joshua R Edwards
- College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Malek El Muayed
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
36
|
Rozenberg JM, Kamynina M, Sorokin M, Zolotovskaia M, Koroleva E, Kremenchutckaya K, Gudkov A, Buzdin A, Borisov N. The Role of the Metabolism of Zinc and Manganese Ions in Human Cancerogenesis. Biomedicines 2022; 10:biomedicines10051072. [PMID: 35625809 PMCID: PMC9139143 DOI: 10.3390/biomedicines10051072] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022] Open
Abstract
Metal ion homeostasis is fundamental for life. Specifically, transition metals iron, manganese and zinc play a pivotal role in mitochondrial metabolism and energy generation, anti-oxidation defense, transcriptional regulation and the immune response. The misregulation of expression or mutations in ion carriers and the corresponding changes in Mn2+ and Zn2+ levels suggest that these ions play a pivotal role in cancer progression. Moreover, coordinated changes in Mn2+ and Zn2+ ion carriers have been detected, suggesting that particular mechanisms influenced by both ions might be required for the growth of cancer cells, metastasis and immune evasion. Here, we present a review of zinc and manganese pathophysiology suggesting that these ions might cooperatively regulate cancerogenesis. Zn and Mn effects converge on mitochondria-induced apoptosis, transcriptional regulation and the cGAS-STING signaling pathway, mediating the immune response. Both Zn and Mn influence cancer progression and impact treatment efficacy in animal models and clinical trials. We predict that novel strategies targeting the regulation of both Zn and Mn in cancer will complement current therapeutic strategies.
Collapse
Affiliation(s)
- Julian Markovich Rozenberg
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Correspondence:
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Maksim Sorokin
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Marianna Zolotovskaia
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- OmicsWay Corporation, Walnut, CA 91789, USA
| | - Elena Koroleva
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
| | - Kristina Kremenchutckaya
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
| | - Alexander Gudkov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
| | - Anton Buzdin
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.K.); (A.G.)
- OmicsWay Corporation, Walnut, CA 91789, USA
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Oncobox Ltd., 121205 Moscow, Russia
| | - Nicolas Borisov
- Moscow Institute of Physics and Technology, National Research University, 141700 Moscow, Russia; (M.S.); (M.Z.); (E.K.); (K.K.); (A.B.); (N.B.)
- OmicsWay Corporation, Walnut, CA 91789, USA
| |
Collapse
|
37
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
38
|
Wilcox JM, Consoli DC, Paffenroth KC, Spitznagel BD, Calipari ES, Bowman AB, Harrison FE. Manganese-induced hyperactivity and dopaminergic dysfunction depend on age, sex and YAC128 genotype. Pharmacol Biochem Behav 2022; 213:173337. [PMID: 35063467 PMCID: PMC8833139 DOI: 10.1016/j.pbb.2022.173337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
Manganese (Mn) is an essential micronutrient but is neurotoxic in excess. Environmental and genetic factors influence vulnerability to Mn toxicity, including sex, age, and the autosomal dominant mutation that causes Huntington disease (HD). To better understand the differential effects of Mn in wild-type (WT) versus YAC128 mice, we examined impacts of Mn exposure across different ages and sexes on disease-relevant behavioral tasks and dopamine dynamics. Young (3-week) and aged (12-month) WT and YAC128 mice received control (70 ppm) or high (2400 ppm) Mn diet for 8 weeks followed by a battery of behavioral tasks. In young female WT mice, high Mn diet induced hyperactivity across two independent behavioral tasks. Changes in the expression of tyrosine hydroxylase (TH) were consistent with the behavioral data in young females such that elevated TH in YAC128 on control diet was decreased by high Mn diet. Aged YAC128 mice showed the expected disease-relevant behavioral impairments in females and decreased TH expression, but we observed no significant effects of Mn diet in either genotype of the aged group. Fast-scan cyclic voltammetry recorded dopamine release and clearance in the nucleus accumbens of eight-month-old WT and YAC128 mice following acute Mn exposure (3×/1 week subcutaneous injections of 50 mg/kg MnCl[2]-tetrahydrate or saline). In WT mice, Mn exposure led to faster dopamine clearance that resembled saline treated YAC128 mice. Mn treatment increased dopamine release only in YAC128 mice, possibly indirectly correcting the faster dopamine clearance observed in saline treated YAC128 mice. The same exposure paradigm led to decreased dopamine and serotonin and metabolites (3-MT, HVA and 5-HIAA) in striatum and increased glutamate in YAC128 mice but not WT mice. These studies confirm an adverse effect of Mn in young, female WT animals and support a role for Mn exposure in stabilizing dopaminergic dysfunction and motivated behavior in early HD.
