1
|
Sas DJ, Bakkaloglu SA, Belostotsky V, Hayes W, Ariceta G, Zhou J, Rawson V. Nedosiran in pediatric patients with PH1 and relatively preserved kidney function, a phase 2 study (PHYOX8). Pediatr Nephrol 2025; 40:1939-1948. [PMID: 39875734 PMCID: PMC12031765 DOI: 10.1007/s00467-025-06675-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/06/2024] [Accepted: 01/05/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Primary hyperoxaluria type 1 (PH1) is an autosomal recessive disorder with dysregulated glyoxylate metabolism in the liver. Oxalate over-production leads to renal stones, progressive kidney damage and renal failure, with potentially life-threatening systemic oxalosis. Nedosiran is a synthetic RNA interference therapy, designed to reduce hepatic lactate dehydrogenase (LDH) to decrease oxalate burden in PH. METHODS Currently, in the PHYOX8 study (NCT05001269), pediatric participants (2-11 years) with PH1 (N = 15) and estimated glomerular filtration rate (eGFR) ≥ 30mL/min/1.73m2 received nedosiran once monthly for 6 months. RESULTS Urinary oxalate:creatinine (Uox:Ucr) levels reduced by 64% on average. Mean Uox:Ucr reduction was 52% at day 60 and ˃60% at day 180. At one or more study visits, 93.3% (N = 14) of participants reached Uox:Ucr < 1.5 × upper limit of normal (ULN), and 53.3% (N = 8) reached ≤ 1.0 × ULN. Median percent change in plasma oxalate (12.0 µmol/L at baseline) to day 180 was -39.23% (n = 10). Average number of kidney stones per participant remained stable, whilst a 10.1% average decrease in summed surface area was observed. Median percent change from baseline in eGFR was 2.5%, indicating preservation of renal function. CONCLUSIONS Nedosiran was well tolerated, with only 3 participants experiencing at least one serious adverse event, none considered treatment-related. The incidence of injection site reactions was 6.7% (1/15 participants). In conclusion, nedosiran treatment led to a significant and sustained reduction of Uox levels in children with PH1. These findings support nedosiran treatment in pediatric patients to reduce Uox and shows promise for limiting PH1-related complications.
Collapse
Affiliation(s)
- David J Sas
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| | | | | | - Wesley Hayes
- Department of Pediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Gema Ariceta
- Pediatric Nephrology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Jing Zhou
- Novo Nordisk A/S, Lexington, MA, USA
| | | |
Collapse
|
2
|
Dindo M, Conter C, Uechi GI, Pampalone G, Ruta L, Pey AL, Rossi L, Laurino P, Magnani M, Cellini B. Engineered Oxalate Decarboxylase Boosts Activity and Stability for Biological Applications. ACS OMEGA 2025; 10:12375-12384. [PMID: 40191304 PMCID: PMC11966277 DOI: 10.1021/acsomega.4c11434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
Oxalate decarboxylase (OxDC) from Bacillus subtilis is a Mn-dependent hexameric enzyme that converts oxalate to carbon dioxide and formate. Recently, OxDC has attracted the interest of the scientific community due to its biotechnological and medical applications for the treatment of hyperoxaluria, a group of pathologic conditions associated with excessive oxalate urinary excretion caused by either increased endogenous production or increased exogenous absorption. The fact that OxDC displays optimum pH in the acidic range represents a big limitation for most biotechnological applications involving processes occurring at neutral pH, where the activity and stability of the enzyme are remarkably reduced. Here, through bioinformatics-guided protein engineering followed by combinatorial mutagenesis and analyses of activity and thermal stability, we identified a double mutant of OxDC endowed with enhanced catalytic efficiency and stability under physiological conditions. The obtained engineered form of OxDC offers a potential tool for improved intestinal oxalate degradation in hyperoxaluria patients.
Collapse
Affiliation(s)
- Mirco Dindo
- Department
of Medicine and Surgery, Section of Physiology and Biochemistry, University of Perugia, 06132 Perugia, Italy
- Protein
Engineering and Evolution Unit, Okinawa
Institute of Science and Technology (OIST), Onna, Okinawa 904-0495, Japan
| | - Carolina Conter
- Center
of Cooperative Research in Biosciences (CIC bioGUNE) Basque Research
and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Gen-Ichiro Uechi
- Protein
Engineering and Evolution Unit, Okinawa
Institute of Science and Technology (OIST), Onna, Okinawa 904-0495, Japan
| | - Gioena Pampalone
- Department
of Medicine and Surgery, Section of Physiology and Biochemistry, University of Perugia, 06132 Perugia, Italy
| | - Luana Ruta
- Department
of Medicine and Surgery, Section of Physiology and Biochemistry, University of Perugia, 06132 Perugia, Italy
| | - Angel L. Pey
- Department
de Quimica Fisica, Unidad de Excelencia en Quimica Aplicada a Biomedicina
y Medioambiente e Instituto de Biotecnologia, Universidad de Granada, Granada 18071, Spain
| | - Luigia Rossi
- Department
of Biomolecular Sciences, University of
Urbino “Carlo Bo”, Urbino 61029, Italy
| | - Paola Laurino
- Protein
Engineering and Evolution Unit, Okinawa
Institute of Science and Technology (OIST), Onna, Okinawa 904-0495, Japan
- Institute
of Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mauro Magnani
- Department
of Biomolecular Sciences, University of
Urbino “Carlo Bo”, Urbino 61029, Italy
| | - Barbara Cellini
- Department
of Medicine and Surgery, Section of Physiology and Biochemistry, University of Perugia, 06132 Perugia, Italy
| |
Collapse
|
3
|
Wanders RJA, Groothoff JW, Deesker LJ, Salido E, Garrelfs SF. Human glyoxylate metabolism revisited: New insights pointing to multi-organ involvement with implications for siRNA-based therapies in primary hyperoxaluria. J Inherit Metab Dis 2025; 48:e12817. [PMID: 39582099 PMCID: PMC11670150 DOI: 10.1002/jimd.12817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Glyoxylate is a toxic metabolite because of its rapid conversion into oxalate, as catalyzed by the ubiquitous enzyme lactate dehydrogenase. This requires the presence of efficient glyoxylate detoxification systems in multiple subcellular compartments, as glyoxylate is produced in peroxisomes, mitochondria, and the cytosol. Alanine glyoxylate aminotransferase (AGT) and glyoxylate reductase/hydroxypyruvate reductase (GRHPR) are the key enzymes involved in glyoxylate detoxification. Bi-allelic mutations in the genes coding for these enzymes cause primary hyperoxaluria type 1 (PH1) and 2 (PH2), respectively. Glyoxylate is derived from various sources, including 4-hydroxyproline, which is degraded in mitochondria, generating pyruvate and glyoxylate, as catalyzed by the mitochondrial enzyme 4-hydroxy-2-oxoglutarate aldolase (HOGA); however, counterintuitively, a defect in HOGA1 is the molecular basis of primary hyperoxaluria type 3 (PH3). Irrespective of its underlying cause, hyperoxaluria in humans leads to nephrocalcinosis, recurrent urolithiasis, and kidney damage, which may culminate in kidney failure requiring combined liver-kidney transplantation in severely affected patients. In the past few years, therapeutic options, especially for primary hyperoxaluria type 1 (PH1), have greatly been improved thanks to the introduction of two RNAi-based therapies that inhibit either the production of glycolate oxidase (lumasiran) or lactate dehydrogenase (nedosiran). While lumasiran only targets PH1 patients, nedosiran was specifically developed to target all three subtypes of PH. Inspired by the findings reported in the literature that nedosiran effectively reduced urinary oxalate excretion in PH1 patients but not in PH2 or PH3 patients, we have now revisited glyoxylate metabolism in humans and performed a thorough literature study which revealed that glyoxylate/oxalate metabolism is not confined to the liver but instead involves multiple different organs. This new view on glyoxylate/oxalate metabolism in humans may well explain the disappointing results of nedosiran in PH2 and PH3, and provides new clues for the future generation of new therapeutic strategies for PH2 and PH3.
Collapse
Affiliation(s)
- Ronald J. A. Wanders
- Department of Pediatric NephrologyEmma Children's HospitalAmsterdamThe Netherlands
- Department of Laboratory Medicine, Laboratory Genetic Metabolic DiseasesAmsterdam UMCAmsterdamThe Netherlands
| | - Jaap W. Groothoff
- Department of Pediatric NephrologyEmma Children's HospitalAmsterdamThe Netherlands
| | - Lisa J. Deesker
- Department of Pediatric NephrologyEmma Children's HospitalAmsterdamThe Netherlands
| | - Eduardo Salido
- Hospital Universitario de Canarias, Centre for Biomedical Research on rare Diseases (CIBERER)Universidad La LagunaTenerifeSpain
| | - Sander F. Garrelfs
- Department of Pediatric NephrologyEmma Children's HospitalAmsterdamThe Netherlands
| |
Collapse
|
4
|
Cellini B, Baum MA, Frishberg Y, Groothoff JW, Harris PC, Hulton SA, Knauf F, Knight J, Lieske JC, Lowther WT, Moochhala S, Nazzal L, Tasian GE, Whittamore JM, Sas DJ. Opportunities in Primary and Enteric Hyperoxaluria at the Cross-Roads Between the Clinic and Laboratory. Kidney Int Rep 2024; 9:3083-3096. [PMID: 39534212 PMCID: PMC11551133 DOI: 10.1016/j.ekir.2024.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/26/2024] [Indexed: 11/16/2024] Open
Abstract
Hyperoxaluria is a condition in which there is a pathologic abundance of oxalate in the urine through either hepatic overproduction (primary hyperoxaluria [PH]) or excessive enteric absorption of dietary oxalate (enteric hyperoxaluria [EH]). Severity can vary with the most severe forms causing kidney failure and extrarenal manifestations. To address the current challenges and innovations in hyperoxaluria, the 14th International Hyperoxaluria Workshop convened in Perugia, Italy, bringing together international experts for focused presentation and discussion. The objective of the following report was to disseminate an overview of the proceedings and provide substrate for further thought. The format of this paper follows the format of the meeting, addressing, "PH type 1" (PH1) first, followed by "surgery, genetics, and ethics in PH", then "PH types 2 and 3," (PH2 and PH3) and, finally, "EH." Each session began with presentations of the current clinical challenges, followed by discussion of the latest advances in basic and translational research, and concluded with interactive discussions about prioritizing the future of research in the field to best serve the need of the patients.
