1
|
Zhao F, Tie N, Kwok LY, Ma T, Wang J, Man D, Yuan X, Li H, Pang L, Shi H, Ren S, Yu Z, Shen X, Li H, Zhang H. Baseline gut microbiome as a predictive biomarker of response to probiotic adjuvant treatment in gout management. Pharmacol Res 2024; 209:107445. [PMID: 39396767 DOI: 10.1016/j.phrs.2024.107445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Gout is characterized by dysregulation of uric acid (UA) metabolism, and the gut microbiota may serve as a regulatory target. This two-month randomized, double-blind, placebo-controlled trial aimed to investigate the additional benefits of coadministering Probio-X alongside febuxostat. A total of 160 patients with gout were randomly assigned to either the probiotic group (n = 120; Probio-X [1 ×1011 CFU/day] with febuxostat) or the placebo group (n = 40; placebo material with febuxostat). Coadministration of Probio-X significantly decreased serum UA levels and the rate of acute gout attacks (P < 0.05). Based on achieving a target sUA level (360 μmol/L) after the intervention, the probiotic group was further subdivided into probiotic-responsive (ProA; n = 54) and probiotic-unresponsive (ProB; n = 66) subgroups. Post-intervention clinical indicators, metagenomic, and metabolomic changes in the ProB and placebo groups were similar, but differed from those in the ProA group, which exhibited significantly lower levels of acute gout attack, gout impact score, serum indicators (UA, XOD, hypoxanthine, and IL-1β), and fecal gene abundances of UA-producing pathways (KEGG orthologs of K13479 and K01487; gut metabolic modules for formate conversion and lactose and galactose degradation). Additionally, the ProA group showed significantly higher levels (P < 0.05) of gut SCFAs-producing bacteria and UA-related metabolites (xanthine, hypoxanthine, bile acids) after the intervention. Finally, we established a gout metagenomic classifier to predict probiotic responsiveness based on subjects' baseline gut microbiota composition. Our results indicate that probiotic-driven therapeutic responses are highly individual, with the probiotic-responsive cohort benefitting significantly from probiotic coadministration.
Collapse
Affiliation(s)
- Feiyan Zhao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Ning Tie
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Hohhot, Inner Mongolia, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Teng Ma
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Jing Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Hohhot, Inner Mongolia, China
| | - Dafu Man
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Hohhot, Inner Mongolia, China
| | - Xiangzheng Yuan
- Physical examination center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Huiyun Li
- Department of Rheumatology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Lixia Pang
- Department of Rheumatology and Immunology, Hulunbuir People's Hospital, Hohhot, Inner Mongolia, China
| | - Hui Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Inner Mongolia Medical University, Baotou, Inner Mongolia, China
| | - Shuiming Ren
- Department of Rheumatology and Immunology, Ordos School of Clinical Medicine, Inner Mongolia Medical University, Ordos, Inner Mongolia, China
| | - Zhongjie Yu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Xin Shen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Hongbin Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Hohhot, Inner Mongolia, China.
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
2
|
Phadke I, Pouzolles M, Machado A, Moraly J, Gonzalez-Menendez P, Zimmermann VS, Kinet S, Levine M, Violet PC, Taylor N. Vitamin C deficiency reveals developmental differences between neonatal and adult hematopoiesis. Front Immunol 2022; 13:898827. [PMID: 36248829 PMCID: PMC9562198 DOI: 10.3389/fimmu.2022.898827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
Hematopoiesis, a process that results in the differentiation of all blood lineages, is essential throughout life. The production of 1x1012 blood cells per day, including 200x109 erythrocytes, is highly dependent on nutrient consumption. Notably though, the relative requirements for micronutrients during the perinatal period, a critical developmental window for immune cell and erythrocyte differentiation, have not been extensively studied. More specifically, the impact of the vitamin C/ascorbate micronutrient on perinatal as compared to adult hematopoiesis has been difficult to assess in animal models. Even though humans cannot synthesize ascorbate, due to a pseudogenization of the L-gulono-γ-lactone oxidase (GULO) gene, its generation from glucose is an ancestral mammalian trait. Taking advantage of a Gulo-/- mouse model, we show that ascorbic acid deficiency profoundly impacts perinatal hematopoiesis, resulting in a hypocellular bone marrow (BM) with a significant reduction in hematopoietic stem cells, multipotent progenitors, and hematopoietic progenitors. Furthermore, myeloid progenitors exhibited differential sensitivity to vitamin C levels; common myeloid progenitors and megakaryocyte-erythrocyte progenitors were markedly reduced in Gulo-/- pups following vitamin C depletion in the dams, whereas granulocyte-myeloid progenitors were spared, and their frequency was even augmented. Notably, hematopoietic cell subsets were rescued by vitamin C repletion. Consistent with these data, peripheral myeloid cells were maintained in ascorbate-deficient Gulo-/- pups while other lineage-committed hematopoietic cells were decreased. A reduction in B cell numbers was associated with a significantly reduced humoral immune response in ascorbate-depleted Gulo-/- pups but not adult mice. Erythropoiesis was particularly sensitive to vitamin C deprivation during both the perinatal and adult periods, with ascorbate-deficient Gulo-/- pups as well as adult mice exhibiting compensatory splenic differentiation. Furthermore, in the pathological context of hemolytic anemia, vitamin C-deficient adult Gulo-/- mice were not able to sufficiently increase their erythropoietic activity, resulting in a sustained anemia. Thus, vitamin C plays a pivotal role in the maintenance and differentiation of hematopoietic progenitors during the neonatal period and is required throughout life to sustain erythroid differentiation under stress conditions.
Collapse
Affiliation(s)
- Ira Phadke
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alice Machado
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Josquin Moraly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie S. Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| |
Collapse
|
3
|
Fatima T, Jacobsson LT, Kern S, Zettergren A, Blennow K, Zetterberg H, Johansson L, Dehlin M, Skoog I. Association between serum urate and CSF markers of Alzheimer's disease pathology in a population-based sample of 70-year-olds. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12241. [PMID: 34934798 PMCID: PMC8652407 DOI: 10.1002/dad2.12241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The relationship between urate and biomarkers for Alzheimer's disease (AD) pathophysiology has not been investigated. METHODS We examined whether serum concentration of urate was associated with cerebrospinal fluid biomarkers, amyloid beta (Aβ)42, Aβ40, phosphorylated tau (p-tau), total tau (t-tau), neurofilament light (NfL), and Aβ42/Aβ40 ratio, in cognitively unimpaired 70-year-old individuals from Gothenburg, Sweden. We also evaluated whether possible associations were modulated by the apolipoprotein E (APOE) ε4 allele. RESULTS Serum urate was positively associated with Aβ42 in males (β = 0.55 pg/mL, P = .04). There was a positive urate-APOE ε4 interaction (1.24 pg/mL, P interaction = .02) in relation to Aβ42 association. The positive urate and Aβ42 association strengthened in male APOE ε4 carriers (β = 1.28 pg/mL, P = .01). DISCUSSION The positive association between urate and Aβ42 in cognitively healthy men may suggest a protective effect of urate against deposition of amyloid protein in the brain parenchyma, and in the longer term, maybe against AD dementia.
Collapse
Affiliation(s)
- Tahzeeb Fatima
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical SciencesLundSection of RheumatologyLund UniversityLundSweden
| | - Lennart T.H. Jacobsson
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Silke Kern
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anna Zettergren
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Lena Johansson
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Mats Dehlin
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Ingmar Skoog
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Li Z, Hoshino Y, Tran L, Gaucher EA. Phylogenetic articulation of uric acid evolution in mammals and how it informs a therapeutic uricase. Mol Biol Evol 2021; 39:6413644. [PMID: 34718698 PMCID: PMC8760943 DOI: 10.1093/molbev/msab312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The role of uric acid during primate evolution has remained elusive ever since it was discovered over 100 years ago that humans have unusually high levels of the small molecule in our serum. It has been difficult to generate a neutral or adaptive explanation in part because the uricase enzyme evolved to become a pseudogene in apes thus masking typical signals of sequence evolution. Adding to the difficulty is a lack of clarity on the functional role of uric acid in apes. One popular hypothesis proposes that uric acid is a potent antioxidant that increased in concentration to compensate for the lack of vitamin C synthesis in primate species ∼65 million years ago (Mya). Here, we have expanded on our previous work with resurrected ancient uricase proteins to better resolve the reshaping of uricase enzymatic activity prior to ape evolution. Our results suggest that the pivotal death-knell to uricase activity occurred between 20-30 Mya despite small sequential modifications to its catalytic efficiency for the tens of millions of years since primates lost their ability to synthesize vitamin C, and thus the two appear uncorrelated. We also use this opportunity to demonstrate how molecular evolution can contribute to biomedicine by presenting ancient uricases to human immune cells that assay for innate reactivity against foreign antigens. A highly stable and highly catalytic ancient uricase is shown to elicit a lower immune response in more human haplotypes than other uricases currently in therapeutic development.