Collapse
Affiliation(s)
- Jordyn M. Wilcox
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN,corresponding author: Jordyn M. Wilcox, PhD, , 2215 Garland Ave, Medical Research Building IV, 7445, Nashville, TN 37232
| | - David C. Consoli
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | | | - Brittany D. Spitznagel
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Erin S. Calipari
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN,Departments of Pharmacology, Molecular Physiology and Biophysics, Psychiatry and Behavioral Sciences; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN
| | - Fiona E. Harrison
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| |
Collapse
|
39
|
Zhang M, Zhu L, Wang H, Hao Y, Zhang Q, Zhao C, Bao X. A novel homozygous SLC39A14 variant in an infant with hypermanganesemia and a review of the literature. Front Pediatr 2022; 10:949651. [PMID: 36733764 PMCID: PMC9886663 DOI: 10.3389/fped.2022.949651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Manganese (Mn) is an essential trace metal necessary for good health; however, excessive amounts in the body are neurotoxic. To date, three genes (SLC30A10, SLC39A8, and SLC39A14) have been discovered to cause inborn errors in Mn metabolism in humans. As very rare diseases, the clinical features require further clarification. METHODS A male Chinese patient who mainly presented with hypermanganesemia and progressive parkinsonism-dystonia was recruited for this study. We collected and analyzed clinical information, performed whole-exome sequencing (WES), and reviewed the relevant literature. RESULTS The motor-developmental milestones of the patient were delayed at the age of 4 months, followed by rapidly progressive dystonia. The patient displayed elevated Mn concentrations in blood and urine, and brain magnetic resonance imaging (MRI) showed symmetrical hyperintensity on T1-weighted images and hypointensity on T2-weighted images in multiple regions. A novel homozygous variant of the SLC39A14 gene (c.1058T > G, p.L353R) was identified. The patient was treated with disodium calcium edetate chelation (Na2CaEDTA). Three months later, mild improvement in clinical manifestation, blood Mn levels, and brain MRI was observed. To date, 15 patients from 10 families have been reported with homozygous mutations of SLC39A14, with a mean age of onset of 14.9 months. The common initial symptom is motor regression or developmental milestone delay, with a disease course for nearly all patients involving development of progressive generalized dystonia and loss of ambulation before treatment. Additionally, hypermanganesemia manifests as Mn values ranging from 4- to 25-fold higher than normal baseline levels, along with brain MRI results similar to those observed in the recruited patient. Nine SLC39A14 variants have been identified. Seven patients have been treated with Na2CaEDTA, and only one patient achieved obvious clinical improvement. CONCLUSION We identified a novel SLC39A14 mutation related to autosomal recessive hypermanganesemia with dystonia-2, which is a very rare disease. Patients present motor regression or delay of developmental milestones and develop progressive generalized dystonia. Chelation therapy with Na2CaEDTA appears to effectively chelate Mn and increase urinary Mn excretion in some cases; however, clinical response varies. The outcome of the disease was unsatisfactory. This study expands the genetic spectrum of this disease.
Collapse
Affiliation(s)
- Meijiao Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Liping Zhu
- Department of Pediatrics, Linyi People's Hospital, Linyi, China
| | - Huiping Wang
- Department of Neurology, Children's Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Ying Hao
- Department of Pediatrics, Yuhuangding Hospital, Yantai, China
| | - Qingping Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chunyan Zhao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xinhua Bao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
40
|
Fujishiro H, Kambe T. Manganese transport in mammals by zinc transporter family proteins, ZNT and ZIP. J Pharmacol Sci 2021; 148:125-133. [PMID: 34924116 DOI: 10.1016/j.jphs.2021.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Manganese (Mn) is an essential trace element required for various biological processes. However, excess Mn causes serious side effects in humans, including parkinsonism. Thus, elucidation of Mn homeostasis at the systemic, cellular, and molecular levels is important. Many metal transporters and channels can be involved in the transport and homeostasis of Mn, and an increasing body of evidence shows that several zinc (Zn) transporters belonging to the ZIP and ZNT families, specifically, ZNT10, ZIP8, and ZIP14, play pivotal roles in Mn metabolism. Mutations in the genes encoding these transporter proteins are associated with congenital disorders related to dysregulated Mn homeostasis in humans. Moreover, single nucleotide polymorphisms of ZIP8 are associated with multiple clinical phenotypes. In this review, we discuss the recent literature on the structural and biochemical features of ZNT10, ZIP8, and ZIP14, including transport mechanisms, regulation of expression, and pathophysiological functions. Because a disturbance in Mn homeostasis is closely associated with a variety of phenotypes and risk of human diseases, these transporters constitute a significant target for drug development. An understanding of the roles of these key transporters in Mn metabolism should provide new insights into pharmacological applications of their inhibitors and enhancers in human diseases.
