1
|
Sampah MES, Moore H, Ahmad R, Duess J, Lu P, Lopez C, Steinway S, Scheese D, Raouf Z, Tsuboi K, Ding J, Caputo C, McFarland M, Fulton WB, Wang S, Wang M, Prindle T, Gazit V, Rubin DC, Alaish S, Sodhi CP, Hackam DJ. Xenotransplanted human organoids identify transepithelial zinc transport as a key mediator of intestinal adaptation. Nat Commun 2024; 15:8613. [PMID: 39375337 PMCID: PMC11458589 DOI: 10.1038/s41467-024-52216-6] [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: 01/19/2024] [Accepted: 08/30/2024] [Indexed: 10/09/2024] Open
Abstract
Short bowel syndrome (SBS) leads to severe morbidity and mortality. Intestinal adaptation is crucial in improving outcomes. To understand the human gene pathways associated with adaptation, we perform single-cell transcriptomic analysis of human small intestinal organoids explanted from mice with experimental SBS. We show that transmembrane ion pathways, specifically the transepithelial zinc transport pathway genes SLC39A4 and SLC39A5, are upregulated in SBS. This discovery is corroborated by an external dataset, bulk RT-qPCR, and Western blots. Oral zinc supplementation is shown to improve survival and weight gain of SBS mice and increase the proliferation of intestinal crypt cells in vitro. Finally, we identify the upregulation of SLC39A5 and associated transcription factor KLF5 in biopsied intestinal tissue specimens from patients with SBS. Thus, we identify zinc supplementation as a potential therapy for SBS and describe a xenotransplantation model that provides a platform for discovery in other intestinal diseases.
Collapse
Affiliation(s)
- Maame Efua S Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Hannah Moore
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raheel Ahmad
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Johannes Duess
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Peng Lu
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla Lopez
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Steve Steinway
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Zachariah Raouf
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jeffrey Ding
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Connor Caputo
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Madison McFarland
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William B Fulton
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanxia Wang
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meghan Wang
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas Prindle
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vered Gazit
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Alaish
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
2
|
Krupp KT, Yaeger JDW, Ledesma LJ, Withanage MHH, Gale JJ, Howe CB, Allen TJ, Sathyanesan M, Newton SS, Summers CH. Single administration of a psychedelic [(R)-DOI] influences coping strategies to an escapable social stress. Neuropharmacology 2024; 252:109949. [PMID: 38636726 PMCID: PMC11073902 DOI: 10.1016/j.neuropharm.2024.109949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/08/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Psychedelic compounds have potentially rapid, long-lasting anxiolytic, antidepressive and anti-inflammatory effects. We investigated whether the psychedelic compound (R)-2,5-dimethoxy-4-iodoamphetamine [(R)-DOI], a selective 5-HT2A receptor partial agonist, decreases stress-related behavior in male mice exposed to repeated social aggression. Additionally, we explored the likelihood that these behavioral changes are related to anti-inflammatory properties of [(R)-DOI]. Animals were subjected to the Stress Alternatives Model (SAM), an escapable social stress paradigm in which animals develop reactive coping strategies - remaining in the SAM arena (Stay) with a social aggressor, or dynamically initiated stress coping strategies that involve utilizing the escape holes (Escape) to avoid aggression. Mice expressing these behavioral phenotypes display behaviors like those in other social aggression models that separate animals into stress-vulnerable (as for Stay) or stress-resilient (as for Escape) groups, which have been shown to have distinct inflammatory responses to social stress. These results show that Stay animals have heightened cytokine gene expression, and both Stay and Escape mice exhibit plasma and neural concentrations of the inflammatory cytokine tumor necrosis factor-α (TNFα) compared to unstressed control mice. Additionally, these results suggest that a single administration of (R)-DOI to Stay animals in low doses, can increase stress coping strategies such as increasing attention to the escape route, promoting escape behavior, and reducing freezing during socially aggressive interaction in the SAM. Lower single doses of (R)-DOI, in addition to shifting behavior to suggest anxiolytic effects, also concomitantly reduce plasma and limbic brain levels of the inflammatory cytokine TNFα.
Collapse
Affiliation(s)
- Kevin T Krupp
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Jazmine D W Yaeger
- Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, 57105, USA; Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Leighton J Ledesma
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, 57105, USA
| | | | - J J Gale
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Chase B Howe
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA
| | - Trevor J Allen
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Samuel S Newton
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Cliff H Summers
- Department of Biology, University of South Dakota, Vermillion, SD, 57069, USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD, 57105, USA.
| |
Collapse
|
3
|
Gopal R, Tutuncuoglu E, Bakalov V, Wasserloos K, Li H, Lemley D, DeVito LJ, Constantinesco NJ, Reed DS, McHugh KJ, Chinnappan B, Andreas AR, Maloy A, Bain D, Alcorn JF, Pitt BR, Kaynar AM. Zinc deficiency enhances sensitivity to influenza A associated bacterial pneumonia in mice. Physiol Rep 2024; 12:e15902. [PMID: 38163670 PMCID: PMC10758336 DOI: 10.14814/phy2.15902] [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: 06/27/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Although zinc deficiency (secondary to malnutrition) has long been considered an important contributor to morbidity and mortality of infectious disease (e.g. diarrhea disorders), epidemiologic data (including randomized controlled trials with supplemental zinc) for such a role in lower respiratory tract infection are somewhat ambiguous. In the current study, we provide the first preclinical evidence demonstrating that although diet-induced acute zinc deficiency (Zn-D: ~50% decrease) did not worsen infection induced by either influenza A (H1N1) or methicillin-resistant staph aureus (MRSA), Zn-D mice were sensitive to the injurious effects of superinfection of H1N1 with MRSA. Although the mechanism underlying the sensitivity of ZnD mice to combined H1N1/MRSA infection is unclear, it was noteworthy that this combination exacerbated lung injury as shown by lung epithelial injury markers (increased BAL protein) and decreased genes related to epithelial integrity in Zn-D mice (surfactant protein C and secretoglobins family 1A member 1). As bacterial pneumonia accounts for 25%-50% of morbidity and mortality from influenza A infection, zinc deficiency may be an important pathology component of respiratory tract infections.
Collapse
Affiliation(s)
- Radha Gopal
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Egemen Tutuncuoglu
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Veli Bakalov
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Medicine InstituteAllegheny Health NetworkPittsburghPennsylvaniaUSA
| | - Karla Wasserloos
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - HuiHua Li
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of PathologyUniversity of WisconsinMadisonWisconsinUSA
| | - David Lemley
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - Louis J. DeVito
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | | | - Douglas S. Reed
- Center for Vaccine ResearchUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kevin J. McHugh
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Baskaran Chinnappan
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Alexis R. Andreas
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Abigail Maloy
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Daniel Bain
- Department of Geology and Planetary ScienceUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John F. Alcorn
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Bruce R. Pitt
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ata Murat Kaynar
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Anesthesiology and Perioperative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
4
|
Prado Y, Aravena D, Gatica S, Llancalahuen FM, Aravena C, Gutiérrez-Vera C, Carreño LJ, Cabello-Verrugio C, Simon F. From genes to systems: The role of food supplementation in the regulation of sepsis-induced inflammation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166909. [PMID: 37805092 DOI: 10.1016/j.bbadis.2023.166909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Systemic inflammation includes a widespread immune response to a harmful stimulus that results in extensive systemic damage. One common example of systemic inflammation is sepsis, which is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Under the pro-inflammatory environment of sepsis, oxidative stress contributes to tissue damage due to dysfunctional microcirculation that progressively causes the failure of multiple organs that ultimately triggers death. To address the underlying inflammatory condition in critically ill patients, progress has been made to assess the beneficial effects of dietary supplements, which include polyphenols, amino acids, fatty acids, vitamins, and minerals that are recognized for their immuno-modulating, anticoagulating, and analgesic properties. Therefore, we aimed to review and discuss the contribution of food-derived supplementation in the regulation of inflammation from gene expression to physiological responses and summarize the precedented potential of current therapeutic approaches during systemic inflammation.
Collapse
Affiliation(s)
- Yolanda Prado
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Sebastian Gatica
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe M Llancalahuen
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cristobal Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Chile
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Chile
| | - Claudio Cabello-Verrugio
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile.
| |
Collapse
|
5
|
Knez M, Stangoulis JCR. Dietary Zn deficiency, the current situation and potential solutions. Nutr Res Rev 2023; 36:199-215. [PMID: 37062532 DOI: 10.1017/s0954422421000342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Zinc (Zn) deficiency is a worldwide problem, and this review presents an overview of the magnitude of Zn deficiency with a particular emphasis on present global challenges, current recommendations for Zn intake, and factors that affect dietary requirements. The challenges of monitoring Zn status are clarified together with the discussion of relevant Zn bioaccessibility and bioavailability issues. Modern lifestyle factors that may exacerbate Zn deficiency and new strategies of reducing its effects are presented. Biofortification, as a potentially useful strategy for improving Zn status in sensitive populations, is discussed. The review proposes potential actions that could deliver promising results both in terms of monitoring dietary and physiological Zn status as well as in alleviating dietary Zn deficiency in affected populations.
Collapse
Affiliation(s)
- Marija Knez
- College of Science and Engineering, Flinders University, GPO Box 2100, AdelaideSA5001, Australia
- Center of Research Excellence in Nutrition and Metabolism, University of Belgrade, Institute for Medical Research, National Institute of the Republic of Serbia, 11000Belgrade, Serbia
| | - James C R Stangoulis
- College of Science and Engineering, Flinders University, GPO Box 2100, AdelaideSA5001, Australia
| |
Collapse
|
6
|
Mairinger E, Wessolly M, Buderath P, Borchert S, Henrich L, Mach P, Steinborn J, Kimming R, Jasani B, Schmid KW, Bankfalvi A, Mairinger FD. Tumor cell cytoplasmic metallothionein expression associates with differential tumor immunogenicity and prognostic outcome in high-grade serous ovarian carcinoma. Front Oncol 2023; 13:1252700. [PMID: 38023247 PMCID: PMC10663300 DOI: 10.3389/fonc.2023.1252700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background The underlying mechanism of high T-cell presence as a favorable prognostic factor in high-grade serous ovarian carcinoma (HGSOC) is not yet understood. In addition to immune cells, various cofactors are essential for immune processes. One of those are metallothioneins (MTs), metal-binding proteins comprising various isoforms. MTs play a role in tumor development and drug resistance. Moreover, MTs influence inflammatory processes by regulating zinc homeostasis. In particular, T-cell function and polarization are particularly susceptible to changes in zinc status. The aim of the present study was to investigate a possible role of MT-mediated immune response and its association with prognostic outcome in ovarian cancer. Methods A retrospective study was conducted on a clinically well-characterized cohort of 24 patients with HGSOC treated at the University Hospital of Essen. Gene expression patterns for anti-cancer immunogenicity-related targets were performed using the NanoString nCounter platform for digital gene expression analysis with the appurtenant PanCancer Immune Profiling panel, consisting of 770 targets and 30 reference genes. Tumor-associated immunohistochemical MT protein expression was evaluated using a semi-quantitative four-tier Immunohistochemistry (IHC) scoring. Results MT immunoexpression was detected in 43% (10/23) of all HGSOC samples. MT immunoexpression levels showed a significant association to survival, leading to prolonged progression-free and overall survival in positively stained tumors. Furthermore, T-cell receptor signaling gene signature showed a strong activation in MT-positive tumors. Activated downstream signaling cascades resulting in elevated interferon-gamma expression with a shift in the balance between T helper cells (TH1 and TH2) could be observed in the MT-positive subgroup. In addition, a higher expression pattern of perforin and several granzymes could be detected, overall suggestive of acute, targeted anti-cancer immune response in MT-positive samples. Conclusion This is the first study combining broad, digital mRNA screening of anti-tumor immune response-associated genes and their relation to MT-I/II in ovarian cancer. MT overexpression is associated with molecular characteristics of an anti-cancer immune response and is a strong prognostic marker in ovarian HGSOC. The observed immune cell activation associated with tumor MT expression comprises but is not limited to T cells and natural killer cells.