Collapse
Affiliation(s)
- Ze Li
- Georgia State University, Department of Biology, Atlanta, GA U.S.A
| | - Yosuke Hoshino
- Georgia State University, Department of Biology, Atlanta, GA U.S.A
| | - Lily Tran
- Georgia State University, Department of Biology, Atlanta, GA U.S.A
| | - Eric A Gaucher
- Georgia State University, Department of Biology, Atlanta, GA U.S.A
| |
Collapse
|
5
|
de Lima Balico L, Gaucher EA. CRISPR-Cas9-mediated reactivation of the uricase pseudogene in human cells prevents acute hyperuricemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:578-584. [PMID: 34589279 PMCID: PMC8463316 DOI: 10.1016/j.omtn.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/09/2021] [Indexed: 12/20/2022]
Abstract
The utility of CRISPR-Cas9 to repair or reverse diseased states that arise from recent genetic mutations in the human genome is now widely appreciated. The use of CRISPR to "design" the outcomes of biology is challenged by both specialized ethicists and the general public. Less of a focus, however, is the ability of CRISPR to provide metabolic supplements or prophylactic molecules that improve long-term human health by overwriting ancient evolutionary events. Here, we use CRISPR to genomically integrate a functional uricase gene that encodes an enzymatically active protein into the human genome. These uricase-producing cells are able to reduce or even eliminate high concentrations of exogenous uric acid despite the enzyme being localized to peroxisomes. Our evolutionary engineered cells represent the first instance of the primate ape lineage expressing a functional uricase encoded in the genome within the last 20 million years. We anticipate that human cells expressing uricase will help prevent hyperuricemia (including gout) as well as hypertension and will help protect against fatty liver disease in the future.
Collapse
Affiliation(s)
- Lais de Lima Balico
- Department of Biology, Georgia State University, 100 Piedmont Ave., Atlanta, GA, 30303, USA
| | - Eric A Gaucher
- Department of Biology, Georgia State University, 100 Piedmont Ave., Atlanta, GA, 30303, USA
| |
Collapse
|
6
|
Agnoletti D, Cicero AFG, Borghi C. The Impact of Uric Acid and Hyperuricemia on Cardiovascular and Renal Systems. Cardiol Clin 2021; 39:365-376. [PMID: 34247750 DOI: 10.1016/j.ccl.2021.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The description of gout dates back almost 5000 years, and scientific interest in uric acid increased when it was found to be involved in the pathogenesis of gout. Since then, many basic and clinical studies have assessed the implications of uric acid for the oxidative system, inflammation, and cardiovascular and renal outcomes. Uric acid-lowering therapy failed to improve clinical hard outcomes in asymptomatic hyperuricemia, and it is retained in symptomatic hyperuricemia. Dietary and lifestyle modifications are critical to manage hyperuricemia. More studies are warranted to investigate the role of uric acid-lowering drugs on cardiovascular outcomes.
Collapse
Affiliation(s)
- Davide Agnoletti
- Internal Medicine Department, IRCCS Sacro Cuore Hospital, Viale Luigi Rizzardi 4, Negrar di Valpolicella (VR) 37024, Italy
| | - Arrigo F G Cicero
- Medical and Surgical Sciences Department, University of Bologna, Via Albertoni 15, Bologna 40138, Italy
| | - Claudio Borghi
- Medical and Surgical Sciences Department, University of Bologna, Via Albertoni 15, Bologna 40138, Italy.
| |
Collapse
|
7
|
Carn D, Lanaspa MA, Benner SA, Andrews P, Dudley R, Andres-Hernando A, Tolan DR, Johnson RJ. The role of thrifty genes in the origin of alcoholism: A narrative review and hypothesis. Alcohol Clin Exp Res 2021; 45:1519-1526. [PMID: 34120350 PMCID: PMC8429132 DOI: 10.1111/acer.14655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 01/21/2023]
Abstract
In this narrative review, we present the hypothesis that key mutations in two genes, occurring 15 and 10 million years ago (MYA), were individually and then collectively adaptive for ancestral humans during periods of starvation, but are maladaptive in modern civilization (i.e., "thrifty genes"), with the consequence that these genes not only increase our risk today for obesity, but also for alcoholism. Both mutations occurred when ancestral apes were experiencing loss of fruit availability during periods of profound climate change or environmental upheaval. The silencing of uricase (urate oxidase) activity 15 MYA enhanced survival by increasing the ability for fructose present in dwindling fruit to be stored as fat, a consequence of enhanced uric acid production during fructose metabolism that stimulated lipogenesis and blocked fatty acid oxidation. Likewise, a mutation in class IV alcohol dehydrogenase ~10 MYA resulted in a remarkable 40-fold increase in the capacity to oxidize ethanol (EtOH), which allowed our ancestors to ingest fallen, fermenting fruit. In turn, the EtOH ingested could activate aldose reductase that stimulates the conversion of glucose to fructose, while uric acid produced during EtOH metabolism could further enhance fructose production and metabolism. By aiding survival, these mutations would have allowed our ancestors to generate more fat, primarily from fructose, to survive changing habitats due to the Middle Miocene disruption and also during the late-Miocene aridification of East Africa. Unfortunately, the enhanced ability to metabolize and utilize EtOH may now be acting to increase our risk for alcoholism, which may be yet another consequence of once-adaptive thrifty genes.
Collapse
Affiliation(s)
| | - Miguel A. Lanaspa
- Division of Nephrology, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Steven A. Benner
- The Foundation for Applied Molecular Evolution, Alachua, FL, USA
| | - Peter Andrews
- Department of Earth Sciences, Natural History Museum, London, UK
| | - Robert Dudley
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ana Andres-Hernando
- Division of Nephrology, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Dean R. Tolan
- Department of Biochemistry, Boston University, Boston, MA, USA
| | - Richard J. Johnson
- Division of Nephrology, University of Colorado Anschutz Medical Center, Aurora, CO, USA,The Rocky Mountain VA Medical Center, Aurora CO, USA
| |
Collapse
|
8
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Hyperuricemia-induced endothelial insulin resistance: the nitric oxide connection. Pflugers Arch 2021; 474:83-98. [PMID: 34313822 DOI: 10.1007/s00424-021-02606-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/12/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Hyperuricemia, defined as elevated serum concentrations of uric acid (UA) above 416 µmol L-1, is related to the development of cardiometabolic disorders, probably via induction of endothelial dysfunction. Hyperuricemia causes endothelial dysfunction via induction of cell apoptosis, oxidative stress, and inflammation; however, it's interfering with insulin signaling and decreased endothelial nitric oxide (NO) availability, resulting in the development of endothelial insulin resistance, which seems to be a major underlying mechanism for hyperuricemia-induced endothelial dysfunction. Here, we elaborate on how hyperuricemia induces endothelial insulin resistance through the disruption of insulin-stimulated endothelial NO synthesis. High UA concentrations decrease insulin-induced NO synthesis within the endothelial cells by interfering with insulin signaling at either the receptor or post-receptor levels (i.e., proximal and distal steps). At the proximal post-receptor level, UA impairs the function of the insulin receptor substrate (IRS) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) in the insulin signaling pathway. At the distal level, high UA concentrations impair endothelial NO synthase (eNOS)-NO system by decreasing eNOS expression and activity as well as by direct inactivation of NO. Clinically, UA-induced endothelial insulin resistance is translated into impaired endothelial function, impaired NO-dependent vasodilation, and the development of systemic insulin resistance. UA-lowering drugs may improve endothelial function in subjects with hyperuricemia.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA.,Graduate Program in Biology, City University of New York Graduate Center, New York, NY, 10016, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, P.O. Box: 19395-4763, VelenjakTehran, Iran.
| |
Collapse
|
9
|
Rocha LS, Silva BPD, Correia TML, Silva RPD, Meireles DDA, Pereira R, Netto LES, Meotti FC, Queiroz RF. Peroxiredoxin AhpC1 protects Pseudomonas aeruginosa against the inflammatory oxidative burst and confers virulence. Redox Biol 2021; 46:102075. [PMID: 34315109 PMCID: PMC8327333 DOI: 10.1016/j.redox.2021.102075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium in patients with cystic fibrosis and hospital acquired infections. It presents a plethora of virulence factors and antioxidant enzymes that help to subvert the immune system. In this study, we identified the 2-Cys peroxiredoxin, alkyl-hydroperoxide reductase C1 (AhpC1), as a relevant scavenger of oxidants generated during inflammatory oxidative burst and a mechanism of P. aeruginosa (PA14) escaping from killing. Deletion of AhpC1 led to a higher sensitivity to hypochlorous acid (HOCl, IC50 3.2 ± 0.3 versus 19.1 ± 0.2 μM), hydrogen peroxide (IC50 91.2 ± 0.3 versus 496.5 ± 6.4 μM) and the organic peroxide urate hydroperoxide. ΔahpC1 strain was more sensitive to the killing by isolated neutrophils and less virulent in a mice model of infection. All mice intranasally instilled with ΔahpC1 survived as long as they were monitored (15 days), whereas 100% wild-type and ΔahpC1 complemented with ahpC1 gene (ΔahpC1 attB:ahpC1) died within 3 days. A significantly lower number of colonies was detected in the lung and spleen of ΔahpC1-infected mice. Total leucocytes, neutrophils, myeloperoxidase activity, pro-inflammatory cytokines, nitrite production and lipid peroxidation were much lower in lungs or bronchoalveolar liquid of mice infected with ΔahpC1. Purified AhpC neutralized the inflammatory organic peroxide, urate hydroperoxide, at a rate constant of 2.3 ± 0.1 × 106 M-1s-1, and only the ΔahpC1 strain was sensitive to this oxidant. Incubation of neutrophils with uric acid, the urate hydroperoxide precursor, impaired neutrophil killing of wild-type but improved the killing of ΔahpC1. Hyperuricemic mice presented higher levels of serum cytokines and succumbed much faster to PA14 infection when compared to normouricemic mice. In summary, ΔahpC1 PA14 presented a lower virulence, which was attributed to a poorer ability to neutralize the oxidants generated by inflammatory oxidative burst, leading to a more efficient killing by the host. The enzyme is particularly relevant in detoxifying the newly reported inflammatory organic peroxide, urate hydroperoxide.