Collapse
Affiliation(s)
- Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
41
|
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: 7] [Impact Index Per Article: 1.8] [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
|
42
|
Liu X, Rong P. Recent Advances of Manganese-Based Hybrid Nanomaterials for Cancer Precision Medicine. Front Oncol 2021; 11:707618. [PMID: 34722253 PMCID: PMC8548572 DOI: 10.3389/fonc.2021.707618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022] Open
Abstract
Cancer precision medicine (CPM) could tailor the best treatment for individual cancer patients, while imaging techniques play important roles in its application. With the characteristics of noninvasion, nonionized, radiation-free, multidimensional imaging function, and real-time monitoring, magnetic resonance imaging (MRI) is an effective way for early tumor detection, and it has become a tower of strength in CPM imaging techniques. Due to linkage with nephrogenic systemic fibrosis (NSF), gadolinium (Gd)-based contrast agent (CA), which was long used in MRI, has been restricted by the Food and Drug Administration (FDA). In this review, we would like to introduce the manganese (Mn)-based CAs that could significantly increase the safety of MRI CAs by realizing more superior performance and functions simultaneously in the diagnosis and treatment of tumors. Also, recent advances in Mn-based hybrid nanomaterials for CPM are summarized and discussed.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China.,Postdoctoral Research Station of Clinical Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengfei Rong
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
Reinert JP, Forbes LD. Manganese Toxicity Associated With Total Parenteral Nutrition: A Review. J Pharm Technol 2021; 37:260-266. [PMID: 34753157 PMCID: PMC8404746 DOI: 10.1177/87551225211023686] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To review hypermanganesemia-induced toxicities in adult patients receiving parenteral nutrition (PN) therapy. Data Sources: A comprehensive literature review was conducted from June 2020 to May 2021 on PubMED, MEDLINE, Scopus, ProQuest, the Cumulative Index of Nursing and Allied Health Literature (CINAHL), and Web of Science. Study Selection and Data Extraction: Keyword and Boolean phrase searches were conducted using the following terminology: "manganese" OR "manganesemia" OR "manganism" or "hypermanganesemia" AND "total parenteral nutrition" OR "PN" or "parenteral nutrition" AND "toxicity" OR "accumulation." Appropriate filters, including "humans" and "English" and NOT "reviews," were utilized on all databases to improve search outcomes. Data Synthesis: A total of 4 reports detailing hypermanganesemia in 57 patient encounters were included in this review. Significant heterogeneity exists with regard to the duration of manganese supplementation and the dose of manganese. Toxicity associated with manganese was observed in as few as 15 days. The dose of manganese, though likely governed by content in commercially available products, may regularly exceed the recommendations of clinical guidelines and should be limited to 55 µg/day. Select patients with underlying malignancy, those with significant and prolonged Vitamin D deficiency, or those who have acquired a SLC30A10 genetic mutation may be at an increased risk of developing manganese toxicity. Conclusions: Clinicians must be cognizant of the concentration of trace elements added to PN, as manganese, and perhaps other biometals, may accumulate when dosed above the recommended daily allowances.
Collapse
Affiliation(s)
- Justin P. Reinert
- Bon Secours Mercy Health St. Vincent Medical Center, Toledo, OH, USA
- The University of Texas at Tyler, TX, USA
| | - Laramie D. Forbes
- Bon Secours Mercy Health St. Vincent Medical Center, Toledo, OH, USA
| |
Collapse
|
44
|
Manganese systemic distribution is modulated in vivo during tumor progression and affects tumor cell migration and invasion in vitro. Sci Rep 2021; 11:15833. [PMID: 34349175 PMCID: PMC8338990 DOI: 10.1038/s41598-021-95190-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/22/2021] [Indexed: 11/23/2022] Open
Abstract
Metastatic disease remains the leading cause of death in cancer and understanding the mechanisms involved in tumor progression continues to be challenging. This work investigates the role of manganese in tumor progression in an in vivo model of tumor growth. Our data revealed that manganese accumulates within primary tumors and secondary organs as manganese-rich niches. Consequences of such phenomenon were investigated, and we verified that short-term changes in manganese alter cell surface molecules syndecan-1 and β1-integrin, enhance collective cell migration and invasive behavior. Long-term increased levels of manganese do not affect cell growth and viability but enhance cell migration. We also observed that manganese is secreted from tumor cells in extracellular vesicles, rather than in soluble form. Finally, we describe exogenous glycosaminoglycans that counteract manganese effects on tumor cell behavior. In conclusion, our analyses describe manganese as a central element in tumor progression by accumulating in Mn-rich niches in vivo, as well as in vitro, affecting migration and extracellular vesicle secretion in vitro. Manganese accumulation in specific regions of the organism may not be a common ground for all cancers, nevertheless, it represents a new aspect of tumor progression that deserves special attention.