Collapse
Affiliation(s)
- Elena Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Michael Wessolly
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Paul Buderath
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Sabrina Borchert
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Larissa Henrich
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Julia Steinborn
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Rainer Kimming
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Bharat Jasani
- Department of Pathology, Targos - A Discovery Life Sciences Company, Kassel, Germany
| | | | - Agnes Bankfalvi
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
7
|
Kijima K, Ono G, Kobayakawa K, Saiwai H, Hara M, Yoshizaki S, Yokota K, Saito T, Tamaru T, Iura H, Haruta Y, Kitade K, Utsunomiya T, Konno D, Edgerton VR, Liu CY, Sakai H, Maeda T, Kawaguchi K, Matsumoto Y, Okada S, Nakashima Y. Zinc deficiency impairs axonal regeneration and functional recovery after spinal cord injury by modulating macrophage polarization via NF-κB pathway. Front Immunol 2023; 14:1290100. [PMID: 38022538 PMCID: PMC10666775 DOI: 10.3389/fimmu.2023.1290100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating disease that results in permanent paralysis. Currently, there is no effective treatment for SCI, and it is important to identify factors that can provide therapeutic intervention during the course of the disease. Zinc, an essential trace element, has attracted attention as a regulator of inflammatory responses. In this study, we investigated the effect of zinc status on the SCI pathology and whether or not zinc could be a potential therapeutic target. Methods We created experimental mouse models with three different serum zinc concentration by changing the zinc content of the diet. After inducing contusion injury to the spinal cord of three mouse models, we assessed inflammation, apoptosis, demyelination, axonal regeneration, and the number of nuclear translocations of NF-κB in macrophages by using qPCR and immunostaining. In addition, macrophages in the injured spinal cord of these mouse models were isolated by flow cytometry, and their intracellular zinc concentration level and gene expression were examined. Functional recovery was assessed using the open field motor score, a foot print analysis, and a grid walk test. Statistical analysis was performed using Wilcoxon rank-sum test and ANOVA with the Tukey-Kramer test. Results In macrophages after SCI, zinc deficiency promoted nuclear translocation of NF-κB, polarization to pro-inflammatory like phenotype and expression of pro-inflammatory cytokines. The inflammatory response exacerbated by zinc deficiency led to worsening motor function by inducing more apoptosis of oligodendrocytes and demyelination and inhibiting axonal regeneration in the lesion site compared to the normal zinc condition. Furthermore, zinc supplementation after SCI attenuated these zinc-deficiency-induced series of responses and improved motor function. Conclusion We demonstrated that zinc affected axonal regeneration and motor functional recovery after SCI by negatively regulating NF-κB activity and the subsequent inflammatory response in macrophages. Our findings suggest that zinc supplementation after SCI may be a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Utsunomiya
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - V. Reggie Edgerton
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
- Institut Guttmann. Hospital de Neurorehabilitació, Institut Universitari adscrit a la Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Charles Y. Liu
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
| | - Hiroaki Sakai
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Fukushima Medical University, Fukushima, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Vatsalya V, Royer AJ, Jha SK, Parthasarathy R, Tiwari H, Feng W, Ramchandani VA, Kirpich IA, McClain CJ. Drinking and laboratory biomarkers, and nutritional status characterize the clinical presentation of early-stage alcohol-associated liver disease. Adv Clin Chem 2023; 114:83-108. [PMID: 37268335 DOI: 10.1016/bs.acc.2023.02.001] [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] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chronic and heavy alcohol consumption is commonly observed in alcohol use disorder (AUD). AUD often leads to alcohol-associated organ injury, including alcohol-associated liver disease (ALD). Approximately 10-20% of patients with AUD progress to ALD. Progression of ALD from the development phase to more advanced states involve the interplay of several pathways, including nutritional alterations. Multiple pathologic processes have been identified in the progression and severity of ALD. However, there are major gaps in the characterization and understanding of the clinical presentation of early-stage ALD as assessed by clinical markers and laboratory measures. Several Institutions and Universities, including the University of Louisville, in collaboration with the National Institutes of Health, have published a series of manuscripts describing early-stage ALD over the past decade. Here, we comprehensively describe early-stage ALD using the liver injury and drinking history markers, and the laboratory biomarkers (with a focus on nutrition status) that are uniquely involved in the development and progression of early-stage ALD.
Collapse
Affiliation(s)
- Vatsalya Vatsalya
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; National Institute on Alcohol Abuse and Alcoholism, NIAAA, NIH, Bethesda, MD, United States; Robley Rex VA Medical Center, Louisville, KY, United States.
| | - Amor J Royer
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Suman Kumar Jha
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Ranganathan Parthasarathy
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Harsh Tiwari
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Wenke Feng
- Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States
| | - Vijay A Ramchandani
- National Institute on Alcohol Abuse and Alcoholism, NIAAA, NIH, Bethesda, MD, United States
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States; Department of Microbiology and Immunology, University of Louisville, Louisville KY United States
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States; Robley Rex VA Medical Center, Louisville, KY, United States
| |
Collapse
|
9
|
Zhu L, An P, Zhao W, Xia Y, Qi J, Luo J, Luo Y. Low Zinc Alleviates the Progression of Thoracic Aortic Dissection by Inhibiting Inflammation. Nutrients 2023; 15:1640. [PMID: 37049478 PMCID: PMC10096567 DOI: 10.3390/nu15071640] [Citation(s) in RCA: 2] [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: 03/04/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Vascular inflammation triggers the development of thoracic aortic dissection (TAD). Zinc deficiency could dampen tissue inflammation. However, the role of zinc as a nutritional intervention in the progression of TAD remains elusive. In this study, we employed a classical β-aminopropionitrile monofumarate (BAPN)-induced TAD model in mice treated with low zinc and observed that the TAD progression was greatly ameliorated under low zinc conditions. Our results showed that low zinc could significantly improve aortic dissection and rupture (BAPN + low zinc vs. BAPN, 36% vs. 100%) and reduce mortality (BAPN + low zinc vs. BAPN, 22% vs. 57%). Mechanically, low zinc attenuated the infiltration of macrophages and inhibited the expression of inflammatory cytokines, suppressed the phenotype switch of vascular smooth muscle cells from contractile to synthetic types, and eventually alleviated the development of TAD. In conclusion, this study suggested that low zinc may serve as a potential nutritional intervention approach for TAD prevention.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Peng An
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Wenting Zhao
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yi Xia
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jingyi Qi
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Junjie Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yongting Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
10
|
Oner P, Er B, Orhan C, Sahin K. Combination of Phycocyanin, Zinc, and Selenium Improves Survival Rate and Inflammation in the Lipopolysaccharide-Galactosamine Mouse Model. Biol Trace Elem Res 2023; 201:1377-1387. [PMID: 36175742 DOI: 10.1007/s12011-022-03433-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/21/2022] [Indexed: 02/07/2023]
Abstract
Sepsis is related to systemic inflammation and oxidative stress, the primary causes of death in intensive care units. Severe functional abnormalities in numerous organs can arise due to sepsis, with acute lung damage being the most common and significant morbidity. Spirulina, blue-green algae with high protein, vitamins, phycocyanin, and antioxidant content, shows anti-inflammatory properties by decreasing the release of cytokines. In addition, zinc (Zn) and selenium (Se) act as an antioxidant by inhibiting the oxidation of macromolecules, as well as the inhibition of the inflammatory response. The current study aimed to examine the combined properties of Zn, Se, and phycocyanin oligopeptides (ZnSePO) against lipopolysaccharide-D-galactosamine (LPS-GalN)-induced septic lung injury through survival rate, inflammatory, and histopathological changes in Balb/c mice. A total of 30 mice were allocated into three groups: normal control, LPS-GalN (100 ng of LPS plus 8 mg of D-galactosamine), LPS-GalN + ZnSePO (ZnPic, 52.5 µg/mL; SeMet, 0.02 µg/mL; and phycocyanin oligopeptide (PO), 2.00 mg/mL; at 1 h before the injection of LPS-GalN). Lung tissue from mice revealed noticeable inflammatory reactions and typical interstitial fibrosis after the LPS-GalN challenge. LPS-GalN-induced increased mortality rate and levels of IL-1, IL-6, IL-10, TGF-β, TNF-α, and NF-κB in lung tissue. Moreover, treatment of septic mice LPS-GalN + ZnSePO reduced mortality rates and inflammatory responses. ZnSePO considerably influenced tissue cytokine levels, contributing to its capacity to minimize acute lung injury (ALI) and pulmonary inflammation and prevent pulmonary edema formation in LPS-GalN-injected mice. In conclusion, ZnSePO treatment enhanced the survival rate of endotoxemia mice via improving inflammation and oxidative stress, indicating a possible therapeutic effect for patients with septic infections.
Collapse
Affiliation(s)
- Pinar Oner
- Department of Microbiology, Fethi Sekin City Hospital, Elazig, Turkey
| | - Besir Er
- Division of Biology, Faculty of Science, Firat University, 23119, Elazig, Turkey
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, 23119, Elazig, Turkey
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, 23119, Elazig, Turkey.
| |
Collapse
|
11
|
Ballambattu VB, Gurugubelli KR. Neonatal sepsis: Recent advances in pathophysiology and management. VIRAL, PARASITIC, BACTERIAL, AND FUNGAL INFECTIONS 2023:503-513. [DOI: 10.1016/b978-0-323-85730-7.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Wang F, Zhong J, Zhang R, Sun Y, Dong Y, Wang M, Sun C. Zinc and COVID-19: Immunity, Susceptibility, Severity and Intervention. Crit Rev Food Sci Nutr 2022; 64:1969-1987. [PMID: 36094452 DOI: 10.1080/10408398.2022.2119932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic and continuing emergence of viral mutants, there has been a lack of effective treatment methods. Zinc maintains immune function, with direct and indirect antiviral activities. Zinc nutritional status is a critical factor in antiviral immune responses. Importantly, COVID-19 and zinc deficiency overlap in high-risk population. Hence, the potential effect of zinc as a preventive and adjunct therapy for COVID-19 is intriguing. Here, this review summarizes the immune and antiviral function of zinc, the relationship between zinc levels, susceptibility, and severity of COVID-19, and the effect of zinc supplementation on COVID-19. Existing studies have confirmed that zinc deficiency was associated with COVID-19 susceptibility and severity. Zinc supplementation plays a potentially protective role in enhancing immunity, decreasing susceptibility, shortening illness duration, and reducing the severity of COVID-19. We recommend that zinc levels should be monitored, particularly in COVID-19 patients, and zinc as a preventive and adjunct therapy for COVID-19 should be considered for groups at risk of zinc deficiency to reduce susceptibility and disease severity.
