1
|
Stalter EJ, Verhofste SL, Dagle JM, Steinbach EJ, Ten Eyck P, Wendt L, Segar JL, Harshman LA. Somatic growth outcomes in response to an individualized neonatal sodium supplementation protocol. J Perinatol 2025; 45:305-311. [PMID: 39420073 DOI: 10.1038/s41372-024-02141-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Evaluate the impact of a sodium (Na) supplementation protocol based upon urine Na concentration on growth parameters and morbidities. STUDY DESIGN Retrospective cohort study of infants 260/7-336/7 weeks gestational age (GA) cared for before (2012-15, n = 310) and after (2016-20, n = 382) implementation of the protocol. Within- and between-group changes over time were assessed using repeated measures generalized linear models. RESULTS For infants 260/7-296/7 weeks GA, utilization of the protocol was associated with increased mean body weight z-score at 8-weeks postnatal age, increased mean head circumference z-score at 16-weeks postnatal age, and decreased time on mechanical ventilation (all p < 0.02). No impact on growth was identified for infants 30-336/7 weeks GA. Incidences of hypertension, hypernatremia, bronchopulmonary dysplasia, necrotizing enterocolitis, and culture positive sepsis were unaffected by the protocol. CONCLUSION Protocolized Na supplementation is associated with improved growth and reduced time on invasive mechanical ventilation in extremely preterm infants without increasing incidence of morbidities.
Collapse
Affiliation(s)
- Elliot J Stalter
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa, IA, USA
| | - Silvia L Verhofste
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa, IA, USA
| | - John M Dagle
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa, IA, USA
| | - Emily J Steinbach
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa, IA, USA
| | - Patrick Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa, IA, USA
| | - Linder Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa, IA, USA
| | - Jeffrey L Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lyndsay A Harshman
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA.
| |
Collapse
|
2
|
Lindström NO, Vanslambrouck JM. Patterning the nephron: Forming an axial polarity with distal and proximal specialization. Curr Top Dev Biol 2025; 163:83-103. [PMID: 40254351 DOI: 10.1016/bs.ctdb.2025.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Nephron formation and patterning are driven by complex cell biology. Progenitors migrate, transition into epithelia, and generate an axial epithelial polarity with distinct transcriptional signatures, regulating virtually all physiologies of the maturing kidney post birth. Here we review current insights into mammalian nephrogenesis and discuss how the nephron forms and patterns along its proximal-distal axis during embryonic and fetal development. Genetic pathways that are necessary for this process are discussed and integrated into the cell biology and morphogenetic programs underpinning nephrogenesis. Together, these views outline a developmental blueprint for replicating nephron formation in vitro.
Collapse
Affiliation(s)
- Nils Olof Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California.
| | - Jessica May Vanslambrouck
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
3
|
Cosgun ZC, Burgmaier K, Zeiher M, Weber A, Klein R, Aydin A, Kribs A, Mehler K, Habbig S. Urinary Output of Very Low Birth Weight Infants during the First Weeks of Life. Neonatology 2024; 122:244-250. [PMID: 39616995 DOI: 10.1159/000542755] [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: 07/26/2024] [Accepted: 11/19/2024] [Indexed: 04/03/2025]
Abstract
INTRODUCTION Daily urinary output (UOP) serves as important tool to identify acute kidney injury (AKI) in preterm infants. However, reference values for UOP, especially stratified for gestational age (GA), are missing. METHODS This retrospective single-center study assessed UOP during the first 28 days of life in 128 very low birth weight (VLBW) infants. RESULTS VLBW infants exhibit a highly dynamic daily UOP profile in the first 28 days of life with a maximum at day 12 with 4.78 mL/kg bodyweight/h. In the subcohort of 64 extremely low gestational age neonates (ELGANs), the highest UOP is measured during the second week of life. Infants born before 24 weeks of gestation have significantly higher UOP than more mature infants. CONCLUSION UOP is dynamic in the postnatal period and differs significantly between GA cohorts in the subgroup of ELGANs. These data might point to an adaption of the UOP threshold for neonatal AKI in preterm infants.
