1
|
Walraven T, Busch M, Wang J, Donkers JM, Duijvestein M, van de Steeg E, Kramer NI, Bouwmeester H. Elevated risk of adverse effects from foodborne contaminants and drugs in inflammatory bowel disease: a review. Arch Toxicol 2024; 98:3519-3541. [PMID: 39249550 PMCID: PMC11489187 DOI: 10.1007/s00204-024-03844-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
The global burden of Inflammatory bowel disease (IBD) has been rising over the last decades. IBD is an intestinal disorder with a complex and largely unknown etiology. The disease is characterized by a chronically inflamed gastrointestinal tract, with intermittent phases of exacerbation and remission. This compromised intestinal barrier can contribute to, enhance, or even enable the toxicity of drugs, food-borne chemicals and particulate matter. This review discusses whether the rising prevalence of IBD in our society warrants the consideration of IBD patients as a specific population group in toxicological safety assessment. Various in vivo, ex vivo and in vitro models are discussed that can simulate hallmarks of IBD and may be used to study the effects of prevalent intestinal inflammation on the hazards of these various toxicants. In conclusion, risk assessments based on healthy individuals may not sufficiently cover IBD patient safety and it is suggested to consider this susceptible subgroup of the population in future toxicological assessments.
Collapse
Affiliation(s)
- Tom Walraven
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| | - Mathias Busch
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Joanne M Donkers
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Marjolijn Duijvestein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Nynke I Kramer
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
2
|
Choi EK, Aring L, Peng Y, Correia AB, Lieberman AP, Iwase S, Seo YA. Neuronal SLC39A8 deficiency impairs cerebellar development by altering manganese homeostasis. JCI Insight 2024; 9:e168440. [PMID: 39435657 PMCID: PMC11530126 DOI: 10.1172/jci.insight.168440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 09/04/2024] [Indexed: 10/23/2024] Open
Abstract
Solute carrier family 39, member 8 (SLC39A8), is a transmembrane transporter that mediates the cellular uptake of zinc, iron, and manganese (Mn). Human genetic studies document the involvement of SLC39A8 in Mn homeostasis, brain development, and function. However, the role and pathophysiological mechanisms of SLC39A8 in the central nervous system remain elusive. We generated Slc39a8 neuron-specific knockout (Slc39a8-NSKO) mice to study SLC39A8 function in neurons. The Slc39a8-NSKO mice displayed markedly decreased Mn levels in the whole brain and brain regions, especially the cerebellum. Radiotracer studies using 54Mn revealed that Slc39a8-NSKO mice had impaired brain uptake of Mn. Slc39a8-NSKO cerebellums exhibited morphological defects and abnormal dendritic arborization of Purkinje cells. Reduced neurogenesis and increased apoptotic cell death occurred in the cerebellar external granular layer of Slc39a8-NSKO mice. Brain Mn deficiency in Slc39a8-NSKO mice was associated with motor dysfunction. Unbiased RNA-Seq analysis revealed downregulation of key pathways relevant to neurodevelopment and synaptic plasticity, including cAMP signaling pathway genes. We further demonstrated that Slc39a8 was required for the optimal transcriptional response to the cAMP-mediated signaling pathway. In summary, our study highlighted the essential roles of SLC39A8 in brain Mn uptake and cerebellum development and functions.
Collapse
Affiliation(s)
- Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Yujie Peng
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | | | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Peng Z, Liao Y, Yang W, Liu L. Metal(loid)-gut microbiota interactions and microbiota-related protective strategies: A review. ENVIRONMENT INTERNATIONAL 2024; 192:109017. [PMID: 39317009 DOI: 10.1016/j.envint.2024.109017] [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: 07/03/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Human exposure to metal(loid)s has dramatically increased over the past five decades, which has triggered public concern worldwide. Recently, gut microbiota has been considered a target for metal(loid)s, and some literature has reviewed the interactions between gut microbiota and heavy metal(loid)s (HMs) with high toxicity. However, whether there is an interaction between gut microbiota and metal(loid)s with essential roles or some normal functions are far from clear to date. Importantly, in addition to traditional probiotics that have been clarified to alleviate the adverse effect of HMs on the body, some novel probiotics, prebiotics, synbiotics, and postbiotics may also exhibit comparable or even better abilities of metal(loid) remediation. In this review, we mainly outline and discuss recent research findings on the metal(loid)-gut microbiota interactions and microbiota-related protective strategies.
Collapse
Affiliation(s)
- Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
| |
Collapse
|
4
|
Briggs K, Tomar V, Ollberding N, Haberman Y, Bourgonje AR, Hu S, Chaaban L, Sunuwar L, Weersma RK, Denson LA, Melia JMP. Crohn's Disease-Associated Pathogenic Mutation in the Manganese Transporter ZIP8 Shifts the Ileal and Rectal Mucosal Microbiota Implicating Aberrant Bile Acid Metabolism. Inflamm Bowel Dis 2024; 30:1379-1388. [PMID: 38289995 PMCID: PMC11291615 DOI: 10.1093/ibd/izae003] [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: 09/27/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND A pathogenic mutation in the manganese transporter ZIP8 (A391T; rs13107325) increases the risk of Crohn's disease. ZIP8 regulates manganese homeostasis and given the shared need for metals between the host and resident microbes, there has been significant interest in alterations of the microbiome in carriers of ZIP8 A391T. Prior studies have not examined the ileal microbiome despite associations between ileal disease and ZIP8 A391T. METHODS Here, we used the Pediatric Risk Stratification Study (RISK) cohort to perform a secondary analysis of 16S ribosomal RNA gene sequencing data obtained from ileal and rectal mucosa to study associations between ZIP8 A391T carrier status and microbiota composition. RESULTS We found sequence variants mapping to Veillonella were decreased in the ileal mucosa of ZIP8 A391T carriers. Prior human studies have demonstrated the sensitivity of Veillonella to bile acid abundance. We therefore hypothesized that bile acid homeostasis is differentially regulated in carriers of ZIP8 A391T. Using a mouse model of ZIP8 A391T, we demonstrate an increase in total bile acids in the liver and stool and decreased fibroblast growth factor 15 (Fgf15) signaling, consistent with our hypothesis. We confirmed dysregulation of FGF19 in the 1000IBD cohort, finding that plasma FGF19 levels are lower in ZIP8 A391T carriers with ileocolonic Crohn's disease. CONCLUSIONS In the search for genotype-specific therapeutic paradigms for patients with Crohn's disease, these data suggest targeting the FGF19 pathway in ZIP8 A391T carriers. Aberrant bile acid metabolism may precede development of Crohn's disease and prioritize study of the interactions between manganese homeostasis, bile acid metabolism and signaling, and complicated ileal Crohn's disease.
Collapse
Affiliation(s)
- Kristi Briggs
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vartika Tomar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas Ollberding
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yael Haberman
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Sheba Medical Center, Tel-Hashomer, affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lara Chaaban
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laxmi Sunuwar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lee A Denson
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joanna M P Melia
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
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
|
6
|
Choi EK, Rajendiran TM, Soni T, Park JH, Aring L, Muraleedharan CK, Garcia-Hernandez V, Kamada N, Samuelson LC, Nusrat A, Iwase S, Seo YA. The manganese transporter SLC39A8 links alkaline ceramidase 1 to inflammatory bowel disease. Nat Commun 2024; 15:4775. [PMID: 38839750 PMCID: PMC11153611 DOI: 10.1038/s41467-024-49049-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/17/2024] [Indexed: 06/07/2024] Open
Abstract
The metal ion transporter SLC39A8 is associated with physiological traits and diseases, including blood manganese (Mn) levels and inflammatory bowel diseases (IBD). The mechanisms by which SLC39A8 controls Mn homeostasis and epithelial integrity remain elusive. Here, we generate Slc39a8 intestinal epithelial cell-specific-knockout (Slc39a8-IEC KO) mice, which display markedly decreased Mn levels in blood and most organs. Radiotracer studies reveal impaired intestinal absorption of dietary Mn in Slc39a8-IEC KO mice. SLC39A8 is localized to the apical membrane and mediates 54Mn uptake in intestinal organoid monolayer cultures. Unbiased transcriptomic analysis identifies alkaline ceramidase 1 (ACER1), a key enzyme in sphingolipid metabolism, as a potential therapeutic target for SLC39A8-associated IBDs. Importantly, treatment with an ACER1 inhibitor attenuates colitis in Slc39a8-IEC KO mice by remedying barrier dysfunction. Our results highlight the essential roles of SLC39A8 in intestinal Mn absorption and epithelial integrity and offer a therapeutic target for IBD associated with impaired Mn homeostasis.
Collapse
Affiliation(s)
- Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Thekkelnaycke M Rajendiran
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jin-Ho Park
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | | | | | - Nobuhiko Kamada
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Linda C Samuelson
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Song C, Wu J, Wu J, Wang F. MnO 2 and roflumilast-loaded probiotic membrane vesicles mitigate experimental colitis by synergistically augmenting cAMP in macrophage. J Nanobiotechnology 2024; 22:294. [PMID: 38807127 PMCID: PMC11131305 DOI: 10.1186/s12951-024-02558-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is one chronic and relapsing inflammatory bowel disease. Macrophage has been reputed as one trigger for UC. Recently, phosphodiesterase 4 (PDE4) inhibitors, for instance roflumilast, have been regarded as one latent approach to modulating macrophage in UC treatment. Roflumilast can decelerate cyclic adenosine monophosphate (cAMP) degradation, which impedes TNF-α synthesis in macrophage. However, roflumilast is devoid of macrophage-target and consequently causes some unavoidable adverse reactions, which restrict the utilization in UC. RESULTS Membrane vesicles (MVs) from probiotic Escherichia coli Nissle 1917 (EcN 1917) served as a drug delivery platform for targeting macrophage. As model drugs, roflumilast and MnO2 were encapsulated in MVs (Rof&MnO2@MVs). Roflumilast inhibited cAMP degradation via PDE4 deactivation and MnO2 boosted cAMP generation by activating adenylate cyclase (AC). Compared with roflumilast, co-delivery of roflumilast and MnO2 apparently produced more cAMP and less TNF-α in macrophage. Besides, Rof&MnO2@MVs could ameliorate colitis in mouse model and regulate gut microbe such as mitigating pathogenic Escherichia-Shigella and elevating probiotic Akkermansia. CONCLUSIONS A probiotic-based nanoparticle was prepared for precise codelivery of roflumilast and MnO2 into macrophage. This biomimetic nanoparticle could synergistically modulate cAMP in macrophage and ameliorate experimental colitis.