Collapse
Affiliation(s)
- Fan Wang
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, China
| | - Jiayi Zhong
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Rui Zhang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Yongzhi Sun
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Yingran Dong
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Maoqing Wang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Changhao Sun
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Rossi RE, Chen J, Caplin ME. The Role of Diet and Supplements in the Prevention and Progression of COVID-19: Current Knowledge and Open Issues. Prev Nutr Food Sci 2022; 27:137-149. [PMID: 35919576 PMCID: PMC9309075 DOI: 10.3746/pnf.2022.27.2.137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 01/08/2023] Open
Abstract
A healthy diet and dietary supplements have gained attention as potential co-adjuvants in managing and preventing coronavirus disease 2019 (COVID-19). This paper critically reviews the current evidence regarding the impact of diet and supplements on the prevention and progression of COVID-19. According to available data, a healthy diet and normal weight are considered protective factors. Regarding dietary supplementation, the most robust results from human studies are for vitamin C, which appears to decrease inflammatory markers and suppress cytokine storm. A small, randomized trial showed that a high dose of vitamin D significantly reduced the need for intensive care unit treatment of patients requiring hospitalization for COVID-19. According to retrospective human studies, there is limited evidence for vitamin E and selenium supplements. Animal studies have investigated the effects of green tea and curcumin. Xanthohumol and probiotics, interesting for their antiviral, anti-inflammatory, and immunoregulatory properties, need formal clinical study. In summary, there is promising evidence supporting the role of diet and supplements as co-adjuvants in the treatment of COVID-19. Further studies and properly designed clinical trials are necessary to draw more robust conclusions; however, it is not unreasonable to take a pragmatic approach and promote the use of appropriate diet and supplements to counter the effects of COVID-19, ideally with a mechanism to assess outcomes.
Collapse
Affiliation(s)
- Roberta Elisa Rossi
- Hepatology and Hepato-Pancreatic-Biliary Surgery and Liver Transplantation, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, MI 20133, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, MI 20122, Italy
| | - Jie Chen
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510275, China
| | - Martyn Evan Caplin
- Centre for Gastroenterology, Royal Free Hospital, London NW3 2QG, UK
- Division of Medicine, Faculty of Medical Sciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
14
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
15
|
Xia W, Li C, Zhao D, Xu L, Kuang M, Yao X, Hu H. The Impact of Zinc Supplementation on Critically Ill Patients With Acute Kidney Injury: A Propensity Score Matching Analysis. Front Nutr 2022; 9:894572. [PMID: 35769374 PMCID: PMC9234667 DOI: 10.3389/fnut.2022.894572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/17/2022] [Indexed: 12/05/2022] Open
Abstract
Background Zinc is an essential trace element involved in multiple metabolic processes. Acute kidney injury (AKI) is associated with low plasma zinc, but outcomes with zinc supplementation in critically ill patients with AKI remain unknown. Our objective was to investigate the effectiveness of zinc supplementation in this patient population. Methods Critically ill patients with AKI were identified from the Medical Informative Mart for Intensive Care IV database. Prosperity score matching (PSM) was applied to match patients receiving zinc treatment to those without zinc treatment. The association between zinc sulfate use and in-hospital mortality and 30-day mortality, need for renal replacement therapy (RRT), and length of stay was determined by logistic regression and Cox proportional hazards modeling. Results A total of 9,811 AKI patients were included in the study. PSM yielded 222 pairs of patients who received zinc treatment and those who did not. Zinc supplementation was associated with reduced in-hospital mortality (HR = 0.48 (95% CI: 0.28, 0.83) P = 0.009) and 30-day mortality (HR = 0.51 (95% CI, 0.30, 0.86) P = 0.012). In the subgroup analysis, zinc use was associated with reduced in-hospital mortality in patients with stage 1 AKI and those with sepsis. Conclusions Zinc supplementation was associated with improved survival in critically ill patients with AKI. The supplementation was especially effective in those with stage 1 AKI and sepsis. These results need to be verified in randomized controlled trials.
Collapse
Affiliation(s)
- Wenkai Xia
- Department of Nephrology, The Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Chenyu Li
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Danyang Zhao
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lingyu Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Meisi Kuang
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Xiajuan Yao
- Department of Nephrology, The Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China
| | - Hong Hu
- Department of Nephrology, The Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China
- *Correspondence: Hong Hu
| |
Collapse
|
16
|
Liu X, Ali MK, Dua K, Xu R. The Role of Zinc in the Pathogenesis of Lung Disease. Nutrients 2022; 14:nu14102115. [PMID: 35631256 PMCID: PMC9143957 DOI: 10.3390/nu14102115] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 12/26/2022] Open
Abstract
Lung diseases, such as asthma, chronic obstructive pulmonary diseases (COPD), and cystic fibrosis (CF), are among the leading causes of mortality and morbidity globally. They contribute to substantial economic burdens on society and individuals. Currently, only a few treatments are available to slow the development and progression of these diseases. Thus, there is an urgent unmet need to develop effective therapies to improve quality of life and limit healthcare costs. An increasing body of clinical and experimental evidence suggests that altered zinc and its regulatory protein levels in the systemic circulation and in the lungs are associated with these disease’s development and progression. Zinc plays a crucial role in human enzyme activity, making it an essential trace element. As a cofactor in metalloenzymes and metalloproteins, zinc involves a wide range of biological processes, such as gene transcription, translation, phagocytosis, and immunoglobulin and cytokine production in both health and disease. Zinc has gained considerable interest in these lung diseases because of its anti-inflammatory, antioxidant, immune, and metabolic modulatory properties. Here we highlight the role and mechanisms of zinc in the pathogenesis of asthma, COPD, CF, acute respiratory distress syndrome, idiopathic pulmonary fibrosis, and pulmonary hypertension.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China;
| | - Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA;
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA 94305, USA
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia;
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | - Ran Xu
- Department of Thoracic Surgery, Shengjing Hospital, China Medical University, Shenyang 110022, China
- Correspondence: ; Tel.: +86-189-4025-8514
| |
Collapse
|
17
|
Liang J, Huang G, Liu X, Taghavifar F, Liu N, Wang Y, Deng N, Yao C, Xie T, Kulur V, Dai K, Burman A, Rowan SC, Weigt SS, Belperio J, Stripp B, Parks WC, Jiang D, Noble PW. The ZIP8/SIRT1 axis regulates alveolar progenitor cell renewal in aging and idiopathic pulmonary fibrosis. J Clin Invest 2022; 132:157338. [PMID: 35389887 PMCID: PMC9151700 DOI: 10.1172/jci157338] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/05/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractType 2 alveolar epithelial cells (AEC2s) function as progenitor cells in the lung. We have shown previously that failure of AEC2 regeneration results in progressive lung fibrosis in mice and is a cardinal feature of idiopathic pulmonary fibrosis (IPF). In this study, we identified a deficiency of a specific zinc transporter SLC39A8 (ZIP8) in AEC2s from both IPF lungs and lungs of old mice. Loss of ZIP8 expression was associated with impaired renewal capacity of AEC2s and enhanced lung fibrosis. ZIP8 regulation of AEC2 progenitor function was dependent on SIRT1. Replenishment with exogenous zinc and SIRT1 activation promoted self-renewal and differentiation of AEC2s from lung tissues of IPF patients and old mice. Deletion of Zip8 in AEC2s in mice impaired AEC2 renewal, increased susceptibility of the mice to bleomycin injury, and the mice developed spontaneous lung fibrosis. Therapeutic strategies to restore zinc metabolism and appropriate SIRT1 signaling could improve AEC2 progenitor function and mitigate ongoing fibrogenesis.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Guanling Huang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Xue Liu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Forough Taghavifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Ningshan Liu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Yizhou Wang
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Nan Deng
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Changfu Yao
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Ting Xie
- Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Vrishika Kulur
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Kristy Dai
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Ankita Burman
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Simon C Rowan
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - S Samuel Weigt
- Department of Medicine, UCLA, Los Angeles, United States of America
| | - John Belperio
- Department of Medicine, UCLA, Los Angeles, United States of America
| | - Barry Stripp
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Dianhua Jiang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Paul W Noble
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, United States of America
| |
Collapse
|
18
|
Wessels I, Rolles B, Slusarenko AJ, Rink L. Zinc deficiency as a possible risk factor for increased susceptibility and severe progression of Corona Virus Disease 19. Br J Nutr 2022; 127:214-232. [PMID: 33641685 PMCID: PMC8047403 DOI: 10.1017/s0007114521000738] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/07/2021] [Accepted: 02/21/2021] [Indexed: 01/08/2023]
Abstract
The importance of Zn for human health becomes obvious during Zn deficiency. Even mild insufficiencies of Zn cause alterations in haematopoiesis and immune functions, resulting in a proinflammatory phenotype and a disturbed redox metabolism. Although immune system malfunction has the most obvious effect, the functions of several tissue cell types are disturbed if Zn supply is limiting. Adhesion molecules and tight junction proteins decrease, while cell death increases, generating barrier dysfunction and possibly organ failure. Taken together, Zn deficiency both weakens the resistance of the human body towards pathogens and at the same time increases the danger of an overactive immune response that may cause tissue damage. The case numbers of Corona Virus Disease 19 (COVID-19) are still increasing, which is causing enormous problems for health systems and economies. There is an urgent need to reduce both the number of severe cases and the resulting deaths. While therapeutic options are still under investigation, and first vaccines have been approved, cost-effective ways to reduce the likelihood of or even prevent infection, and the transition from mild symptoms to more serious detrimental disease, are highly desirable. Nutritional supplementation might be an effective option to achieve these aims. In this review, we discuss known Zn deficiency effects in the context of an infection with Severe Acute Respiratory Syndrome-Coronavirus-2 and its currently known pathogenic mechanisms and elaborate on how severe pre-existing Zn deficiency may pre-dispose patients to a severe progression of COVID-19. First published clinical data on the association of Zn homoeostasis with COVID-19 and registered studies in progress are listed.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074Aachen, Germany
| | - Benjamin Rolles
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074Aachen, Germany
| | - Alan J. Slusarenko
- Department of Plant Physiology, RWTH Aachen University, Worringer Weg 1, 52074Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074Aachen, Germany
| |
Collapse
|
19
|
Ho E, Wong CP, King JC. Impact of zinc on DNA integrity and age-related inflammation. Free Radic Biol Med 2022; 178:391-397. [PMID: 34921929 DOI: 10.1016/j.freeradbiomed.2021.12.256] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/22/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023]
Abstract
Dr. Bruce Ames was a pioneer in understanding the role of oxidative stress and DNA damage, and in the 1990s began to make connections between micronutrient deficiencies and DNA damage. Zinc is an essential micronutrient for human health and a key component for the function of numerous cellular processes. In particular, zinc plays a critical role in cellular antioxidant defense, the maintenance of DNA integrity and is also essential for the normal development and function of the immune system. This review highlights the work helping connect zinc deficiency to oxidative stress, susceptibility to DNA damage and chronic inflammation that was initiated while working with Dr. Ames. This review outlines the body of work in this area, from cells to humans. The article also reviews the unique challenges of maintaining zinc status as we age and the interplay between zinc deficiency and age-related inflammation and immune dysfunction. Several micronutrient deficiencies, including zinc deficiency, can drastically affect the risk of many chronic diseases and underscores the importance of adequate nutrition for healthy aging.