Collapse
Affiliation(s)
- Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Michelle A. Baum
- Department of Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Yaacov Frishberg
- Division of Pediatric Nephrology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Jaap W. Groothoff
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Peter C. Harris
- Division of Nephrology and Hypertension and Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sally A. Hulton
- Department of Nephrology, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Felix Knauf
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - John Knight
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John C. Lieske
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - W. Todd Lowther
- Center for Structural Biology, Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Lama Nazzal
- Nephrology Division, NYU Langone Health and NYU Grossman School of Medicine, New York, New York, USA
| | - Gregory E. Tasian
- Division of Pediatric Urology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jonathan M. Whittamore
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research UT Southwestern Medical Center, Dallas, Texas, USA
| | - David J. Sas
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic Children’s Center, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Messing M, Torres JA, Holznecht N, Weimbs T. Trigger Warning: How Modern Diet, Lifestyle, and Environment Pull the Trigger on Autosomal Dominant Polycystic Kidney Disease Progression. Nutrients 2024; 16:3281. [PMID: 39408247 PMCID: PMC11479178 DOI: 10.3390/nu16193281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Understanding chronic kidney disease (CKD) through the lens of evolutionary biology highlights the mismatch between our Paleolithic-optimized genes and modern diets, which led to the dramatically increased prevalence of CKD in modern societies. In particular, the Standard American Diet (SAD), high in carbohydrates and ultra-processed foods, causes conditions like type 2 diabetes (T2D), chronic inflammation, and hypertension, leading to CKD. Autosomal dominant polycystic kidney disease (ADPKD), a genetic form of CKD, is characterized by progressive renal cystogenesis that leads to renal failure. This review challenges the fatalistic view of ADPKD as solely a genetic disease. We argue that, just like non-genetic CKD, modern dietary practices, lifestyle, and environmental exposures initiate and accelerate ADPKD progression. Evidence shows that carbohydrate overconsumption, hyperglycemia, and insulin resistance significantly impact renal health. Additionally, factors like dehydration, electrolyte imbalances, nephrotoxin exposure, gastrointestinal dysbiosis, and renal microcrystal formation exacerbate ADPKD. Conversely, carbohydrate restriction, ketogenic metabolic therapy (KMT), and antagonizing the lithogenic risk show promise in slowing ADPKD progression. Addressing disease triggers through dietary modifications and lifestyle changes offers a conservative, non-pharmacological strategy for disease modification in ADPKD. This comprehensive review underscores the urgency of integrating diet and lifestyle factors into the clinical management of ADPKD to mitigate disease progression, improve patient outcomes, and offer therapeutic choices that can be implemented worldwide at low or no cost to healthcare payers and patients.
Collapse
Affiliation(s)
| | | | | | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA; (M.M.); (J.A.T.); (N.H.)
| |
Collapse
|
6
|
Li X, Cunneely OP, Fargue S, Wood KD, Assimos DG, Knight J. 4-hydroxy-2-oxoglutarate metabolism in a mouse model of Primary Hyperoxaluria Type 3. Biochem Biophys Rep 2024; 39:101765. [PMID: 39040543 PMCID: PMC11261398 DOI: 10.1016/j.bbrep.2024.101765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Primary Hyperoxaluria Type 3 (PH3) results from 4-hydroxy-2-oxoglutarate (HOG) aldolase (HOGA) deficiency, which causes an increase in endogenous oxalate synthesis leading to calcium oxalate kidney stone disease. The mechanisms underlying HOG metabolism and increased oxalate synthesis in PH3 are not well understood. We used a Hoga1 knock-out mouse model of PH3 to investigate two aspects of HOG metabolism: reduction to dihydroxyglutarate (DHG), a pathway that may limit oxalate synthesis in PH3, and metabolism to glyoxylate, which is a direct precursor to oxalate. The metabolism of HOG to DHG was highest in liver and kidney cortical tissue, enhanced in the cytosolic compartment of the liver, and preferred NADPH as a cofactor. In the absence of HOGA, HOG to glyoxylate aldolase activity was highest in liver mitoplasts, with no activity present in brain tissue lysates. These findings will assist in the identification of enzymes responsible for the metabolism of HOG to DHG and glyoxylate, which may lead to novel therapeutic approaches to limit oxalate synthesis in those afflicted with PH3.
Collapse
Affiliation(s)
- Xingsheng Li
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Owen P. Cunneely
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sonia Fargue
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyle D. Wood
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dean G. Assimos
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John Knight
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
7
|
Cellini B. A molecular journey on the pathogenesis of primary hyperoxaluria. Curr Opin Nephrol Hypertens 2024; 33:398-404. [PMID: 38602143 PMCID: PMC11139248 DOI: 10.1097/mnh.0000000000000987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Primary hyperoxalurias (PHs) are rare disorders caused by the deficit of liver enzymes involved in glyoxylate metabolism. Their main hallmark is the increased excretion of oxalate leading to the deposition of calcium oxalate stones in the urinary tract. This review describes the molecular aspects of PHs and their relevance for the clinical management of patients. RECENT FINDINGS Recently, the study of PHs pathogenesis has received great attention. The development of novel in vitro and in vivo models has allowed to elucidate how inherited mutations lead to enzyme deficit, as well as to confirm the pathogenicity of newly-identified mutations. In addition, a better knowledge of the metabolic consequences in disorders of liver glyoxylate detoxification has been crucial to identify the key players in liver oxalate production, thus leading to the identification and validation of new drug targets. SUMMARY The research on PHs at basic, translational and clinical level has improved our knowledge on the critical factors that modulate disease severity and the response to the available treatments, leading to the development of new drugs, either in preclinical stage or, very recently, approved for patient treatment.
Collapse
Affiliation(s)
- Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
8
|
Desenclos J, Forté V, Clément C, Daudon M, Letavernier E. Pathophysiology and management of enteric hyperoxaluria. Clin Res Hepatol Gastroenterol 2024; 48:102322. [PMID: 38503362 DOI: 10.1016/j.clinre.2024.102322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/21/2024]
Abstract
Enteric hyperoxaluria is a metabolic disorder resulting from conditions associated with fatty acid malabsorption and characterized by an increased urinary output of oxalate. Oxalate is excessively absorbed in the gut and then excreted in urine where it forms calcium oxalate crystals, inducing kidney stones formation and crystalline nephropathies. Enteric hyperoxaluria is probably underdiagnosed and may silently damage kidney function of patients affected by bowel diseases. Moreover, the prevalence of enteric hyperoxaluria has increased because of the development of bariatric surgical procedures. Therapeutic options are based on the treatment of the underlying disease, limitation of oxalate intakes, increase in calcium salts intakes but also increase in urine volume and correction of hypocitraturia. There are few data regarding the natural evolution of kidney stone events and chronic kidney disease in these patients, and there is a need for new treatments limiting kidney injury by calcium oxalate crystallization.
Collapse
Affiliation(s)
- Jordan Desenclos
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, 4 rue de la Chine, Paris F-75020, France
| | - Valentine Forté
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, 4 rue de la Chine, Paris F-75020, France
| | - Cécile Clément
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, 4 rue de la Chine, Paris F-75020, France
| | - Michel Daudon
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, 4 rue de la Chine, Paris F-75020, France; INSERM, UMR S 1155, Paris F-75020, France
| | - Emmanuel Letavernier
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, 4 rue de la Chine, Paris F-75020, France; INSERM, UMR S 1155, Paris F-75020, France; Sorbonne Université, UMR S 1155, Paris F-75020, France.
| |
Collapse
|
9
|
Naguib S, Mansour LA, Soliman NA, El-Hanafy HM, Fahmy YA, Elmonem MA, Halim RMA. Expanding the Genetic Spectrum of AGXT Gene Variants in Egyptian Patients with Primary Hyperoxaluria Type I. Genet Test Mol Biomarkers 2024; 28:151-158. [PMID: 38657121 DOI: 10.1089/gtmb.2023.0525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Introduction: Approximately 80% of primary hyperoxaluria cases are caused by primary hyperoxaluria type 1 (PH1, OMIM# 259900), which is characterized by pathogenic variants in the AGXT gene, resulting in deficiency of the liver-specific enzyme alanine-glyoxylate aminotransferase (AGT). This leads to increased production of oxalate, which cannot be effectively eliminated from the body, resulting in its accumulation primarily in the kidneys and other organs. Subjects and Methods: This study included 17 PH1 Egyptian patients from 12 unrelated families, recruited from the Inherited Kidney Disease Outpatient Clinic and the Dialysis Units, Cairo University Hospitals, during the period from January 2018 to December 2019, aiming to identify the pathogenic variants in the AGXT gene. Results: Six different variants were detected. These included three frameshift and three missense variants, all found in homozygosity within the respective families. The most common variant was c.121G>A;p.(Gly41Arg) detected in four families, followed by c.725dup;p.(Asp243GlyfsTer12) in three families, c.33dup;p.(Lys12Glnfs156) in two families, and c.731T >C;p.(Ile244Thr), c.33delC;p.(Lys12Argfs34), and c.568G>A;p.(Gly190Arg) detected in one family each. Conclusion: Consanguineous Egyptian families with history of renal stones or renal disease suspicious of primary hyperoxaluria should undergo AGXT genetic sequencing, specifically targeting exons 1 and 7, as variants in these two exons account for >75% of disease-causing variants in Egyptian patients with confirmed PH1.