Collapse
Affiliation(s)
- Chengjun Song
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jiamin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Institution of Drug R&D, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Fangyu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China.
- Department of Gastroenterology and Hepatology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
8
|
Lin Z, Luo W, Zhang K, Dai S. Environmental and Microbial Factors in Inflammatory Bowel Disease Model Establishment: A Review Partly through Mendelian Randomization. Gut Liver 2024; 18:370-390. [PMID: 37814898 PMCID: PMC11096900 DOI: 10.5009/gnl230179] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 10/11/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex condition resulting from environmental, microbial, immunologic, and genetic factors. With the advancement of Mendelian randomization research in IBD, we have gained new insights into the relationship between these factors and IBD. Many animal models of IBD have been developed using different methods, but few studies have attempted to model IBD by combining environmental factors and microbial factors. In this review, we examine how environmental factors and microbial factors affect the development and progression of IBD, and how they interact with each other and with the intestinal microbiota. We also summarize the current methods for creating animal models of IBD and compare their advantages and disadvantages. Based on the latest findings from Mendelian randomization studies on the role of environmental factors in IBD, we discuss which environmental and microbial factors could be used to construct a more realistic and reliable IBD experimental model. We propose that animal models of IBD should consider both environmental and microbial factors to better mimic human IBD pathogenesis and to reveal the underlying mechanisms of IBD at the immune and genetic levels. We highlight the importance of environmental and microbial factors in IBD pathogenesis and offer new perspectives and suggestions for improving experimental animal modeling. Our goal is to create a model that closely resembles the clinical picture of IBD.
Collapse
Affiliation(s)
- Zesheng Lin
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Wenjing Luo
- The Second Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Kaijun Zhang
- Department of Gastroenterology, Guangdong Provincial Geriatrics Institute, Guangzhou, ChinaNational Key Clinical Specialty, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shixue Dai
- Department of Gastroenterology, Guangdong Provincial Geriatrics Institute, Guangzhou, ChinaNational Key Clinical Specialty, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Geriatric Center, National Regional Medical Center, Ganzhou Hospital Affiliated to Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Ganzhou, China
| |
Collapse
|
9
|
Braun T, Feng R, Amir A, Levhar N, Shacham H, Mao R, Hadar R, Toren I, Algavi Y, Abu-Saad K, Zhuo S, Efroni G, Malik A, Picard O, Yavzori M, Agranovich B, Liu TC, Stappenbeck TS, Denson L, Kalter-Leibovici O, Gottlieb E, Borenstein E, Elinav E, Chen M, Ben-Horin S, Haberman Y. Diet-omics in the Study of Urban and Rural Crohn disease Evolution (SOURCE) cohort. Nat Commun 2024; 15:3764. [PMID: 38704361 PMCID: PMC11069498 DOI: 10.1038/s41467-024-48106-6] [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: 10/11/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Crohn disease (CD) burden has increased with globalization/urbanization, and the rapid rise is attributed to environmental changes rather than genetic drift. The Study Of Urban and Rural CD Evolution (SOURCE, n = 380) has considered diet-omics domains simultaneously to detect complex interactions and identify potential beneficial and pathogenic factors linked with rural-urban transition and CD. We characterize exposures, diet, ileal transcriptomics, metabolomics, and microbiome in newly diagnosed CD patients and controls in rural and urban China and Israel. We show that time spent by rural residents in urban environments is linked with changes in gut microbial composition and metabolomics, which mirror those seen in CD. Ileal transcriptomics highlights personal metabolic and immune gene expression modules, that are directly linked to potential protective dietary exposures (coffee, manganese, vitamin D), fecal metabolites, and the microbiome. Bacteria-associated metabolites are primarily linked with host immune modules, whereas diet-linked metabolites are associated with host epithelial metabolic functions.
Collapse
Affiliation(s)
- Tzipi Braun
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Rui Feng
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, Nanning, Guangxi, China
| | - Amnon Amir
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Nina Levhar
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hila Shacham
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ren Mao
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rotem Hadar
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Itamar Toren
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yadid Algavi
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Kathleen Abu-Saad
- Gertner Institute for Epidemiology and Health Policy Research, Tel Hashomer, Israel
| | - Shuoyu Zhuo
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Gilat Efroni
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Alona Malik
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Orit Picard
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Miri Yavzori
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
| | - Bella Agranovich
- Laura and Isaac Perlmutter Metabolomics Center, Technion-Israel Institute of Technology, Bat Galim, Haifa, Israel
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University in St Louis School of Medicine, St. Louis, MO, USA
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lee Denson
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ofra Kalter-Leibovici
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Gertner Institute for Epidemiology and Health Policy Research, Tel Hashomer, Israel
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Bat Galim, Haifa, Israel
| | - Elhanan Borenstein
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Santa Fe Institute, Santa Fe, NM, USA
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome & Cancer Division, German National Cancer Center (DKFZ), Heidelberg, Germany
| | - Minhu Chen
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shomron Ben-Horin
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yael Haberman
- Sheba Medical Center, Tel-Hashomer, Affiliated with the Tel Aviv University, Tel Aviv, Israel.
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Huang W, Das NK, Radyk MD, Keeley T, Quiros M, Jain C, El-Derany MO, Swaminathan T, Dziechciarz S, Greenson JK, Nusrat A, Samuelson LC, Shah YM. Dietary Iron Is Necessary to Support Proliferative Regeneration after Intestinal Injury. J Nutr 2024; 154:1153-1164. [PMID: 38246358 PMCID: PMC11181351 DOI: 10.1016/j.tjnut.2024.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Tissue repair and regeneration in the gastrointestinal system are crucial for maintaining homeostasis, with the process relying on intricate cellular interactions and affected by micro- and macro-nutrients. Iron, essential for various biological functions, plays a dual role in tissue healing by potentially causing oxidative damage and participating in anti-inflammatory mechanisms, underscoring its complex relationship with inflammation and tissue repair. OBJECTIVE The study aimed to elucidate the role of low dietary iron in gastrointestinal tissue repair. METHODS We utilized quantitative iron measurements to assess iron levels in inflamed regions of patients with ulcerative colitis and Crohn's disease. In addition, 3 mouse models of gastrointestinal injury/repair (dextran sulfate sodium-induced colitis, radiation injury, and wound biopsy) were used to assess the effects of low dietary iron on tissue repair. RESULTS We found that levels of iron in inflamed regions of both patients with ulcerative colitis and Crohn's disease are elevated. Similarly, during gastrointestinal repair, iron levels were found to be heightened, specifically in intestinal epithelial cells across the 3 injury/repair models. Mice on a low-iron diet showed compromised tissue repair with reduced proliferation. In standard diet, epithelial cells and the stem cell compartment maintain adequate iron stores. However, during a period of iron deficiency, epithelial cells exhaust their iron reserves, whereas the stem cell compartments maintain their iron pools. During injury, when the stem compartment is disrupted, low iron levels impair proliferation and compromise repair mechanisms. CONCLUSIONS Low dietary iron impairs intestinal repair through compromising the ability of epithelial cells to aid in intestinal proliferation.
Collapse
Affiliation(s)
- Wesley Huang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, United States; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, United States
| | - Nupur K Das
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Megan D Radyk
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Theresa Keeley
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Miguel Quiros
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Chesta Jain
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Marwa O El-Derany
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Thaarini Swaminathan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Sofia Dziechciarz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Joel K Greenson
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
11
|
Apalowo OE, Adegoye GA, Mbogori T, Kandiah J, Obuotor TM. Nutritional Characteristics, Health Impact, and Applications of Kefir. Foods 2024; 13:1026. [PMID: 38611332 PMCID: PMC11011999 DOI: 10.3390/foods13071026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
A global epidemiological shift has been observed in recent decades, characterized by an increase in age-related disorders, notably non-communicable chronic diseases, such as type 2 diabetes mellitus, cardiovascular and neurodegenerative diseases, and cancer. An appreciable causal link between changes in the gut microbiota and the onset of these maladies has been recognized, offering an avenue for effective management. Kefir, a probiotic-enriched fermented food, has gained significance in this setting due to its promising resource for the development of functional or value-added food formulations and its ability to reshape gut microbial composition. This has led to increasing commercial interest worldwide as it presents a natural beverage replete with health-promoting microbes and several bioactive compounds. Given the substantial role of the gut microbiota in human health and the etiology of several diseases, we conducted a comprehensive synthesis covering a total of 33 investigations involving experimental animal models, aimed to elucidate the regulatory influence of bioactive compounds present in kefir on gut microbiota and their potential in promoting optimal health. This review underscores the outstanding nutritional properties of kefir as a central repository of bioactive compounds encompassing micronutrients and amino acids and delineates their regulatory effects at deficient, adequate, and supra-nutritional intakes on the gut microbiota and their broader physiological consequences. Furthermore, an investigation of putative mechanisms that govern the regulatory effects of kefir on the gut microbiota and its connections with various human diseases was discussed, along with potential applications in the food industry.
Collapse
Affiliation(s)
- Oladayo Emmanuel Apalowo
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA; (O.E.A.); (G.A.A.)
| | - Grace Adeola Adegoye
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA; (O.E.A.); (G.A.A.)