Collapse
Affiliation(s)
- Emily Ho
- Linus Pauling Institute, 307 Linus Pauling Science Center, Oregon State University, Corvallis, OR, 97331, USA; School of Biological and Population Health Sciences, 101 Milam Hall, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, 97331, USA.
| | - Carmen P Wong
- Linus Pauling Institute, 307 Linus Pauling Science Center, Oregon State University, Corvallis, OR, 97331, USA; School of Biological and Population Health Sciences, 101 Milam Hall, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Janet C King
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| |
Collapse
|
20
|
Peng-Winkler Y, Büttgenbach A, Rink L, Weßels I. Zinc supplementation prior to heat shock enhances HSP70 synthesis through HSF1 phosphorylation at serine 326 in human peripheral mononuclear cells. Food Funct 2022; 13:9143-9152. [DOI: 10.1039/d2fo01406h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Zinc supplementation prior to heat shock increases HSP70 (Heat shock protein 70) expression, which has cytoprotective effects in tissue cells during inflammation. Effects of zinc deficiency in this regard are...
Collapse
|
21
|
Verschelden G, Noeparast M, Noparast M, Goossens MC, Lauwers M, Cotton F, Michel C, Goyvaerts C, Hites M. Plasma zinc status and hyperinflammatory syndrome in hospitalized COVID-19 patients: An observational study. Int Immunopharmacol 2021; 100:108163. [PMID: 34583122 PMCID: PMC8450071 DOI: 10.1016/j.intimp.2021.108163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Zinc deficiency is associated with impaired antiviral response, cytokine releasing syndrome (CRS), and acute respiratory distress syndrome. Notably, similar complications are being observed during severe SARS-CoV-2 infection. We conducted a prospective, single-center, observational study in a tertiary university hospital (CUB-Hôpital Erasme, Brussels) to address the zinc status, the association between the plasma zinc concentration, development of CRS, and the clinical outcomes in PCR-confirmed and hospitalized COVID-19 patients. One hundred and thirty-nine eligible patients were included between May 2020 and November 2020 (median age of 65 years [IQR = 54, 77]). Our cohort's median plasma zinc concentration was 57 µg/dL (interquartile range [IQR] = 45, 67) compared to 74 µg/dL (IQR = 64, 84) in the retrospective non-COVID-19 control group (N = 1513; p < 0.001). Markedly, the absolute majority of COVID-19 patients (96%) were zinc deficient (<80 µg/dL). The median zinc concentration was lower in patients with CRS compared to those without CRS (-5 µg/dL; 95% CI = -10.5, 0.051; p = 0.048). Among the tested outcomes, zinc concentration is significantly correlated with only the length of hospital stay (rho = -0.19; p = 0.022), but not with mortality or morbidity. As such, our findings do not support the role of zinc as a robust prognostic marker among hospitalized COVID-19 patients who in our cohort presented a high prevalence of zinc deficiency. It might be more beneficial to explore the role of zinc as a biomarker for assessing the risk of developing a tissue-damaging CRS and predicting outcomes in patients diagnosed with COVID-19 at the early stage of the disease.
Collapse
Affiliation(s)
- Gil Verschelden
- Clinic of Infectious Diseases, Cliniques Universitaires de Bruxelles (CUB), Erasme Hospital, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.
| | - Maxim Noeparast
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043 Marburg, Germany; Fonds Wetenschappelijk Onderzoek (FWO) - Vlaanderen, Belgium.
| | - Maryam Noparast
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Maïlis Lauwers
- Department of Clinical Chemistry, Laboratoire Hospitalier Universitaire de Bruxelles (LHUB-ULB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frédéric Cotton
- Department of Clinical Chemistry, Laboratoire Hospitalier Universitaire de Bruxelles (LHUB-ULB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Charlotte Michel
- Department of Microbiology, Laboratoire Hospitalier Universitaire de Bruxelles (LHUB-ULB), Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Maya Hites
- Clinic of Infectious Diseases, Cliniques Universitaires de Bruxelles (CUB), Erasme Hospital, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
22
|
Role of biometals in activation of immune cum inflammatory response in ovine ageing eye: a potential model for understanding human geriatric eye diseases. Biometals 2021; 34:1081-1098. [PMID: 34297243 DOI: 10.1007/s10534-021-00331-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 07/05/2021] [Indexed: 10/25/2022]
Abstract
The present study was designed to evaluate the age-related changes in biometal and antimicrobial peptide (cathelicidin) concentration and their role in oxidative cum pro-inflammatory cascade in an ovine animal model. Clinically healthy ovine (n = 126) were grouped as Group I (n = 55, age = up to 3 years), Group II (n = 52, age = above 3-below 6 years) and Group III (n = 19, age = 6 years above). Samples (aqueous humour and lens of the eye) were collected stored at - 80 °C till further analysis. In aqueous humour, the concentration of zinc (p < 0.001 in group III), copper (p < 0.05 in group II and p < 0.001 group III) and iron (p < 0.05 in group III) were significantly increased compared to group I. While as the concentration of magnesium were significantly decreased in group II (p < 0.001) and group III (p < 0.05) compared to group I. Similarly in eye lens the level of copper remained uniform as no significant change was observed across different age groups, while as significantly elevated levels of iron were observed in group III (p < 0.001) compared to group I. whereas, levels of lens Zinc (p < 0.05 in group II) and magnesium (p < 0.05 in group III and p < 0.001 in group II) were significantly decreased compared to group I. Age-dependent increase in levels of oxidation products which include advanced oxidation protein products (AOPP) in aqueous humour and lenses of group II and group III (p < 0.001) and MDA in aqueous humour of group III (p < 0.05) were found compared to levels recorded in group I. In contrast, levels of antioxidants which include lens vitamin C in group II and group III (p < 0.01) and lens superoxide dismutase (SOD) in group III (p < 0.001) were significantly increased compared to group I. Levels of pro-inflammatory cytokines in aqueous humour revealed significantly (p < 0.001) age-dependent increase in IL-1, IL-6 and TNF-α elevated in group III, and group II as compared to group I, However, cathelicidin level in aqueous humour of group III and group II were significantly (p < 0.001) lower as compared to groups I. Furthermore,the present study observed significant (p < 0.05) metal-metal positive interaction between copper levels in lens with levels of (iron and magnesium) in aqueous humour, levels of Zn in lens with levels of Zn in aqueous humour, levels of Mg in lens with levels of (Cu, Zn and Mg) in aqueous humour. In addition,the present study reports significantly negative interaction between levels of lens Fe with levels of lens magnesium level, aqueous humour magnesium level and levels of copper in aqueous humour. A significantly positive correlation was observed between oxidative markers and pro-inflammatory cytokine levels, while a significant negative correlation was observed between antioxidant defence markers and pro-inflammatory cytokine. These results suggest the essential role of age-related changes in biometal levels, oxidative stress and pro-inflammatory cytokines. These changes might help understand age-related changes in pathogenesis and effective targeting of pathogenetic pathways in ocular diseases.
Collapse
|
23
|
Abstract
Evidence for the importance of zinc for all immune cells and for mounting an efficient and balanced immune response to various environmental stressors has been accumulating in recent years. This article describes the role of zinc in fundamental biological processes and summarizes our current knowledge of zinc's effect on hematopoiesis, including differentiation into immune cell subtypes. In addition, the important role of zinc during activation and function of immune cells is detailed and associated with the specific immune responses to bacteria, parasites, and viruses. The association of zinc with autoimmune reactions and cancers as diseases with increased or decreased immune responses is also discussed. This article provides a broad overview of the manifold roles that zinc, or its deficiency, plays in physiology and during various diseases. Consequently, we discuss why zinc supplementation should be considered, especially for people at risk of deficiency. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany;
| | | | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
24
|
Multinutrient Biofortification of Maize ( Zea mays L.) in Africa: Current Status, Opportunities and Limitations. Nutrients 2021; 13:nu13031039. [PMID: 33807073 PMCID: PMC8004732 DOI: 10.3390/nu13031039] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/21/2022] Open
Abstract
Macro and micronutrient deficiencies pose serious health challenges globally, with the largest impact in developing regions such as subSaharan Africa (SSA), Latin America and South Asia. Maize is a good source of calories but contains low concentrations of essential nutrients. Major limiting nutrients in maize-based diets are essential amino acids such as lysine and tryptophan, and micronutrients such as vitamin A, zinc (Zn) and iron (Fe). Responding to these challenges, separate maize biofortification programs have been designed worldwide, resulting in several cultivars with high levels of provitamin A, lysine, tryptophan, Zn and Fe being commercialized. This strategy of developing single-nutrient biofortified cultivars does not address the nutrient deficiency challenges in SSA in an integrated manner. Hence, development of maize with multinutritional attributes can be a sustainable and cost-effective strategy for addressing the problem of nutrient deficiencies in SSA. This review provides a synopsis of the health challenges associated with Zn, provitamin A and tryptophan deficiencies and link these to vulnerable societies; a synthesis of past and present intervention measures for addressing nutrient deficiencies in SSA; and a discussion on the possibility of developing maize with multinutritional quality attributes, but also with adaptation to stress conditions in SSA.