Collapse
Affiliation(s)
- Zülfü Cem Cosgun
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Kathrin Burgmaier
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Melanie Zeiher
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Anna Weber
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Ruth Klein
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Aynur Aydin
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Angela Kribs
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Katrin Mehler
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Sandra Habbig
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Cheng YJG, Chen CC, Cheng CJ. Postnatal renal tubule development: roles of tubular flow and flux. Curr Opin Nephrol Hypertens 2024; 33:518-525. [PMID: 38913022 PMCID: PMC11290981 DOI: 10.1097/mnh.0000000000001007] [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] [Indexed: 06/25/2024]
Abstract
PURPOSE OF REVIEW Postnatal renal tubule development is critical to adult kidney function. Several postnatal changes regulate the differentiation and proliferation of renal tubular cells. Here, we review the literature and our efforts on thick ascending limb (TAL) development in Bartter syndrome (BS). RECENT FINDINGS Glomerular filtrate quickly increases after birth, imposing fluid shear stress and circumferential stretch on immature renal tubules. Recent studies showed that kidney organoids under flow (superfusion) have better development of tubular structures and the expression of cilia and solute transporters. These effects are likely mediated by mechanosensors, such as cilia and the piezo1 channel. Improved renal oxygenation and sodium pump-dependent active transport can stimulate mitochondrial respiration and biogenesis. The functional coupling between transport and mitochondria ensures ATP supply for energy-demanding reactions in tubular cells, including cell cycle progression and proliferation. We recently discovered that postnatal renal medulla maturation and TAL elongation are impaired in Clc-k2-deficient BS mice. Primary cultured Clc-k2-deficient TAL cells have G1-S transition and proliferation delay. These developmental defects could be part of the early pathogenesis of BS and worsen the phenotype. SUMMARY Understanding how tubular flow and transepithelial ion fluxes regulate renal tubule development may improve the treatment of congenital renal tubulopathies.
Collapse
Affiliation(s)
- Yi-Jing G. Cheng
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
| | - Chien-Chou Chen
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Jen Cheng
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
5
|
Purswani MU, Jacobson DL, DiMeglio LA, Yao TJ, Kopp JB, Van Dyke RB, Yu W, Siberry GK. Phosphaturia in HIV-Exposed Uninfected Neonates Associated with Maternal Use of Tenofovir Disoproxil Fumarate in Late Pregnancy. J Pediatric Infect Dis Soc 2024; 13:396-405. [PMID: 38820092 DOI: 10.1093/jpids/piae054] [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: 09/17/2023] [Accepted: 05/29/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Tenofovir disoproxil fumarate (TDF) is often used in treating pregnant women living with HIV. Third-trimester TDF exposure is associated with a 12% reduction in bone mineral content in HIV-exposed uninfected (HEU) neonates. The potential mechanisms underlying this observation are unknown. METHODS The TDF study enrolled newborns of gestational age ≥36 weeks from the Surveillance Monitoring for Antiretroviral Therapy and Toxicities study based on in utero TDF exposure (TDF use ≥8 weeks in the third trimester vs none). Blood and urine samples were collected cross-sectionally within 30 days of birth to assess renal function (serum creatinine, serum phosphate, eGFR, percent tubular reabsorption of phosphate [PTRP]), and bone turnover (serum parathyroid hormone, 25-OH vitamin D [25(OH)D], and urinary cross-linked N-telopeptide of type 1 collagen). For each biomarker, a LOESS plot was fit using values at age at specimen collection; regression lines over age were fit among samples collected from 4 to 30 days, to compare slopes by TDF exposure. RESULTS Among 141 neonates, 77 were TDF-exposed and 64 TDF-unexposed. Between age 4 and 30 days, PTRP decreased more rapidly in the TDF-exposed compared to the unexposed group with slopes of -0.58 vs -0.08/day (difference -0.50/day [95% CI -0.88, -0.11]). Slopes for 25(OH)D were similar in both groups, but serum levels were lower in TDF-exposed neonates (median [IQR]: 22 [19, 29] vs 26 [22, 37] ng/mL). No differences were observed for other biomarkers. CONCLUSIONS Third-trimester in utero exposure to TDF is associated with increased urinary loss of phosphate and lower serum concentrations of 25(OH)D in HEU neonates.