Collapse
Affiliation(s)
- Somayya Naguib
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Lamiaa A Mansour
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Egyptian Group for Orphan Renal Disease (EGORD), Cairo, Egypt
- Department of Clinical Genetics, Egypt Center for Research and Regenerative Medicine (ECRRM), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hadeel M El-Hanafy
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yosra A Fahmy
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Egyptian Group for Orphan Renal Disease (EGORD), Cairo, Egypt
| | - Mohamed A Elmonem
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Clinical Genetics, Egypt Center for Research and Regenerative Medicine (ECRRM), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Radwa M Abdel Halim
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Clinical and Chemical Pathology, Faculty of Medicine, New Giza University, Giza, Egypt
| |
Collapse
|
10
|
Bons J, Tadeo A, Scott GK, Teramayi F, Tanner JJ, Schilling B, Benz CC, Ellerby LM. Therapeutic targeting of HYPDH/PRODH2 with N-propargylglycine offers a Hyperoxaluria treatment opportunity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166848. [PMID: 37586438 PMCID: PMC10854995 DOI: 10.1016/j.bbadis.2023.166848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
N-propargylglycine prevents 4-hydroxyproline catabolism in mouse liver and kidney. N-propargylglycine is a novel suicide inhibitor of PRODH2 and induces mitochondrial degradation of PRODH2. PRODH2 is selectively expressed in liver and kidney and contributes to primary hyperoxaluria (PH). Preclinical evaluation of N-propargylglycine efficacy as a new PH therapeutic is warranted.
Collapse
Affiliation(s)
- Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Ada Tadeo
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Gary K. Scott
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - John J. Tanner
- Departments of Biochemistry and Chemistry, University of Missouri, Columbia, MO, USA
| | | | | | | |
Collapse
|
11
|
Pang J, Huang Y, Liu Y, Huang W. Applications of ion chromatography in urine analysis: A review. J Chromatogr A 2023; 1706:464231. [PMID: 37517316 DOI: 10.1016/j.chroma.2023.464231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Ion chromatography (IC) plays a crucial role in urine analysis for diverse medical diagnoses. This paper reviews a comprehensive investigation into urine pretreatment techniques, as well as the design and development of IC systems for the measurement of various chemicals. Prior to analysis, urine samples commonly undergo pretreatment procedures such as dilution, filtration, purification, and concentration. These steps effectively eliminate interfering factors and facilitate the accurate and sensitive analysis of ultra-trace components. To separate and quantify different chemical elements or ions present in urine, a range of homemade or commercially available columns coupled with various detectors were employed. This study focuses on the analysis of chemicals such as heavy metals, halogens, pesticides, drugs, and other essential or toxic substances by IC methods.
Collapse
Affiliation(s)
- Jiafeng Pang
- Hubei Key Laboratory of Yangtze Catchment Environmental Aquatic Science, School of Environmental Studies, China University of Geosciences, Wuhan 430078, Hubei, China
| | - Yongming Huang
- Hubei Key Laboratory of Yangtze Catchment Environmental Aquatic Science, School of Environmental Studies, China University of Geosciences, Wuhan 430078, Hubei, China
| | - Yanli Liu
- Hubei Key Laboratory of Yangtze Catchment Environmental Aquatic Science, School of Environmental Studies, China University of Geosciences, Wuhan 430078, Hubei, China
| | - Weixiong Huang
- Hubei Key Laboratory of Yangtze Catchment Environmental Aquatic Science, School of Environmental Studies, China University of Geosciences, Wuhan 430078, Hubei, China.
| |
Collapse
|
12
|
Fargue S, Wood KD, Crivelli JJ, Assimos DG, Oster RA, Knight J. Endogenous Oxalate Synthesis and Urinary Oxalate Excretion. J Am Soc Nephrol 2023; 34:1505-1507. [PMID: 37312251 PMCID: PMC10482058 DOI: 10.1681/asn.0000000000000176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/06/2023] [Indexed: 06/15/2023] Open
Affiliation(s)
- Sonia Fargue
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kyle D. Wood
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joseph J. Crivelli
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dean G. Assimos
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert A. Oster
- Division of Preventive Medicine, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - John Knight
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
13
|
Abstract
Oxalate homeostasis is maintained through a delicate balance between endogenous sources, exogenous supply and excretion from the body. Novel studies have shed light on the essential roles of metabolic pathways, the microbiome, epithelial oxalate transporters, and adequate oxalate excretion to maintain oxalate homeostasis. In patients with primary or secondary hyperoxaluria, nephrolithiasis, acute or chronic oxalate nephropathy, or chronic kidney disease irrespective of aetiology, one or more of these elements are disrupted. The consequent impairment in oxalate homeostasis can trigger localized and systemic inflammation, progressive kidney disease and cardiovascular complications, including sudden cardiac death. Although kidney replacement therapy is the standard method for controlling elevated plasma oxalate concentrations in patients with kidney failure requiring dialysis, more research is needed to define effective elimination strategies at earlier stages of kidney disease. Beyond well-known interventions (such as dietary modifications), novel therapeutics (such as small interfering RNA gene silencers, recombinant oxalate-degrading enzymes and oxalate-degrading bacterial strains) hold promise to improve the outlook of patients with oxalate-related diseases. In addition, experimental evidence suggests that anti-inflammatory medications might represent another approach to mitigating or resolving oxalate-induced conditions.
Collapse
Affiliation(s)
- Theresa Ermer
- Department of Surgery, Division of Thoracic Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Lama Nazzal
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Clarissa Tio
- Division of Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sushrut Waikar
- Department of Medicine, Section of Nephrology, Boston University, Boston, MA, USA
| | - Peter S Aronson
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Felix Knauf
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, USA.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
14
|
Popovics P, Penniston KL. Current research and future directions in non-malignant urologic research - proceedings of the annual CAIRIBU meeting. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:449-461. [PMID: 36636691 PMCID: PMC9831912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/25/2022] [Indexed: 01/14/2023]
Abstract
The Annual Collaborating for the Advancement of Interdisciplinary Research (CAIRIBU) Meeting in 2022 highlighted basic, translational, and clinical non-malignant urology research within five main areas affecting the urinary tract: urinary dysfunction due to prostate disease, microbes and infection, bladder function and physiology, neurology and neuromuscular influences and calculi and obstruction. In this paper, we summarize main findings and future directions outlined by CAIRIBU-affiliated scientists who presented as part of the scientific sessions.
Collapse
Affiliation(s)
- Petra Popovics
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical SchoolVA, USA
| | - Kristina L Penniston
- Department of Urology, University of Wisconsin School of Medicine and Public HealthWI, USA
| |
Collapse
|
15
|
Probiotic Oxalate-Degrading Bacteria: New Insight of Environmental Variables and Expression of the oxc and frc Genes on Oxalate Degradation Activity. Foods 2022; 11:foods11182876. [PMID: 36141002 PMCID: PMC9498451 DOI: 10.3390/foods11182876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Oxalate, a compound produced by many edible plants and as a terminal metabolite in the liver of mammals, is a toxin that has a detrimental role to human health. Humans and other mammals do possess enzymatic systems to degrade oxalate. Moreover, numerous oxalate-degrading bacteria reside in the mammalian gut and, thus, provide an important function for hosts. The current review focuses on the environmental factors that influence the efficacy of probiotic oxalate-degrading bacteria, relative to oxalate metabolism. We describe the mechanism of oxalate catabolism and its consumption by obligate and facultative anaerobic oxalate-degrading bacteria, in both in vitro and in vivo environments. We also explore the environmental variables that impact oxalate degradation. Studies on single species degrade oxalate have not shown a strong impact on oxalate metabolism, especially in high oxalate conditions such as consumption of foods high in oxalate (such as coffee and chocolate for humans or halogeton in animal feed). Considering effective variables which enhance oxalate degradation could be used in application of effective probiotic as a therapeutic tool in individuals with hyperoxaluria. This study indicates probiotics can be considered a good source of naturally occurring oxalate degrading agent in human colon.
Collapse
|
16
|
Kumar P, Yang Z, Lever JM, Chávez MD, Fatima H, Crossman DK, Maynard CL, George JF, Mitchell T. Hydroxyproline stimulates inflammation and reprograms macrophage signaling in a rat kidney stone model. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166442. [PMID: 35562038 PMCID: PMC10101222 DOI: 10.1016/j.bbadis.2022.166442] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022]
Abstract
Meals rich in oxalate are associated with calcium oxalate (CaOx) kidney stone disease. Hydroxy-L-proline (HLP) is an oxalate precursor found in milk and collagen-containing foods. HLP has been shown to induce CaOx crystal formation in rodents. The purpose of this study was to evaluate the effect of HLP induced oxalate levels on inflammation and renal leukocytes during crystal formation. Male Sprague-Dawley rats (6-8 weeks old) were fed a control diet containing no oxalate for 3 days before being randomized to continue the control diet or 5% HLP for up to 28 days. Blood, 24 h urine, and kidneys were collected on Days 0, 7, 14, or 28. Urinary oxalate levels, crystal deposition, and renal macrophage markers were evaluated using ion chromatography-mass spectrometry, immunohistochemistry, and qRT-PCR. Renal leukocytes were assessed using flow cytometry and RNA-sequencing. HLP feeding increased urinary oxalate levels and renal crystal formation in animals within 7 days. HLP also increased renal macrophage populations on Days 14 and 28. Transcriptome analysis revealed that renal macrophages from animals fed HLP for 7 days were involved in inflammatory response and disease, stress response to LPS, oxidative stress, and immune cell trafficking. Renal macrophages isolated on Day 14 were involved in cell-mediated immunological pathways, ion homeostasis, and inflammatory response. Collectively, these findings suggest that HLP-mediated oxalate levels induce markers of inflammation, leukocyte populations, and reprograms signaling pathways in macrophages in a time-dependent manner. Additional studies investigating the significance of oxalate on renal macrophages could aid in our understanding of kidney stone formation.