Collapse
|
25
|
Oyagbemi AA, Ajibade TO, Aboua YG, Gbadamosi IT, Adedapo ADA, Aro AO, Adejumobi OA, Thamahane-Katengua E, Omobowale TO, Falayi OO, Oyagbemi TO, Ogunpolu BS, Hassan FO, Ogunmiluyi IO, Ola-Davies OE, Saba AB, Adedapo AA, Nkadimeng SM, McGaw LJ, Kayoka-Kabongo PN, Oguntibeju OO, Yakubu MA. Potential health benefits of zinc supplementation for the management of COVID-19 pandemic. J Food Biochem 2021; 45:e13604. [PMID: 33458853 PMCID: PMC7995057 DOI: 10.1111/jfbc.13604] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/05/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent for the Coronavirus Disease 2019 (COVID-19). The COVID-19 pandemic has created unimaginable and unprecedented global health crisis. Since the outbreak of COVID-19, millions of dollars have been spent, hospitalization overstretched with increasing morbidity and mortality. All these have resulted in unprecedented global economic catastrophe. Several drugs and vaccines are currently being evaluated, tested, and administered in the frantic efforts to stem the dire consequences of COVID-19 with varying degrees of successes. Zinc possesses potential health benefits against COVID-19 pandemic by improving immune response, minimizing infection and inflammation, preventing lung injury, inhibiting viral replication through the interference of the viral genome transcription, protein translation, attachment, and host infectivity. However, this review focuses on the various mechanisms of action of zinc and its supplementation as adjuvant for vaccines an effective therapeutic regimen in the management of the ravaging COVID-19 pandemic. PRACTICAL APPLICATIONS: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent for the Coronavirus Disease 2019 (COVID-19), has brought unprecedented untold hardship to both developing and developed countries. The global race for vaccine development against COVID-19 continues with success in sight with attendant increasing hospitalization, morbidity, and mortality. Available drugs with anti-inflammatory actions have become alternative to stem the tide of COVID-19 with attendant global financial crises. However, Zinc is known to modulate several physiological functions including intracellular signaling, enzyme function, gustation, and olfaction, as well as reproductive, skeletal, neuronal, and cardiovascular systems. Hence, achieving a significant therapeutic approach against COVID-19 could imply the use of zinc as a supplement together with available drugs and vaccines waiting for emergency authorization to win the battle of COVID-19. Together, it becomes innovative and creative to supplement zinc with currently available drugs and vaccines.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Yapo Guillaume Aboua
- Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek, Namibia
| | | | | | - Abimbola Obemisola Aro
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Pretoria, South Africa
| | - Olumuyiwa Abiola Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Emma Thamahane-Katengua
- Department of Health Information Management, Faculty of Health and Education, Botho University, Gaborone, Botswana
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Olubunmi Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Taiwo Olaide Oyagbemi
- Department of Veterinary Parasitology and Entomology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Blessing Seun Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Fasilat Oluwakemi Hassan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Iyanuoluwa Omolola Ogunmiluyi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Sanah Malomile Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Science, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - Lyndy Joy McGaw
- Phytomedicine Programme, Department of Paraclinical Science, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - Prudence Ngalula Kayoka-Kabongo
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Pretoria, South Africa
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Oxidative Stress Research Centre, Cape Peninsula University of Technology, Bellville, South Africa
| | - Momoh Audu Yakubu
- Vascular Biology Unit, Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Center for Cardiovascular Diseases, Texas Southern University, Houston, TX, USA
| |
Collapse
|
26
|
Su SY, Tang QQ. Altered intestinal microflora and barrier injury in severe acute pancreatitis can be changed by zinc. Int J Med Sci 2021; 18:3050-3058. [PMID: 34400875 PMCID: PMC8364456 DOI: 10.7150/ijms.45980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
To investigate the effect of zinc (Zn) supplementation on intestinal microflora changes and bacterial translocation in rats with severe acute pancreatitis (SAP), the rats were divided into the sham surgery (SS), SAP, SS + Zn, and SAP + Zn groups. Saline (0.1 mL/100g) and 5% sodium taurocholate were injected into the pancreaticobiliary duct of the rats in the SS and SAP + Zn groups, respectively. Intraperitoneal injection of 5 mg/kg Zn was performed immediately after injecting saline or 5% sodium taurocholate into the rats in both groups. Serum amylase and Zn levels, plasma endogenous endotoxin, intestinal permeability, and the positive rate of intestinal bacterial translocation were detected, haematoxylin and eosin (H&E) staining was performed, and the pancreatic tissue scores were calculated for each group. In addition, immunohistochemical (IHC) staining was performed to evaluate the expression of IL-1β and TNF-α. Real-time fluorescence quantitative PCR was used to quantify the gene copy numbers of Escherichia, Bifidobacterium, and Lactobacillus in the cecum. The levels of amylase and plasma endotoxin in the SAP group were significantly higher than those in the SS and SS + Zn groups. Intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were increased in the SAP group. However, the levels of amylase and plasma endotoxin were decreased as a result of zinc supplementation in the SAP group. The expression of IL-1β and TNF-α was also reduced to a greater degree in the SAP + Zn group than in the SAP group. Moreover, alleviated intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were found in the SAP + Zn group. The results of real-time quantitative PCR showed that the gene copy number of Escherichia increased with time, and the gene copy numbers of Lactobacillus and Bifidobacterium decreased over time. Zn supplementation prevented the release of TNF-α and IL-1β, alleviated intestinal permeability and endotoxemia, reduced bacterial translocation, and inhibited changes in pathogenic intestinal flora in rats with SAP.
Collapse
Affiliation(s)
- Shi-Yue Su
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| | - Qin-Qing Tang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| |
Collapse
|
27
|
Kuźmicka W, Manda-Handzlik A, Cieloch A, Mroczek A, Demkow U, Wachowska M, Ciepiela O. Zinc Supplementation Modulates NETs Release and Neutrophils' Degranulation. Nutrients 2020; 13:nu13010051. [PMID: 33375275 PMCID: PMC7823768 DOI: 10.3390/nu13010051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Zinc plays an important physiological role in the entire body, especially in the immune system. It is one of the most abundant microelements in our organism and an essential component of enzymes and antibacterial proteins. Zinc levels were reported to be correlated with the intensity of innate immunity responses, especially those triggered by neutrophils. However, as the results are fragmentary, the phenomenon is still not fully understood and requires further research. In this study, we aimed to perform a comprehensive assessment and study the impact of zinc on several basic neutrophils’ functions in various experimental setups. Human and murine neutrophils were preincubated in vitro with zinc, and then phagocytosis, oxidative burst, degranulation and release of neutrophil extracellular traps (NETs) were analyzed. Moreover, a murine model of zinc deficiency and zinc supplementation was introduced in the study and the functions of isolated cells were thoroughly studied. We showed that zinc inhibits NETs release as well as degranulation in both human and murine neutrophils. Our study revealed that zinc decreases NETs release by inhibiting citrullination of histone H3. On the other hand, studies performed in zinc-deficient mice demonstrated that low zinc levels result in increased release of NETs and enhanced neutrophils degranulation. Overall, it was shown that zinc affects neutrophils’ functions in vivo and in vitro. Proper zinc level is necessary to maintain efficient functioning of the innate immune response.
Collapse
Affiliation(s)
- Weronika Kuźmicka
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091 Warsaw, Poland;
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
| | - Aneta Manda-Handzlik
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
| | - Adrianna Cieloch
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 63 Street, 02-091 Warsaw, Poland
| | - Agnieszka Mroczek
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
| | - Małgorzata Wachowska
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091 Warsaw, Poland; (A.M.-H.); (A.M.); (U.D.); (A.C.)
- Correspondence: (M.W.); (O.C.); Tel.: +48-223179503 (M.W.); +48-225992405 (O.C.)
| | - Olga Ciepiela
- Department of Laboratory Medicine, Medical University of Warsaw, Banacha 1a Street, 02-097 Warsaw, Poland
- Correspondence: (M.W.); (O.C.); Tel.: +48-223179503 (M.W.); +48-225992405 (O.C.)
| |
Collapse
|
28
|
Wessels I, Fischer HJ, Rink L. Update on the multi-layered levels of zinc-mediated immune regulation. Semin Cell Dev Biol 2020; 115:62-69. [PMID: 33323322 DOI: 10.1016/j.semcdb.2020.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 11/15/2022]
Abstract
The significance of zinc for an efficient immune response is well accepted. During zinc deficiency, an increase in the myeloid to lymphoid immune cells ratio was observed. This results in a disturbed balance of pro- and anti-inflammatory processes as well as defects in tolerance during infections. Consequently, instead of efficiently defending the body against invading pathogens, damage of host cells is frequently observed. This explains the increased susceptibility to infections and their severe progression observed for zinc deficient individuals as well as the association of autoimmune diseases with low serum zinc levels. Together with the advances in techniques for investigating cellular development, communication and intracellular metabolism, our understanding of the mechanisms underlying the benefits of zinc for human health and the detriments of zinc deficiency has much improved. As analyses of the zinc status and effects of zinc supplementation were more frequently included into clinical studies, our knowledge of the association of zinc deficiency to a variety of diseases was strongly improved. Still there are several areas in zinc biology that require further in-depth investigation such as the interaction with other nutritional elements, the direct association between zinc transportation, membrane-structure, receptors, and signaling as well as its role in cell degeneration. This article will describe our current understanding of the role of zinc during the immune response focusing on the most recent findings and underlying mechanisms. Research questions that need to be addressed in the future will be discussed as well.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Henrike J Fischer
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany.
| |
Collapse
|
29
|
von Pein JB, Stocks CJ, Schembri MA, Kapetanovic R, Sweet MJ. An alloy of zinc and innate immunity: Galvanising host defence against infection. Cell Microbiol 2020; 23:e13268. [PMID: 32975847 DOI: 10.1111/cmi.13268] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Innate immune cells such as macrophages and neutrophils initiate protective inflammatory responses and engage antimicrobial responses to provide frontline defence against invading pathogens. These cells can both restrict the availability of certain transition metals that are essential for microbial growth and direct toxic concentrations of metals towards pathogens as antimicrobial responses. Zinc is important for the structure and function of many proteins, however excess zinc can be cytotoxic. In recent years, several studies have revealed that innate immune cells can deliver toxic concentrations of zinc to intracellular pathogens. In this review, we discuss the importance of zinc status during infectious disease and the evidence for zinc intoxication as an innate immune antimicrobial response. Evidence for pathogen subversion of this response is also examined. The likely mechanisms, including the involvement of specific zinc transporters that facilitate delivery of zinc by innate immune cells for metal ion poisoning of pathogens are also considered. Precise mechanisms by which excess levels of zinc can be toxic to microorganisms are then discussed, particularly in the context of synergy with other antimicrobial responses. Finally, we highlight key unanswered questions in this emerging field, which may offer new opportunities for exploiting innate immune responses for anti-infective development.
Collapse
Affiliation(s)
- Jessica B von Pein
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Claudia J Stocks
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Mark A Schembri
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, St. Lucia, Queensland, Australia.,IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
30
|
Wang M, Phadke M, Packard D, Yadav D, Gorelick F. Zinc: Roles in pancreatic physiology and disease. Pancreatology 2020; 20:1413-1420. [PMID: 32917512 PMCID: PMC7572834 DOI: 10.1016/j.pan.2020.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Zinc is an essential trace element. Deficiencies are frequently seen with gastrointestinal diseases, including chronic pancreatitis, nutritional deficiency, and reduced intestinal absorption. Additionally, reduced zinc levels have been linked to cellular changes associated with acute pancreatitis such as enhanced inflammation with increased macrophage activation and production of inflammatory cytokines such as IL-1β, impaired autophagy, and modulation of calcium homeostasis. Preliminary data suggest that zinc deficiency may lead to pancreatic injury in animal models. The purpose of this review is to explore the biologic effects of zinc deficiency that could impact pancreatic disease. MESH KEYWORDS: Malnutrition, inflammation, trace element.
Collapse
Affiliation(s)
- Melinda Wang
- Yale School of Medicine, Department of Internal Medicine and VA HealthCare System, CT, USA
| | - Madhura Phadke
- Yale School of Medicine, Department of Internal Medicine and VA HealthCare System, CT, USA
| | - Daniel Packard
- Yale School of Medicine, Department of Internal Medicine and VA HealthCare System, CT, USA
| | - Dhiraj Yadav
- University of Pittsburgh, Department of Medicine, USA
| | - Fred Gorelick
- Yale School of Medicine, Department of Internal Medicine and VA HealthCare System, CT, USA.
| |
Collapse
|
31
|
Abstract
Background and objectives: Sepsis is defined as a life-threatening organ dysfunction syndrome, which occurs when the body's immune response to infection is impaired. The aim of the present study was to investigate serum Iron, Copper, Zinco, Cobalt, Chromium, Selenium, Vanadium, Nickel, Cadmium, and Aliminium levels in patients with sepsis.Materials and methods: This prospective and observational study was conducted at a tertiary care university hospital of Turkey from 2015 to 2016, and comprised patients with sepsis. Serum concentrations of 10 elements were analyzed using inductively coupled plasma mass spectrometry. Analyses were performed at the laboratory of Düzce University Scientific and Technological Research Application and Research Center. A total of 87 participants (52 men, 35 women; average age, 74.11 ± 14.26) were enrolled.Results: When evaluated in terms of trace elements, a significant difference was noted between the sepsis and control groups in terms of the levels of the five elements. Chromium, Iron, Nickel, Copper, and Cadmium levels were significantly higher in the sepsis group.Conclusion: Our study indicated in particular, Iron, Copper, Chromium, Nickel, and Cadmium levels were elevated in patients with sepsis.