Collapse
Affiliation(s)
- Leonardo Silva Rocha
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil
| | | | - Thiago M L Correia
- Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil
| | | | - Diogo de Abreu Meireles
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Rafael Pereira
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil; Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia, Brazil
| | - Luis Eduardo Soares Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Flavia Carla Meotti
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil.
| | - Raphael Ferreira Queiroz
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Departamento de Ciências Naturais, Universidade Estadual do Sudoeste da Bahia, Brazil.
| |
Collapse
|
10
|
Braga TT, Foresto-Neto O, Camara NOS. The role of uric acid in inflammasome-mediated kidney injury. Curr Opin Nephrol Hypertens 2021; 29:423-431. [PMID: 32452918 DOI: 10.1097/mnh.0000000000000619] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Uric acid is produced after purine nucleotide degradation, upon xanthine oxidase catalytic action. In the evolutionary process, humans lost uricase, an enzyme that converts uric acid into allantoin, resulting in increased serum uric acid levels that may vary according to dietary ingestion, pathological conditions, and other factors. Despite the controversy over the inflammatory role of uric acid in its soluble form, crystals of uric acid are able to activate the NLRP3 inflammasome in different tissues. Uric acid, therefore, triggers hyperuricemic-related disease such as gout, metabolic syndrome, and kidney injuries. The present review provides an overview on the role of uric acid in the inflammasome-mediated kidney damage. RECENT FINDINGS Hyperuricemia is present in 20-35% of patients with chronic kidney disease. However, whether this increased circulating uric acid is a risk factor or just a biomarker of renal and cardiovascular injuries has become a topic of intense discussion. Despite these conflicting views, several studies support the idea that hyperuricemia is indeed a cause of progression of kidney disease, with a putative role for soluble uric acid in activating renal NLRP3 inflammasome, in reprograming renal and immune cell metabolism and, therefore, in promoting kidney inflammation/injury. SUMMARY Therapies aiming to decrease uric acid levels prevent renal NLRP3 inflammasome activation and exert renoprotective effects in experimental kidney diseases. However, further clinical studies are needed to investigate whether reduced circulating uric acid can also inhibit the inflammasome and be beneficial in human conditions.
Collapse
Affiliation(s)
- Tarcio Teodoro Braga
- Department of Basic Pathology, Federal University of Parana, Curitiba, PR.,Carlos Chagas Institute - Fiocruz-Parana, Curitiba
| | - Orestes Foresto-Neto
- Nephrology Division, Federal University of São Paulo.,Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, SP, Brazil
| | - Niels Olsen Saraiva Camara
- Nephrology Division, Federal University of São Paulo.,Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, SP, Brazil
| |
Collapse
|
11
|
Gonzalez-Menendez P, Romano M, Yan H, Deshmukh R, Papoin J, Oburoglu L, Daumur M, Dumé AS, Phadke I, Mongellaz C, Qu X, Bories PN, Fontenay M, An X, Dardalhon V, Sitbon M, Zimmermann VS, Gallagher PG, Tardito S, Blanc L, Mohandas N, Taylor N, Kinet S. An IDH1-vitamin C crosstalk drives human erythroid development by inhibiting pro-oxidant mitochondrial metabolism. Cell Rep 2021; 34:108723. [PMID: 33535038 PMCID: PMC9169698 DOI: 10.1016/j.celrep.2021.108723] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/26/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The metabolic changes controlling the stepwise differentiation of hematopoietic stem and progenitor cells (HSPCs) to mature erythrocytes are poorly understood. Here, we show that HSPC development to an erythroid-committed proerythroblast results in augmented glutaminolysis, generating alpha-ketoglutarate (αKG) and driving mitochondrial oxidative phosphorylation (OXPHOS). However, sequential late-stage erythropoiesis is dependent on decreasing αKG-driven OXPHOS, and we find that isocitrate dehydrogenase 1 (IDH1) plays a central role in this process. IDH1 downregulation augments mitochondrial oxidation of αKG and inhibits reticulocyte generation. Furthermore, IDH1 knockdown results in the generation of multinucleated erythroblasts, a morphological abnormality characteristic of myelodysplastic syndrome and congenital dyserythropoietic anemia. We identify vitamin C homeostasis as a critical regulator of ineffective erythropoiesis; oxidized ascorbate increases mitochondrial superoxide and significantly exacerbates the abnormal erythroblast phenotype of IDH1-downregulated progenitors, whereas vitamin C, scavenging reactive oxygen species (ROS) and reprogramming mitochondrial metabolism, rescues erythropoiesis. Thus, an IDH1-vitamin C crosstalk controls terminal steps of human erythroid differentiation.
Collapse
Affiliation(s)
- Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| | - Manuela Romano
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Hongxia Yan
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; New York Blood Center, New York, NY, USA
| | - Ruhi Deshmukh
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Julien Papoin
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Leal Oburoglu
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marie Daumur
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Anne-Sophie Dumé
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Ira Phadke
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA
| | - Cédric Mongellaz
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Xiaoli Qu
- New York Blood Center, New York, NY, USA
| | - Phuong-Nhi Bories
- Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Michaela Fontenay
- Laboratory of Excellence GR-Ex, Paris 75015, France; Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Xiuli An
- New York Blood Center, New York, NY, USA
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Patrick G Gallagher
- Departments of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Lionel Blanc
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | | | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA.
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| |
Collapse
|
12
|
Packer M. Uric Acid Is a Biomarker of Oxidative Stress in the Failing Heart: Lessons Learned from Trials With Allopurinol and SGLT2 Inhibitors. J Card Fail 2020; 26:977-984. [PMID: 32890737 DOI: 10.1016/j.cardfail.2020.08.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022]
Abstract
Hyperuricemia increases the risk of heart failure, and higher levels of serum uric acid are seen in patients who have worse ventricular function, functional capacity, and prognosis. Heart failure is also accompanied by an upregulation of xanthine oxidase, the enzyme that catalyzes the formation of uric acid and a purported source of reactive oxygen species. However, the available evidence does not support the premise that either uric acid or the activation of xanthine oxidase has direct injurious effects on the heart in the clinical setting. Xanthine oxidase inhibitors (allopurinol and oxypurinol) have had little benefit and may exert detrimental effects in patients with chronic heart failure in randomized controlled trials, and the more selective and potent inhibitor febuxostat increases the risk of cardiovascular death more than allopurinol. Instead, the available evidence indicates that changes in xanthine oxidase and uric acid are biomarkers of oxidative stress (particularly in heart failure) and that xanthine oxidase may provide an important source of nitric oxide that quenches the injurious effects of reactive oxygen species. A primary determinant of the cellular redox state is nicotinamide adenine dinucleotide, whose levels drive an inverse relationship between xanthine oxidase and sirtuin-1, a nutrient deprivation sensor that exerts important antioxidant and cardioprotective effects. Interestingly, sodium-glucose cotransporter 2 inhibitors induce a state of nutrient deprivation that includes activation of sirtuin-1, suppression of xanthine oxidase, and lowering of serum uric acid. The intermediary role of sirtuin-1 in both uric acid-lowering and cardioprotection may explain why, in mediation analyses of large-scale cardiovascular trials, the effect of sodium-glucose cotransporter 2 inhibitors to decrease serum uric acid is a major predictor of the ability of these drugs to decrease serious heart failure events.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas and Imperial College, London, UK.
| |
Collapse
|
13
|
Dissanayake LV, Spires DR, Palygin O, Staruschenko A. Effects of uric acid dysregulation on the kidney. Am J Physiol Renal Physiol 2020; 318:F1252-F1257. [PMID: 32223309 DOI: 10.1152/ajprenal.00066.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recently, research has redirected its interests in uric acid (UA) from gout, an inflammatory disease in joints, to groups of closely interrelated pathologies associated with cardiovascular and kidney dysfunction. Many epidemiological, clinical, and experimental studies have shown that UA may play a role in the pathophysiology of the cardiorenal syndrome continuum; however, it is still unclear if it is a risk factor or a causal role. Hyperuricemia has been well studied in the past two decades, revealing mechanistic insights into UA homeostasis. Likewise, some epidemiological and experimental evidence suggests that hypouricemia can lead to cardiorenal pathologies. The goal of this review is to highlight why studying both hyperuricemia and hypouricemia is warranted as well as to summarize the relevance of UA to kidney function.