Collapse
Affiliation(s)
- Parveen Kumar
- Department of Urology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Zhengqin Yang
- Department of Nephrology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Jeremie M Lever
- Department of Nephrology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Miranda D Chávez
- Department of Urology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Huma Fatima
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL, USA
| | - David K Crossman
- Department of Medicine, University of Alabama Birmingham, Birmingham, AL, USA
| | - Craig L Maynard
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL, USA
| | - James F George
- Department of Nephrology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Tanecia Mitchell
- Department of Urology, University of Alabama Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
In vivo CRISPR-Cas9 inhibition of hepatic LDH as treatment of primary hyperoxaluria. Mol Ther Methods Clin Dev 2022; 25:137-146. [PMID: 35402636 PMCID: PMC8971349 DOI: 10.1016/j.omtm.2022.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/14/2022] [Indexed: 12/26/2022]
Abstract
Genome-editing strategies, especially CRISPR-Cas9 systems, have substantially increased the efficiency of innovative therapeutic approaches for monogenic diseases such as primary hyperoxalurias (PHs). We have previously demonstrated that inhibition of glycolate oxidase using CRISPR-Cas9 systems represents a promising therapeutic option for PH type I (PH1). Here, we extended our work evaluating the efficacy of liver-specific inhibition of lactate dehydrogenase (LDH), a key enzyme responsible for converting glyoxylate to oxalate; this strategy would not be limited to PH1, being applicable to other PH subtypes. In this work, we demonstrate a liver-specific inhibition of LDH that resulted in a drastic reduction of LDH levels in the liver of PH1 and PH3 mice after a single-dose delivery of AAV8 vectors expressing the CRISPR-Cas9 system, resulting in reduced urine oxalate levels and kidney damage without signs of toxicity. Deep sequencing analysis revealed that this approach was safe and specific, with no off-targets detected in the liver of treated animals and no on-target/off-tissue events. Altogether, our data provide evidence that in vivo genome editing using CRISPR-Cas9 systems would represent a valuable tool for improved therapeutic approaches for PH.
Collapse
|
18
|
Belostotsky R, Frishberg Y. Catabolism of Hydroxyproline in Vertebrates: Physiology, Evolution, Genetic Diseases and New siRNA Approach for Treatment. Int J Mol Sci 2022; 23:ijms23021005. [PMID: 35055190 PMCID: PMC8779045 DOI: 10.3390/ijms23021005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Hydroxyproline is one of the most prevalent amino acids in animal proteins. It is not a genetically encoded amino acid, but, rather, it is produced by the post-translational modification of proline in collagen, and a few other proteins, by prolyl hydroxylase enzymes. Although this post-translational modification occurs in a limited number of proteins, its biological significance cannot be overestimated. Considering that hydroxyproline cannot be re-incorporated into pro-collagen during translation, it should be catabolized following protein degradation. A cascade of reactions leads to production of two deleterious intermediates: glyoxylate and hydrogen peroxide, which need to be immediately converted. As a result, the enzymes involved in hydroxyproline catabolism are located in specific compartments: mitochondria and peroxisomes. The particular distribution of catabolic enzymes in these compartments, in different species, depends on their dietary habits. Disturbances in hydroxyproline catabolism, due to genetic aberrations, may lead to a severe disease (primary hyperoxaluria), which often impairs kidney function. The basis of this condition is accumulation of glyoxylate and its conversion to oxalate. Since calcium oxalate is insoluble, children with this rare inherited disorder suffer from progressive kidney damage. This condition has been nearly incurable until recently, as significant advances in substrate reduction therapy using small interference RNA led to a breakthrough in primary hyperoxaluria type 1 treatment.
Collapse
|
19
|
Popkov VA, Zharikova AA, Demchenko EA, Andrianova NV, Zorov DB, Plotnikov EY. Gut Microbiota as a Source of Uremic Toxins. Int J Mol Sci 2022; 23:ijms23010483. [PMID: 35008909 PMCID: PMC8745165 DOI: 10.3390/ijms23010483] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 12/20/2022] Open
Abstract
Uremic retention solutes are the compounds that accumulate in the blood when kidney excretory function is impaired. Some of these compounds are toxic at high concentrations and are usually known as “uremic toxins”. The cumulative detrimental effect of uremic toxins results in numerous health problems and eventually mortality during acute or chronic uremia, especially in end-stage renal disease. More than 100 different solutes increase during uremia; however, the exact origin for most of them is still debatable. There are three main sources for such compounds: exogenous ones are consumed with food, whereas endogenous ones are produced by the host metabolism or by symbiotic microbiota metabolism. In this article, we identify uremic retention solutes presumably of gut microbiota origin. We used database analysis to obtain data on the enzymatic reactions in bacteria and human organisms that potentially yield uremic retention solutes and hence to determine what toxins could be synthesized in bacteria residing in the human gut. We selected biochemical pathways resulting in uremic retention solutes synthesis related to specific bacterial strains and revealed links between toxin concentration in uremia and the proportion of different bacteria species which can synthesize the toxin. The detected bacterial species essential for the synthesis of uremic retention solutes were then verified using the Human Microbiome Project database. Moreover, we defined the relative abundance of human toxin-generating enzymes as well as the possibility of the synthesis of a particular toxin by the human metabolism. Our study presents a novel bioinformatics approach for the elucidation of the origin of both uremic retention solutes and uremic toxins and for searching for the most likely human microbiome producers of toxins that can be targeted and used for the therapy of adverse consequences of uremia.
Collapse
Affiliation(s)
- Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Anastasia A. Zharikova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Evgenia A. Demchenko
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
| | - Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.A.P.); (A.A.Z.); (E.A.D.); (N.V.A.); (D.B.Z.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Correspondence: ; Tel.: +7-(495)939-59-44
| |
Collapse
|
20
|
Garrelfs SF, van Harskamp D, Peters-Sengers H, van den Akker CH, Wanders RJ, Wijburg FA, van Goudoever JB, Groothoff JW, Schierbeek H, Oosterveld MJ. Endogenous Oxalate Production in Primary Hyperoxaluria Type 1 Patients. J Am Soc Nephrol 2021; 32:3175-3186. [PMID: 34686543 PMCID: PMC8638398 DOI: 10.1681/asn.2021060729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Primary hyperoxaluria type 1 (PH1) is an inborn error of glyoxylate metabolism, characterized by increased endogenous oxalate production. The metabolic pathways underlying oxalate synthesis have not been fully elucidated, and upcoming therapies require more reliable outcome parameters than the currently used plasma oxalate levels and urinary oxalate excretion rates. We therefore developed a stable isotope infusion protocol to assess endogenous oxalate synthesis rate and the contribution of glycolate to both oxalate and glycine synthesis in vivo . METHODS Eight healthy volunteers and eight patients with PH1 (stratified by pyridoxine responsiveness) underwent a combined primed continuous infusion of intravenous [1- 13 C]glycolate, [U- 13 C 2 ]oxalate, and, in a subgroup, [D 5 ]glycine. Isotopic enrichment of 13 C-labeled oxalate and glycolate were measured using a new gas chromatography-tandem mass spectrometry (GC-MS/MS) method. Stable isotope dilution and incorporation calculations quantified rates of appearance and synthetic rates, respectively. RESULTS Total daily oxalate rates of appearance (mean [SD]) were 2.71 (0.54), 1.46 (0.23), and 0.79 (0.15) mmol/d in patients who were pyridoxine unresponsive, patients who were pyridoxine responsive, and controls, respectively ( P =0.002). Mean (SD) contribution of glycolate to oxalate production was 47.3% (12.8) in patients and 1.3% (0.7) in controls. Using the incorporation of [1- 13 C]glycolate tracer in glycine revealed significant conversion of glycolate into glycine in pyridoxine responsive, but not in patients with PH1 who were pyridoxine unresponsive. CONCLUSIONS This stable isotope infusion protocol could evaluate efficacy of new therapies, investigate pyridoxine responsiveness, and serve as a tool to further explore glyoxylate metabolism in humans.
Collapse
Affiliation(s)
- Sander F. Garrelfs
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Dewi van Harskamp
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ronald J.A. Wanders
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frits A. Wijburg
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Jaap W. Groothoff
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Henk Schierbeek
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michiel J.S. Oosterveld
- Emma’s Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Milliner DS, Lieske JC. Back to the Future: The Role of Metabolic Studies in Therapeutic Advances. J Am Soc Nephrol 2021; 32:2980-2982. [PMID: 36734820 PMCID: PMC8638383 DOI: 10.1681/asn.2021101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Dawn S. Milliner
- Divison of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - John C. Lieske
- Divison of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
22
|
Li X, Fargue S, Challa AK, Poore W, Knight J, Wood KD. Generation of a GLO-2 deficient mouse reveals its effects on liver carbonyl and glutathione levels. Biochem Biophys Rep 2021; 28:101138. [PMID: 34584990 PMCID: PMC8453187 DOI: 10.1016/j.bbrep.2021.101138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Hydroxyacylglutathione hydrolase (aka as GLO-2) is a component of the glyoxalase pathway involved in the detoxification of the reactive oxoaldehydes, glyoxal and methylglyoxal. These reactive metabolites have been linked to a variety of pathological conditions, including diabetes, cancer and heart disease and may be involved in the aging process. The objective of this study was to generate a mouse model deficient in GLO-2 to provide insight into the function of GLO-2 and to determine if it is potentially linked to endogenous oxalate synthesis which could influence urinary oxalate excretion. METHODS A GLO-2 knock out mouse was generated using CRISPR/Cas 9 techniques. Tissue and 24-h urine samples were collected under baseline conditions from adult male and female animals for biochemical analyses, including chromatographic measurement of glycolate, oxalate, glyoxal, methylglyoxal, D-lactate, ascorbic acid and glutathione levels. RESULTS The GLO-2 KO animals developed normally and there were no changes in 24-h urinary oxalate excretion, liver levels of methylglyoxal, glyoxal, ascorbic acid and glutathione, or plasma d-lactate levels. GLO-2 deficient males had lower plasma glycolate levels than wild type males while this relationship was not observed in females. CONCLUSIONS The lack of a unique phenotype in a GLO-2 KO mouse model under baseline conditions is consistent with recent evidence, suggesting a functional glyoxalase pathway is not required for optimal health. A lower plasma glycolate in male GLO-2 KO animals suggests glyoxal production may be a significant contributor to circulating glycolate levels, but not to endogenous oxalate synthesis.