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | - Teresia Mbogori
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | - Jayanthi Kandiah
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | | |
Collapse
|
12
|
Liu Y, Li S, Huang Z, Dai H, Shi F, Lv Z. Dietary collagen peptide-chelated trace elements supplementation for breeder hens improves the intestinal health of chick offspring. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:174-183. [PMID: 37612258 DOI: 10.1002/jsfa.12938] [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: 02/02/2023] [Revised: 08/01/2023] [Accepted: 08/24/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Dietary supplementation with trace elements zinc (Zn), iron (Fe) and manganese (Mn) could promote intestinal development and improve intestinal health. There are, however, few studies examining the possibility that maternal original Zn, Fe and Mn could regulate intestinal development and barrier function in the offspring. This study aimed to investigate how the intestinal growth and barrier function of breeder offspring were affected by collagen peptide-chelated trace elements (PTE; Zn, Fe, Mn). RESULTS PTE supplementation in the diet of breeder hens increased the concentrations of Zn, Fe and Mn in egg yolk. Maternal PTE supplementation improved morphological parameters of the intestine (villi height, crypt depth and villi height/crypt depth) and upregulated the mRNA expression level of leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) in the ileum of chick embryos. Furthermore, maternal PTE effect improved villi height/crypt depth of offspring at 1 and 14 days of age, and upregulated Lgr5, Claudin-3 and E-cadherin mRNA expression in the broiler ileum. Additionally, PTE treatment could enhance the intestinal microbial diversity of offspring. Maternal PTE supplementation increased the relative abundance of Clostridiales at the genus level and decreased the relative abundance of Enterococcus in newborn offspring. Moreover, maternal PTE supplementation ameliorated the elevated nuclear factor kappa B, toll-like receptor 4 and interleukin 1β mRNA expression in the ileum of offspring caused by LPS challenge. CONCLUSION Maternal PTE supplementation could promote intestinal development and enhance the intestinal barrier function of chicken offspring. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yongfa Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Simeng Li
- Aksu Vocational and Technical College, Aksu, China
| | - Zhenwu Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hongjian Dai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Xue X, Liu Z, Liang Y, Kwon YY, Liu R, Martin D, Hui S. Glutathione peroxidase 4 suppresses manganese-dependent oxidative stress to reduce colorectal tumorigenesis. RESEARCH SQUARE 2024:rs.3.rs-3837925. [PMID: 38260380 PMCID: PMC10802749 DOI: 10.21203/rs.3.rs-3837925/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The role of glutathione peroxidase 4 (GPX4) in ferroptosis and various cancers is well-established; however, its specific contribution to colorectal cancer has been unclear. Surprisingly, in a genetic mouse model of colon tumors, the deletion of GPX4 specifically in colon epithelial cells increased tumor burden but decreased oxidized glutathione. Notably, this specific GPX4 deletion did not enhance susceptibility to dextran sodium sulfate (DSS)-induced colitis in mice with varied iron diets but showed vulnerability in mice with a vitamin E-deficient diet. Additionally, a high manganese diet heightened susceptibility, while a low manganese diet reduced DSS-induced colitis in colon epithelial-specific GPX4-deficient mice. Strikingly, the low manganese diet also significantly reduced colorectal cancer formation in both colon epithelial-specific GPX4-deficient and wildtype mice. Mechanistically, antioxidant proteins, especially manganese-dependent superoxide dismutase (MnSOD or SOD2), correlated with disease severity. Treatment with tempol, a superoxide dismutase mimetic radical scavenger, suppressed GPX4 deficiency-induced colorectal tumors. In conclusion, the study elucidates the critical role of GPX4 in inhibiting colorectal cancer progression by regulating oxidative stress in a manganese-dependent manner. The findings underscore the intricate interactions between GPX4, dietary factors, and their collective influence on colorectal cancer development, providing potential insights for personalized therapeutic strategies.
Collapse
|
14
|
Shi JH, Chen YX, Feng Y, Yang X, Lin J, Wang T, Wei CC, Ma XH, Yang R, Cao D, Zhang H, Xie X, Xie Z, Zhang WJ. Fructose overconsumption impairs hepatic manganese homeostasis and ammonia disposal. Nat Commun 2023; 14:7934. [PMID: 38040719 PMCID: PMC10692208 DOI: 10.1038/s41467-023-43609-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Arginase, a manganese (Mn)-dependent enzyme, is indispensable for urea generation and ammonia disposal in the liver. The potential role of fructose in Mn and ammonia metabolism is undefined. Here we demonstrate that fructose overconsumption impairs hepatic Mn homeostasis and ammonia disposal in male mice. Fructose overexposure reduces liver Mn content as well as its activity of arginase and Mn-SOD, and impairs the clearance of blood ammonia under liver dysfunction. Mechanistically, fructose activates the Mn exporter Slc30a10 gene transcription in the liver in a ChREBP-dependent manner. Hepatic overexpression of Slc30a10 can mimic the effect of fructose on liver Mn content and ammonia disposal. Hepatocyte-specific deletion of Slc30a10 or ChREBP increases liver Mn contents and arginase activity, and abolishes their responsiveness to fructose. Collectively, our data establish a role of fructose in hepatic Mn and ammonia metabolism through ChREBP/Slc30a10 pathway, and postulate fructose dietary restriction for the prevention and treatment of hyperammonemia.
Collapse
Affiliation(s)
- Jian-Hui Shi
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Yu-Xia Chen
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Yingying Feng
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaohang Yang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jie Lin
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Ting Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Chun-Chun Wei
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Xian-Hua Ma
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Rui Yang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Dongmei Cao
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Hai Zhang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Zhifang Xie
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Weiping J Zhang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China.
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Mitchell SB, Thorn TL, Lee MT, Kim Y, Comrie JMC, Bai ZS, Johnson EL, Aydemir TB. Metal transporter SLC39A14/ZIP14 modulates regulation between the gut microbiome and host metabolism. Am J Physiol Gastrointest Liver Physiol 2023; 325:G593-G607. [PMID: 37873588 PMCID: PMC10887856 DOI: 10.1152/ajpgi.00091.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Metal transporter SLC39A14/ZIP14 is localized on the basolateral side of the intestine, functioning to transport metals from blood to intestine epithelial cells. Deletion of Slc39a14/Zip14 causes spontaneous intestinal permeability with low-grade chronic inflammation, mild hyperinsulinemia, and greater body fat with insulin resistance in adipose. Importantly, antibiotic treatment reverses the adipocyte phenotype of Slc39a14/Zip14 knockout (KO), suggesting a potential gut microbial role in the metabolic alterations in the Slc39a14/Zip14 KO mice. Here, we investigated the hypothesis that increased intestinal permeability and subsequent metabolic alterations in the absence of Zip14 could be in part due to alterations in gut microbial composition. Dietary metals have been shown to be involved in the regulation of gut microbial diversity and composition. However, studies linking the action of intestinal metal transporters to gut microbial regulation are lacking. We showed the influence of deletion of metal transporter Slc39a14/Zip14 on gut microbiome composition and how ZIP14-linked changes to gut microbiome community composition are correlated with changes in host metabolism. Deletion of Slc39a14/Zip14 generated Zn-deficient epithelial cells and luminal content in the entire intestinal tract, a shift in gut microbial composition that partially overlapped with changes previously associated with obesity and inflammatory bowel disease (IBD), increased the fungi/bacteria ratio in the gut microbiome, altered the host metabolome, and shifted host energy metabolism toward glucose utilization. Collectively, our data suggest a potential predisease microbial susceptibility state dependent on host gene Slc39a14/Zip14 that contributes to intestinal permeability, a common trait of IBD, and metabolic disorders such as obesity and type 2 diabetes.NEW & NOTEWORTHY Metal dyshomeostasis, intestinal permeability, and gut dysbiosis are emerging signatures of chronic disorders, including inflammatory bowel diseases, type-2 diabetes, and obesity. Studies in reciprocal regulations between host intestinal metal transporters genes and gut microbiome are scarce. Our research revealed a potential predisease microbial susceptibility state dependent on the host metal transporter gene, Slc39a14/Zip14, that contributes to intestinal permeability providing new insight into understanding host metal transporter gene-microbiome interactions in developing chronic disease.
Collapse
Affiliation(s)
- Samuel B Mitchell
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Trista L Thorn
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Min-Ting Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Yongeun Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Janine M C Comrie
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Zi Shang Bai
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Elizabeth L Johnson
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Tolunay B Aydemir
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| |
Collapse
|
16
|
Song WX, Yu ZH, Ren XF, Chen JH, Chen X. Role of micronutrients in inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2023; 31:711-731. [DOI: 10.11569/wcjd.v31.i17.711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an autoimmune intestinal disease that includes ulcerative colitis, Crohn's disease, and indeterminate colitis. Patients with IBD are often at risk for malnutrition, including micronutrient deficiencies, due to dietary restrictions and poor intestinal absorption. Micronutrients, including vitamins and minerals, play an important role in the human body's metabolism and maintenance of tissue functions. This article reviews the role of micronutrients in IBD. Micronutrients can affect the occurrence and progression of IBD by regulating immunity, intestinal flora, oxidative stress, intestinal barrier function, and other aspects. Monitoring and timely supplementation of micronutrients are important to delay progression and improve clinical symptoms in IBD patients.