Collapse
Affiliation(s)
- Murli U Purswani
- Division of Pediatric Infectious Disease, BronxCare Health System, Icahn School of Medicine at Mount Sinai, Bronx, New York, USA
| | - Denise L Jacobson
- Center for Biostatistics in AIDS Research, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | - Linda A DiMeglio
- Division of Pediatric Endocrinology and Diabetology, Riley Hospital for Children at IU Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tzy-Jyun Yao
- Center for Biostatistics in AIDS Research, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, NIDDK, NIH, Bethesda, Maryland, USA
| | - Russell B Van Dyke
- Section of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Wendy Yu
- Center for Biostatistics in AIDS Research, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | - George K Siberry
- Prevention Care and Treatment Division, Office of HIV/AIDS, United States Agency for International Development, Washington, District of Columbia, USA
| |
Collapse
|
6
|
Meesters K, Balbas-Martinez V, Allegaert K, Downes KJ, Michelet R. Personalized Dosing of Medicines for Children: A Primer on Pediatric Pharmacometrics for Clinicians. Paediatr Drugs 2024; 26:365-379. [PMID: 38755515 DOI: 10.1007/s40272-024-00633-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/18/2024]
Abstract
The widespread use of drugs for unapproved purposes remains common in children, primarily attributable to practical, ethical, and financial constraints associated with pediatric drug research. Pharmacometrics, the scientific discipline that involves the application of mathematical models to understand and quantify drug effects, holds promise in advancing pediatric pharmacotherapy by expediting drug development, extending applications, and personalizing dosing. In this review, we delineate the principles of pharmacometrics, and explore its clinical applications and prospects. The fundamental aspect of any pharmacometric analysis lies in the selection of appropriate methods for quantifying pharmacokinetics and pharmacodynamics. Population pharmacokinetic modeling is a data-driven method ('top-down' approach) to approximate population-level pharmacokinetic parameters, while identifying factors contributing to inter-individual variability. Model-informed precision dosing is increasingly used to leverage population pharmacokinetic models and patient data, to formulate individualized dosing recommendations. Physiologically based pharmacokinetic models integrate physicochemical drug properties with biological parameters ('bottom-up approach'), and is particularly valuable in situations with limited clinical data, such as early drug development, assessing drug-drug interactions, or adapting dosing for patients with specific comorbidities. The effective implementation of these complex models hinges on strong collaboration between clinicians and pharmacometricians, given the pivotal role of data availability. Promising advancements aimed at improving data availability encompass innovative techniques such as opportunistic sampling, minimally invasive sampling approaches, microdialysis, and in vitro investigations. Additionally, ongoing research efforts to enhance measurement instruments for evaluating pharmacodynamics responses, including biomarkers and clinical scoring systems, are expected to significantly bolster our capacity to understand drug effects in children.
Collapse
Affiliation(s)
- Kevin Meesters
- Department of Pediatrics, University of British Columbia, 4480 Oak Street, Vancouver, BC, V6H 3V4, Canada.
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
| | | | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Kevin J Downes
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
- qPharmetra LLC, Berlin, Germany
| |
Collapse
|
7
|
Romano J. Macropod Pediatrics. Vet Clin North Am Exot Anim Pract 2024; 27:245-261. [PMID: 38097494 DOI: 10.1016/j.cvex.2023.11.007] [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: 04/08/2024]
Abstract
Macropods belong to the marsupial family Macropodidae, which includes animals such as kangaroos and wallabies. Macropod offspring are highly altricial at birth and require specialized care and environmental conditions for healthy development. The care and management of pediatric macropods poses a challenge due to the unique physiology and reproductive strategy of macropods. In order to successfully work with pediatric macropods, clinical veterinarians should have knowledge of species-specific husbandry, normal postnatal development, and common medical conditions/treatments. With limited information available on macropod pediatric medicine, further research is warranted to improve the care and management of these animals in human care.
Collapse
Affiliation(s)
- Jon Romano
- Department of Veterinary Clinical Sciences, Exotics and Lab Animal Medicine, Long Island University College of Veterinary Medicine, 720 Northern Boulevard, Brookville, NY 11548, USA.
| |
Collapse
|
8
|
Segar JL, Jetton JG. Fluid and electrolyte management in the neonate and what can go wrong. Curr Opin Pediatr 2024; 36:198-203. [PMID: 37962361 PMCID: PMC10932865 DOI: 10.1097/mop.0000000000001308] [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] [Indexed: 11/15/2023]
Abstract
PURPOSE OF REVIEW This review highlights recent advances in understanding fluid and electrolyte homeostasis during the newborn period, including heightened recognition of fluid overload and acute kidney injury contributing to poor clinical outcomes. Particular attention is given towards the care of extremely preterm infants. RECENT FINDINGS Emerging data demonstrate (i) disproportionally large transepidermal water loss in the extremely preterm population, (ii) the relationship between postnatal weight loss (negative fluid balance) and improved outcomes, (iii) the frequency and negative effects of dysnatremias early in life, (iv) the role of sodium homeostasis in optimizing postnatal growth, and (v) the deleterious effects of fluid overload and acute kidney injury. SUMMARY As clinicians care for an increasing number of preterm infants, understanding progress in approaches to fluid and electrolyte management and avoidance of fluid overload states will improve the care and outcomes of this vulnerable population. Further translational and clinical studies are needed to address remaining knowledge gaps and improve current approaches to fluid and electrolyte management.