Collapse
|
45
|
Prajapati M, Conboy HL, Hojyo S, Fukada T, Budnik B, Bartnikas TB. Biliary excretion of excess iron in mice requires hepatocyte iron import by Slc39a14. J Biol Chem 2021; 297:100835. [PMID: 34051234 PMCID: PMC8214222 DOI: 10.1016/j.jbc.2021.100835] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022] Open
Abstract
Iron is essential for erythropoiesis and other biological processes, but is toxic in excess. Dietary absorption of iron is a highly regulated process and is a major determinant of body iron levels. Iron excretion, however, is considered a passive, unregulated process, and the underlying pathways are unknown. Here we investigated the role of metal transporters SLC39A14 and SLC30A10 in biliary iron excretion. While SLC39A14 imports manganese into the liver and other organs under physiological conditions, it imports iron under conditions of iron excess. SLC30A10 exports manganese from hepatocytes into the bile. We hypothesized that biliary excretion of excess iron would be impaired by SLC39A14 and SLC30A10 deficiency. We therefore analyzed biliary iron excretion in Slc39a14-and Slc30a10-deficient mice raised on iron-sufficient and -rich diets. Bile was collected surgically from the mice, then analyzed with nonheme iron assays, mass spectrometry, ELISAs, and an electrophoretic assay for iron-loaded ferritin. Our results support a model in which biliary excretion of excess iron requires iron import into hepatocytes by SLC39A14, followed by iron export into the bile predominantly as ferritin, with iron export occurring independently of SLC30A10. To our knowledge, this is the first report of a molecular determinant of mammalian iron excretion and can serve as basis for future investigations into mechanisms of iron excretion and relevance to iron homeostasis.
Collapse
Affiliation(s)
- Milankumar Prajapati
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Heather L Conboy
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Shintaro Hojyo
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Toshiyuki Fukada
- Department of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Faculty of Arts and Sciences, Division of Science, Harvard University, Cambridge, Massachusetts, USA
| | - Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
46
|
Wu Y, Wei G, Zhao N. Restriction of Manganese Intake Prevents the Onset of Brain Manganese Overload in Zip14-/- Mice. Int J Mol Sci 2021; 22:ijms22136773. [PMID: 34202493 PMCID: PMC8268934 DOI: 10.3390/ijms22136773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023] Open
Abstract
As a newly identified manganese transport protein, ZIP14 is highly expressed in the small intestine and liver, which are the two principal organs involved in regulating systemic manganese homeostasis. Loss of ZIP14 function leads to manganese overload in both humans and mice. Excess manganese in the body primarily affects the central nervous system, resulting in irreversible neurological disorders. Therefore, to prevent the onset of brain manganese accumulation becomes critical. In this study, we used Zip14−/− mice as a model for ZIP14 deficiency and discovered that these mice were born without manganese loading in the brain, but started to hyper-accumulate manganese within 3 weeks after birth. We demonstrated that decreasing manganese intake in Zip14−/− mice was effective in preventing manganese overload that typically occurs in these animals. Our results provide important insight into future studies that are targeted to reduce the onset of manganese accumulation associated with ZIP14 dysfunction in humans.
Collapse
|
47
|
The Potential Roles of Blood-Brain Barrier and Blood-Cerebrospinal Fluid Barrier in Maintaining Brain Manganese Homeostasis. Nutrients 2021; 13:nu13061833. [PMID: 34072120 PMCID: PMC8227615 DOI: 10.3390/nu13061833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
Manganese (Mn) is a trace nutrient necessary for life but becomes neurotoxic at high concentrations in the brain. The brain is a “privileged” organ that is separated from systemic blood circulation mainly by two barriers. Endothelial cells within the brain form tight junctions and act as the blood–brain barrier (BBB), which physically separates circulating blood from the brain parenchyma. Between the blood and the cerebrospinal fluid (CSF) is the choroid plexus (CP), which is a tissue that acts as the blood–CSF barrier (BCB). Pharmaceuticals, proteins, and metals in the systemic circulation are unable to reach the brain and spinal cord unless transported through either of the two brain barriers. The BBB and the BCB consist of tightly connected cells that fulfill the critical role of neuroprotection and control the exchange of materials between the brain environment and blood circulation. Many recent publications provide insights into Mn transport in vivo or in cell models. In this review, we will focus on the current research regarding Mn metabolism in the brain and discuss the potential roles of the BBB and BCB in maintaining brain Mn homeostasis.