Collapse
Affiliation(s)
- Wen-Xuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zi-Han Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiang-Feng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ji-Hua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
17
|
Ma Y, Fei Y, Ding S, Jiang H, Fang J, Liu G. Trace metal elements: a bridge between host and intestinal microorganisms. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1976-1993. [PMID: 37528296 DOI: 10.1007/s11427-022-2359-4] [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: 02/15/2023] [Accepted: 04/23/2023] [Indexed: 08/03/2023]
Abstract
Trace metal elements, such as iron, copper, manganese, and zinc, are essential nutrients for biological processes. Although their intake demand is low, they play a crucial role in cell homeostasis as the cofactors of various enzymes. Symbiotic intestinal microorganisms compete with their host for the use of trace metal elements. Moreover, the metabolic processes of trace metal elements in the host and microorganisms affect the organism's health. Supplementation or the lack of trace metal elements in the host can change the intestinal microbial community structure and function. Functional changes in symbiotic microorganisms can affect the host's metabolism of trace metal elements. In this review, we discuss the absorption and transport processes of trace metal elements in the host and symbiotic microorganisms and the effects of dynamic changes in the levels of trace metal elements on the intestinal microbial community structure. We also highlight the participation of trace metal elements as enzyme cofactors in the host immune process. Our findings indicate that the host uses metal nutrition immunity or metal poisoning to resist pathogens and improve immunity.
Collapse
Affiliation(s)
- Yong Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Yanquan Fei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Hongmei Jiang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China.
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| |
Collapse
|
18
|
Makevic V, Milovanovich ID, Popovac N, Janković S, Janković V, Stefanović S, Bukumiric Z, de Luka SR. Oligoelements in serum and intestinal tissue of pediatric IBD patients. J Trace Elem Med Biol 2023; 79:127239. [PMID: 37302217 DOI: 10.1016/j.jtemb.2023.127239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) develops through complex interplay of genetic, microbial, immune, and environmental factors. Trace elements alterations are commonly present in IBD and may have influence on IBD development. Heavy metal pollution is one of the major environmental issues nowadays and IBD incidence is rising in countries where industry starts to develop. Metals are implicated in processes that are connected to IBD pathogenesis. AIM The aim of this study was to investigate toxic and trace element levels in pediatric population of IBD patients both in serum and intestinal mucosa. MATERIALS AND METHODS This prospective study enrolled children newly diagnosed with IBD in University children's hospital in Belgrade. Concentrations of thirteen elements: Al, As, Ca, Cd, Cr, Cu, Fe, K, Mg, Mn, Na, Se and Zn in serum and intestinal mucosa of 17 newly diagnosed children with IBD (10 Crohn's disease (CD) and 7ulcerative colitis (UC)) and 10 controls were assessed using inductively coupled plasma mass spectrometry (ICP-MS). Intestinal mucosa samples were taken from terminal ileum and six different colon segments (cecum, ascending colon, colon transversum, descending and sigmoid colon and rectum). RESULTS The results demonstrated significant alterations in serum and intestinal mucosa concentrations of investigated elements. Serum iron was significantly decreased in IBD and CD group, compared to controls while serum Cu significantly differed between three investigated groups with highest concentration observed in CD children. Serum manganese was the highest in the UC subgroup. Terminal ileums of IBD patients contained significantly lower amount of Cu, Mg, Mn and Zn with Mn being significantly decreased also in CD patients compared to control. IBD patients' caecum contained significantly less Mg and Cu while colon transversum tissue samples from IBD and Crohn's patients contained significantly more chromium than controls. Moreover, sigmoid colon of IBD patients were poorer in Mg than controls (p < 0.05). Colon Al, As and Cd were significantly reduced in IBD, and UC children compared to control. Correlations of investigated elements in CD and UC groups were different from controls. Biochemical and clinical parameters showed correlation with element concentrations in intestines. CONCLUSION Sera of CD, UC and control children significantly differ in Fe, Cu and Mn levels. Serum manganese was the highest in the UC subgroup creating the most prominent and only significant difference between UC and CD subgroups. Terminal ileum of IBD patients contained significantly lower amount of majority of investigated essential trace elements and toxic elements were significantly reduced in colon of IBD and UC patients. Investigation of macro- and microelement alterations in children and adults has potential to further elucidate IBD pathogenesis.
Collapse
Affiliation(s)
- Vedrana Makevic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotica1, 11000 Belgrade, Serbia.
| | | | - Nevena Popovac
- University Children's Hospital, Tiršova 10, 11000 Belgrade, Serbia.
| | - Saša Janković
- Institute of Meat Hygiene and Technology, Kaćanskog 13, 11000 Belgrade, Serbia.
| | - Vesna Janković
- Institute of Meat Hygiene and Technology, Kaćanskog 13, 11000 Belgrade, Serbia.
| | - Srdjan Stefanović
- Institute of Meat Hygiene and Technology, Kaćanskog 13, 11000 Belgrade, Serbia.
| | - Zoran Bukumiric
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Dr Subotica 15, 11000 Belgrade, Serbia.
| | - Silvio R de Luka
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotica1, 11000 Belgrade, Serbia.
| |
Collapse
|
19
|
Šik Novak K, Bogataj Jontez N, Petelin A, Hladnik M, Baruca Arbeiter A, Bandelj D, Pražnikar J, Kenig S, Mohorko N, Jenko Pražnikar Z. Could Gut Microbiota Composition Be a Useful Indicator of a Long-Term Dietary Pattern? Nutrients 2023; 15:2196. [PMID: 37432336 DOI: 10.3390/nu15092196] [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: 03/23/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 07/12/2023] Open
Abstract
Despite the known effects of diet on gut microbiota composition, not many studies have evaluated the relationship between distinct dietary patterns and gut microbiota. The aim of our study was to determine whether gut microbiota composition could be a useful indicator of a long-term dietary pattern. We collected data from 89 subjects adhering to omnivorous, vegetarian, vegan, and low-carbohydrate, high-fat diet that were equally distributed between groups and homogenous by age, gender, and BMI. Gut microbiota composition was analyzed with a metabarcoding approach using V4 hypervariable region of the 16S rRNA gene. K-means clustering of gut microbiota at the genus level was performed and the nearest neighbor classifier was applied to predict microbiota clustering classes. Our results suggest that gut microbiota composition at the genus level is not a useful indicator of a subject's dietary pattern, with the exception of a vegan diet that is represented by a high abundance of Prevotella 9. Based on our model, a combination of 26 variables (anthropometric measurements, serum biomarkers, lifestyle factors, gastrointestinal symptoms, psychological factors, specific nutrients intake) is more important to predict an individual's microbiota composition cluster, with 91% accuracy, than the dietary intake alone. Our findings could serve to develop strategies to educate individuals about changes of some modifiable lifestyle factors, aiming to classify them into clusters with favorable health markers, independent of their dietary pattern.
Collapse
Affiliation(s)
- Karin Šik Novak
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Nives Bogataj Jontez
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Ana Petelin
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Matjaž Hladnik
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, 6000 Koper, Slovenia
| | - Alenka Baruca Arbeiter
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, 6000 Koper, Slovenia
| | - Dunja Bandelj
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, 6000 Koper, Slovenia
| | - Jure Pražnikar
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, 6000 Koper, Slovenia
| | - Saša Kenig
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Nina Mohorko
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Zala Jenko Pražnikar
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| |
Collapse
|
20
|
Wu Y, Liu C, Dong W. Adjunctive therapeutic effects of micronutrient supplementation in inflammatory bowel disease. Front Immunol 2023; 14:1143123. [PMID: 37077923 PMCID: PMC10106602 DOI: 10.3389/fimmu.2023.1143123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Growing evidence suggests that micronutrient status may have some impact on the course of inflammatory bowel disease (IBD). However, micronutrient deficiencies are easily overlooked during the treatment of IBD patients. There have been many studies on micronutrient supplementation, in which several clinical trials have been conducted targeting vitamin D and iron, but the current research is still preliminary for other vitamins and minerals. This review provides an overview of the adjunctive therapeutic effects of micronutrient supplementation in IBD, to summarize the available evidence, draw the attention of clinicians to micronutrient monitoring and supplementation in patients with IBD, and also provide some perspectives for future research directions.
Collapse
|
21
|
Huynh U, Zastrow ML. Metallobiology of Lactobacillaceae in the gut microbiome. J Inorg Biochem 2023; 238:112023. [PMID: 36270041 PMCID: PMC9888405 DOI: 10.1016/j.jinorgbio.2022.112023] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022]
Abstract
Lactobacillaceae are a diverse family of lactic acid bacteria found in the gut microbiota of humans and many animals. These bacteria exhibit beneficial effects on intestinal health, including modulating the immune system and providing protection against pathogens, and many species are frequently used as probiotics. Gut bacteria acquire essential metal ions, like iron, zinc, and manganese, through the host diet and changes to the levels of these metals are often linked to alterations in microbial community composition, susceptibility to infection, and gastrointestinal diseases. Lactobacillaceae are frequently among the organisms increased or decreased in abundance due to changes in metal availability, yet many of the molecular mechanisms underlying these changes have yet to be defined. Metal requirements and metallotransporters have been studied in some species of Lactobacillaceae, but few of the mechanisms used by these bacteria to respond to metal limitation or excess have been investigated. This review provides a current overview of these mechanisms and covers how iron, zinc, and manganese impact Lactobacillaceae in the gut microbiota with an emphasis on their biochemical roles, requirements, and homeostatic mechanisms in several species.