Collapse
Affiliation(s)
- Jeffrey L. Segar
- Sections of Neonatology, Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI53226
| | - Jennifer G. Jetton
- Pediatric Nephrology, Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI53226
| |
Collapse
|
9
|
Benjamin JI, Pollock DM. Current perspective on circadian function of the kidney. Am J Physiol Renal Physiol 2024; 326:F438-F459. [PMID: 38134232 PMCID: PMC11207578 DOI: 10.1152/ajprenal.00247.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023] Open
Abstract
Behavior and function of living systems are synchronized by the 24-h rotation of the Earth that guides physiology according to time of day. However, when behavior becomes misaligned from the light-dark cycle, such as in rotating shift work, jet lag, and even unusual eating patterns, adverse health consequences such as cardiovascular or cardiometabolic disease can arise. The discovery of cell-autonomous molecular clocks expanded interest in regulatory systems that control circadian physiology including within the kidney, where function varies along a 24-h cycle. Our understanding of the mechanisms for circadian control of physiology is in the early stages, and so the present review provides an overview of what is known and the many gaps in our current understanding. We include a particular focus on the impact of eating behaviors, especially meal timing. A better understanding of the mechanisms guiding circadian function of the kidney is expected to reveal new insights into causes and consequences of a wide range of disorders involving the kidney, including hypertension, obesity, and chronic kidney disease.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
10
|
Harshman L, Stalter E, Verhofste S, Dagle J, Steinbach E, Eyck PT, Wendt L, Segar J. Somatic growth outcomes in response to an individualized neonatal sodium supplementation protocol. RESEARCH SQUARE 2024:rs.3.rs-3911085. [PMID: 38405851 PMCID: PMC10889073 DOI: 10.21203/rs.3.rs-3911085/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Objective Evaluate the impact of a sodium (Na) supplementation protocol based upon urine Na concentration on growth parameters and morbidities. Study Design Retrospective cohort study of infants 260/7-336/7 weeks gestational age (GA) cared for before (2012-15, n = 225) and after (2016-20, n = 157) implementation of the protocol. Within- and between-group changes over time were assessed using repeated measures generalized linear models. Results For infants 260/7-296/7 weeks GA, utilization of the protocol was associated with increased mean body weight z-score at 8-weeks postnatal age, increased mean head circumference z-score at 16-weeks postnatal age, and decreased time on mechanical ventilation (all p < 0.02). No impact on growth was identified for infants 30-336/7 weeks GA. Incidences of hypertension, hypernatremia, bronchopulmonary dysplasia, and culture positive sepsis were unaffected by the protocol. Conclusion Protocolized Na supplementation results in improved growth and reduced time on invasive mechanical ventilation in extremely preterm infants without increasing incidence of morbidities.
Collapse
|
11
|
Alikhani R, Pai MP. Reconsideration of the current models of estimated kidney function-based drug dose adjustment in older adults: The role of biological age. Clin Transl Sci 2023; 16:2095-2105. [PMID: 37702349 PMCID: PMC10651638 DOI: 10.1111/cts.13643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Human lifespan has increased from a median of 46.5 years in 1950 to 71.7 years in 2022. As people age, one of the inevitable consequences is a decline in kidney function and glomerular filtration rate (GFR) which can have direct or indirect effects on the pharmacokinetic and pharmacodynamic profiles of many drugs. Numerous equations have been developed to generate estimated GFR (eGFR) using the two principal biomarkers: serum creatinine and serum cystatin C. However, the trajectory of changes with aging is dissimilar in these equations. In addition, there is recognition that chronological age (lifespan) often does not reflect biological age (healthspan) as an essential parameter in kidney function equations. In the past decade, there has been an increasing interest in quantifying biological age and new commercially available assays have entered the marketplace. In this narrative review, we illustrate how dominant equations of eGFR model the fractional change in this parameter very differently across chronological age. In addition, we review various biological age indicators (aging clocks) and challenges to their application in clinical practice. Importantly, by comparing vancomycin's mean clearance as a drug with limited metabolism and unchanged elimination between two age milestones in some recent population pharmacokinetic models, we show how efforts to quantify kidney function in older adults optimally remain under-explored, particularly those at the upper end of their lifespan. We also propose considering new models that integrate biological age as a new pathway to improve precision drug dosing in older adults.