Collapse
Affiliation(s)
| | - Denisha R Spires
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
14
|
Mosiichuk N, Grujic D, Woliński J, Podpryatov S, Podpriatov S, Szczurek P, Yatsenko T, Shmihel H, Drahanchuk O, Pierzynowski S, Goncharova Pierzynowska K. Uricemia in juvenile pigs model: effect of nephrectomy and potassium oxonate. JOURNAL OF ANIMAL AND FEED SCIENCES 2019. [DOI: 10.22358/jafs/110436/2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
15
|
Li L, Song Q, Yang X. Lack of Associations between Elevated Serum Uric Acid and Components of Metabolic Syndrome Such as Hypertension, Dyslipidemia, and T2DM in Overweight and Obese Chinese Adults. J Diabetes Res 2019; 2019:3175418. [PMID: 31871945 PMCID: PMC6913180 DOI: 10.1155/2019/3175418] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 01/01/2023] Open
Abstract
The overweight and obese population experiences a higher occurrence of both hyperuricemia and metabolic syndrome. The present study was to explore the relationship between serum uric acid and metabolic syndrome-related risk factors among 409 obese Chinese adults (254 women and 155 men) with >24 kg/m2 BMI. Based on sex-specific reference ranges, 233 (57%) patients showed elevated serum uric acid. A total of 15 attributes were selected to assess the associations between elevated serum uric acid and components of metabolic syndrome, including serum uric acid, total cholesterol, HDL-C, LDL-C, triglyceride, systolic blood pressure, fasting blood glucose, glycosylated hemoglobin, HOMA-IR, alanine aminotransferase, creatinine, urine microalbumin, muscle mass amount, BMI, and age. Among the participants stratified into three groups of grade I, grade II, and grade III obesity, as well as among the participants stratified into male and female groups, univariate correlation analysis identified a negative association (P < 0.01) for age, positive associations (P < 0.01) for BMI, muscle mass, alanine aminotransferase, and creatinine. The stepwise multivariate logistic regression proved similar associations for age, BMI, creatinine, and alanine aminotransferase. No significant associations were testified between serum uric acid levels and cholesterol, HDL-C, LDL-C, triglyceride, fasting blood glucose, glycosylated hemoglobin, HOMA-IR, and urine microalbumin. Factor analysis illustrated that 15 attributes could be grouped into two common factors and five individual factors. A common underlying factor was identified among uric acid, muscle mass, creatinine, alanine aminotransferase, and BMI. The results indicate that serum uric acid has no apparent association with metabolic syndromes that are commonly characterized by hypertension, dyslipidemia, and T2DM.
Collapse
Affiliation(s)
- Li Li
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Qifa Song
- Ningbo Municipal Centre for Disease Control and Prevention, Ningbo, Zhejiang, China
| | - Xi Yang
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
16
|
Dissociation between urate and blood pressure in mice and in people with early Parkinson's disease. EBioMedicine 2018; 37:259-268. [PMID: 30415890 PMCID: PMC6284456 DOI: 10.1016/j.ebiom.2018.10.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/05/2018] [Accepted: 10/12/2018] [Indexed: 02/02/2023] Open
Abstract
Background Epidemiological, laboratory and clinical studies have established an association between elevated urate and high blood pressure (BP). However, the inference of causality remains controversial. A naturally occurring antioxidant, urate may also be neuroprotective, and urate-elevating treatment with its precursor inosine is currently under clinical development as a potential disease-modifying strategy for Parkinson's disease (PD). Methods Our study takes advantage of a recently completed phase II trial evaluating oral inosine in de novo non-disabling early PD with no major cardiovascular and nephrological conditions, and of three lines of genetically engineered mice: urate oxidase (UOx) global knockout (gKO), conditional KO (cKO), and transgenic (Tg) mice with markedly elevated, mildly elevated, and substantially reduced serum urate, respectively, to systematically investigate effects of urate-modifying manipulation on BP. Findings Among clinical trial participants, change in serum urate but not changes in systolic, diastolic and orthostatic BP differed by treatment group. There was no positive correlation between urate elevations and changes in systolic, diastolic and orthostatic BP ((p = .05 (in inverse direction), 0.30 and 0.63, respectively)). Between UOx gKO, cKO, or Tg mice and their respective wildtype littermates there were no significant differences in systolic or diastolic BP or in their responses to BP-regulating interventions. Interpretation Our complementary preclinical and human studies of urate modulation in animal models and in generally healthy early PD do not support a hypertensive effect of urate elevation or an association between urate and BP. Fund U.S. Department of Defense, RJG Foundation, Michael J. Fox Foundation LEAPS program, National Institutes of Health, American Federation for Aging Research, Parkinson's Disease Foundation Advancing Parkinson's Therapies initiative.
Collapse
|
17
|
The prevalence of thyroid nodules in northwest China and its correlation with metabolic parameters and uric acid. Oncotarget 2018; 8:41555-41562. [PMID: 28107199 PMCID: PMC5522227 DOI: 10.18632/oncotarget.14720] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 01/03/2017] [Indexed: 11/25/2022] Open
Abstract
This study aimed to estimate the prevalence of thyroid nodules (TN) and investigate its correlation with metabolic parameters, especially uric acid (UA) in northwest Chinese population. We conducted a large cross-sectional survey with 67,781 residents (33,020 men, 34,761 women), aged from 18 to 86 years in Shanxi, China, from January 2012 to December 2014. A thyroid ultrasound examination was performed with number and size of nodules being recorded. Metabolic parameters including body mass index (BMI), blood pressure (BP), triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), high-density lipoprotein cholesterol (HDL-c), fasting glucose (FG), and uric acid (UA) were also examined. Our study revealed that approximately 30.7% of men and 39.9% of women in Northwest China had TN, about half of which were multi-nodularity and a quarter of their TN larger than 1 cm. The prevalence of TN increased with aging and increasing BMI, and metabolic disorders, which also related to the increased incident of multi-nodularity and larger TN. Serum UA appeared to be a protective factor for TN in men older than 30 years, but a risk factor in both men younger than 30 years and women older than 30 years. This phenomenon needs to be further investigated.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The complexity of gout continues to unravel with each new investigation. Gout sits at the intersection of multiple intrinsically complex processes, and its prevalence, impact on healthcare costs, and association with important co-morbidities make it increasingly relevant. The association between gout and type 2 diabetes, hypertension, hyperlipidemia, cardiovascular disease, renal disease, and obesity suggest that either gout, or its necessary precursor hyperuricemia, may play an important role in the manifestations of the metabolic syndrome. In this review, we analyze the complex interconnections between gout and metabolic syndrome, by reviewing gout's physiologic and epidemiologic relationships with its major co-morbidities. RECENT FINDINGS Increasing evidence supports gout's association with metabolic syndrome. More specifically, both human studies and animal models suggest that hyperuricemia may play a role in promoting inflammation, hypertension and cardiovascular disease, adipogenesis and lipogenesis, insulin and glucose dysregulation, and liver disease. Fructose ingestion is associated with increased rates of hypertension, weight gain, impaired glucose tolerance, and dyslipidemia and is a key driver of urate biosynthesis. AMP kinase (AMPK) is a central regulator of processes that tend to mitigate against the metabolic syndrome. Within hepatocytes, leukocytes, and other cells, a fructose/urate metabolic loop drives key inhibitors of AMPK, including AMP deaminase and fructokinase, that may tilt the balance toward metabolic syndrome progression. Preliminary evidence suggests that agents that block the intracellular synthesis of urate may restore AMPK activity and help maintain metabolic homeostasis. Gout is both an inflammatory and a metabolic disease. With further investigation of urate's role, the possibility of proper gout management additionally mitigating metabolic syndrome is an evolving and important question.
Collapse
Affiliation(s)
- Gabrielle E Thottam
- Department of Medicine, Roger Williams Medical Center, Providence, RI, USA.,Crystal Diseases Study Group, Division of Rheumatology, Department of Medicine, New York University School of Medicine, 301 East 17th Street, Suite 1410, New York, NY, 10003, USA
| | - Svetlana Krasnokutsky
- Crystal Diseases Study Group, Division of Rheumatology, Department of Medicine, New York University School of Medicine, 301 East 17th Street, Suite 1410, New York, NY, 10003, USA.,Rheumatology Section, Department of Medicine, VA New Harbor Health Care System, New York Campus, New York, NY, USA
| | - Michael H Pillinger
- Crystal Diseases Study Group, Division of Rheumatology, Department of Medicine, New York University School of Medicine, 301 East 17th Street, Suite 1410, New York, NY, 10003, USA. .,Rheumatology Section, Department of Medicine, VA New Harbor Health Care System, New York Campus, New York, NY, USA.
| |
Collapse
|
19
|
Molino-Lova R, Sofi F, Pasquini G, Vannetti F, Del Ry S, Vassalle C, Clerici M, Sorbi S, Macchi C. Higher uric acid serum levels are associated with better muscle function in the oldest old: Results from the Mugello Study. Eur J Intern Med 2017; 41:39-43. [PMID: 28342714 DOI: 10.1016/j.ejim.2017.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Sarcopenia is the progressive loss of muscle mass and strength that occurs with advancing age and plays a pivotal role in the causal pathway leading to frailty, disability and, eventually, to death among older persons. As oxidative damage of muscle proteins has been shown to be a relevant contributory factor, in this study we hypothesized that uric acid (UA), a powerful endogenous antioxidant, might exert a protective effect on muscle function in the oldest old and we tested our hypothesis in a group of nonagenarians who participated in the Mugello Study. METHODS 239 subjects, 73 men and 166 women, mean age 92.8years±SD 3.1, underwent the assessment of UA serum level and isometric handgrip strength, a widely used clinical measure of sarcopenia. RESULTS Mean UA serum level was 5.69mg/dL±SD 1.70 and mean handgrip strength was 15.0kg±SD 6.9. After adjusting for relevant confounders, higher UA serum levels remained independent positive predictors of isometric handgrip strength (β 1.24±SE(β) 0.43, p=0.005). CONCLUSION Our results show that higher UA serum levels are associated with better muscle function in the oldest old and, accordingly, might slow down the progression of sarcopenia.