Collapse
Affiliation(s)
- Uyen Huynh
- Department of Chemistry, University of Houston, Houston, TX, USA
| | | |
Collapse
|
22
|
Amerikanou C, Karavoltsos S, Gioxari A, Tagkouli D, Sakellari A, Papada E, Kalogeropoulos N, Forbes A, Kaliora AC. Clinical and inflammatory biomarkers of inflammatory bowel diseases are linked to plasma trace elements and toxic metals; new insights into an old concept. Front Nutr 2022; 9:997356. [PMID: 36570124 PMCID: PMC9780073 DOI: 10.3389/fnut.2022.997356] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Inflammatory bowel diseases (IBD) are chronic immune-mediated diseases, mainly represented by Crohn's disease (CD) and ulcerative colitis (UC). Several environmental factors have been proposed to contribute to disease pathogenesis, amongst which are metals. These can affect the immune system and may be associated with IBD. The aim of the present cross-sectional study was to investigate blood levels of metals in IBD patients and to examine possible associations with clinical and inflammatory disease markers. Methods In total, 76 CD patients, 39 UC patients and 38 healthy controls were included. Blood and stool samples were collected. Metals were quantified in plasma samples using inductively coupled plasma mass spectrometry. Results There were more abnormalities of circulating metals in CD than in UC when compared to healthy controls. CD: Concentrations of the essential trace elements zinc and selenium were lower in CD patients than the controls. Chromium was negatively associated with serum IL-6 (Beta: -3.558, p = 0.011), and caesium with fecal calprotectin (Beta: -0.481, p = 0.038) and serum IL-10 (Beta: -1.912, p = 0.050). In contrast, copper was positively associated with C-reactive protein (Beta: 2.548 × 102, p = 0.033). UC: In UC, a negative association of iron with serum myeloperoxidase levels (Beta: -1.270 × 103, p = 0.044) was detected. Thallium, a hazardous metal, however, was positively associated with disease activity (Beta: 3.899, p = < 0.01). Conclusion In conclusion, our study offers new insights into the relations of metals with IBD. Further research should focus on the evaluation of the above associations and potential underlying mechanisms.
Collapse
Affiliation(s)
- Charalampia Amerikanou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece,Charalampia Amerikanou ;
| | - Sotirios Karavoltsos
- Laboratory of Environmental Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Aristea Gioxari
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece,Department of Nutritional Science and Dietetics, University of Peloponnese, Tripolis, Greece
| | - Dimitra Tagkouli
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Aikaterini Sakellari
- Laboratory of Environmental Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathia Papada
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece,Division of Medicine, University College London, London, United Kingdom
| | - Nick Kalogeropoulos
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Alastair Forbes
- Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Andriana C. Kaliora
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece,*Correspondence: Andriana C. Kaliora ;
| |
Collapse
|
23
|
Santana-Codina N, del Rey MQ, Kapner KS, Zhang H, Gikandi A, Malcolm C, Poupault C, Kuljanin M, John KM, Biancur DE, Chen B, Das NK, Lowder KE, Hennessey CJ, Huang W, Yang A, Shah YM, Nowak JA, Aguirre AJ, Mancias JD. NCOA4-Mediated Ferritinophagy Is a Pancreatic Cancer Dependency via Maintenance of Iron Bioavailability for Iron-Sulfur Cluster Proteins. Cancer Discov 2022; 12:2180-2197. [PMID: 35771492 PMCID: PMC9437572 DOI: 10.1158/2159-8290.cd-22-0043] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDAC) depend on autophagy for survival; however, the metabolic substrates that autophagy provides to drive PDAC progression are unclear. Ferritin, the cellular iron storage complex, is targeted for lysosomal degradation (ferritinophagy) by the selective autophagy adaptor NCOA4, resulting in release of iron for cellular utilization. Using patient-derived and murine models of PDAC, we demonstrate that ferritinophagy is upregulated in PDAC to sustain iron availability, thereby promoting tumor progression. Quantitative proteomics reveals that ferritinophagy fuels iron-sulfur cluster protein synthesis to support mitochondrial homeostasis. Targeting NCOA4 leads to tumor growth delay and prolonged survival but with the development of compensatory iron acquisition pathways. Finally, enhanced ferritinophagy accelerates PDAC tumorigenesis, and an elevated ferritinophagy expression signature predicts for poor prognosis in patients with PDAC. Together, our data reveal that the maintenance of iron homeostasis is a critical function of PDAC autophagy, and we define NCOA4-mediated ferritinophagy as a therapeutic target in PDAC. SIGNIFICANCE Autophagy and iron metabolism are metabolic dependencies in PDAC. However, targeted therapies for these pathways are lacking. We identify NCOA4-mediated selective autophagy of ferritin ("ferritinophagy") as upregulated in PDAC. Ferritinophagy supports PDAC iron metabolism and thereby tumor progression and represents a new therapeutic target in PDAC. See related commentary by Jain and Amaravadi, p. 2023. See related article by Ravichandran et al., p. 2198. This article is highlighted in the In This Issue feature, p. 2007.
Collapse
Affiliation(s)
- Naiara Santana-Codina
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Maria Quiles del Rey
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Huan Zhang
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ajami Gikandi
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Callum Malcolm
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Clara Poupault
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kristen M. John
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Douglas E. Biancur
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Brandon Chen
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Nupur K. Das
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kristen E. Lowder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Connor J. Hennessey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wesley Huang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yatrik M. Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Jonathan A. Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Joseph D. Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Dong PF, Jin C, Lian CY, Wang L, Wang ZY. Enhanced Extracellular Matrix Degradation in Growth Plate Contributes to Manganese Deficiency-Induced Tibial Dyschondroplasia in Broiler Chicks. Biol Trace Elem Res 2022; 200:3326-3335. [PMID: 34546491 DOI: 10.1007/s12011-021-02921-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/08/2021] [Indexed: 11/26/2022]
Abstract
Manganese (Mn) is a crucial trace element for poultry nutrition, and its deficiency compromises tibial cartilage development, leading to perosis and a higher incidence of slipped tendon. Tibial dyschondroplasia (TD) is a metabolic cartilage disease characterized by disruption of endochondral bone formation, which is closely related to extracellular matrix (ECM) degradation, in which Mn deficiency plays an important role. Previous studies have confirmed the role of matrix metalloproteinases (MMPs) in the pathogenesis of TD, but whether dysregulated ECM degradation and MMP expression profiles in growth plate are involved in Mn deficiency-induced avian TD has not been fully elucidated yet. Thus, this study was conducted to clarify these issues. Firstly, we successfully established TD model induced by Mn deficiency in broiler chicks. Mn deficiency decreased the number of chondrocytes, contents of proteoglycan, and type II collagen in tibial growth plate, demonstrating the tibial growth plate damage with enhanced ECM degradation. Also, Mn deficiency inhibited the Nrf2 signaling pathway and enhanced the protein levels of NLRP3, active caspase-1, and active IL-1β in tibial growth plate, indicating the oxidative stress and inflammatory response in Mn deficiency-induced TD. Additionally, upregulated expression levels of MMPs (MMP1, 9, and 13) were observed in tibial growth plate of Mn deficiency group. In summary, these findings suggest that Mn deficiency-enhanced ECM degradation is involved in avian TD, which may be correlated with oxidative stress, inflammatory response, and upregulation of MMPs.
Collapse
Affiliation(s)
- Peng-Fei Dong
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Cong Jin
- Yishui Animal Disease Control Center, 690 Chang'an Street, Shandong Province, 276400, Lin'yi City, China
| | - Cai-Yu Lian
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Lin Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
| | - Zhen-Yong Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
| |
Collapse
|
25
|
Xue H, Song W, Wang Z, Wang Q. Ulva prolifera Polysaccharide–Manganese Alleviates Inflammation and Regulates Microbiota Composition in Dextran Sulfate Sodium-Induced Colitis Mice. Front Microbiol 2022; 13:916552. [PMID: 35722338 PMCID: PMC9205199 DOI: 10.3389/fmicb.2022.916552] [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: 04/09/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Manganese (Mn) deficiency exacerbates colitis symptoms, whereas diet supplemented with inorganic Mn merely maintains colon length in experimental colitis. In the present study, a new form of Mn, Ulva prolifera polysaccharide cheated-Mn (PMn) was used and its treatment effects on dextran sulfate sodium (DSS)-induced colitis were investigated. Male C57BL/6 mice were orally administrated with 3.5% DSS to induce colitis. Then, the colitis mice were treated with PBS or PMn for 7 days. The results showed that PMn administration help retrieve the body weight loss and intestinal morphology damage, and alleviate apoptosis and inflammatory responses in colitis mice. Moreover, PMn administration decreased intestinal infiltration as indicated by decreased concentration of myeloperoxidase and eosinophil peroxidase. Importantly, PMn retrieved the increased abundance of Firmicutes and the decreased abundance of Bacteroidetes caused by DSS, suggested its beneficial roles in regulating microbiota composition in mice with colon inflammation. Gut microbiota composition at the genus level in the mice administrated with PMn was similar to those in control mice, whereas they were clearly distinct from DSS-treated mice. These results support the potential therapeutic role of PMn in the treatment of intestinal colitis and microbes may play critical roles in mediating its effects.
Collapse
Affiliation(s)
- Haoran Xue
- Department of Clinical Laboratory, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Wei Song
- Ministry of Natural Resources (MNR) Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Zongling Wang
- Ministry of Natural Resources (MNR) Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital (Jinan) of Shandong University, Jinan, China
- *Correspondence: Qian Wang
| |
Collapse
|
26
|
Das NK, Jain C, Sankar A, Schwartz AJ, Santana-Codina N, Solanki S, Zhang Z, Ma X, Parimi S, Rui L, Mancias JD, Shah YM. Modulation of the HIF2α-NCOA4 axis in enterocytes attenuates iron loading in a mouse model of hemochromatosis. Blood 2022; 139:2547-2552. [PMID: 34990508 PMCID: PMC9029091 DOI: 10.1182/blood.2021013452] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/22/2021] [Indexed: 11/20/2022] Open
Abstract
Intestinal iron absorption is activated during increased systemic demand for iron. The best-studied example is iron deficiency anemia, which increases intestinal iron absorption. Interestingly, the intestinal response to anemia is very similar to that of iron overload disorders, as both the conditions activate a transcriptional program that leads to a hyperabsorption of iron via the transcription factor hypoxia-inducible factor 2α (HIF2α). However, pathways for selective targeting of intestine-mediated iron overload remain unknown. Nuclear receptor coactivator 4 (NCOA4) is a critical cargo receptor for autophagic breakdown of ferritin and the subsequent release of iron, in a process termed ferritinophagy. Our work demonstrates that NCOA4-mediated intestinal ferritinophagy is integrated into systemic iron demand via HIF2α. To demonstrate the importance of the intestinal HIF2α/ferritinophagy axis in systemic iron homeostasis, whole-body and intestine-specific NCOA4-/- mouse lines were generated and assessed. The analyses revealed that the intestinal and systemic response to iron deficiency was not altered after disruption of intestinal NCOA4. However, in a mouse model of hemochromatosis, ablation of intestinal NCOA4 was protective against iron overload. Therefore, NCOA4 can be selectively targeted for the management of iron overload disorders without disrupting the physiological processes involved in the response to systemic iron deficiency.