Collapse
Affiliation(s)
- İdris Akkaş
- Department of Infectious Diseases, Oltu State Hospital, Ministry of Health, Erzurum, Turkey
| | - Nevin Ince
- Department of Infectious Diseases and Clinical Microbiology, Duzce University Faculty of Medicine, Duzce, Turkey
| | - Mehmet Ali Sungur
- Department of Biostatistics, Düzce University Faculty of Medicine, Düzce, Turkey
| |
Collapse
|
32
|
Wu J, Meng QH. Current understanding of the metabolism of micronutrients in chronic alcoholic liver disease. World J Gastroenterol 2020; 26:4567-4578. [PMID: 32884217 PMCID: PMC7445863 DOI: 10.3748/wjg.v26.i31.4567] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) remains an important health problem worldwide. Perturbation of micronutrients has been broadly reported to be a common characteristic in patients with ALD, given the fact that micronutrients often act as composition or coenzymes of many biochemical enzymes responsible for the inflammatory response, oxidative stress, and cell proliferation. Mapping the metabolic pattern and the function of these micronutrients is a prerequisite before targeted intervention can be delivered in clinical practice. Recent years have registered a significant improvement in our understanding of the role of micronutrients on the pathogenesis and progression of ALD. However, how and to what extent these micronutrients are involved in the pathophysiology of ALD remains largely unknown. In the current study, we provide a review of recent studies that investigated the imbalance of micronutrients in patients with ALD with a focus on zinc, iron, copper, magnesium, selenium, vitamin D and vitamin E, and determine how disturbances in micronutrients relates to the pathophysiology of ALD. Overall, zinc, selenium, vitamin D, and vitamin E uniformly exhibited a deficiency, and iron demonstrated an elevated trend. While for copper, both an elevation and deficiency were observed from existing literature. More importantly, we also highlight several challenges in terms of low sample size, study design discrepancies, sample heterogeneity across studies, and the use of machine learning approaches.
Collapse
Affiliation(s)
- Jing Wu
- Department of Critical Care Medicine of Liver Disease, Beijing You-An Hospital, Capital Medical University, Beijing 100069, China
| | - Qing-Hua Meng
- Department of Critical Care Medicine of Liver Disease, Beijing You-An Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
33
|
Mayor-Ibarguren A, Busca-Arenzana C, Robles-Marhuenda Á. A Hypothesis for the Possible Role of Zinc in the Immunological Pathways Related to COVID-19 Infection. Front Immunol 2020; 11:1736. [PMID: 32754165 PMCID: PMC7365859 DOI: 10.3389/fimmu.2020.01736] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023] Open
|
34
|
Skalny AV, Rink L, Ajsuvakova OP, Aschner M, Gritsenko VA, Alekseenko SI, Svistunov AA, Petrakis D, Spandidos DA, Aaseth J, Tsatsakis A, Tinkov AA. Zinc and respiratory tract infections: Perspectives for COVID‑19 (Review). Int J Mol Med 2020; 46:17-26. [PMID: 32319538 PMCID: PMC7255455 DOI: 10.3892/ijmm.2020.4575] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
In view of the emerging COVID‑19 pandemic caused by SARS‑CoV‑2 virus, the search for potential protective and therapeutic antiviral strategies is of particular and urgent interest. Zinc is known to modulate antiviral and antibacterial immunity and regulate inflammatory response. Despite the lack of clinical data, certain indications suggest that modulation of zinc status may be beneficial in COVID‑19. In vitro experiments demonstrate that Zn2+ possesses antiviral activity through inhibition of SARS‑CoV RNA polymerase. This effect may underlie therapeutic efficiency of chloroquine known to act as zinc ionophore. Indirect evidence also indicates that Zn2+ may decrease the activity of angiotensin‑converting enzyme 2 (ACE2), known to be the receptor for SARS‑CoV‑2. Improved antiviral immunity by zinc may also occur through up‑regulation of interferon α production and increasing its antiviral activity. Zinc possesses anti‑inflammatory activity by inhibiting NF‑κB signaling and modulation of regulatory T‑cell functions that may limit the cytokine storm in COVID‑19. Improved Zn status may also reduce the risk of bacterial co‑infection by improving mucociliary clearance and barrier function of the respiratory epithelium, as well as direct antibacterial effects against S. pneumoniae. Zinc status is also tightly associated with risk factors for severe COVID‑19 including ageing, immune deficiency, obesity, diabetes, and atherosclerosis, since these are known risk groups for zinc deficiency. Therefore, Zn may possess protective effect as preventive and adjuvant therapy of COVID‑19 through reducing inflammation, improvement of mucociliary clearance, prevention of ventilator‑induced lung injury, modulation of antiviral and antibacterial immunity. However, further clinical and experimental studies are required.
Collapse
Affiliation(s)
- Anatoly V. Skalny
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
- Yaroslavl State University, 150003 Yaroslavl, Russia
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, D-52062 Aachen, Germany
| | - Olga P. Ajsuvakova
- Yaroslavl State University, 150003 Yaroslavl, Russia
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, 460000 Orenburg, Russia
| | - Michael Aschner
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Viktor A. Gritsenko
- Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, 460000 Orenburg
| | - Svetlana I. Alekseenko
- I.I. Mechnikov North-Western State Medical University, 191015 St. Petersburg
- K.A. Rauhfus Children's City Multidisciplinary Clinical Center for High Medical Technologies, 191000 St. Petersburg, Russia
| | - Andrey A. Svistunov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
| | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71409 Heraklion, Greece
| | - Jan Aaseth
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
- Research Department, Innlandet Hospital Trust, 3159894 Brumunddal, Norway
| | - Aristidis Tsatsakis
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
- Center of Toxicology Science and Research
| | - Alexey A. Tinkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow
- Yaroslavl State University, 150003 Yaroslavl, Russia
- Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, 460000 Orenburg
| |
Collapse
|
35
|
Hassan A, Sada KK, Ketheeswaran S, Dubey AK, Bhat MS. Role of Zinc in Mucosal Health and Disease: A Review of Physiological, Biochemical, and Molecular Processes. Cureus 2020; 12:e8197. [PMID: 32572355 PMCID: PMC7302722 DOI: 10.7759/cureus.8197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Zinc is an essential trace element of all highly proliferating cells in the human body. It is essential to the development and growth of all organisms. Zinc plays a critical role in modulating resistance to infectious agents and reduces the duration, severity, and risk of diarrheal disease via improved regeneration of intestinal epithelium, improved absorption of water and electrolytes, increased levels of brush border enzymes, and, possibly, an enhancement in the immune response allowing better clearance of pathogens. On the cellular level, zinc finger motifs play various roles including diverse functions that involve specific gene expression for ion channels throughout the body. It maintains the function and the structure of the membrane barrier by contributing to host defense, which is particularly crucial in the intestines due to the continuous exposure to noxious agents and pathogens. Zinc deficiency is characterized by impaired immune function, loss of appetite, and growth retardation. More severe cases cause diarrhea, delayed sexual maturation, hair loss, eye and skin lesions, impotence and hypogonadism in males, as well as weight loss, taste abnormalities, delayed healing of wounds, and mental lethargy. The objective of this study is a critical review of the molecular and genetic regulation of zinc in various cellular processes and organs, the association between zinc and diarrheal disease, the recommended dietary zinc intake, and the effects of zinc deficiency on the human body.
Collapse
Affiliation(s)
- Abbas Hassan
- Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine, Chicago, USA
| | | | | | - Arun Kumar Dubey
- Pharmacology, Xavier University School of Medicine, Oranjestad, ABW
| | - Malpe Surekha Bhat
- Biochemistry and Genetics, American University School of Medicine, Oranjestad, ABW
| |
Collapse
|
36
|
Wessels I, Pupke JT, von Trotha KT, Gombert A, Himmelsbach A, Fischer HJ, Jacobs MJ, Rink L, Grommes J. Zinc supplementation ameliorates lung injury by reducing neutrophil recruitment and activity. Thorax 2020; 75:253-261. [PMID: 31915307 DOI: 10.1136/thoraxjnl-2019-213357] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 11/25/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Zinc is well known for its anti-inflammatory effects, including regulation of migration and activity of polymorphonuclear neutrophils (PMN). Zinc deficiency is associated with inflammatory diseases such as acute lung injury (ALI). As deregulated neutrophil recruitment and their hyper-activation are hallmarks of ALI, benefits of zinc supplementation on the development of lipopolysaccharides (LPS)-induced ALI were tested. METHODS 64 C57Bl/6 mice, split into eight groups, were injected with 30 µg zinc 24 hours before exposure to aerosolised LPS for 4 hours. Zinc homoeostasis was characterised measuring serum and lung zinc concentrations as well as metallothionein-1 expression. Recruitment of neutrophils to alveolar, interstitial and intravascular space was assessed using flow cytometry. To determine the extent of lung damage, permeability and histological changes and the influx of protein into the bronchoalveolar lavage fluid were measured. Inflammatory status and PMN activity were evaluated via tumour necrosis factor α levels and formation of neutrophil extracellular traps. The effects of zinc supplementation prior to LPS stimulation on activation of primary human granulocytes and integrity of human lung cell monolayers were assessed as well. RESULTS Injecting zinc 24 hours prior to LPS-induced ALI indeed significantly decreased the recruitment of neutrophils to the lungs and prevented their hyperactivity and thus lung damage was decreased. Results from in vitro investigations using human cells suggest the transferability of the finding to human disease, which remains to be tested in more detail. CONCLUSION Zinc supplementation attenuated LPS-induced lung injury in a murine ALI model. Thus, the usage of zinc-based strategies should be considered to prevent detrimental consequences of respiratory infection and lung damage in risk groups.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Johanna Theresa Pupke
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Klaus-Thilo von Trotha
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany.,Department of Vascular Surgery, Marienhospital Aachen, Aachen, Nordrhein-Westfalen, Germany
| | - Alexander Gombert
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Anika Himmelsbach
- Department of Cardiology, Medical Clinic I, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Henrike Josephine Fischer
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Michael J Jacobs
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Jochen Grommes
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany .,Department of Vascular Surgery, Rhein-Maas Klinikum GmbH, Wurselen, Nordrhein-Westfalen, Germany
| |
Collapse
|
37
|
Magrone T, Jirillo E. Sepsis: From Historical Aspects to Novel Vistas. Pathogenic and Therapeutic Considerations. Endocr Metab Immune Disord Drug Targets 2020; 19:490-502. [PMID: 30857516 DOI: 10.2174/1871530319666181129112708] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Sepsis is a clinical condition due to an infectious event which leads to an early hyper-inflammatory phase followed by a status of tolerance or immune paralysis. Hyper-inflammation derives from a massive activation of immune (neutrophils, monocytes/macrophages, dendritic cells and lymphocytes) and non-immune cells (platelets and endothelial cells) in response to Gram-negative and Gram-positive bacteria and fungi. DISCUSSION A storm of pro-inflammatory cytokines and reactive oxygen species accounts for the systemic inflammatory response syndrome. In this phase, bacterial clearance may be associated with a severe organ failure development. Tolerance or compensatory anti-inflammatory response syndrome (CARS) depends on the production of anti-inflammatory mediators, such as interleukin-10, secreted by T regulatory cells. However, once triggered, CARS, if prolonged, may also be detrimental to the host, thus reducing bacterial clearance. CONCLUSION In this review, the description of pathogenic mechanisms of sepsis is propaedeutic to the illustration of novel therapeutic attempts for the prevention or attenuation of experimental sepsis as well as of clinical trials. In this direction, inhibitors of NF-κB pathway, cell therapy and use of dietary products in sepsis will be described in detail.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
38
|
Abstract
Metals are essential components in all forms of life required for the function of nearly half of all enzymes and are critically involved in virtually all fundamental biological processes. Especially, the transition metals iron (Fe), zinc (Zn), manganese (Mn), nickel (Ni), copper (Cu) and cobalt (Co) are crucial micronutrients known to play vital roles in metabolism as well due to their unique redox properties. Metals carry out three major functions within metalloproteins: to provide structural support, to serve as enzymatic cofactors, and to mediate electron transportation. Metal ions are also involved in the immune system from metal allergies to nutritional immunity. Within the past decade, much attention has been drawn to the roles of metal ions in the immune system, since increasing evidence has mounted to suggest that metals are critically implicated in regulating both the innate immune sensing of and the host defense against invading pathogens. The importance of ions in immunity is also evidenced by the identification of various immunodeficiencies in patients with mutations in ion channels and transporters. In addition, cancer immunotherapy has recently been conclusively demonstrated to be effective and important for future tumor treatment, although only a small percentage of cancer patients respond to immunotherapy because of inadequate immune activation. Importantly, metal ion-activated immunotherapy is becoming an effective and potential way in tumor therapy for better clinical application. Nevertheless, we are still in a primary stage of discovering the diverse immunological functions of ions and mechanistically understanding the roles of these ions in immune regulation. This review summarizes recent advances in the understanding of metal-controlled immunity. Particular emphasis is put on the mechanisms of innate immune stimulation and T cell activation by the essential metal ions like calcium (Ca2+), zinc (Zn2+), manganese (Mn2+), iron (Fe2+/Fe3+), and potassium (K+), followed by a few unessential metals, in order to draw a general diagram of metalloimmunology.