Collapse
Affiliation(s)
- Radin Alikhani
- Department of Clinical Pharmacy, College of PharmacyUniversity of MichiganAnn ArborMichiganUSA
| | - Manjunath P. Pai
- Department of Clinical Pharmacy, College of PharmacyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
12
|
Sakkongviseth W, Sommai K, Sumboonnanonda A, Pattaragarn A, Supavekin S, Piyaphanee N, Lomjansook K, Thunsiribuddhichai Y, Chaiyapak T. Dysnatremia and subsequent sodium level changes following various intravenous treatments in infants with acute gastroenteritis. Eur J Pediatr 2023; 182:4741-4748. [PMID: 37581625 DOI: 10.1007/s00431-023-05151-5] [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: 05/25/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/16/2023]
Abstract
Acute gastroenteritis is one of the main causes of electrolyte imbalance in infants. We aimed to determine the frequency of and factors associated with dysnatremia at presentation and establish the ideal intravenous treatment scheme. The records of hospitalized infants aged 1-12 months with community-acquired acute gastroenteritis between January 2017 and March 2021 were retrospectively reviewed. Factors associated with dysnatremia at presentation were analyzed by multivariable logistic regression analysis. Subsequent sodium levels 4-24 h after intravenous fluid treatments, which were categorized into 2 groups, were determined in the subgroup of infants with normal sodium levels at presentation. A total of 347 infants with a median age of 8.0 (5.0-10.0) months were included. The frequency of dysnatremia at presentation was 14% (hyponatremia 12% and hypernatremia 2.0%). Severe dehydration was associated with dysnatremia at presentation (p = 0.048). Among 68 infants with normal sodium levels at presentation, the median sodium change was highest in the 5% dextrose in saline group, with changes of + 3 (0.5-5) and + 1 (- 2 to 2) mmol/L in infants who received 5% dextrose in saline and 5% dextrose in 1/3-1/2 saline, respectively (p = 0.001). Four out of 47 infants (8.5%) developed hyponatremia while receiving 5% dextrose in 1/3-1/2 saline. None of those who received 5% dextrose in saline developed subsequent dysnatremia. Conclusion: The frequency of dysnatremia at presentation among infants with acute gastroenteritis was 14%. Severe dehydration was associated with dysnatremia at presentation, so electrolyte levels need to be assessed in these patients. The use of isotonic solution did not promote acquired dysnatremia. This study supports once more that current guidelines recommending isotonic solution for children, and, especially, infant rehydration, are important also for infants in Thailand. What is Known: • There were a wide variation in the incidence of dysnatremia at presentation in children with acute gastroenteritis in previous pediatric series. • The AAP guidelines recommend using isotonic solution in children with acute illness from 28 days to 18 years of age to prevent acquired hyponatremia. What is New: • The incidence of dysnatremia at presentation in infants with acute gastroenteritis was 14% (hyponatremia 12% and hypernatremia 2.0%). • The use of isotonic solution did not promote acquired dysnatremia in infants with acute gastroenteritis.
Collapse
Affiliation(s)
- Wattanaporn Sakkongviseth
- Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokwan Sommai
- Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Achra Sumboonnanonda
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anirut Pattaragarn
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suroj Supavekin
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nuntawan Piyaphanee
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kraisoon Lomjansook
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yarnarin Thunsiribuddhichai
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanaporn Chaiyapak
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
13
|
Cremonesi A, Meili D, Rassi A, Poms M, Tavazzi B, Škopová V, Häberle J, Zikánová M, Hersberger M. Improved diagnostics of purine and pyrimidine metabolism disorders using LC-MS/MS and its clinical application. Clin Chem Lab Med 2023; 61:1792-1801. [PMID: 37011034 DOI: 10.1515/cclm-2022-1236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/27/2023] [Indexed: 04/04/2023]
Abstract
OBJECTIVES To develop a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to quantify 41 different purine and pyrimidine (PuPy) metabolites in human urine to allow detection of most known disorders in this metabolic pathway and to determine reference intervals. METHODS Urine samples were diluted with an aqueous buffer to minimize ion suppression. For detection and quantification, liquid chromatography was combined with electrospray ionization, tandem mass spectrometry and multiple reaction monitoring. Transitions and instrument settings were established to quantify 41 analytes and nine stable-isotope-labeled internal standards (IS). RESULTS The established method is precise (intra-day CV: 1.4-6.3%; inter-day CV: 1.3-15.2%), accurate (95.2% external quality control results within ±2 SD and 99.0% within ±3 SD; analyte recoveries: 61-121%), sensitive and has a broad dynamic range to quantify normal and pathological metabolite concentrations within one run. All analytes except aminoimidazole ribonucleoside (AIr) are stable before, during and after sample preparation. Moreover, analytes are not affected by five cycles of freeze-thawing (variation: -5.6 to 7.4%), are stable in thymol (variation: -8.4 to 12.9%) and the lithogenic metabolites also in HCl conserved urine. Age-dependent reference intervals from 3,368 urine samples were determined and used to diagnose 11 new patients within 7 years (total performed tests: 4,206). CONCLUSIONS The presented method and reference intervals enable the quantification of 41 metabolites and the potential diagnosis of up to 25 disorders of PuPy metabolism.