Collapse
Affiliation(s)
- Xingsheng Li
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sonia Fargue
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Anil Kumar Challa
- Department of Genetics University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - William Poore
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - John Knight
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kyle D. Wood
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
23
|
Gianmoena K, Gasparoni N, Jashari A, Gabrys P, Grgas K, Ghallab A, Nordström K, Gasparoni G, Reinders J, Edlund K, Godoy P, Schriewer A, Hayen H, Hudert CA, Damm G, Seehofer D, Weiss TS, Boor P, Anders HJ, Motrapu M, Jansen P, Schiergens TS, Falk-Paulsen M, Rosenstiel P, Lisowski C, Salido E, Marchan R, Walter J, Hengstler JG, Cadenas C. Epigenomic and transcriptional profiling identifies impaired glyoxylate detoxification in NAFLD as a risk factor for hyperoxaluria. Cell Rep 2021; 36:109526. [PMID: 34433051 DOI: 10.1016/j.celrep.2021.109526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Epigenetic modifications (e.g. DNA methylation) in NAFLD and their contribution to disease progression and extrahepatic complications are poorly explored. Here, we use an integrated epigenome and transcriptome analysis of mouse NAFLD hepatocytes and identify alterations in glyoxylate metabolism, a pathway relevant in kidney damage via oxalate release-a harmful waste product and kidney stone-promoting factor. Downregulation and hypermethylation of alanine-glyoxylate aminotransferase (Agxt), which detoxifies glyoxylate, preventing excessive oxalate accumulation, is accompanied by increased oxalate formation after metabolism of the precursor hydroxyproline. Viral-mediated Agxt transfer or inhibiting hydroxyproline catabolism rescues excessive oxalate release. In human steatotic hepatocytes, AGXT is also downregulated and hypermethylated, and in NAFLD adolescents, steatosis severity correlates with urinary oxalate excretion. Thus, this work identifies a reduced capacity of the steatotic liver to detoxify glyoxylate, triggering elevated oxalate, and provides a mechanistic explanation for the increased risk of kidney stones and chronic kidney disease in NAFLD patients.
Collapse
Affiliation(s)
- Kathrin Gianmoena
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Nina Gasparoni
- Department of Genetics, Saarland University, 66123 Saarbrücken, Germany
| | - Adelina Jashari
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Philipp Gabrys
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Katharina Grgas
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Ahmed Ghallab
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany; Department of Forensic and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Karl Nordström
- Department of Genetics, Saarland University, 66123 Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, 66123 Saarbrücken, Germany
| | - Jörg Reinders
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Karolina Edlund
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Patricio Godoy
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Alexander Schriewer
- Department of Analytical Chemistry, Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | - Heiko Hayen
- Department of Analytical Chemistry, Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | - Christian A Hudert
- Department of Pediatric Gastroenterology, Hepatology and Metabolic Diseases, Charité-University Medicine Berlin, 13353 Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany; Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany; Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Thomas S Weiss
- University Children Hospital (KUNO), University Hospital Regensburg, 93053 Regensburg, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Clinic of RWTH Aachen, 52074 Aachen, Germany
| | - Hans-Joachim Anders
- Department of Medicine IV, Renal Division, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Manga Motrapu
- Department of Medicine IV, Renal Division, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Peter Jansen
- Maastricht Centre for Systems Biology, University of Maastricht, 6229 Maastricht, the Netherlands
| | - Tobias S Schiergens
- Biobank of the Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology (IKMB), Kiel University and University Hospital Schleswig Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology (IKMB), Kiel University and University Hospital Schleswig Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Clivia Lisowski
- Institute of Experimental Immunology, University Hospital Bonn, Rheinische-Friedrich-Wilhelms University Bonn, 53127 Bonn, Germany
| | - Eduardo Salido
- Hospital Universitario de Canarias, Universidad La Laguna, CIBERER, 38320 Tenerife, Spain
| | - Rosemarie Marchan
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, 66123 Saarbrücken, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany
| | - Cristina Cadenas
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), 44139 Dortmund, Germany.
| |
Collapse
|
24
|
Crivelli JJ, Wood KD, Assimos DG. Is It Time to Retire the Low-Oxalate Diet? No! J Endourol 2021; 35:1435-1437. [PMID: 34409855 DOI: 10.1089/end.2021.0576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Joseph J Crivelli
- Department of Urology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Kyle D Wood
- Department of Urology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Dean G Assimos
- Department of Urology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
25
|
Uebanso T, Suyama M, Shimohata T, Mawatari K, Takahashi A. Effect of Vitamin B2-Deficient Diet on Hydroxyproline- or Obesity-Induced Hyperoxaluria in Mice. Mol Nutr Food Res 2021; 65:e2100226. [PMID: 34110671 DOI: 10.1002/mnfr.202100226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/16/2021] [Indexed: 12/17/2022]
Abstract
SCOPE Hyperoxaluria is a major cause of kidney stone disease. Around half of the oxalate in mammals is supplied from the diet and the other half is endogenously synthesized from glyoxylate. Reduction of hepatic glycolate oxidase (GO) activity is one approach to reduce endogenous production of oxalate. However, there are currently few effective dietary approaches to reduce hepatic GO activity. METHODS AND RESULTS In the present study, it is investigated whether restriction of dietary vitamin B2 (VB2) can reduce hepatic GO activity and oxalate excretion in mice with hyperoxaluria induce by hydroxyproline (Hyp) or obesity. It is found that VB2 restriction significantly reduces hepatic GO activity in both the Hyp- and obesity-induced model of hyperoxaluria in mice. However, VB2 restriction reduces urinary oxalate excretion only in the Hyp-treated mice and not the obese mice. This difference could be due to the contribution of endogenous oxalate production that manifests as increased hepatic GO activity in Hyp-treated mice but not obese mice. CONCLUSION Together these results suggest that VB2 restriction could be a new dietary approach to improve hyperoxaluria when endogenous production of oxalate is increased.
Collapse
Affiliation(s)
- Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Mai Suyama
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503, Japan
| |
Collapse
|
26
|
Pozdzik A, David C, Vekeman J, Tielens F, Daudon M. Lanthanum carbonate to control plasma and urinary oxalate level in type 1 primary hyperoxaluria? IJU Case Rep 2021; 4:235-238. [PMID: 34258537 PMCID: PMC8255283 DOI: 10.1002/iju5.12296] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION The therapy to reduce urinary oxalate excretion in primary hyperoxaluria type 1 is still required. CASE PRESENTATION A 37-year-old hemodialyzed man suffered from systemic oxalosis secondary to primary hyperoxaluria type 1 exhibited a drastic plasma oxalate decrease from 110 to 22 µmol/L two months after adjunction of lanthanum carbonate to classical treatment (intensive hemodialysis with pyridoxine). A 34-year-old woman with normal kidney function presented 10 years of bilateral kidney stones due to primary hyperoxaluria type 1 [hyperoxaluria (109.2 mg/24 h), plasma oxalate (56.0 µmol/L)]. The oxalate level remained uncontrolled despite of low oxalate-normal calcium diet, pyridoxine and increased water intake though the lanthanum carbonate adjunction resulted in significant decrease in plasma oxalate and oxaluria. CONCLUSION We report the lanthanum efficacy in reducing circulating and urinary oxalate levels in type 1 primary hyperoxaluria. Possible mechanism of observed falls in oxalate concentration would be a decrease in the intestinal absorption of oxalate.
Collapse
Affiliation(s)
- Agnieszka Pozdzik
- Department of Nephrology and DialysisKidney Stone ClinicCentre Hospitalier UniversitaireBrugmann HospitalBrusselsBelgium
- Faculty of MedicineUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | - Cristina David
- Department of Nephrology and DialysisKidney Stone ClinicCentre Hospitalier UniversitaireBrugmann HospitalBrusselsBelgium
| | - Jelle Vekeman
- General Chemistry (ALGC)Materials Modelling GroupVrije Universiteit BrusselsBrusselsBelgium
| | - Frederik Tielens
- General Chemistry (ALGC)Materials Modelling GroupVrije Universiteit BrusselsBrusselsBelgium
| | - Michel Daudon
- Service des explorations fonctionnelles multidisciplinairesTenon HospitalAP_HPParisFrance
- INSERM UMRS 1155Université Pierre et Marie Curie‐Paris VI‐Sorbonne UniversitésParisFrance
| |
Collapse
|
27
|
Siener R. Nutrition and Kidney Stone Disease. Nutrients 2021; 13:1917. [PMID: 34204863 PMCID: PMC8229448 DOI: 10.3390/nu13061917] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
The prevalence of kidney stone disease is increasing worldwide. The recurrence rate of urinary stones is estimated to be up to 50%. Nephrolithiasis is associated with increased risk of chronic and end stage kidney disease. Diet composition is considered to play a crucial role in urinary stone formation. There is strong evidence that an inadequate fluid intake is the major dietary risk factor for urolithiasis. While the benefit of high fluid intake has been confirmed, the effect of different beverages, such as tap water, mineral water, fruit juices, soft drinks, tea and coffee, are debated. Other nutritional factors, including dietary protein, carbohydrates, oxalate, calcium and sodium chloride can also modulate the urinary risk profile and contribute to the risk of kidney stone formation. The assessment of nutritional risk factors is an essential component in the specific dietary therapy of kidney stone patients. An appropriate dietary intervention can contribute to the effective prevention of recurrent stones and reduce the burden of invasive surgical procedures for the treatment of urinary stone disease. This narrative review has intended to provide a comprehensive and updated overview on the role of nutrition and diet in kidney stone disease.
Collapse
Affiliation(s)
- Roswitha Siener
- University Stone Center, Department of Urology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
28
|
Borin JF, Knight J, Holmes RP, Joshi S, Goldfarb DS, Loeb S. Plant-Based Milk Alternatives and Risk Factors for Kidney Stones and Chronic Kidney Disease. J Ren Nutr 2021; 32:363-365. [PMID: 34045136 PMCID: PMC8611107 DOI: 10.1053/j.jrn.2021.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Patients with kidney stones are counseled to eat a diet low in animal protein, sodium, and oxalate and rich in fruits and vegetables, with a modest amount of calcium, usually from dairy products. Restriction of sodium, potassium, and oxalate may also be recommended in patients with chronic kidney disease. Recently, plant-based diets have gained popularity owing to health, environmental, and animal welfare considerations. Our objective was to compare concentrations of ingredients important for kidney stones and chronic kidney disease in popular brands of milk alternatives. DESIGN AND METHODS Sodium, calcium, and potassium contents were obtained from nutrition labels. The oxalate content was measured by ion chromatography coupled with mass spectrometry. RESULTS The calcium content is highest in macadamia followed by soy, almond, rice, and dairy milk; it is lowest in cashew, hazelnut, and coconut milk. Almond milk has the highest oxalate concentration, followed by cashew, hazelnut, and soy. Coconut and flax milk have undetectable oxalate levels; coconut milk also has comparatively low sodium, calcium, and potassium, while flax milk has the most sodium. Overall, oat milk has the most similar parameters to dairy milk (moderate calcium, potassium and sodium with low oxalate). Rice, macadamia, and soy milk also have similar parameters to dairy milk. CONCLUSION As consumption of plant-based dairy substitutes increases, it is important for healthcare providers and patients with renal conditions to be aware of their nutritional composition. Oat, macadamia, rice, and soy milk compare favorably in terms of kidney stone risk factors with dairy milk, whereas almond and cashew milk have more potential stone risk factors. Coconut milk may be a favorable dairy substitute for patients with chronic kidney disease based on low potassium, sodium, and oxalate. Further study is warranted to determine the effect of plant-based milk alternatives on urine chemistry.