Collapse
Affiliation(s)
- Chenguang Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rui Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xiaoming Wei
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Mengze Lv
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
39
|
Knoell DL, Smith DA, Sapkota M, Heires AJ, Hanson CK, Smith LM, Poole JA, Wyatt TA, Romberger DJ. Insufficient zinc intake enhances lung inflammation in response to agricultural organic dust exposure. J Nutr Biochem 2019; 70:56-64. [PMID: 31153019 PMCID: PMC10575612 DOI: 10.1016/j.jnutbio.2019.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/18/2019] [Accepted: 04/25/2019] [Indexed: 11/15/2022]
Abstract
Organic dust exposure particularly within hog confinement facilities is a significant cause of airway inflammation and lung disease. In a cohort of Midwestern veterans with COPD and agricultural work exposure we observed reduced zinc intakes which were associated with decreased lung function. Because insufficient zinc intake is common within the U.S. and a potent modulator of innate immune function, we sought to determine whether deficits in zinc intake would impact the airway inflammatory response to hog confinement facility dust extract (HDE). Adult male C57BL/6 mice were randomized to zinc deficient or matched zinc sufficient diets for 3 weeks and subsequently treated with intranasal HDE inhalation or saline once or daily for 3 weeks while maintained on specific diets. Lavage fluid and lung tissue was collected. Conditions of zinc deficiency were also studied in macrophages exposed to HDE. Single and repetitive HDE inhalation exposure resulted in increased influx of total cells and neutrophils, increased mediator hyper-responsiveness (TNFα, IL-6, CXCL1, and amphiregulin), and enhanced tissue pathology that was more pronounced in zinc deficient mice compared to normal dietary counterparts. Airway inflammation was most pronounced in zinc deficient mice treated with repetitive HDE for 3 weeks. Similarly, macrophages maintained in a zinc deficient environment exhibited increased CXCL1 and IL-23 production as a result of increased NF-κB activation. Conclusion: Given the relatively high incidence of dietary deficiencies in agriculture workers, we anticipate that zinc intake, or a lack thereof, may play an important role in modulating the host response to organic dust exposure.
Collapse
Affiliation(s)
- Daren L Knoell
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Deandra A Smith
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Muna Sapkota
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Art J Heires
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| | - Corrine K Hanson
- The University of Nebraska Medical Center College of Allied Health, Omaha, NE 68198.
| | - Lynette M Smith
- The University of Nebraska Medical Center College of Public Health, Omaha, NE 68198
| | - Jill A Poole
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| | - Todd A Wyatt
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198; The University of Nebraska Medical Center College of Public Health, Omaha, NE 68198; VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105.
| | - Debra J Romberger
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| |
Collapse
|
40
|
He Y, Yuan X, Zuo H, Li X, Sun Y, Feng A. Berberine induces ZIP14 expression and modulates zinc redistribution to protect intestinal mucosal barrier during polymicrobial sepsis. Life Sci 2019; 233:116697. [PMID: 31351968 DOI: 10.1016/j.lfs.2019.116697] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/12/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022]
Abstract
AIMS The present study investigated if berberine might induce Zrt-Irt-like protein 14 (ZIP14) and affect zinc redistribution to protect intestinal barrier in sepsis. MAIN METHODS Rodent model of sepsis was induced by cecal ligation and puncture (CLP). Plasma endotoxin was assayed by LAL test and plasma zinc was measured by flame atomic spectrophotometer. Gut mucosal permeability was determined by plasma FITC-dextran. Zinc content and ZIP14 mRNA in gut mucosa were assayed by spectrophotometer and qRT-PCR, respectively. Tight junction integrity of Caco-2 was evaluated by transepithelial electrical resistance (TEER). Tight junction (TJ) protein expression was detected by Western blotting. KEY FINDINGS Berberine and zinc gluconate pretreatment to CLP rats improved survival rate, reduced plasma endotoxin level, alleviated hypozincemia, increased zinc accumulation and ZIP14 mRNA expression in the intestinal mucosa. Berberine and zinc gluconate pretreatment decreased CLP-elicited intestinal hyperpermeability to FITC-dextran. These effects of berberine in vivo were abolished by AG1024. In vitro, lipopolysaccharide (LPS) repressed zinc transfer into Caco-2 cells exposed to zinc gluconate. Berberine and IGF-I treatment increased ZIP14 protein expression and promoted zinc transfer into Caco-2 cells exposed to zinc gluconate plus LPS. Berberine treatment induced TJ protein (claudin-1 and occludin) and raised TEER in LPS-treated Caco-2 cells. These effects of berberine in vitro were partially inhibited by ZIP14 siRNA. SIGNIFICANCE The present study reveals that berberine induces ZIP14 expression and affects zinc re- distribution to protect intestinal barrier in sepsis, which is partially linked with the activation of IGF-I signaling.
Collapse
Affiliation(s)
- Yan He
- Department of Oncological Radiotherapy, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China
| | - Xiaoming Yuan
- Department of Gastrointestinal Surgery, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China
| | - Hao Zuo
- Department of Gastrointestinal Surgery, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China
| | - Xiangwei Li
- Department of Gastrointestinal Surgery, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China
| | - Ying Sun
- Department of Gastrointestinal Surgery, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China
| | - Aiwen Feng
- Department of Gastrointestinal Surgery, Affiliated Huai'an First Hospital, Nanjing Medical University, Huaian City, Jiangsu Province, PR China.
| |
Collapse
|
41
|
Uropathogenic Escherichia coli employs both evasion and resistance to subvert innate immune-mediated zinc toxicity for dissemination. Proc Natl Acad Sci U S A 2019; 116:6341-6350. [PMID: 30846555 PMCID: PMC6442554 DOI: 10.1073/pnas.1820870116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is responsible for most urinary tract infections and is also a frequent cause of sepsis, thus necessitating an understanding of UPEC-mediated subversion of innate immunity. The role of zinc in the innate immune response against UPEC infection, and whether this pathogen counters this response, has not been examined. Here we demonstrate, both in vitro and in vivo, that UPEC both evades and resists innate immune-mediated zinc toxicity to persist and disseminate within the host. Moreover, we have defined the set of UPEC genes conferring zinc resistance and have developed highly selective E. coli reporter systems to track zinc toxicity. These innovative approaches substantially enhance our understanding of immune-mediated metal ion toxicity and bacterial pathogenesis. Toll-like receptor (TLR)-inducible zinc toxicity is a recently described macrophage antimicrobial response used against bacterial pathogens. Here we investigated deployment of this pathway against uropathogenic Escherichia coli (UPEC), the major cause of urinary tract infections. Primary human macrophages subjected EC958, a representative strain of the globally disseminated multidrug-resistant UPEC ST131 clone, to zinc stress. We therefore used transposon-directed insertion site sequencing to identify the complete set of UPEC genes conferring protection against zinc toxicity. Surprisingly, zinc-susceptible EC958 mutants were not compromised for intramacrophage survival, whereas corresponding mutants in the nonpathogenic E. coli K-12 strain MG1655 displayed significantly reduced intracellular bacterial loads within human macrophages. To investigate whether the intramacrophage zinc stress response of EC958 reflected the response of only a subpopulation of bacteria, we generated and validated reporter systems as highly specific sensors of zinc stress. Using these tools we show that, in contrast to MG1655, the majority of intramacrophage EC958 evades the zinc toxicity response, enabling survival within these cells. In addition, EC958 has a higher tolerance to zinc than MG1655, with this likely being important for survival of the minor subset of UPEC cells exposed to innate immune-mediated zinc stress. Indeed, analysis of zinc stress reporter strains and zinc-sensitive mutants in an intraperitoneal challenge model in mice revealed that EC958 employs both evasion and resistance against zinc toxicity, enabling its dissemination to the liver and spleen. We thus demonstrate that a pathogen of global significance uses multiple mechanisms to effectively subvert innate immune-mediated zinc poisoning for systemic spread.
Collapse
|
42
|
Sociodemographic, Electrophysiological, and Biochemical Profiles in Children with Attention Deficit Hyperactivity Disorder and/or Epilepsy. Behav Neurol 2018; 2018:8932817. [PMID: 30631381 PMCID: PMC6305032 DOI: 10.1155/2018/8932817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/12/2018] [Indexed: 01/11/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is among the most prevalent neurobehavioral disorders affecting children worldwide. The prevalence of ADHD is higher in children with epilepsy. Despite the plethora of conducted work, the precise cause of ADHD is not identified yet. We studied here the sociodemographic, clinical, electrophysiological, and biochemical profiles of children with ADHD, epilepsy, and ADHD with epilepsy. Subjects were divided into 4 groups (25 child/group): I—control, II—ADHD, III—epilepsy, and IV—ADHD with epilepsy. Male to female ratio was significantly (p < 0.05) higher in the ADHD (3.1) and ADHD with epilepsy (2.1) groups when compared to the control (1.08) or epilepsy (1.08) groups. Positive family history was significantly evident in patients with epilepsy and ADHD with epilepsy, but not in the control or ADHD groups. Speech development was significantly delayed in the ADHD and ADHD with epilepsy groups. EEG abnormalities were detected in patients with ADHD (12%) and ADHD with epilepsy (68%). Focal frontal activities were significantly detectable in the ADHD (100%) and ADHD with epilepsy (77.8%) groups, whereas focal temporal activity was significantly present in the epilepsy (83.3%) group. Serum ferritin was significantly lower in the ADHD group (110.27 ± 6.64 ηg/ml) when compared to the control (134.23 ± 14.82 ηg/ml), epilepsy (159.66 ± 33.17 ηg/ml), and ADHD with epilepsy (203.04 ± 50.64 ηg/ml) groups. Serum zinc was significantly higher in the ADHD, epilepsy, and ADHD with epilepsy groups (236.63 ± 20.89, 286.74 ± 43.84, and 229.95 ± 67.34 μg/dl, respectively), when compared to the control group (144.21 ± 17.40 μg/dl). Serum adenosine deaminase was insignificantly different among the groups. Our results indicate that gender and family history are significant moderators in the aetiology of ADHD and epilepsy or their comorbidity. We also demonstrated that EEG could be central in the assessment of ADHD with epilepsy cases. Serum ferritin and zinc alteration may contribute significantly in ADHD and epilepsy pathophysiology.