Collapse
Affiliation(s)
- Nupur K Das
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Chesta Jain
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Amanda Sankar
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Andrew J Schwartz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Naiara Santana-Codina
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | - Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Zhiguo Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Xiaoya Ma
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Sanjana Parimi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA; and
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
27
|
Huynh U, Qiao M, King J, Trinh B, Valdez J, Haq M, Zastrow ML. Differential Effects of Transition Metals on Growth and Metal Uptake for Two Distinct Lactobacillus Species. Microbiol Spectr 2022; 10:e0100621. [PMID: 35080431 PMCID: PMC8791193 DOI: 10.1128/spectrum.01006-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
Lactobacillus is a genus of Gram-positive bacteria and comprises a major part of the lactic acid bacteria group that converts sugars to lactic acid. Lactobacillus species found in the gut microbiota are considered beneficial to human health and commonly used in probiotic formulations, but their molecular functions remain poorly defined. Microbes require metal ions for growth and function and must acquire them from the surrounding environment. Therefore, lactobacilli need to compete with other gut microbes for these nutrients, although their metal requirements are not well-understood. Indeed, the abundance of lactobacilli in the microbiota is frequently affected by dietary intake of essential metals like zinc, manganese, and iron, but few studies have investigated the role of metals, especially zinc, in the physiology and metabolism of Lactobacillus species. Here, we investigated metal uptake by quantifying total cellular metal contents and compared how transition metals affect the growth of two distinct Lactobacillus species, Lactobacillus plantarum ATCC 14917 and Lactobacillus acidophilus ATCC 4356. When grown in rich or metal-limited medium, both species took up more manganese, zinc, and iron compared with other transition metals measured. Distinct zinc-, manganese- and iron-dependent patterns were observed in the growth kinetics for these species and while certain levels of each metal promoted the growth kinetics of both Lactobacillus species, the effects depend significantly on the culture medium and growth conditions. IMPORTANCE The gastrointestinal tract contains trillions of microorganisms, which are central to human health. Lactobacilli are considered beneficial microbiota members and are often used in probiotics, but their molecular functions, and especially those which are metal-dependent, remain poorly defined. Abundance of lactobacilli in the microbiota is frequently affected by dietary intake of essential metals like manganese, zinc, and iron, but results are complex, sometimes contradictory, and poorly predictable. There is a significant need to understand how host diet and metabolism will affect the microbiota, given that changes in microbiota composition are linked with disease and infection. The significance of our research is in gaining insight to how metals distinctly affect individual Lactobacillus species, which could lead to novel therapeutics and improved medical treatment. Growth kinetics and quantification of metal contents highlights how distinct species can respond differently to varied metal availability and provide a foundation for future molecular and mechanistic studies.
Collapse
Affiliation(s)
- Uyen Huynh
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Muxin Qiao
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - John King
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Brittany Trinh
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Juventino Valdez
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Marium Haq
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Melissa L. Zastrow
- Department of Chemistry, University of Houston, Houston, Texas, United States
| |
Collapse
|
28
|
Aring L, Choi EK, Kopera H, Lanigan T, Iwase S, Klionsky DJ, Seo YA. A neurodegeneration gene, WDR45, links impaired ferritinophagy to iron accumulation. J Neurochem 2021; 160:356-375. [PMID: 34837396 PMCID: PMC8811950 DOI: 10.1111/jnc.15548] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/18/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a clinically and genetically heterogeneous group of neurodegenerative diseases characterized by the abnormal accumulation of brain iron and the progressive degeneration of the nervous system. One of the recently identified subtypes of NBIA is β‐propeller protein‐associated neurodegeneration (BPAN). BPAN is caused by de novo mutations in the WDR45/WIPI4 (WD repeat domain 45) gene. WDR45 is one of the four mammalian homologs of yeast Atg18, a regulator of autophagy. WDR45 deficiency in BPAN patients and animal models may result in defects in autophagic flux. However, how WDR45 deficiency leads to brain iron overload remains unclear. To elucidate the role of WDR45, we generated a WDR45‐knockout (KO) SH‐SY5Y neuroblastoma cell line using CRISPR‐Cas9‐mediated genome editing. Using these cells, we demonstrated that the non‐TF (transferrin)‐bound iron pathway dominantly mediated the accumulation of iron. Moreover, the loss of WDR45 led to defects in ferritinophagy, a form of autophagy that degrades the iron storage protein ferritin. We showed that impaired ferritinophagy contributes to iron accumulation in WDR45‐KO cells. Iron accumulation was also detected in the mitochondria, which was accompanied by impaired mitochondrial respiration, elevated reactive oxygen species, and increased cell death. Thus, our study links WDR45 to specific iron acquisition pathways and ferritinophagy.
Collapse
Affiliation(s)
- Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Huira Kopera
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA.,Vector Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas Lanigan
- Vector Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, Michigan, USA.,Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel J Klionsky
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Pajarillo EAB, Lee E, Kang DK. Trace metals and animal health: Interplay of the gut microbiota with iron, manganese, zinc, and copper. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:750-761. [PMID: 34466679 PMCID: PMC8379138 DOI: 10.1016/j.aninu.2021.03.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022]
Abstract
Metals such as iron, manganese, copper, and zinc are recognized as essential trace elements. These trace metals play critical roles in development, growth, and metabolism, participating in various metabolic processes by acting as cofactors of enzymes or providing structural support to proteins. Deficiency or toxicity of these metals can impact human and animal health, giving rise to a number of metabolic and neurological disorders. Proper breakdown, absorption, and elimination of these trace metals is a tightly regulated process that requires crosstalk between the host and these micronutrients. The gut is a complex system that serves as the interface between these components, but other factors that contribute to this delicate interaction are not well understood. The gut is home to trillions of microorganisms and microbial genes (the gut microbiome) that can regulate the metabolism and transport of micronutrients and contribute to the bioavailability of trace metals through their assimilation from food sources or by competing with the host. Furthermore, deficiency or toxicity of these metals can modulate the gut microenvironment, including microbiota, nutrient availability, stress, and immunity. Thus, understanding the role of the gut microbiota in the metabolism of manganese, iron, copper, and zinc, as well as in heavy metal deficiencies and toxicities, and vice versa, may provide insight into developing improved or alternative therapeutic strategies to address emerging health concerns. This review describes the current understanding of how the gut microbiome and trace metals interact and affect host health, particularly in pigs.
Collapse
Affiliation(s)
- Edward Alain B. Pajarillo
- Department of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee 32307, FL, USA
| | - Eunsook Lee
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
30
|
Tinkov AA, Martins AC, Avila DS, Gritsenko VA, Skalny AV, Santamaria A, Lee E, Bowman AB, Aschner M. Gut Microbiota as a Potential Player in Mn-Induced Neurotoxicity. Biomolecules 2021; 11:1292. [PMID: 34572505 PMCID: PMC8469589 DOI: 10.3390/biom11091292] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Manganese (Mn) is an essential metal, which at high exposures causes neurotoxic effects and neurodegeneration. The neurotoxic effects of Mn are mediated by neuroinflammation, oxidative and endoplasmic reticulum stress, mitochondrial dysfunction, and other mechanisms. Recent findings have demonstrated the potential impact of Mn overexposure on gut microbiota dysbiosis, which is known to contribute to neurodegeneration via secretion of neuroactive and proinflammatory metabolites. Therefore, in this review, we discuss the existing data on the impact of Mn exposure on gut microbiota biodiversity, bacterial metabolite production, and gut wall permeability regulating systemic levels. Recent data have demonstrated that Mn exposure may affect gut microbiota biodiversity by altering the abundance of Shiegella, Ruminococcus, Dorea, Fusicatenibacter, Roseburia, Parabacteroides, Bacteroidetes, Firmicutes, Ruminococcaceae, Streptococcaceae, and other bacterial phyla. A Mn-induced increase in Bacteroidetes abundance and a reduced Firmicutes/Bacteroidetes ratio may increase lipopolysaccharide levels. Moreover, in addition to increased systemic lipopolysaccharide (LPS) levels, Mn is capable of potentiating LPS neurotoxicity. Due to the high metabolic activity of intestinal microflora, Mn-induced perturbations in gut microbiota result in a significant alteration in the gut metabolome that has the potential to at least partially mediate the biological effects of Mn overexposure. At the same time, a recent study demonstrated that healthy microbiome transplantation alleviates Mn-induced neurotoxicity, which is indicative of the significant role of gut microflora in the cascade of Mn-mediated neurotoxicity. High doses of Mn may cause enterocyte toxicity and affect gut wall integrity through disruption of tight junctions. The resulting increase in gut wall permeability further promotes increased translocation of LPS and neuroactive bacterial metabolites to the systemic blood flow, ultimately gaining access to the brain and leading to neuroinflammation and neurotransmitter imbalance. Therefore, the existing data lead us to hypothesize that gut microbiota should be considered as a potential target of Mn toxicity, although more detailed studies are required to characterize the interplay between Mn exposure and the gut, as well as its role in the pathogenesis of neurodegeneration and other diseases.