Collapse
Affiliation(s)
- R Molino-Lova
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy
| | - F Sofi
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy.
| | - G Pasquini
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy
| | - F Vannetti
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy
| | - S Del Ry
- Institute of Clinical Physiology, CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - C Vassalle
- Gabriele Monasterio Tuscany Foundation, CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - M Clerici
- IRCCS Don Gnocchi Foundation, Via Alfonso Capecelatro 66, 20148 Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Via Fratelli Cervi 93, 20090, Segrate, Milan, Italy
| | - S Sorbi
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy; Dept. Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| | - C Macchi
- IRCCS Don Gnocchi Foundation, Via di Scandicci 269, 50143 Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| |
Collapse
|
20
|
Kaneko C, Ogura J, Sasaki S, Okamoto K, Kobayashi M, Kuwayama K, Narumi K, Iseki K. Fructose suppresses uric acid excretion to the intestinal lumen as a result of the induction of oxidative stress by NADPH oxidase activation. Biochim Biophys Acta Gen Subj 2016; 1861:559-566. [PMID: 27913188 DOI: 10.1016/j.bbagen.2016.11.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/08/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND A high intake of fructose increases the risk for hyperuricemia. It has been reported that long-term fructose consumption suppressed renal uric acid excretion and increased serum uric acid level. However, the effect of single administration of fructose on excretion of uric acid has not been clarified. METHODS We used male Wistar rats, which were orally administered fructose (5g/kg). Those rats were used in each experiment at 12h after administration. RESULTS Single administration of fructose suppressed the function of ileal uric acid excretion and had no effect on the function of renal uric acid excretion. Breast cancer resistance protein (BCRP) predominantly contributes to intestinal excretion of uric acid as an active homodimer. Single administration of fructose decreased BCRP homodimer level in the ileum. Moreover, diphenyleneiodonium (DPI), an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox), recovered the suppression of the function of ileal uric acid excretion and the Bcrp homodimer level in the ileum of rats that received single administration of fructose. CONCLUSIONS Single administration of fructose decreases in BCRP homodimer level, resulting in the suppression the function of ileal uric acid excretion. The suppression of the function of ileal uric acid excretion by single administration of fructose is caused by the activation of Nox. The results of our study provide a new insight into the mechanism of fructose-induced hyperuricemia.
Collapse
Affiliation(s)
- Chihiro Kaneko
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Jiro Ogura
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Shunichi Sasaki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Keisuke Okamoto
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Department of Pharmacy, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan.
| | - Kaori Kuwayama
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan; Department of Pharmacy, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan.
| |
Collapse
|
21
|
Li H, Qin X, Xie D, Tang G, Zhang Y, Li J, Hou F, Wang X, Huo Y, Xu X. Effects of combined enalapril and folic acid therapy on the serum uric acid levels in hypertensive patients: a multicenter, randomized, double-blind, parallel-controlled clinical trial. Intern Med 2015; 54:17-24. [PMID: 25742888 DOI: 10.2169/internalmedicine.54.2931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective The efficacy of combined treatment consisting of enalapril and folic acid (FA) was compared to that of enalapril alone in reducing the serum uric acid (UA) levels in adult hypertensive patients in China. Methods Patients with mild to moderate hypertension (n=480) were randomly assigned to one of three treatment groups: (1) 10 mg enalapril (control group), (2) 10 mg enalapril plus 0.4 mg FA (low-FA group) or (3) 10 mg enalapril plus 0.8 mg FA (high-FA group) daily for eight weeks. The primary outcome was the UA ratio (week 8 UA: baseline UA). Results The final analysis included 450 patients (43.1% men, 27-75 years of age). An adjusted multivariable regression analysis revealed no significant differences in the UA ratio between the three groups after eight weeks of treatment. In the subgroup analysis stratified according to the baseline UA level, the high-FA group demonstrated a significantly greater UA-lowering response among the patients with an elevated baseline UA concentration (UA ≥310 μmol/L) [median UA ratio (25th percentile, 75th percentile): 0.94 (0.83, 1.01)], compared with that observed in the control group [0.97 (0.90, 1.00), p=0.025]. Similar results were found in the participants with baseline hyperuricemia (HUA; UA: men >420 μmol/L, women >350 μmol/L). Conclusion In this sample of adult hypertensive patients, the administration of a daily dose of 10 mg of enalapril combined with 0.8 mg of FA had a greater beneficial effect on the serum UA levels than did that of 10 mg of enalapril alone in patients with either an elevated UA concentration or HUA.
Collapse
Affiliation(s)
- Haibo Li
- Institute of Biomedicine, Anhui Medical University, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Li M, Yang D, Mei L, Yuan L, Xie A, Yuan J. Screening and characterization of purine nucleoside degrading lactic acid bacteria isolated from Chinese sauerkraut and evaluation of the serum uric acid lowering effect in hyperuricemic rats. PLoS One 2014; 9:e105577. [PMID: 25184445 PMCID: PMC4153548 DOI: 10.1371/journal.pone.0105577] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/16/2014] [Indexed: 02/07/2023] Open
Abstract
Hyperuricemia is well known as the cause of gout. In recent years, it has also been recognized as a risk factor for arteriosclerosis, cerebrovascular and cardiovascular diseases, and nephropathy in diabetic patients. Foods high in purine compounds are more potent in exacerbating hyperuricemia. Therefore, the development of probiotics that efficiently degrade purine compounds is a promising potential therapy for the prevention of hyperuricemia. In this study, fifty-five lactic acid bacteria isolated from Chinese sauerkraut were evaluated for the ability to degrade inosine and guanosine, the two key intermediates in purine metabolism. After a preliminary screening based on HPLC, three candidate strains with the highest nucleoside degrading rates were selected for further characterization. The tested biological characteristics of candidate strains included acid tolerance, bile tolerance, anti-pathogenic bacteria activity, cell adhesion ability, resistance to antibiotics and the ability to produce hydrogen peroxide. Among the selected strains, DM9218 showed the best probiotic potential compared with other strains despite its poor bile resistance. Analysis of 16S rRNA sequences showed that DM9218 has the highest similarity (99%) to Lactobacillus plantarum WCFS1. The acclimated strain DM9218-A showed better resistance to 0.3% bile salt, and its survival in gastrointestinal tract of rats was proven by PCR-DGGE. Furthermore, the effects of DM9218-A in a hyperuricemia rat model were evaluated. The level of serum uric acid in hyperuricemic rat can be efficiently reduced by the intragastric administration of DM9218-A (P<0.05). The preventive treatment of DM9218-A caused a greater reduction in serum uric acid concentration in hyperuricemic rats than the later treatment (P<0.05). Our results suggest that DM9218-A may be a promising candidate as an adjunctive treatment in patients with hyperuricemia during the onset period of disease. DM9218-A also has potential as a probiotic in the prevention of hyperuricemia in the normal population.