Collapse
|
43
|
Essential Role of Zinc and Zinc Transporters in Myeloid Cell Function and Host Defense against Infection. J Immunol Res 2018; 2018:4315140. [PMID: 30417019 PMCID: PMC6207864 DOI: 10.1155/2018/4315140] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential micronutrient known to play a vital role in host defense against pathogens. Diets that are deficient in zinc lead to impaired immunity and delayed recovery from and worse outcomes following infection. Sustained insufficient zinc intake leads to dysregulation of the innate immune response and increases susceptibility to infection whereas zinc supplementation in at-risk populations has been shown to restore host defense and reduce pathogen-related morbidity and mortality. Upon infection, zinc deficiency leads to increased pathology due to imbalance in key signaling networks that result in excessive inflammation and collateral tissue damage. In particular, zinc impacts macrophage function, a critical front-line cell in host defense, in addition to other immune cells. Deficits in zinc adversely impact macrophage function resulting in dysregulation of phagocytosis, intracellular killing, and cytokine production. An additional work in this field has revealed a vital role for several zinc transporter proteins that are required for proper bioredistribution of zinc within mononuclear cells to achieve an optimal immune response against invading microorganisms. In this review, we will discuss the most recent developments regarding zinc's role in innate immunity and protection against pathogen invasion.
Collapse
|
44
|
|
45
|
Banupriya N, Bhat BV, Benet BD, Catherine C, Sridhar MG, Parija SC. Short Term Oral Zinc Supplementation among Babies with Neonatal Sepsis for Reducing Mortality and Improving Outcome - A Double-Blind Randomized Controlled Trial. Indian J Pediatr 2018; 85:5-9. [PMID: 28891027 DOI: 10.1007/s12098-017-2444-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/31/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To evaluate the efficacy of short term zinc supplementation on the mortality rate and neurodevelopment outcome in neonates with sepsis at 12 mo corrected age. METHODS The clinical trial was undertaken in the neonatal intensive care unit of JIPMER during the time period from September 2013 through December 2016. Neonates with clinical manifestations of sepsis who exhibited two positive screening tests (microESR, C- reactive protein, band cell count) were included and randomized into no zinc and zinc group. The intervention was zinc sulfate monohydrate given at a dose of 3 mg/kg twice a day orally for 10 d along with standard antibiotics. The no zinc group was on antibiotic treatment. Blood samples from both groups were collected at baseline and after day 10. Babies were carefully discharged from the hospital. The babies were followed up till 12 mo corrected age using DASII (Development Assessment Scale for Indian Infants). RESULTS At the time of enrolment, patient characteristics were similar in both the groups. The mortality rate was significantly higher in no zinc compared to zinc group (5 vs. 13; P = 0.04). Although motor development quotient was similar, mental development quotient was significantly better among babies who received zinc supplementation. CONCLUSIONS Short term zinc supplementation of newborns with sepsis reduces mortality and improves mental development quotient at 12 mo of age.
Collapse
Affiliation(s)
- Newton Banupriya
- Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, 605006, India
| | - Ballambattu Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, 605006, India.
| | - Bosco Dhas Benet
- Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, 605006, India
| | - Christina Catherine
- Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, 605006, India
| | - Magadi Gopalakrishna Sridhar
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, India
| | - Subhash Chandra Parija
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education & Research, Pondicherry, India
| |
Collapse
|
46
|
Mechanistic insights into the protective impact of zinc on sepsis. Cytokine Growth Factor Rev 2017; 39:92-101. [PMID: 29279185 DOI: 10.1016/j.cytogfr.2017.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022]
Abstract
Sepsis, a systemic inflammation as a response to a bacterial infection, is a huge unmet medical need. Data accumulated over the last decade suggest that the nutritional status of patients as well as composition of their gut microbiome, are strongly linked with the risk to develop sepsis, the severity of the disease and prognosis. In particular, the essential micronutrient zinc is essential in the resistance against sepsis and has shown to be protective in animal models as well as in human patients. The potential mechanisms by which zinc protects in sepsis are discussed in this review paper: we will focus on the inflammatory response, chemotaxis, phagocytosis, immune response, oxidative stress and modulation of the microbiome. A full understanding of the mechanism of action of zinc may open new preventive and therapeutic interventions in sepsis.
Collapse
|
47
|
Tang Z, Wei Z, Wen F, Wu Y. Efficacy of zinc supplementation for neonatal sepsis: a systematic review and meta-analysis. J Matern Fetal Neonatal Med 2017; 32:1213-1218. [PMID: 29103346 DOI: 10.1080/14767058.2017.1402001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background: Zinc supplementation has some potential in treating neonatal sepsis. We conduct a systematic review and meta-analysis to explore the efficacy of zinc supplementation for neonatal sepsis. Methods: PubMed, Embase, Web of science, EBSCO, and Cochrane Library databases are systematically searched. Randomized controlled trials (RCTs) assessing the efficacy of zinc supplementation in neonatal sepsis are included. Two investigators independently search articles, extract the data, and assessed the quality of included studies. Meta-analysis is performed using the random-effect model. Results: Four RCTs involving 986 patients are included in the meta-analysis. Overall, compared with control intervention in neonatal sepsis, zinc supplementation is able to significantly reduce mortality rate (risk ratio (RR) = 0.48; 95% confidence intervals (CIs) = 0.25-0.94; p = .03) and improve serum zinc (mean difference (MD) = 81.97; 95% CI = 34.57-129.37; p = .0007), but has no remarkable influence on hospital stay (MD = -4.51; 95% CI = -15.08 to 6.05; p = .40) and the number of expired patients (RR = 0.63; 95% CI = 0.24-1.65; p = .35). Conclusions: Zinc supplementation may significantly reduce mortality rate and improve serum zinc in neonatal sepsis, but has no substantial influence on hospital stay and the number of expired patients.
Collapse
Affiliation(s)
- Zhijun Tang
- a Department of Respiratory Disease , Nanchuan People's Hospital of Chongqing Medical University , Chongqing , China
| | - Zonghui Wei
- a Department of Respiratory Disease , Nanchuan People's Hospital of Chongqing Medical University , Chongqing , China
| | - Fei Wen
- a Department of Respiratory Disease , Nanchuan People's Hospital of Chongqing Medical University , Chongqing , China
| | - Yongdei Wu
- a Department of Respiratory Disease , Nanchuan People's Hospital of Chongqing Medical University , Chongqing , China
| |
Collapse
|
48
|
Wessels I, Maywald M, Rink L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017; 9:E1286. [PMID: 29186856 PMCID: PMC5748737 DOI: 10.3390/nu9121286] [Citation(s) in RCA: 404] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 12/27/2022] Open
Abstract
After the discovery of zinc deficiency in the 1960s, it soon became clear that zinc is essential for the function of the immune system. Zinc ions are involved in regulating intracellular signaling pathways in innate and adaptive immune cells. Zinc homeostasis is largely controlled via the expression and action of zinc "importers" (ZIP 1-14), zinc "exporters" (ZnT 1-10), and zinc-binding proteins. Anti-inflammatory and anti-oxidant properties of zinc have long been documented, however, underlying mechanisms are still not entirely clear. Here, we report molecular mechanisms underlying the development of a pro-inflammatory phenotype during zinc deficiency. Furthermore, we describe links between altered zinc homeostasis and disease development. Consequently, the benefits of zinc supplementation for a malfunctioning immune system become clear. This article will focus on underlying mechanisms responsible for the regulation of cellular signaling by alterations in zinc homeostasis. Effects of fast zinc flux, intermediate "zinc waves", and late homeostatic zinc signals will be discriminated. Description of zinc homeostasis-related effects on the activation of key signaling molecules, as well as on epigenetic modifications, are included to emphasize the role of zinc as a gatekeeper of immune function.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Martina Maywald
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| |
Collapse
|
49
|
Zinc is a critical regulator of placental morphogenesis and maternal hemodynamics during pregnancy in mice. Sci Rep 2017; 7:15137. [PMID: 29123159 PMCID: PMC5680205 DOI: 10.1038/s41598-017-15085-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/20/2017] [Indexed: 01/20/2023] Open
Abstract
Zinc is an essential micronutrient in pregnancy and zinc deficiency impairs fetal growth. We used a mouse model of moderate zinc deficiency to investigate the physiological mechanisms by which zinc is important to placental morphogenesis and the maternal blood pressure changes during pregnancy. A 26% reduction in circulating zinc (P = 0.005) was exhibited in mice fed a moderately zinc-deficient diet. Zinc deficiency in pregnancy resulted in an 8% reduction in both near term fetal and placental weights (both P < 0.0001) indicative of disrupted placental development and function. Detailed morphological analysis confirmed changes to the placental labyrinth microstructure. Continuous monitoring of maternal mean arterial pressure (MAP) revealed a late gestation decrease in the zinc-deficient dams. Differential expression of a number of regulatory genes within maternal kidneys supported observations on MAP changes in gestation. Increased MAP late in gestation is required to maintain perfusion of multiple placentas within rodent pregnancies. Decreased MAP within the zinc-deficient dams implies reduced blood flow and nutrient delivery to the placenta. These findings show that adequate zinc status is required for correct placental morphogenesis and appropriate maternal blood pressure adaptations to pregnancy. We conclude that insufficient maternal zinc intake from before and during pregnancy is likely to impact in utero programming of offspring growth and development largely through effects to the placenta and maternal cardiovascular system.
Collapse
|
50
|
Crowell KT, Kelleher SL, Soybel DI, Lang CH. Marginal dietary zinc deprivation augments sepsis-induced alterations in skeletal muscle TNF-α but not protein synthesis. Physiol Rep 2017; 4:4/21/e13017. [PMID: 27811170 PMCID: PMC5112495 DOI: 10.14814/phy2.13017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/04/2016] [Indexed: 12/11/2022] Open
Abstract
Severe zinc deficiency is associated with an increased systemic inflammatory response and mortality after sepsis. However, the impact of mild zinc deficiency, which is more common in populations with chronic illnesses and sepsis, is unknown. In this study, we hypothesized that marginal dietary Zn deprivation (ZM) would amplify tissue inflammation and exacerbate the sepsis-induced decrease in muscle protein synthesis. Adult male C57BL/6 mice were fed a zinc-adequate (ZA) or ZM diet (30 or 10 mg Zn/kg, respectively) over 4 weeks, peritonitis was induced by cecal ligation and puncture (CLP), and mice were examined at either 24 h (acute) or 5 days (chronic) post-CLP Acute sepsis decreased the in vivo rate of skeletal muscle protein synthesis and the phosphorylation of the mTOR substrate 4E-BP1. Acutely, sepsis increased TNF-α and IL-6 mRNA in muscle, and the increase in TNF-α was significantly greater in ZM mice. However, muscle protein synthesis and 4E-BP1 phosphorylation returned to baseline 5 days post-CLP in both ZA and ZM mice. Protein degradation via markers of the ubiquitin proteasome pathway was increased in acute sepsis, yet only MuRF1 mRNA was increased in chronic sepsis and ZM amplified this elevation. Our data suggest that mild zinc deficiency increases TNF-α in muscle acutely after sepsis but does not significantly modulate the rate of muscle protein synthesis.
Collapse
Affiliation(s)
- Kristen T Crowell
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Shannon L Kelleher
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania.,Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania.,Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania.,Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - David I Soybel
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania.,Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania.,Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Charles H Lang
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania .,Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| |
Collapse
|