Collapse
|
48
|
Generation of a Polyclonal Antibody against the Mouse Metal Transporter ZIP8. Antibodies (Basel) 2021; 10:antib10020016. [PMID: 33919173 PMCID: PMC8167614 DOI: 10.3390/antib10020016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 11/25/2022] Open
Abstract
ZIP8 is a newly identified metal transporter. In human patients, mutations in ZIP8 result in severe manganese deficiency, suggesting a critical role for ZIP8 in regulating systemic manganese homeostasis. In mice, the deletion of ZIP8 recapitulates the symptoms of patients with ZIP8 mutations. However, further studies using mouse models to examine ZIP8′s function were hindered by the lack of suitable antibodies to detect endogenous ZIP8 protein. In this study, we report the design, generation, and validation of a polyclonal antibody against mouse ZIP8. We have demonstrated that the newly generated antibody can be reliably used in immunoblotting analysis to detect endogenous ZIP8 protein in mouse tissues. The successful generation and validation of anti-mouse ZIP8 antibody provide opportunities to further examine the function and regulation of this metal transporter. In addition, our study may provide valuable insights into the future development of antibodies targeting polytopic membrane proteins.
Collapse
|
49
|
Zhang L, Xiao S, Kang X, Sun T, Zhou C, Xu Z, Du M, Zhang Y, Wang G, Liu Y, Zhang D, Gong M. Metabolic Conversion and Removal of Manganese Ferrite Nanoparticles in RAW264.7 Cells and Induced Alteration of Metal Transporter Gene Expression. Int J Nanomedicine 2021; 16:1709-1724. [PMID: 33688187 PMCID: PMC7936572 DOI: 10.2147/ijn.s289707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/10/2021] [Indexed: 12/26/2022] Open
Abstract
Background Manganese Ferrite Nanoparticles (Mn-IONPs) are widely used in biomedical field and their cytotoxicity has been initially explored, but the mechanism remains obscure. The nano-bio interactions are believed to be crucial for cytotoxicity mechanism, while little data have been acquired. Methods Mn-IONPs were synthesized by thermal decomposition of acetylacetonate precursor. After physicochemical characterization, we analyzed the metabolic conversion and removal of Mn-IONPs in RAW264.7 cells by Prussian blue staining, TEM, HRTEM and elemental quantitative analysis, followed by gene expression evaluation using quantitative RT-PCR. Results Mn-IONPs were successfully synthesized. Both the uptake and cytotoxicity of Mn-IONPs on RAW264.7 cells were time- and dose-dependent. After internalized, Mn-IONPs were passed to daughter cells with passages on. Meanwhile, Mn-IONPs were exocytosed and digested to metal ions and further excreted out, resulted in the labeling rate and ions contents decreased gradually. As ion influx related genes, the expressions of ZIP14, IRP2, FtH and DMT1 were suppressed within 24 hours but overexpressed to a plateau at the 48th hour in a dose-dependent manner. At the 72nd hour, ZIP14 and DMT1 mRNA levels decreased toward normal, while IRP2 and FtH kept up-regulated. As efflux related genes, FPN, SLC30A10 and Hamp2 genes were up-regulated within 24–72 hours; SPCA1 was suppressed at the 24th and 72nd hour, while overexpressed at the 48th hour. All the efflux related genes’ mRNA had a dose-dependent increasing manner at the corresponding time points. Conclusion Mn-IONPs showed time- and dose-dependent cytotoxicity and cell labeling rate in RAW264.7 cells. Accompanying with the intracellular catabolic breakdown and exocytosis of Mn-IONPs, RAW264.7 cells also secreted and re-uptook manganese and iron ions to maintain intracellular homeostasis in the succeeding passages. And the metabolic conversion of Mn-IONPs in RAW264.7 cells can affect the expression of ZIP14, DMT1, FPN, SLC30A10, IRP2, FtH, Hamp2 and SPCA1 genes.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Shilin Xiao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xun Kang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zhongsheng Xu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Mengmeng Du
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ya Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Guangxian Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yun Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|