Collapse
Affiliation(s)
- James F Borin
- Department of Urology, New York University and New York Harbor VA Healthcare System, New York, New York.
| | - John Knight
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ross P Holmes
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shivam Joshi
- Division of Nephrology, Department of Medicine, New York University and New York Harbor VA Healthcare System, New York, New York
| | - David S Goldfarb
- Division of Nephrology, Department of Medicine, New York University and New York Harbor VA Healthcare System, New York, New York
| | - Stacy Loeb
- Department of Urology, New York University and New York Harbor VA Healthcare System, New York, New York
| |
Collapse
|
29
|
Letavernier E, Daudon M. [Effect of stiripentol on urine oxalate excretion]. Nephrol Ther 2021; 17S:S95-S99. [PMID: 33910706 DOI: 10.1016/j.nephro.2020.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 02/03/2020] [Indexed: 11/16/2022]
Abstract
Oxalate is a metabolite promoting the formation of calcium oxalate crystals in urine. Hyperoxaluria is a feature of genetic diseases, known as primary hyperoxaluria, leading to chronic kidney disease. Ethylene glycol poisoning induces the crystallization of calcium oxalate crystals in renal tubules, promoting acute renal failure. Urine oxalate results from glyoxylate transformation to oxalate in the liver, due to lactate dehydrogenase (LDH) activity, especially the LDH-5 isoenzyme. Genetic RNA interference therapy targeting lactate dehydrogenase lowers urine oxalate excretion in murine models. Stiripentol is a drug inhibiting neuronal LDH-5 isoenzyme activity. We hypothesized that stiripentol would also reduce hepatic oxalate production and urine oxalate excretion. In vitro Stiripentol decreases oxalate synthesis by hepatocytes. In vivo, stiripentol decreases urine oxalate excretion in rats and protects kidney tissue and function against ethylene glycol intoxication and hydroxyproline-induced calcium oxalate crystalline nephropathy. The use of stiripentol in clinical practice deserves further clinical studies.
Collapse
Affiliation(s)
- Emmanuel Letavernier
- Sorbonne Université, 4, rue de la Chine, 75020 Paris, France; Inserm, UMR S 1155 Maladies fréquentes et rares: des mécanismes moléculaires à la médecine personnalisée, 4, rue de la Chine, 75020 Paris, France; Service des explorations fonctionnelles multidisciplinaires, hôpital Tenon, 4, rue de la Chine, 75020 Paris France.
| | - Michel Daudon
- Sorbonne Université, 4, rue de la Chine, 75020 Paris, France; Inserm, UMR S 1155 Maladies fréquentes et rares: des mécanismes moléculaires à la médecine personnalisée, 4, rue de la Chine, 75020 Paris, France; Service des explorations fonctionnelles multidisciplinaires, hôpital Tenon, 4, rue de la Chine, 75020 Paris France
| |
Collapse
|
30
|
Kumar P, Patel M, Oster RA, Yarlagadda V, Ambrosetti A, Assimos DG, Mitchell T. Dietary Oxalate Loading Impacts Monocyte Metabolism and Inflammatory Signaling in Humans. Front Immunol 2021; 12:617508. [PMID: 33732242 PMCID: PMC7959803 DOI: 10.3389/fimmu.2021.617508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/11/2021] [Indexed: 12/27/2022] Open
Abstract
Diet has been associated with several metabolic diseases and may impact immunity. Increased consumption of meals with high oxalate content may stimulate urinary calcium oxalate (CaOx) crystals, which are precursors to CaOx kidney stones. We previously reported that CaOx stone formers have decreased monocyte cellular bioenergetics compared to healthy participants and oxalate reduces monocyte metabolism and redox status in vitro. The purpose of this study was to investigate whether dietary oxalate loading impacts monocyte cellular bioenergetics, mitochondrial complex activity, and inflammatory signaling in humans. Healthy participants (n = 40; 31.1 ± 1.3 years) with a BMI of 24.9 ± 0.6 kg/m2 consumed a controlled low oxalate diet for 3 days before drinking a blended preparation of fruits and vegetables containing a large amount of oxalate. Blood and urine were collected before (pre-oxalate) and for 5 h after the oxalate load to assess urinary oxalate levels, monocyte cellular bioenergetics and mitochondrial complex activity, and plasma cytokine/chemokine levels. Urinary oxalate levels significantly increased in post-oxalate samples compared to pre-oxalate samples. Monocyte cellular bioenergetics, mitochondrial complex I activity, and plasma cytokine and chemokine levels were altered to varying degrees within the study cohort. We demonstrate for the first time that dietary oxalate loading may impact monocyte metabolism and immune response in a cohort of healthy adults, but these response are variable. Further studies are warranted to understand oxalate mediated mechanisms on circulating monocytes and how this potentially influences CaOx kidney stone formation.
Collapse
Affiliation(s)
- Parveen Kumar
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mikita Patel
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert A Oster
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vidhush Yarlagadda
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Adam Ambrosetti
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Dean G Assimos
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Oxalate is a metabolic end-product promoting the formation of calcium oxalate crystals in urine. Massive urine oxalate excretion occurs in genetic diseases, mainly primary hyperoxaluria type I and II, threatening renal function. Ethylene glycol poisoning may induce the precipitation of calcium oxalate crystals in renal tubules, leading to acute renal failure. In both cases, oxalate results from glyoxylate transformation to oxalate in the liver, by lactate dehydrogenase (LDH) enzymes, especially the LDH-5 isoenzyme. The purpose of the review is to highlight LDH as a potential therapeutic target according to recent publications. RECENT FINDINGS Genetic therapy targeting LDH metabolism decreases urine oxalate excretion in rodents. Stiripentol is an antiepileptic drug that has been shown recently to inhibit neuronal LDH-5 isoenzyme. Stiripentol was hypothesized to reduce hepatic oxalate production and urine oxalate excretion. In vitro, stiripentol decreases oxalate synthesis by hepatocytes. In vivo, stiripentol oral administration decreases urine oxalate excretion in rats and protects renal function and renal tissue against ethylene glycol intoxication and chronic calcium oxalate crystalline nephropathy. SUMMARY The use of stiripentol in-vitro and in-vivo highlights that targeting hepatic LDH by pharmacological or genetic tools may decrease oxalate synthesis, deserving clinical studies.
Collapse
|
32
|
Small Molecule-Based Enzyme Inhibitors in the Treatment of Primary Hyperoxalurias. J Pers Med 2021; 11:jpm11020074. [PMID: 33513899 PMCID: PMC7912158 DOI: 10.3390/jpm11020074] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Primary hyperoxalurias (PHs) are a group of inherited alterations of the hepatic glyoxylate metabolism. PHs classification based on gene mutations parallel a variety of enzymatic defects, and all involve the harmful accumulation of calcium oxalate crystals that produce systemic damage. These geographically widespread rare diseases have a deep impact in the life quality of the patients. Until recently, treatments were limited to palliative measures and kidney/liver transplants in the most severe forms. Efforts made to develop pharmacological treatments succeeded with the biotechnological agent lumasiran, a siRNA product against glycolate oxidase, which has become the first effective therapy to treat PH1. However, small molecule drugs have classically been preferred since they benefit from experience and have better pharmacological properties. The development of small molecule inhibitors designed against key enzymes of glyoxylate metabolism is on the focus of research. Enzyme inhibitors are successful and widely used in several diseases and their pharmacokinetic advantages are well known. In PHs, effective enzymatic targets have been determined and characterized for drug design and interesting inhibitory activities have been achieved both in vitro and in vivo. This review describes the most recent advances towards the development of small molecule enzyme inhibitors in the treatment of PHs, introducing the multi-target approach as a more effective and safe therapeutic option.
Collapse
|
33
|
Plasma oxalate and eGFR are correlated in primary hyperoxaluria patients with maintained kidney function-data from three placebo-controlled studies. Pediatr Nephrol 2021; 36:1785-1793. [PMID: 33515281 PMCID: PMC8172484 DOI: 10.1007/s00467-020-04894-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/06/2020] [Accepted: 12/03/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND In patients with primary hyperoxaluria (PH), endogenous oxalate overproduction increases urinary oxalate excretion, leading to compromised kidney function and often kidney failure. Highly elevated plasma oxalate (Pox) is associated with systemic oxalate deposition in patients with PH and severe chronic kidney disease (CKD). The relationship between Pox and estimated glomerular filtration rate (eGFR) in patients with preserved kidney function, however, is not well established. Our analysis aimed to investigate a potential correlation between these parameters in PH patients from three randomized, placebo-controlled trials (studies OC3-DB-01, OC3-DB-02, and OC5-DB-01). METHODS Baseline data from patients with a PH diagnosis (type 1, 2, or 3) and eGFR > 40 mL/min/1.73 m2 were analyzed for a correlation between eGFR and Pox using Spearman's rank and Pearson's correlation coefficients. Data were analyzed by individual study and additionally were pooled for Studies OC3-DB-02 and OC5-DB-01 in which the same Pox assay was used. RESULTS A total of 106 patients were analyzed. A statistically significant inverse Spearman's correlation between eGFR and Pox was observed across all analyses; correlation coefficients were - 0.44 in study OC3-DB-01, - 0.55 in study OC3-DB-02, - 0.51 in study OC5-DB-01, and - 0.49 in the pooled studies (p < 0.0064). CONCLUSIONS Baseline evaluations showed a moderate and statistically significant inverse correlation between eGFR and Pox in patients with PH already at early stages of CKD (stages 1-3b), demonstrating that a correlation is present before substantial loss in kidney function occurs.