Collapse
Affiliation(s)
- Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - David Meili
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anahita Rassi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Poms
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Barbara Tavazzi
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Václava Škopová
- Research Unit for Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marie Zikánová
- Research Unit for Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Hersberger
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Leopoldino RWD, Marques DP, Rocha LC, Fernandes FEM, Oliveira AG, Martins RR. Temporal profile of adverse drug reactions and associated clinical factors: a prospective observational study in a neonatal intensive care unit. BMJ Open 2023; 13:e073304. [PMID: 37553191 PMCID: PMC10414078 DOI: 10.1136/bmjopen-2023-073304] [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: 03/02/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023] Open
Abstract
OBJECTIVE Although adverse drug reactions (ADRs) are quite common in hospitalised neonates, pharmacovigilance activities in this public are still incipient. This study aims to characterise ADRs in neonates in a neonatal intensive care unit (NICU), identifying causative drugs, temporal profile and associated factors. DESIGN Prospective observational study. SETTING NICU of a public maternity hospital in Natal/Brazil. PARTICIPANTS All neonates admitted to the NICU for more than 24 hours and using at least one medication were followed up during the time of hospitalisation. PRIMARY OUTCOME MEASURES Incidence rate and risk factors for ADRs. The ADRs were detected by an active search in electronic medical records and analysis of spontaneous reports in the hospital pharmacovigilance system. RESULTS Six hundred neonates were included in the study, where 118 neonates had a total of 186 ADRs. The prevalence of ADRs at the NICU was 19.7% (95% CI 16.7% to 23.0%). The most common ADRs were tachycardia (30.6%), polyuria (9.1%) and hypokalaemia (8.6%). Tachycardia (peak incidence rate: 57.1 ADR/1000 neonates) and hyperthermia (19.1 ADR/1000 neonates) predominated during the first 5 days of hospitalisation. The incidence rate of polyuria and hypokalaemia increased markedly after the 20th day, with both reaching a peak of 120.0 ADR/1000 neonates. Longer hospitalisation time (OR 0.018, 95% CI 0.007 to 0.029; p<0.01) and number of prescribed drugs (OR 0.127, 95% CI 0.075 to 0.178; p<0.01) were factors associated with ADRs. CONCLUSION ADRs are very common in NICU, with tachycardia and hyperthermia predominant in the first week of hospitalisation and polyuria and hypokalaemia from the third week onwards.
Collapse
Affiliation(s)
- Ramon Weyler Duarte Leopoldino
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Daniel Paiva Marques
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Luan Carvalho Rocha
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Flavia Evelyn Medeiros Fernandes
- School Maternity Januário Cicco (MEJC/EBSERH), Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Antonio Gouveia Oliveira
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Rand Randall Martins
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| |
Collapse
|
15
|
Kamianowska M, Rybi-Szumińska A, Kamianowska A, Maciejczyk M, Sołomianko K, Koput A, Wasilewska A. The Urinary Concentration of Trefoil Factor 3 (TFF3) in the Term and Preterm Neonates. J Clin Med 2023; 12:4936. [PMID: 37568337 PMCID: PMC10419516 DOI: 10.3390/jcm12154936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Distinguishing between a pathologic state and renal development is important in neonatology. Because the assessment of serum creatinine in neonates is not reliable, better biomarkers are needed. Trefoil factor 3 (TFF3) is proposed as a biomarker of kidney injury. The study aimed to assess its urinary concentration in healthy term and stable preterm neonates. MATERIAL AND METHODS The study included 80 term and 20 preterm neonates born in the Department of Perinatology of the University Clinical Hospital in Bialystok. Urine was obtained from the term neonates on the 1st day of life and from the preterm neonates on the 1st, 8th, 15th and 22nd day of life. The urinary concentration of TFF3 was determined using a commercially available immunoassay and was normalized for the urinary creatinine concentration (cr.). RESULTS The values of TFF3/cr. were higher in the preterm than in the term neonates (p < 0.05) (median (Q1-Q3): 1486.85 (614.92-3559.18) and 317.29 (68.07-671.40) ng/mg cr.). They did not differ in the subsequent days of the preterm neonates' lives. The ROC curve for TFF3/cr. in the preterm and term neonates showed AUC = 0.751 (cut-off value = 1684.25 ng/mg cr.). CONCLUSIONS Prematurity is associated with higher urinary excretion of TFF3. Male gender is associated with an increased urinary TFF3 excretion in term neonates.