Collapse
Affiliation(s)
- Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Daiana Silva Avila
- Laboratory of Biochemistry and Toxicoology in Caenorhabditis elegans, Universidade Federal do Pampa, Campus Uruguaiana, BR-472 Km 592, Uruguaiana 97500-970, RS, Brazil;
| | - Victor A. Gritsenko
- Institute of Cellular and Intracellular Symbiosis, Ural Branch of the Russian Academy of Sciences, Pionerskaya st 11, 460000 Orenburg, Russia;
| | - Anatoly V. Skalny
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Laboratory of Medical Elementology, KG Razumovsky Moscow State University of Technologies and Management, 109004 Moscow, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico;
| | - Eunsook Lee
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Michael Aschner
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|
31
|
Gîlcă-Blanariu GE, Coroabă A, Ciocoiu M, Trifan A, Dimofte G, Diaconescu S, Afrăsânie VA, Balan GG, Pinteală T, Ștefănescu G. Hair EDX Analysis-A Promising Tool for Micronutrient Status Evaluation of Patients with IBD? Nutrients 2021; 13:2572. [PMID: 34444730 PMCID: PMC8399661 DOI: 10.3390/nu13082572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 02/05/2023] Open
Abstract
Micronutrient deficiencies can arise in various conditions, including inflammatory bowel diseases (IBD), and diagnosing these deficiencies can be challenging in the absence of specific clinical signs. The aim of this study was to evaluate the status of various trace elements hair concentration in IBD patients compared to a healthy control group and to identify potential correlations between the micronutrient status and relevant parameters related to disease activity. The concentrations of iron, magnesium, calcium, zinc, copper, manganese, selenium and sulfur in the hair of 37 IBD patients with prior diagnosed IBD (12 Crohn's disease and 25 ulcerative colitis) and 31 healthy controls were evaluated by Energy Dispersive X-Ray spectroscopy (EDX). Significant differences in hair concentration profile of studied trace elements were identified for IBD patients compared to healthy controls. A significantly decreased hair concentration of iron, magnesium, calcium and selenium and a significantly increased sulfur hair concentration were observed in IBD patients at the time of evaluation. A decreased hair calcium concentration (r = -0.772, p = 0.003) and an increased sulfur concentration (r = 0.585, p = 0.046) were significantly correlated with disease activity. Conclusion: Hair mineral and trace elements evaluation may contribute to a proper evaluation of their status in IBD patients and improving the management of nutritional status of IBD patients.
Collapse
Affiliation(s)
- Georgiana-Emmanuela Gîlcă-Blanariu
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
| | - Adina Coroabă
- Petru Poni Institute of Macromolecular Chemistry, 700487 Iași, Romania;
| | - Manuela Ciocoiu
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
| | - Anca Trifan
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
- Sf Spiridon County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Gabriel Dimofte
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
| | - Smaranda Diaconescu
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
| | - Vlad-Adrian Afrăsânie
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
| | - Gheorghe G. Balan
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
- Sf Spiridon County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Tudor Pinteală
- Sf Spiridon County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Gabriela Ștefănescu
- Faculty of Medicine, Grigore T Popa University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (M.C.); (A.T.); (G.D.); (V.-A.A.); (G.G.B.); (G.Ș.)
- Sf Spiridon County Clinical Emergency Hospital, 700111 Iași, Romania;
| |
Collapse
|
32
|
Sub-chronic exposure to PhIP induces oxidative damage and DNA damage, and disrupts the amino acid metabolism in the colons of Wistar rats. Food Chem Toxicol 2021; 153:112249. [PMID: 33945839 DOI: 10.1016/j.fct.2021.112249] [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] [Received: 12/25/2020] [Revised: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
Heterocyclic amines (HCAs) are a group of mutagenic compounds produced during thermal processing of protein-rich foods. One of the most abundant HCAs, 2-amino-1-methyl-6-phenylimidazo[4,5-b] pyridine (PhIP) has potential carcinogenic and mutagenic effects on human organs, especially the colon. This study aimed to explore the toxic effects of PhIP on amino acid metabolism in the colon of Wistar rats using RNA-seq and LC-MS/MS. Exposure to PhIP for 4 weeks induced oxidative damage and DNA damage in the colons, and disrupted the expression of related genes involved in tryptophan metabolism, beta(β)-alanine metabolism, valine, leucine, and isoleucine degradation, and glutathione metabolic pathways. Moreover, the levels of fecal metabolites related to amino acid metabolism were affected by PhIP. Cumulatively, these results indicate that PhIP can induce colonic oxidative injury and disorders related to amino acid metabolism, thereby providing a new theoretical basis for the study of PhIP toxicity.
Collapse
|
33
|
Recent Advances in Understanding the Influence of Zinc, Copper, and Manganese on the Gastrointestinal Environment of Pigs and Poultry. Animals (Basel) 2021; 11:ani11051276. [PMID: 33946674 PMCID: PMC8145729 DOI: 10.3390/ani11051276] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Pigs and poultry, similar to humans, need regular consumption of zinc, copper, and manganese for normal functioning. To ensure adequate dietary intake, and prevent deficiency, their diets are supplemented with sufficient, often excessive, levels of these minerals or even at higher levels, which have been associated with improvements in their health and/or growth. However, if provided in excess, mineral quantities beyond those required are simply excreted from the animal, which is associated with negative consequences for the environment and even the development of antimicrobial resistance. Therefore, it is of great interest to better understand the dynamics of zinc, copper, and manganese in the intestine of pigs and poultry following consumption of supplemented diets, and how the requirements and benefits related to these minerals can be optimized and negative impacts minimized. The intestine of pigs and poultry contains vast numbers of microorganisms, notably bacteria, that continually interact with, and influence, their host. This review explores the influence of zinc, copper, and manganese on these interactions and how novel forms of these minerals have the potential to maximize their delivery and benefits, while limiting any negative consequences. Abstract Zinc, copper, and manganese are prominent essential trace (or micro) minerals, being required in small, but adequate, amounts by pigs and poultry for normal biological functioning. Feed is a source of trace minerals for pigs and poultry but variable bioavailability in typical feed ingredients means that supplementation with low-cost oxides and sulphates has become common practice. Such trace mineral supplementation often provides significant ‘safety margins’, while copper and zinc have been supplemented at supra-nutritional (or pharmacological) levels to improve health and/or growth performance. Regulatory mechanisms ensure that much of this oversupply is excreted by the host into the environment, which can be toxic to plants and microorganisms or promote antimicrobial resistance in microbes, and thus supplying trace minerals more precisely to pigs and poultry is necessary. The gastrointestinal tract is thus central to the maintenance of trace mineral homeostasis and the provision of supra-nutritional or pharmacological levels is associated with modification of the gut environment, such as the microbiome. This review, therefore, considers recent advances in understanding the influence of zinc, copper, and manganese on the gastrointestinal environment of pigs and poultry, including more novel, alternative sources seeking to maintain supra-nutritional benefits with minimal environmental impact.
Collapse
|
34
|
Li C, Chen M, He X, Ouyang D. A mini-review on ion fluxes that regulate NLRP3 inflammasome activation. Acta Biochim Biophys Sin (Shanghai) 2021; 53:131-139. [PMID: 33355638 DOI: 10.1093/abbs/gmaa155] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Indexed: 12/15/2022] Open
Abstract
The activation of NLR family pyrin domain containing 3 (NLRP3) inflammasome can be induced by a wide spectrum of activators. This is unlikely achieved by the binding of different activators directly to the NLRP3 protein itself, as the activators found so far show different forms of chemical structures. Previous studies have shown that these activators can induce potassium ion (K+) and chloride ion (Cl-) efflux, calcium (Ca2+) and other ion mobilization, mitochondrial dysfunction, and lysosomal disruption, all of which are believed to cause NLRP3 inflammasome activation; how these events are induced by the activators and how they coordinate with each other in inducing the NLRP3 inflammasome activation are not fully understood. Increasing evidence suggests that the coordinated change of intracellular ion concentrations may be a common mechanism for the NLRP3 activation by different activators. In this mini-review, we present a brief summary of the current knowledge about how different ionic flows (including K+, sodium ion, Ca2+, magnesium ion, manganese ion, zinc ion, iron ion, and Cl-) are involved in regulating the NLRP3 inflammasome activation in macrophages.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Mingye Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xianhui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
35
|
Kundra P, Rachmühl C, Lacroix C, Geirnaert A. Role of Dietary Micronutrients on Gut Microbial Dysbiosis and Modulation in Inflammatory Bowel Disease. Mol Nutr Food Res 2021. [DOI: 10.1002/mnfr.201901271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Palni Kundra
- Laboratory of Food Biotechnology Institute of Food Nutrition and Health Schmelzbergstrasse 7 Zürich 8092 Switzerland
| | - Carole Rachmühl
- Laboratory of Food Biotechnology Institute of Food Nutrition and Health Schmelzbergstrasse 7 Zürich 8092 Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology Institute of Food Nutrition and Health Schmelzbergstrasse 7 Zürich 8092 Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology Institute of Food Nutrition and Health Schmelzbergstrasse 7 Zürich 8092 Switzerland
| |
Collapse
|
36
|
Seo YA, Choi EK, Aring L, Paschall M, Iwase S. Transcriptome Analysis of the Cerebellum of Mice Fed a Manganese-Deficient Diet. Front Genet 2020; 11:558725. [PMID: 33408735 PMCID: PMC7780674 DOI: 10.3389/fgene.2020.558725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Manganese (Mn), primarily acquired through diet, is required for brain function and development. Epidemiological studies have found an association between both low and high levels of Mn and impaired neurodevelopment in children. Recent genetic studies have revealed that patients with congenital Mn deficiency display severe psychomotor disability and cerebral and cerebellar atrophy. Although the impact of Mn on gene expression is beginning to be appreciated, Mn-dependent gene expression remains to be explored in vertebrate animals. The goal of this study was to use a mouse model to define the impact of a low-Mn diet on brain metal levels and gene expression. We interrogated gene expression changes in the Mn-deficient mouse brain at the genome-wide scale by RNA-seq analysis of the cerebellum of mice fed low or normal Mn diets. A total of 137 genes were differentially expressed in Mn-deficient cerebellums compared with Mn-adequate cerebellums (Padj < 0.05). Mn-deficient mice displayed downregulation of key pathways involved with "focal adhesion," "neuroactive ligand-receptor interaction," and "cytokine-cytokine receptor interaction" and upregulation of "herpes simplex virus 1 infection," "spliceosome," and "FoxO signaling pathway." Reactome pathway analysis identified upregulation of the splicing-related pathways and transcription-related pathways, as well as downregulation of "metabolism of carbohydrate," and "extracellular matrix organization," and "fatty acid metabolism" reactomes. The recurrent identifications of splicing-related pathways suggest that Mn deficiency leads to upregulation of splicing machineries and downregulation of diverse biological pathways.