Collapse
|
34
|
Crivelli JJ, Mitchell T, Knight J, Wood KD, Assimos DG, Holmes RP, Fargue S. Contribution of Dietary Oxalate and Oxalate Precursors to Urinary Oxalate Excretion. Nutrients 2020; 13:nu13010062. [PMID: 33379176 PMCID: PMC7823532 DOI: 10.3390/nu13010062] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 02/07/2023] Open
Abstract
Kidney stone disease is increasing in prevalence, and the most common stone composition is calcium oxalate. Dietary oxalate intake and endogenous production of oxalate are important in the pathophysiology of calcium oxalate stone disease. The impact of dietary oxalate intake on urinary oxalate excretion and kidney stone disease risk has been assessed through large cohort studies as well as smaller studies with dietary control. Net gastrointestinal oxalate absorption influences urinary oxalate excretion. Oxalate-degrading bacteria in the gut microbiome, especially Oxalobacter formigenes, may mitigate stone risk through reducing net oxalate absorption. Ascorbic acid (vitamin C) is the main dietary precursor for endogenous production of oxalate with several other compounds playing a lesser role. Renal handling of oxalate and, potentially, renal synthesis of oxalate may contribute to stone formation. In this review, we discuss dietary oxalate and precursors of oxalate, their pertinent physiology in humans, and what is known about their role in kidney stone disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Ross P. Holmes
- Correspondence: ; Tel.: +1-(205)-996-8765; Fax: +1-(205)-934-4933
| | | |
Collapse
|
35
|
Wood KD, Freeman BL, Killian ME, Lai WS, Assimos D, Knight J, Fargue S. Effect of alanine supplementation on oxalate synthesis. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165981. [PMID: 33002578 DOI: 10.1016/j.bbadis.2020.165981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 11/26/2022]
Abstract
The Primary Hyperoxalurias (PH) are rare disorders of metabolism leading to excessive endogenous synthesis of oxalate and recurring calcium oxalate kidney stones. Alanine glyoxylate aminotransferase (AGT), deficient in PH type 1, is a key enzyme in limiting glyoxylate oxidation to oxalate. The affinity of AGT for its co-substrate, alanine, is low suggesting that its metabolic activity could be sub-optimal in vivo. To test this hypothesis, we examined the effect of L-alanine supplementation on oxalate synthesis in cell culture and in mouse models of Primary Hyperoxaluria Type 1 (Agxt KO), Type 2 (Grhpr KO) and in wild-type mice. Our results demonstrated that increasing L-alanine in cells decreased synthesis of oxalate and increased viability of cells expressing GO and AGT when incubated with glycolate. In both wild type and Grhpr KO male and female mice, supplementation with 10% dietary L-alanine significantly decreased urinary oxalate excretion ~30% compared to baseline levels. This study demonstrates that increasing the availability of L-alanine can increase the metabolic efficiency of AGT and reduce oxalate synthesis.
Collapse
Affiliation(s)
- Kyle D Wood
- University of Alabama at Birmingham, Department of Urology, Birmingham, AL, United States of America
| | - Brian L Freeman
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - Mary E Killian
- University of Tennessee Health Science Center, Department of Urology, Memphis, TN, United States of America
| | - Win Shun Lai
- University of Texas Medical Branch, Division of Urology, Galveston, TX, United States of America
| | - Dean Assimos
- University of Alabama at Birmingham, Department of Urology, Birmingham, AL, United States of America
| | - John Knight
- University of Alabama at Birmingham, Department of Urology, Birmingham, AL, United States of America
| | - Sonia Fargue
- University of Alabama at Birmingham, Department of Urology, Birmingham, AL, United States of America.
| |
Collapse
|
36
|
Dietary Oxalate Intake and Kidney Outcomes. Nutrients 2020; 12:nu12092673. [PMID: 32887293 PMCID: PMC7551439 DOI: 10.3390/nu12092673] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022] Open
Abstract
Oxalate is both a plant-derived molecule and a terminal toxic metabolite with no known physiological function in humans. It is predominantly eliminated by the kidneys through glomerular filtration and tubular secretion. Regardless of the cause, the increased load of dietary oxalate presented to the kidneys has been linked to different kidney-related conditions and injuries, including calcium oxalate nephrolithiasis, acute and chronic kidney disease. In this paper, we review the current literature on the association between dietary oxalate intake and kidney outcomes.
Collapse
|
37
|
Kumar P, Patel M, Thomas V, Knight J, Holmes RP, Mitchell T. Dietary Oxalate Induces Urinary Nanocrystals in Humans. Kidney Int Rep 2020; 5:1040-1051. [PMID: 32647761 PMCID: PMC7335953 DOI: 10.1016/j.ekir.2020.04.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Introduction Crystalluria is thought to be associated with kidney stone formation and can occur when urine becomes supersaturated with calcium, oxalate, and phosphate. The principal method used to identify urinary crystals is microscopy, with or without a polarized light source. This method can detect crystals above 1 μm in diameter (microcrystals). However, analyses of calcium oxalate kidney stones have indicated that crystallite components in these calculi are 50–100 nm in diameter. Recent studies have suggested that nanocrystals (<200 nm) elicit more injury to renal cells compared to microcrystals. The purpose of this study was to determine whether (i) urinary nanocrystals can be detected and quantified by nanoparticle tracking analysis (NTA, a high-resolution imaging technology), (ii) early-void urine samples from healthy subjects contain calcium nanocrystals, and (iii) a dietary oxalate load increases urinary nanocrystal formation. Methods Healthy subjects consumed a controlled low-oxalate diet for 3 days before a dietary oxalate load. Urinary crystals were isolated by centrifugation and assessed using NTA before and 5 hours after the oxalate load. The morphology and chemical composition of crystals was assessed using electron microscopy, Fourier-transform infrared spectroscopy (FTIR), and ion chromatography-mass spectrometry (IC–MS). Results Urinary calcium oxalate nanocrystals were detected in pre-load samples and increased substantially following the oxalate load. Conclusion These findings indicate that NTA can quantify urinary nanocrystals and that meals rich in oxalate can promote nanocrystalluria. NTA should provide valuable insight about the role of nanocrystals in kidney stone formation.
Collapse
Affiliation(s)
- Parveen Kumar
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mikita Patel
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Vinoy Thomas
- Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John Knight
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ross P Holmes
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
38
|
van Harskamp D, Garrelfs SF, Oosterveld MJS, Groothoff JW, van Goudoever JB, Schierbeek H. Development and Validation of a New Gas Chromatography-Tandem Mass Spectrometry Method for the Measurement of Enrichment of Glyoxylate Metabolism Analytes in Hyperoxaluria Patients Using a Stable Isotope Procedure. Anal Chem 2020; 92:1826-1832. [PMID: 31867958 PMCID: PMC6977104 DOI: 10.1021/acs.analchem.9b03670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Primary
hyperoxalurias (PH) are inborn errors of glyoxylate metabolism
characterized by an increase in endogenous oxalate production. Oxalate
overproduction may cause calcium-oxalate crystal formation leading
to kidney stones, nephrocalcinosis, and ultimately kidney failure.
Twenty-four hour urine oxalate excretion is an inaccurate measure
for endogenous oxalate production in PH patients and not applicable
in those with kidney failure. Treatment efficacy cannot be assessed
with this measure during clinical trials. We describe the development
and validation of a gas chromatography–tandem mass spectrometry
method to analyze the samples obtained following a stable isotope
infusion protocol of 13C2-oxalate and 1-13C-glycolate in both healthy individuals and PH patients.
Isotopic enrichments of plasma oxalate, glycolate, and glyoxylate
were measured on a gas chromatography–triple quadrupole mass
spectrometry system using ethylhydroxylamine and N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide
(MTBSTFA) for analyte derivatization. Method precision was good for
oxalate and glycolate (coefficients of variation [CV] were <6.3%
and <4.2% for inter- and intraday precision, respectively) and
acceptable for glyoxylate (CV <18.3% and <6.7% for inter- and
intraday precision, respectively). The enrichment curves were linear
over the specified range. Sensitivity was sufficient to accurately
analyze enrichments. This new method allowed calculation of kinetic
features of these metabolites, thus enabling a detailed analysis of
the various pathways involved in glyoxylate metabolism. The method
will further enhance the investigation of the metabolic PH derangements,
provides a tool to accurately assess the therapeutic efficacy of new
promising therapeutic interventions for PH, and could serve as a clinical
tool to improve personalized therapeutic strategies.
Collapse
Affiliation(s)
- Dewi van Harskamp
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| | - Sander F Garrelfs
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| | - Michiel J S Oosterveld
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| | - Jaap W Groothoff
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| | - Johannes B van Goudoever
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| | - Henk Schierbeek
- Amsterdam UMC, University of Amsterdam , Vrije Universiteit, Emma Children's Hospital, Amsterdam , Meibergdreef 9 , 1105AZ Amsterdam , The Netherlands
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The review of potential therapies in the treatment of hyperoxaluria is timely, given the current excitement with clinical trials and the mounting evidence of the importance of oxalate in both kidney stone and chronic kidney disease. RECENT FINDINGS Given the significant contribution of both endogenous and dietary oxalate to urinary oxalate excretions, it is not surprising therapeutic targets are being studied in both pathways. This article covers the existing data on endogenous and dietary oxalate and the current targets in these pathways. SUMMARY In the near future, there will likely be therapies targeting both endogenous and dietary oxalate, especially in subsets of kidney stone formers.