Collapse
Affiliation(s)
- Monika Kamianowska
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Agnieszka Rybi-Szumińska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Aleksandra Kamianowska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomic, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Katarzyna Sołomianko
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Alicja Koput
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Anna Wasilewska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| |
Collapse
|
16
|
Balázs G, Balajthy A, Seri I, Hegyi T, Ertl T, Szabó T, Röszer T, Papp Á, Balla J, Gáll T, Balla G. Prevention of Chronic Morbidities in Extremely Premature Newborns with LISA-nCPAP Respiratory Therapy and Adjuvant Perinatal Strategies. Antioxidants (Basel) 2023; 12:1149. [PMID: 37371878 DOI: 10.3390/antiox12061149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Less invasive surfactant administration techniques, together with nasal continuous airway pressure (LISA-nCPAP) ventilation, an emerging noninvasive ventilation (NIV) technique in neonatology, are gaining more significance, even in extremely premature newborns (ELBW), under 27 weeks of gestational age. In this review, studies on LISA-nCPAP are compiled with an emphasis on short- and long-term morbidities associated with prematurity. Several perinatal preventative and therapeutic investigations are also discussed in order to start integrated therapies as numerous organ-saving techniques in addition to lung-protective ventilations. Two thirds of immature newborns can start their lives on NIV, and one third of them never need mechanical ventilation. With adjuvant intervention, these ratios are expected to be increased, resulting in better outcomes. Optimized cardiopulmonary transition, especially physiologic cord clamping, could have an additively beneficial effect on patient outcomes gained from NIV. Organ development and angiogenesis are strictly linked not only in the immature lung and retina, but also possibly in the kidney, and optimized interventions using angiogenic growth factors could lead to better morbidity-free survival. Corticosteroids, caffeine, insulin, thyroid hormones, antioxidants, N-acetylcysteine, and, moreover, the immunomodulatory components of mother's milk are also discussed as adjuvant treatments, since immature newborns deserve more complex neonatal interventions.
Collapse
Affiliation(s)
- Gergely Balázs
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - András Balajthy
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - István Seri
- First Department of Pediatrics, School of Medicine, Semmelweis University, 1083 Budapest, Hungary
- Keck School of Medicine of USC, Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Thomas Hegyi
- Department of Pediatrics, Division of Neonatology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Tibor Ertl
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Papp
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Gáll
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Araya BR, Ziegler AA, Grobe CC, Grobe JL, Segar JL. Sodium and Growth in Preterm Infants: A Review. NEWBORN (CLARKSVILLE, MD.) 2023; 2:142-147. [PMID: 37614871 PMCID: PMC10445331 DOI: 10.5005/jp-journals-11002-0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Aim This article is intended to review the relationship between sodium homeostasis and growth, outline reasons why preterm infants may become sodium deficient, and share data from our group and others regarding the potential benefits of dietary sodium supplementation. Background Despite tremendous efforts over the past 20 years to optimize neonatal nutrition, postnatal growth failure in preterm infants remains a significant problem. Compelling associations have been identified between in-hospital growth failure and cardiometabolic and neurodevelopmental disorders, heightening the need to further identify the optimal nutritional needs of preterm infants. Results The impact of sodium deficiency may have on somatic growth is poorly studied and reported upon within the human literature. In contrast, animal studies dating back almost 100 years highlight the nutritional importance of dietary sodium. Sodium homeostasis during early postnatal life is understudied and underappreciated by neonatologists. Conclusion Insufficient sodium intake during early life is likely a critical yet underappreciated contributor to growth failure. Total body sodium depletion may be an important risk factor driving complications of premature birth. Clinical significance Increased awareness of sodium homeostasis in preterm infants may improve outcomes in this population. Sodium intake recommendations are provided based on the interpretation of currently available literature.