Collapse
Affiliation(s)
- Ming Li
- Department of Microecology, School of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Dianbin Yang
- Department of Microecology, School of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Lu Mei
- Department of Microecology, School of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Yuan
- Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ao Xie
- Department of Microecology, School of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Jieli Yuan
- Department of Microecology, School of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
- * E-mail:
| |
Collapse
|
23
|
Tovchiga OV, Shtrygol’ SY. Uric acid and central nervous system functioning (a literature review). ACTA ACUST UNITED AC 2014. [DOI: 10.1134/s2079086414030086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
24
|
Structure and function of BCRP, a broad specificity transporter of xenobiotics and endobiotics. Arch Toxicol 2014; 88:1205-48. [DOI: 10.1007/s00204-014-1224-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/06/2014] [Indexed: 12/20/2022]
|
25
|
Cicerchi C, Li N, Kratzer J, Garcia G, Roncal-Jimenez CA, Tanabe K, Hunter B, Rivard CJ, Sautin YY, Gaucher EA, Johnson RJ, Lanaspa MA. Uric acid-dependent inhibition of AMP kinase induces hepatic glucose production in diabetes and starvation: evolutionary implications of the uricase loss in hominids. FASEB J 2014; 28:3339-50. [PMID: 24755741 DOI: 10.1096/fj.13-243634] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Reduced AMP kinase (AMPK) activity has been shown to play a key deleterious role in increased hepatic gluconeogenesis in diabetes, but the mechanism whereby this occurs remains unclear. In this article, we document that another AMP-dependent enzyme, AMP deaminase (AMPD) is activated in the liver of diabetic mice, which parallels with a significant reduction in AMPK activity and a significant increase in intracellular glucose accumulation in human HepG2 cells. AMPD activation is induced by a reduction in intracellular phosphate levels, which is characteristic of insulin resistance and diabetic states. Increased gluconeogenesis is mediated by reduced TORC2 phosphorylation at Ser171 by AMPK in these cells, as well as by the up-regulation of the rate-limiting enzymes PEPCK and G6Pc. The mechanism whereby AMPD controls AMPK activation depends on the production of a specific AMP downstream metabolite through AMPD, uric acid. In this regard, humans have higher uric acid levels than most mammals due to a mutation in uricase, the enzyme involved in uric acid degradation in most mammals, that developed during a period of famine in Europe 1.5 × 10(7) yr ago. Here, working with resurrected ancestral uricases obtained from early hominids, we show that their expression on HepG2 cells is enough to blunt gluconeogenesis in parallel with an up-regulation of AMPK activity. These studies identify a key role AMPD and uric acid in mediating hepatic gluconeogenesis in the diabetic state, via a mechanism involving AMPK down-regulation and overexpression of PEPCK and G6Pc. The uricase mutation in the Miocene likely provided a survival advantage to help maintain glucose levels under conditions of near starvation, but today likely has a role in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Christina Cicerchi
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Nanxing Li
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - James Kratzer
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA; and
| | - Gabriela Garcia
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Katsuyuki Tanabe
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Brandi Hunter
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Yuri Y Sautin
- School of Medicine, University of Florida, Gainesville, Florida, USA
| | - Eric A Gaucher
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA; and
| | - Richard J Johnson
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Miguel A Lanaspa
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA;
| |
Collapse
|
26
|
Abraham A, Drory VE. Influence of serum uric acid levels on prognosis and survival in amyotrophic lateral sclerosis: a meta-analysis. J Neurol 2014; 261:1133-8. [DOI: 10.1007/s00415-014-7331-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/23/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022]
|
27
|
Evolutionary history and metabolic insights of ancient mammalian uricases. Proc Natl Acad Sci U S A 2014; 111:3763-8. [PMID: 24550457 DOI: 10.1073/pnas.1320393111] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Uricase is an enzyme involved in purine catabolism and is found in all three domains of life. Curiously, uricase is not functional in some organisms despite its role in converting highly insoluble uric acid into 5-hydroxyisourate. Of particular interest is the observation that apes, including humans, cannot oxidize uric acid, and it appears that multiple, independent evolutionary events led to the silencing or pseudogenization of the uricase gene in ancestral apes. Various arguments have been made to suggest why natural selection would allow the accumulation of uric acid despite the physiological consequences of crystallized monosodium urate acutely causing liver/kidney damage or chronically causing gout. We have applied evolutionary models to understand the history of primate uricases by resurrecting ancestral mammalian intermediates before the pseudogenization events of this gene family. Resurrected proteins reveal that ancestral uricases have steadily decreased in activity since the last common ancestor of mammals gave rise to descendent primate lineages. We were also able to determine the 3D distribution of amino acid replacements as they accumulated during evolutionary history by crystallizing a mammalian uricase protein. Further, ancient and modern uricases were stably transfected into HepG2 liver cells to test one hypothesis that uricase pseudogenization allowed ancient frugivorous apes to rapidly convert fructose into fat. Finally, pharmacokinetics of an ancient uricase injected in rodents suggest that our integrated approach provides the foundation for an evolutionarily-engineered enzyme capable of treating gout and preventing tumor lysis syndrome in human patients.
Collapse
|
28
|
Johnson RJ, Stenvinkel P, Martin SL, Jani A, Sánchez-Lozada LG, Hill JO, Lanaspa MA. Redefining metabolic syndrome as a fat storage condition based on studies of comparative physiology. Obesity (Silver Spring) 2013; 21:659-64. [PMID: 23401356 PMCID: PMC3660463 DOI: 10.1002/oby.20026] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 07/02/2012] [Indexed: 01/20/2023]
Abstract
OBJECTIVE The metabolic syndrome refers to a constellation of signs including abdominal obesity, elevated serum triglycerides, low HDL-cholesterol, elevated blood pressure, and insulin resistance. Today approximately one third of the adult population has the metabolic syndrome. While there is little doubt that the signs constituting the metabolic syndrome frequently cluster, much controversy exists over the definition, pathogenesis, or clinical utility. DESIGN AND METHODS Here we present evidence from the field of comparative physiology that the metabolic syndrome is similar to the biological process that animals engage to store fat in preparation for periods of food shortage. RESULTS We propose that the metabolic syndrome be changed to fat storage condition to more clearly align with its etiology. Obesity in humans is likely the consequences of both genetic predisposition (driven in part by thrifty genes) and environment. Recent studies suggest that the loss of the uricase gene may be one factor that predisposes humans to obesity today. CONCLUSION Understanding the process animals engage to switch from a lean insulin-sensitive to an obese insulin-resistant state may provide novel insights into the cause of obesity and diabetes in humans, and unique opportunities for reversing their pathology.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Yu R, Schellhorn HE. Recent applications of engineered animal antioxidant deficiency models in human nutrition and chronic disease. J Nutr 2013; 143:1-11. [PMID: 23173175 DOI: 10.3945/jn.112.168690] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Dietary antioxidants are essential nutrients that inhibit the oxidation of biologically important molecules and suppress the toxicity of reactive oxygen or nitrogen species. When the total antioxidant capacity is insufficient to quench these reactive species, oxidative damage occurs and contributes to the onset and progression of chronic diseases, such as neurodegenerative diseases, cardiovascular diseases, and cancer. However, epidemiological studies that examine the relationship between antioxidants and disease outcome can only identify correlative associations. Additionally, many antioxidants also have prooxidant effects. Thus, clinically relevant animal models of antioxidant function are essential for improving our understanding of the role of antioxidants in the pathogenesis of complex diseases as well as evaluating the therapeutic potential and risks of their supplementation. Recent progress in gene knockout mice and virus-based gene expression has potentiated these areas of study. Here, we review the current genetically modified animal models of dietary antioxidant function and their clinical relevance in chronic diseases. This review focuses on the 3 major antioxidants in the human body: vitamin C, vitamin E, and uric acid. We examine genetic models of vitamin C synthesis (guinea pig, Osteogenic Disorder Shionogi rat, Gulo(-/-) and SMP30(-/-) mouse mutants) and transport (Slc23a1(-/-) and Slc23a2(-/-) mouse mutants), vitamin E transport (Ttpa(-/-) mouse mutant), and uric acid synthesis (Uox(-/-) mouse mutant). The application of these models to current research goals is also discussed.
Collapse
Affiliation(s)
- Rosemary Yu
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
30
|
Pascual Izuel JM. [Uricemia and metabolic syndrome in patients with arterial hypertension]. Rev Clin Esp 2012; 212:440-1. [PMID: 22762923 DOI: 10.1016/j.rce.2012.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/14/2012] [Indexed: 11/18/2022]
|
31
|
Cebollada J, Gimeno J. Ácido úrico como factor de riesgo cardiovascular. HIPERTENSION Y RIESGO VASCULAR 2012. [DOI: 10.1016/j.hipert.2012.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Ahmad P, Ashraf M, Younis M, Hu X, Kumar A, Akram NA, Al-Qurainy F. Role of transgenic plants in agriculture and biopharming. Biotechnol Adv 2011; 30:524-40. [PMID: 21959304 DOI: 10.1016/j.biotechadv.2011.09.006] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 08/23/2011] [Accepted: 09/12/2011] [Indexed: 11/29/2022]
Abstract
At present, environmental degradation and the consistently growing population are two main problems on the planet earth. Fulfilling the needs of this growing population is quite difficult from the limited arable land available on the globe. Although there are legal, social and political barriers to the utilization of biotechnology, advances in this field have substantially improved agriculture and human life to a great extent. One of the vital tools of biotechnology is genetic engineering (GE) which is used to modify plants, animals and microorganisms according to desired needs. In fact, genetic engineering facilitates the transfer of desired characteristics into other plants which is not possible through conventional plant breeding. A variety of crops have been engineered for enhanced resistance to a multitude of stresses such as herbicides, insecticides, viruses and a combination of biotic and abiotic stresses in different crops including rice, mustard, maize, potato, tomato, etc. Apart from the use of GE in agriculture, it is being extensively employed to modify the plants for enhanced production of vaccines, hormones, etc. Vaccines against certain diseases are certainly available in the market, but most of them are very costly. Developing countries cannot afford the disease control through such cost-intensive vaccines. Alternatively, efforts are being made to produce edible vaccines which are cheap and have many advantages over the commercialized vaccines. Transgenic plants generated for this purpose are capable of expressing recombinant proteins including viral and bacterial antigens and antibodies. Common food plants like banana, tomato, rice, carrot, etc. have been used to produce vaccines against certain diseases like hepatitis B, cholera, HIV, etc. Thus, the up- and down-regulation of desired genes which are used for the modification of plants have a marked role in the improvement of genetic crops. In this review, we have comprehensively discussed the role of genetic engineering in generating transgenic lines/cultivars of different crops with improved nutrient quality, biofuel production, enhanced production of vaccines and antibodies, increased resistance against insects, herbicides, diseases and abiotic stresses as well as the safety measures for their commercialization.