Collapse
Affiliation(s)
- Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Molly Paschall
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Shigeki Iwase
- Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
37
|
A missense variant in SLC39A8 confers risk for Crohn's disease by disrupting manganese homeostasis and intestinal barrier integrity. Proc Natl Acad Sci U S A 2020; 117:28930-28938. [PMID: 33139556 PMCID: PMC7682327 DOI: 10.1073/pnas.2014742117] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
SLC39A8 A391T exhibits remarkable pleiotropic effects on multiple conditions, including cardiovascular diseases, Parkinson’s disease, and Crohn’s disease. However, how this single coding variant impacts such a wide range of pathologies has not been investigated. We generated Slc39a8 A391T knockin mice and show that they exhibit severe Mn deficiency in the colon, and impaired intestinal barrier integrity due to glycoprotein barrier structure defects, leading to indolent inflammation that can prime further inflammation driven by epithelial injury. Thus, we highlight the importance of Mn in gut homeostasis, and mechanistically unravel how A391T impacts intestinal barrier integrity. Common genetic variants interact with environmental factors to impact risk of heritable diseases. A notable example of this is a single-nucleotide variant in the Solute Carrier Family 39 Member 8 (SLC39A8)geneencoding the missense variant A391T, which is associated with a variety of traits ranging from Parkinson’s disease and neuropsychiatric disease to cardiovascular and metabolic diseases and Crohn’s disease. The remarkable extent of pleiotropy exhibited by SLC39A8 A391T raises key questions regarding how a single coding variant can contribute to this diversity of clinical outcomes and what is the mechanistic basis for this pleiotropy. Here, we generate a murine model for the Slc39a8 A391T allele and demonstrate that these mice exhibit Mn deficiency in the colon associated with impaired intestinal barrier function and epithelial glycocalyx disruption. Consequently, Slc39a8 A391T mice exhibit increased sensitivity to epithelial injury and pathological inflammation in the colon. Taken together, our results link a genetic variant with a dietary trace element to shed light on a tissue-specific mechanism of disease risk based on impaired intestinal barrier integrity.
Collapse
|
38
|
Sunuwar L, Frkatović A, Sharapov S, Wang Q, Neu HM, Wu X, Haritunians T, Wan F, Michel S, Wu S, Donowitz M, McGovern D, Lauc G, Sears C, Melia J. Pleiotropic ZIP8 A391T implicates abnormal manganese homeostasis in complex human disease. JCI Insight 2020; 5:140978. [PMID: 32897876 PMCID: PMC7605523 DOI: 10.1172/jci.insight.140978] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
ZIP8 is a metal transporter with a role in manganese (Mn) homeostasis. A common genetic variant in ZIP8 (rs13107325; A391T) ranks in the top 10 of pleiotropic SNPs identified in GWAS; A391T has associations with an increased risk of schizophrenia, obesity, Crohn’s disease, and reduced blood Mn. Here, we used CRISPR/Cas9-mediated knockin (KI) to generate a mouse model of ZIP8 A391T (Zip8 393T-KI mice). Recapitulating the SNP association with blood Mn, blood Mn was reduced in Zip8 393T-KI mice. There was restricted abnormal tissue Mn homeostasis, with decreases in liver and kidney Mn and a reciprocal increase in biliary Mn, providing in vivo evidence of hypomorphic Zip8 function. Upon challenge in a chemically induced colitis model, male Zip8 393T-KI mice exhibited enhanced disease susceptibility. ZIP8 391-Thr associated with reduced triantennary plasma N-glycan species in a population-based cohort to define a genotype-specific glycophenotype hypothesized to be linked to Mn-dependent glycosyltransferase activity. This glycophenotype was maintained in a cohort of patients with Crohn’s disease. These data and the pleiotropic disease associations with ZIP8 391-Thr suggest underappreciated roles of Mn homeostasis in complex human disease. Abnormal manganese homeostasis is implicated by a GWAS disease-associated SNP, rs13107325 (ZIP8 A391T), studied in a knockin mouse model and human N-glycome analyses.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Sodbo Sharapov
- Laboratory of Glycogenomics, Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Qinchuan Wang
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heather M Neu
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Xinqun Wu
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology and.,Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sarah Michel
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Shaoguang Wu
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dermot McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | - Cynthia Sears
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joanna Melia
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
39
|
Foligné B, George F, Standaert A, Garat A, Poiret S, Peucelle V, Ferreira S, Sobry H, Muharram G, Lucau‐Danila A, Daniel C. High‐dose dietary supplementation with zinc prevents gut inflammation: Investigation of the role of metallothioneins and beyond by transcriptomic and metagenomic studies. FASEB J 2020; 34:12615-12633. [DOI: 10.1096/fj.202000562rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Benoît Foligné
- Univ. Lille, INSERM, CHU Lille, U1286 ‐ Infinite ‐ Institute for Translational Research in Inflammation Lille France
| | - Fanny George
- Univ. Lille, INSERM, CHU Lille, U1286 ‐ Infinite ‐ Institute for Translational Research in Inflammation Lille France
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483‐IMPECS‐IMPact de l'Environnement Chimique sur la Santé humaine Lille France
| | - Annie Standaert
- Univ. Lille, INSERM, CHU Lille, U1286 ‐ Infinite ‐ Institute for Translational Research in Inflammation Lille France
| | - Anne Garat
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483‐IMPECS‐IMPact de l'Environnement Chimique sur la Santé humaine Lille France
- CHU Lille, Unité Fonctionnelle de Toxicologie Lille France
| | - Sabine Poiret
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 ‐ CIIL ‐ Center for Infection and Immunity of Lille Lille France
| | - Véronique Peucelle
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 ‐ CIIL ‐ Center for Infection and Immunity of Lille Lille France
| | | | - Hélène Sobry
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 ‐ CIIL ‐ Center for Infection and Immunity of Lille Lille France
| | - Ghaffar Muharram
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 ‐ CIIL ‐ Center for Infection and Immunity of Lille Lille France
| | - Anca Lucau‐Danila
- BIOECOAGRO INRAe, UArtois, ULiege, ULille, ULCO, UPJV, YNCREA, Institut Charles Viollette Lille France
| | - Catherine Daniel
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 ‐ CIIL ‐ Center for Infection and Immunity of Lille Lille France
| |
Collapse
|
40
|
Zhang H, Pan S, Zhang K, Michiels J, Zeng Q, Ding X, Wang J, Peng H, Bai J, Xuan Y, Su Z, Bai S. Impact of Dietary Manganese on Intestinal Barrier and Inflammatory Response in Broilers Challenged with Salmonella Typhimurium. Microorganisms 2020; 8:microorganisms8050757. [PMID: 32443502 PMCID: PMC7285304 DOI: 10.3390/microorganisms8050757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/10/2020] [Accepted: 05/16/2020] [Indexed: 11/16/2022] Open
Abstract
Growing concern for public health and food safety has prompted a special interest in developing nutritional strategies for removing waterborne and foodborne pathogens, including Salmonella. Strong links between manganese (Mn) and intestinal barrier or immune function hint that dietary Mn supplementation is likely to be a promising approach to limit the loads of pathogens in broilers. Here, we provide evidence that Salmonella Typhimurium (S. Typhimurium, 4 × 108 CFUs) challenge-induced intestinal injury along with systemic Mn redistribution in broilers. Further examining of the effect of dietary Mn treatments (a basal diet plus additional 0, 40, or 100 mg Mn/kg for corresponding to Mn-deficient, control, or Mn-surfeit diet, respectively) on intestinal barrier and inflammation status of broilers infected with S. Typhimurium revealed that birds fed the control and Mn-surfeit diets exhibited improved intestinal tight junctions and microbiota composition. Even without Salmonella infection, dietary Mn deficiency alone increased intestinal permeability by impairing intestinal tight junctions. In addition, when fed the control and Mn-surfeit diets, birds showed decreased Salmonella burdens in cecal content and spleen, with a concomitant increase in inflammatory cytokine levels in spleen. Furthermore, the dietary Mn-supplementation-mediated induction of cytokine production was probably associated with the nuclear factor kappa-B (NF-κB)/hydrogen peroxide (H2O2) pathway, as judged by the enhanced manganese superoxide dismutase activity and the increased H2O2 level in mitochondria, together with the increased mRNA level of NF-κB in spleen. Ingenuity-pathway analysis indicated that acute-phase response pathways, T helper type 1 pathway, and dendritic cell maturation were significantly activated by the dietary Mn supplementation. Our data suggest that dietary Mn supplementation could enhance intestinal barrier and splenic inflammatory response to fight against Salmonella infection in broilers.
Collapse
Affiliation(s)
- Huaiyong Zhang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Shuqin Pan
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Keying Zhang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Joris Michiels
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium;
| | - Qiufeng Zeng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Xuemei Ding
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Jianping Wang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Huanwei Peng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Jie Bai
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Yue Xuan
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Zhuowei Su
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
| | - Shiping Bai
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (S.P.); (K.Z.); (Q.Z.); (X.D.); (J.W.); (H.P.); (J.B.); (Y.X.); (Z.S.)
- Correspondence: ; Tel.: +86-028-86290922
| |
Collapse
|