Collapse
|
40
|
Buchalski B, Wood KD, Challa A, Fargue S, Holmes RP, Lowther WT, Knight J. The effects of the inactivation of Hydroxyproline dehydrogenase on urinary oxalate and glycolate excretion in mouse models of primary hyperoxaluria. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165633. [PMID: 31821850 PMCID: PMC7047938 DOI: 10.1016/j.bbadis.2019.165633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/20/2019] [Accepted: 12/04/2019] [Indexed: 01/18/2023]
Abstract
The major clinical manifestation of the Primary Hyperoxalurias (PH) is increased production of oxalate, as a consequence of genetic mutations that lead to aberrant glyoxylate and hydroxyproline metabolism. Hyperoxaluria can lead to the formation of calcium-oxalate kidney stones, nephrocalcinosis and renal failure. Current therapeutic approaches rely on organ transplants and more recently modifying the pathway of oxalate synthesis using siRNA therapy. We have recently reported that the metabolism of trans-4-hydroxy-L-proline (Hyp), an amino acid derived predominantly from collagen metabolism, is a significant source of oxalate production in individuals with PH2 and PH3. Thus, the first enzyme in the Hyp degradation pathway, hydroxyproline dehydrogenase (HYPDH), represents a promising therapeutic target for reducing endogenous oxalate production in these individuals. This is supported by the observation that individuals with inherited mutations in HYPDH (PRODH2 gene) have no pathological consequences. The creation of mouse models that do not express HYPDH will facilitate research evaluating HYPDH as a target. We describe the phenotype of the Prodh2 knock out mouse model and show that the lack of HYPDH in PH mouse models results in lower levels of urinary oxalate excretion, consistent with our previous metabolic tracer and siRNA-based knockdown studies. The double knockout mouse, Grhpr KO (PH2 model) and Prodh2 KO, prevented calcium-oxalate crystal deposition in the kidney, when placed on a 1% Hyp diet. These observations support the use of the Grhpr KO mice to screen HYPDH inhibitors in vivo. Altogether these data support HYPDH as an attractive therapeutic target for PH2 and PH3 patients.
Collapse
Affiliation(s)
- Brianna Buchalski
- Department of Urology, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL, 35294, United States of America
| | - Kyle D Wood
- Department of Urology, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL, 35294, United States of America
| | - Anil Challa
- Department of Genetics, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL 35294, United States of America
| | - Sonia Fargue
- Department of Urology, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL, 35294, United States of America
| | - Ross P Holmes
- Department of Urology, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL, 35294, United States of America
| | - W Todd Lowther
- Department of Biochemistry, Center for Structural Biology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, United States of America.
| | - John Knight
- Department of Urology, University of Alabama at Birmingham, 720 20(th) Street South, Birmingham, AL, 35294, United States of America.
| |
Collapse
|
41
|
Torres JA, Rezaei M, Broderick C, Lin L, Wang X, Hoppe B, Cowley BD, Savica V, Torres VE, Khan S, Holmes RP, Mrug M, Weimbs T. Crystal deposition triggers tubule dilation that accelerates cystogenesis in polycystic kidney disease. J Clin Invest 2019; 129:4506-4522. [PMID: 31361604 PMCID: PMC6763267 DOI: 10.1172/jci128503] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
The rate of disease progression in autosomal-dominant (AD) polycystic kidney disease (PKD) exhibits high intra-familial variability suggesting that environmental factors may play a role. We hypothesized that a prevalent form of renal insult may accelerate cystic progression and investigated tubular crystal deposition. We report that calcium oxalate (CaOx) crystal deposition led to rapid tubule dilation, activation of PKD-associated signaling pathways, and hypertrophy in tubule segments along the affected nephrons. Blocking mTOR signaling blunted this response and inhibited efficient excretion of lodged crystals. This mechanism of "flushing out" crystals by purposefully dilating renal tubules has not previously been recognized. Challenging PKD rat models with CaOx crystal deposition, or inducing calcium phosphate deposition by increasing dietary phosphorous intake, led to increased cystogenesis and disease progression. In a cohort of ADPKD patients, lower levels of urinary excretion of citrate, an endogenous inhibitor of calcium crystal formation, correlated with increased disease severity. These results suggest that PKD progression may be accelerated by commonly occurring renal crystal deposition which could be therapeutically controlled by relatively simple measures.
Collapse
Affiliation(s)
- Jacob A. Torres
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Mina Rezaei
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Caroline Broderick
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Louis Lin
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| | - Xiaofang Wang
- Mayo Clinic College of Medicine, Division of Nephrology and Hypertension, Rochester, Minnesota, USA
| | - Bernd Hoppe
- University Children’s Hospital Bonn, Division of Pediatric Nephrology, Bonn, Germany
| | - Benjamin D. Cowley
- University of Oklahoma Health Sciences Center, Department of Medicine, Section of Nephrology, Oklahoma City, Oklahoma, USA
| | - Vincenzo Savica
- University of Messina, Department of Clinical and Experimental Medicine, Messina, Italy
| | - Vicente E. Torres
- Mayo Clinic College of Medicine, Division of Nephrology and Hypertension, Rochester, Minnesota, USA
| | - Saeed Khan
- University of Florida, Department of Pathology, Gainesville, Florida, USA
| | | | - Michal Mrug
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - Thomas Weimbs
- University of California Santa Barbara, Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, Santa Barbara, California, USA
| |
Collapse
|
42
|
Stevens JS, Al-Awqati Q. Lactate dehydrogenase 5: identification of a druggable target to reduce oxaluria. J Clin Invest 2019; 129:2201-2204. [PMID: 31107247 DOI: 10.1172/jci128709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Excessive excretion of oxalate in the urine results in the formation of calcium oxalate crystals and subsequent kidney stone formation. Severe forms of hyperoxaluria, including genetic forms and those that result from ethylene glycol poisoning, can result in end-stage renal disease. Therapeutic interventions are limited and often rely on dietary intervention. In this issue of the JCI, Le Dudal and colleagues demonstrate that the lactate dehydrogenase 5 inhibitor (LDH5) stiripentol reduces urinary oxalate excretion. Importantly, stiripentol treatment of a single individual with primary hyperoxaluria reduced the urinary oxalate excretion. Together, these results support further evaluation of LDH5 as a therapeutic target for hyperoxaluria.
Collapse
|
43
|
Bruel A, Bacchetta J, Ginhoux T, Rodier-Bonifas C, Sellier-Leclerc AL, Fromy B, Cochat P, Sigaudo-Roussel D, Dubourg L. Skin microvascular dysfunction as an early cardiovascular marker in primary hyperoxaluria type I. Pediatr Nephrol 2019; 34:319-327. [PMID: 30276532 DOI: 10.1007/s00467-018-4081-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/03/2018] [Accepted: 09/03/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Primary hyperoxaluria type 1 (PH1) is an orphan inborn error of oxalate metabolism leading to hyperoxaluria, progressive renal failure, oxalate deposition, and increased cardiovascular complications. As endothelial dysfunction and arterial stiffness are early markers of cardiovascular risk, we investigated early endothelial and vascular dysfunction in young PH1 patients either under conservative treatment (PH1-Cons) or after combined kidney liver transplantation (PH1-T) in comparison to healthy controls (Cont-H) and patients with a past of renal transplantation (Cont-T). METHODS Skin microvascular function was non-invasively assessed by laser Doppler flowmetry before and after stimulation by current, thermal, or pharmacological (nitroprussiate (SNP) or acetylcholine (Ach)) stimuli in young PH1 patients and controls. RESULTS Seven PH1-Cons (6 F, median age 18.2) and 6 PH1-T (2 F, median age 13.3) were compared to 96 Cont-H (51 F, median age 14.2) and 6 Cont-T (4 F, median age 14.5). The endothelium-independent vasodilatation (SNP) was severely decreased in PH1-T compared to Cont-H. Ach, current-induced vasodilatation (CIV), and thermal response was increased in PH1-Cons and Cont-T compared to controls. CONCLUSIONS PH1-T patients displayed severely decreased smooth muscle capacity to vasodilate. An exacerbated endothelial-dependent vasodilation suggests a role for silent inflammation in the early dysfunction of microcirculation observed in PH1-Cons and Cont-T.
Collapse
Affiliation(s)
- Alexandra Bruel
- Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France.,Service de Pédiatrie, Hôpital Mère et Enfants, Centre hospitalo-universitaire de Nantes, Nantes, France
| | - Justine Bacchetta
- Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France
| | - Tiphanie Ginhoux
- EPICIME-CIC 1407 de Lyon, Inserm, Service de Pharmacologie Clinique, CHU-Lyon, Lyon, France
| | - Christelle Rodier-Bonifas
- Service d'ophtalmologie, Groupement Hospitalier Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Anne-Laure Sellier-Leclerc
- Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France
| | - Bérengère Fromy
- Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Claude Bernard Lyon 1, Villeurbanne, France
| | - Pierre Cochat
- Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Claude Bernard Lyon 1, Villeurbanne, France
| | - Dominique Sigaudo-Roussel
- Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Claude Bernard Lyon 1, Villeurbanne, France
| | - Laurence Dubourg
- Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France. .,Université Claude Bernard Lyon 1, Lyon, France. .,Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Claude Bernard Lyon 1, Villeurbanne, France. .,Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Groupement Hospitalier Edouard Herriot, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
44
|
Affiliation(s)
- Gill Rumsby
- Clinical Biochemistry, UCL Hospitals, London, UK
| | - Sally-Anne Hulton
- Department of Nephrology, Birmingham Women’s and Children’s Hospital NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
45
|
Moya-Garzón MD, Martín Higueras C, Peñalver P, Romera M, Fernandes MX, Franco-Montalbán F, Gómez-Vidal JA, Salido E, Díaz-Gavilán M. Salicylic Acid Derivatives Inhibit Oxalate Production in Mouse Hepatocytes with Primary Hyperoxaluria Type 1. J Med Chem 2018; 61:7144-7167. [DOI: 10.1021/acs.jmedchem.8b00399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- María Dolores Moya-Garzón
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Cristina Martín Higueras
- Hospital Universitario de Canarias, Universidad La Laguna & Center for Rare Diseases (CIBERER), 38320 Tenerife, Spain
| | - Pablo Peñalver
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Manuela Romera
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Miguel X. Fernandes
- Hospital Universitario de Canarias, Universidad La Laguna & Center for Rare Diseases (CIBERER), 38320 Tenerife, Spain
| | - Francisco Franco-Montalbán
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - José A. Gómez-Vidal
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Eduardo Salido
- Hospital Universitario de Canarias, Universidad La Laguna & Center for Rare Diseases (CIBERER), 38320 Tenerife, Spain
| | - Mónica Díaz-Gavilán
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| |
Collapse
|