Collapse
Affiliation(s)
- Parvaiz Ahmad
- Department of Botany, A.S. College, 190008, University of Kashmir, Srinagar, India.
| | | | | | | | | | | | | |
Collapse
|
33
|
Bonastre AM, Sosna M, Bartlett PN. An analysis of the kinetics of oxidation of ascorbate at poly(aniline)-poly(styrene sulfonate) modified microelectrodes. Phys Chem Chem Phys 2011; 13:5365-72. [DOI: 10.1039/c0cp02327b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
34
|
Electrodeposition of PANi films on platinum needle type microelectrodes. Application to the oxidation of ascorbate in human plasma. Anal Chim Acta 2010; 676:1-8. [DOI: 10.1016/j.aca.2010.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/29/2010] [Accepted: 07/05/2010] [Indexed: 11/19/2022]
|
35
|
Gunjaca G, Boban M, Pehlić M, Zemunik T, Budimir D, Kolcić I, Lauc G, Rudan I, Polasek O. Predictive value of 8 genetic loci for serum uric acid concentration. Croat Med J 2010; 51:23-31. [PMID: 20162742 DOI: 10.3325/cmj.2010.51.23] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM To investigate the value of genomic information in prediction of individual serum uric acid concentrations. METHODS Three population samples were investigated: from isolated Adriatic island communities of Vis (n=980) and Korcula (n=944), and from general population of the city of Split (n=507). Serum uric acid concentration was correlated with the genetic risk score based on 8 previously described genes: PDZK1, GCKR, SLC2A9, ABCG2, LRRC16A, SLC17A1, SLC16A9, and SLC22A12, represented by a total of 16 single-nucleotide polymorphisms (SNP). The data were analyzed using classification and regression tree (CART) and general linear modeling. RESULTS The most important variables for uric acid prediction with CART were genetic risk score in men and age in women. The percent of variance for any single SNP in predicting serum uric acid concentration varied from 0.0%-2.0%. The use of genetic risk score explained 0.1%-2.5% of uric acid variance in men and 3.9%-4.9% in women. The highest percent of variance was obtained when age, sex, and genetic risk score were used as predictors, with a total of 30.9% of variance in pooled analysis. CONCLUSION Despite overall low percent of explained variance, uric acid seems to be among the most predictive human quantitative traits based on the currently available SNP information. The use of genetic risk scores is a valuable approach in genetic epidemiology and increases the predictability of human quantitative traits based on genomic information compared with single SNP approach.
Collapse
Affiliation(s)
- Grgo Gunjaca
- University of Split School of Medicine, Split, Croatia
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Polasek O, Jeroncić I, Mulić R, Klismanic Z, Pehlić M, Zemunik T, Kolcić I. Common variants in SLC17A3 gene affect intra-personal variation in serum uric acid levels in longitudinal time series. Croat Med J 2010; 51:32-9. [PMID: 20162743 DOI: 10.3325/cmj.2010.51.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM To investigate whether intra-personal variation in serum uric acid concentration is influenced by genes that were described to be associated with serum uric acid levels in cross-sectional studies. METHODS The study included 92 participants from the isolated community of the Croatian island of Vis. For each participant, two uric acid concentration measurements were available, one from 2002 and one from 2003. Changes in uric acid concentration were correlated with a set of 8 genes known to affect it: PDZK1, GCKR, SLC2A9, ABCG2, LRRC16A, SLC17A3, SLC16A9, and SLC22A12. RESULTS Thirteen participants (14%) had uric acid concentration change greater than 130 micromol/L. Greater variability of uric acid concentration was recorded in women (coefficient of variation 49% vs 12% in men). Two SNPs belonging to SLC17A3 gene (rs9393672 and rs942379) yielded significant association with serum uric acid concentration changes in women. These two single-nucleotide polymorphisms (SNP) explained 0.2%-1.3% of variance for 2002 or 2003 uric acid measurement and 1.1%-1.8% of variance for the average value of these two measurements. CONCLUSIONS Repeated measurements offer a possibility to enrich the percent of explained variance and contribute to the understanding of the "missing heritability" concept. Although a number of genes have been shown to affect serum uric acid concentration, SLC17A3 seems to have a major role in determination of serum uric acid repeated measurements variation.
Collapse
Affiliation(s)
- Ozren Polasek
- Andrija Stampar School of Public Health, Medical School, University of Zagreb, Rockefellerova 4, 10000 Zagreb, Croatia.
| | | | | | | | | | | | | |
Collapse
|
37
|
Triuret as a potential hypokalemic agent: Structure characterization of triuret and triuret-alkali metal adducts by mass spectrometric techniques. Arch Biochem Biophys 2010; 498:23-34. [PMID: 20371222 DOI: 10.1016/j.abb.2010.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 03/31/2010] [Indexed: 02/07/2023]
Abstract
Triuret (also known as carbonyldiurea, dicarbamylurea, or 2,4-diimidotricarbonic diamide) is a byproduct of purine degradation in living organisms. An abundant triuret precursor is uric acid, whose level is altered in multiple metabolic pathologies. Triuret can be generated via urate oxidation by peroxynitrite, the latter being produced by the reaction of nitric oxide radical with superoxide radical anion. From this standpoint, an excess production of superoxide radical anions could indirectly favor triuret formation; however very little is known about the potential in vivo roles of this metabolite. Triuret's structure is suggestive of its ability to adopt various conformations and act as a flexible ligand for metal ions. In the current study, HPLC-MS/MS, energy-resolved mass spectrometry, selected ion monitoring, collision-induced dissociation, IRMPD spectroscopy, Fourier transform-ion cyclotron resonance mass spectrometry and computational methods were employed to characterize the structure of triuret and its metal complexes, to determine the triuret-alkali metal binding motif, and to evaluate triuret affinity toward alkali metal ions, as well as its affinity for Na(+) and K(+) relative to other organic ligands. The most favored binding motif was determined to be a bidentate chelation of triuret with the alkali metal cation involving two carbonyl oxygens. Using the complexation selectivity method, it was observed that in solution triuret has an increased affinity for potassium ions, compared to sodium and other alkali metal ions. We propose that triuret may act as a potential hypokalemic agent under pathophysiological conditions conducive to its excessive formation and thus contribute to electrolyte disorders. The collision- or photo-induced fragmentation channels of deprotonated and protonated triuret, as well as its alkali metal adducts, are likely to mimic the triuret degradation pathways in vivo.
Collapse
|
38
|
Johnson RJ, Andrews P, Benner SA, Oliver W. Theodore E. Woodward award. The evolution of obesity: insights from the mid-Miocene. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2010; 121:295-308. [PMID: 20697570 PMCID: PMC2917125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
All humans are double knockouts. Humans lack the ability to synthesize vitamin C due to a mutation in L-gulono-lactone oxidase that occurred during the late Eocene, and humans have higher serum uric acid levels due to a mutation in uricase that occurred in the mid Miocene. In this paper we review the hypothesis that these mutations have in common the induction of oxidative stress that may have had prosurvival effects to enhance the effects of fructose to increase fat stores. Fructose was the primary nutrient in fruit which was the main staple of early primates, but this food likely became less available during the global cooling that occurred at the time of these mutations. However, in today's society, the intake of fructose, primarily in the form of added sugars, has skyrocketed, while the intake of natural fruits high in vitamin C has fallen. We suggest that it is the interaction of these genetic changes with diet that is responsible for the obesity epidemic today. Hence, we propose that Neel's thrifty gene hypothesis is supported by these new insights into the mechanisms regulating fructose metabolism.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, CO 80016, USA.
| | | | | | | |
Collapse
|
39
|
Monoamine oxidase A gene polymorphisms and enzyme activity associated with risk of gout in Taiwan aborigines. Hum Genet 2009; 127:223-9. [DOI: 10.1007/s00439-009-0765-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Accepted: 11/02/2009] [Indexed: 10/20/2022]
|
40
|
Improved rapid assay of plasma uric acid by short-end injection capillary zone electrophoresis. Anal Bioanal Chem 2009; 395:2577-82. [DOI: 10.1007/s00216-009-3171-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2009] [Revised: 08/17/2009] [Accepted: 09/18/2009] [Indexed: 01/08/2023]
|
41
|
Hermans MP, Ahn SA, Rousseau MF. Raised natriuretic peptides, big-endothelin-1 and improved beta-cell function in type 2 diabetic males with hyperuricaemia. Diab Vasc Dis Res 2009; 6:190-3. [PMID: 20368210 DOI: 10.1177/1479164109336689] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Urate, a naturally-occurring antioxidant, is a marker/factor for cardiovascular disease. Hyperuricaemia is associated with IR, MetS and endothelial dysfunction. We characterised the associations between neurohormones, uricaemia, and glucose homeostasis in type 2 diabetes mellitus (T2DM) males. Cross-sectional; 705 T2DM males divided into two groups: uric acid < 7.0 mg/dl (normouricaemic; n=476) versus uric acid >or= 7.0 mg/dl (hyperuricaemic; n=229). HOMA beta-cell function (B), insulin sensitivity (S), hyperbolic product (BxS), and (BxS) loss rate were determined alongside neurohormones (Nt-proANP, BNP, Big ET-1 and UII). Mean age and diabetes duration were not different between groups. Hyperuricaemics had more macroangiopathy, total/central adiposity, IR, hypertension, dyslipidemia and MetS prevalence. Nt-proANP and BNP levels were more than twice as high in hyperuricaemics, whereas Big ET-1 and UII were higher by 46% and 14%, respectively. HOMA (BxS) was higher in hyperuricaemics: 31 (16)% vs. 26 (18)% (p=0.0004). BxS loss rate was faster in normouricaemics: 1.36 (0.54)% vs. 1.20 (0.43)%/year(-1) (p<0.0001 ). The proportion with HbA(1C) < 7.0% was 39% (normouricaemics) vs. 49% (hyperuricaemics; p=0.0091). In T2DM males, hyperuricaemia is associated with raised neurohormones together with better beta-cell indices. Urate's dual properties may translate into beneficial (glucose homeostasis) and detrimental (raised neurohormones) effects.
Collapse
Affiliation(s)
- Michel P Hermans
- Endocrinology and Nutrition Unit, Cliniques Universitaires St-Luc, Brussels, Belgium.
| | | | | |
Collapse
|
42
|
Abstract
Although ascorbic acid is an important water-soluble antioxidant and enzyme cofactor in plants and animals, humans and some other species do not synthesize ascorbate due to the lack of the enzyme catalyzing the final step of the biosynthetic pathway, and for them it has become a vitamin. This review focuses on the role of ascorbate in various hydroxylation reactions and in the redox homeostasis of subcellular compartments including mitochondria and endoplasmic reticulum. Recently discovered functions of ascorbate in nucleic acid and histone dealkylation and proteoglycan deglycanation are also summarized. These new findings might delineate a role for ascorbate in the modulation of both pro- and anti-carcinogenic mechanisms. Recent advances and perspectives in therapeutic applications are also reviewed. On the basis of new and earlier observations, the advantages of the lost ability to synthesize ascorbate are pondered. The increasing knowledge of the functions of ascorbate and of its molecular sites of action can mechanistically substantiate a place for ascorbate in the treatment of various diseases.
Collapse
Affiliation(s)
- J Mandl
- Department of Medical Chemistry, Molecular Biology and Patobiochemistry, Semmelweis University Budapest, Budapest, Hungary.
| | | | | |
Collapse
|
43
|
Martí N, Mena P, Cánovas JA, Micol V, Saura D. Vitamin C and the Role of Citrus Juices as Functional Food. Nat Prod Commun 2009. [DOI: 10.1177/1934578x0900400506] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The literature on the content and stability of vitamin C (ascorbic acid, AA) in citrus juices in relation to industrial practices is reviewed. The role of vitamin C from citrus juices in human diet is also reviewed. Citrus fruits and juices are rich in several types of bioactive compounds. Their antioxidant activity and related benefits derive not only from vitamin C but also from other phytochemicals, mainly flavonoids. During juice processing, temperature and oxygen are the main factors responsible for vitamin C losses. Nonthermal processed juices retain higher levels of vitamin C, but economic factors apparently delay the use of such methods in the citrus industry. Regarding packing material, vitamin C in fruit juice is quite stable when stored in metal or glass containers, whereas juice stored in plastic bottles has a much shorter shelf-life. The limiting step for vitamin C absorption in humans is transcellular active transport across the intestinal wall where AA may be oxidized to dehydroascorbic acid (DHAA), which is easily transported across the cell membrane and immediately reduced back to AA by two major pathways. AA bioavailability in the presence of flavonoids has yielded controversial results. Whereas flavonoids seem to inhibit intestinal absorption of AA, some studies have shown that AA in citrus extract was more available than synthetic ascorbic acid alone. DHAA is reported to possess equivalent biological activity to AA, so recent studies often consider the vitamin C activity in the diet as the sum of AA plus DHAA. However, this claimed equivalence should be carefully reexamined. Humans are one of the few species lacking the enzyme (L-gulonolactone oxidase, GLO) to convert glucose to vitamin C. It has been suggested that this is due to a mutation that provided a survival advantage to early primates, since GLO produces toxic H2O2. Furthermore, the high concentration of AA (and DHAA) in neural tissues could have been the key factor that caused primates (vertebrates with relative big brain) to lose the capacity to synthesize vitamin C. Oxidative damage has many pathological implications in human health, and AA may play a central role in maintaining the metabolic antioxidant response. The abundance of citrus juices in the Mediterranean diet may provide the main dietary source for natural vitamin C.
Collapse
Affiliation(s)
- Nuria Martí
- Unidad Mixta JBT Corp.-IBMC. Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Orihuela, Alicante. Spain
| | - Pedro Mena
- Unidad Mixta JBT Corp.-IBMC. Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Orihuela, Alicante. Spain
| | - Jose Antonio Cánovas
- Unidad Mixta JBT Corp.-IBMC. Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Orihuela, Alicante. Spain
| | - Vicente Micol
- Unidad Mixta JBT Corp.-IBMC. Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Orihuela, Alicante. Spain
| | - Domingo Saura
- Unidad Mixta JBT Corp.-IBMC. Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Orihuela, Alicante. Spain
| |
Collapse
|
44
|
|
45
|
Johnson RJ, Perez-Pozo SE, Sautin YY, Manitius J, Sanchez-Lozada LG, Feig DI, Shafiu M, Segal M, Glassock RJ, Shimada M, Roncal C, Nakagawa T. Hypothesis: could excessive fructose intake and uric acid cause type 2 diabetes? Endocr Rev 2009; 30:96-116. [PMID: 19151107 PMCID: PMC2647706 DOI: 10.1210/er.2008-0033] [Citation(s) in RCA: 328] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 12/31/2008] [Indexed: 02/07/2023]
Abstract
We propose that excessive fructose intake (>50 g/d) may be one of the underlying etiologies of metabolic syndrome and type 2 diabetes. The primary sources of fructose are sugar (sucrose) and high fructose corn syrup. First, fructose intake correlates closely with the rate of diabetes worldwide. Second, unlike other sugars, the ingestion of excessive fructose induces features of metabolic syndrome in both laboratory animals and humans. Third, fructose appears to mediate the metabolic syndrome in part by raising uric acid, and there are now extensive experimental and clinical data supporting uric acid in the pathogenesis of metabolic syndrome. Fourth, environmental and genetic considerations provide a potential explanation of why certain groups might be more susceptible to developing diabetes. Finally, we discuss the counterarguments associated with the hypothesis and a potential explanation for these findings. If diabetes might result from excessive intake of fructose, then simple public health measures could have a major impact on improving the overall health of our populace.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Nephrology, Hypertension and Transplantation, University of Florida, P.O. Box 100224, Gainesville, Florida 32620-0224, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Johnson RJ, Sautin YY, Oliver WJ, Roncal C, Mu W, Sanchez-Lozada LG, Rodriguez-Iturbe B, Nakagawa T, Benner SA. Lessons from comparative physiology: could uric acid represent a physiologic alarm signal gone awry in western society? J Comp Physiol B 2009; 179:67-76. [PMID: 18649082 PMCID: PMC2684327 DOI: 10.1007/s00360-008-0291-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 06/18/2008] [Accepted: 06/23/2008] [Indexed: 02/07/2023]
Abstract
Uric acid has historically been viewed as a purine metabolic waste product excreted by the kidney and gut that is relatively unimportant other than its penchant to crystallize in joints to cause the disease gout. In recent years, however, there has been the realization that uric acid is not biologically inert but may have a wide range of actions, including being both a pro- and anti-oxidant, a neurostimulant, and an inducer of inflammation and activator of the innate immune response. In this paper, we present the hypothesis that uric acid has a key role in the foraging response associated with starvation and fasting. We further suggest that there is a complex interplay between fructose, uric acid and vitamin C, with fructose and uric acid stimulating the foraging response and vitamin C countering this response. Finally, we suggest that the mutations in ascorbate synthesis and uricase that characterized early primate evolution were likely in response to the need to stimulate the foraging "survival" response and might have inadvertently had a role in accelerating the development of bipedal locomotion and intellectual development. Unfortunately, due to marked changes in the diet, resulting in dramatic increases in fructose- and purine-rich foods, these identical genotypic changes may be largely responsible for the epidemic of obesity, diabetes and cardiovascular disease in today's society.
Collapse
Affiliation(s)
- Richard J. Johnson
- Division of Nephrology, Hypertension and Transplantation, University of Florida, PO Box 100224, Gainesville, FL 32610, USA, e-mail:
| | - Yuri Y. Sautin
- Division of Nephrology, Hypertension and Transplantation, University of Florida, PO Box 100224, Gainesville, FL 32610, USA, e-mail:
| | | | - Carlos Roncal
- Division of Nephrology, Hypertension and Transplantation, University of Florida, PO Box 100224, Gainesville, FL 32610, USA, e-mail:
| | - Wei Mu
- Division of Nephrology, Hypertension and Transplantation, University of Florida, PO Box 100224, Gainesville, FL 32610, USA, e-mail:
| | | | | | - Takahiko Nakagawa
- Division of Nephrology, Hypertension and Transplantation, University of Florida, PO Box 100224, Gainesville, FL 32610, USA, e-mail:
| | | |
Collapse
|