Collapse
Affiliation(s)
- Benjamin R Araya
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Alisha A Ziegler
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Connie C Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jeffrey L Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
18
|
Trigolet M, Bonsante F, Guignard JP, Gouyon JB, Iacobelli S. Urinary protein to creatinine ratio during the first month of life in very preterm infants-a prospective cohort study (PROTIPREMA). Pediatr Nephrol 2023; 38:721-727. [PMID: 35759001 DOI: 10.1007/s00467-022-05653-8] [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: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Preterm infants have physiological proteinuria and values of urine protein to creatinine ratio (UPr/Cr) are higher compared to full-term infants during the first week of life. Few investigations explored the changes of proteinuria in very preterm infants (VPI, ≤ 31 weeks of gestation) older than a week, and it is unclear whether high and persistent proteinuria is associated with kidney injury in this population. This study aimed to (1) observe the changes of UPr/Cr during the first month of life in VPI and (2) describe clinical and biological variables associated with the changes of UPr/Cr. METHODS Spot urine samples for UPr/Cr were collected on the first day of life (DOL1) and then on DOL2-3, DOL5-6, second week of life (WOL2), WOL3, and WOL4 in VPI cared for in a third-level NICU. We tested the relationship of UPr/Cr with perinatal variables and diseases. RESULTS A total of 1140 urine samples were obtained for 190 infants. UPr/Cr values (mg/mmol) (median with interquartile) at DOL1, DOL2, DOL3, WOL2, WOL3, and WOL4 were, respectively, 191 (114-399), 226 (152-319), 225 (156-350), 282 (200-488), 308 (188-576), and 325 (175-664). At the multivariate analysis, lower gestational age (GA) and increasing postnatal age were the only variables significantly associated with higher UPr/Cr values (p < 0.001). There was wide intra- and interindividual variability in UPr/Cr, especially in infants with higher GA and clinical stability. CONCLUSIONS In VPI, UPr/Cr is higher at lower GA and increases with advancing postnatal age. High persistent proteinuria is not associated with clinical and biological variables reflecting kidney injury during the first month of life. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Marine Trigolet
- Néonatologie, Réanimation Néonatale Et Pédiatrique, CHU La Réunion, Site Sud, Saint Pierre, France
| | - Francesco Bonsante
- Néonatologie, Réanimation Néonatale Et Pédiatrique, CHU La Réunion, Site Sud, Saint Pierre, France
- Centre d'Études Périnatales de L'Océan Indien, Université de La Réunion, UR7388, Saint Pierre, France
| | | | - Jean-Bernard Gouyon
- Centre d'Études Périnatales de L'Océan Indien, Université de La Réunion, UR7388, Saint Pierre, France
| | - Silvia Iacobelli
- Néonatologie, Réanimation Néonatale Et Pédiatrique, CHU La Réunion, Site Sud, Saint Pierre, France.
- Centre d'Études Périnatales de L'Océan Indien, Université de La Réunion, UR7388, Saint Pierre, France.
| |
Collapse
|
19
|
Meesters K, Alemayehu T, Benou S, Buonsenso D, Decloedt EH, Pillay-Fuentes Lorente V, Downes KJ, Allegaert K. Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review. Antibiotics (Basel) 2022; 12:17. [PMID: 36671218 PMCID: PMC9854442 DOI: 10.3390/antibiotics12010017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/25/2022] Open
Abstract
Effective antimicrobial exposure is essential to treat infections and prevent antimicrobial resistance, both being major public health problems in low and middle income countries (LMIC). Delivery of drug concentrations to the target site is governed by dose and pharmacokinetic processes (absorption, distribution, metabolism and excretion). However, specific data on the pharmacokinetics of antimicrobials in children living in LMIC settings are scarce. Additionally, there are significant logistical constraints to therapeutic drug monitoring that further emphasize the importance of understanding pharmacokinetics and dosing in LMIC. Both malnutrition and diarrheal disease reduce the extent of enteral absorption. Multiple antiretrovirals and antimycobacterial agents, commonly used by children in low resource settings, have potential interactions with other antimicrobials. Hypoalbuminemia, which may be the result of malnutrition, nephrotic syndrome or liver failure, increases the unbound concentrations of protein bound drugs that may therefore be eliminated faster. Kidney function develops rapidly during the first years of life and different inflammatory processes commonly augment renal clearance in febrile children, potentially resulting in subtherapeutic drug concentrations if doses are not adapted. Using a narrative review approach, we outline the effects of growth, maturation and comorbidities on maturational and disease specific effects on pharmacokinetics in children in LMIC.
Collapse
Affiliation(s)
- Kevin Meesters
- Department of Pediatrics, BC Children’s Hospital and The University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Tinsae Alemayehu
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Child Health, St. Paul’s Hospital Millennium Medical College, Addis Ababa P.O. Box 1271, Ethiopia
- Division of Infectious Diseases and Travel Medicine, American Medical Center, Addis Ababa P.O. Box 62706, Ethiopia
| | - Sofia Benou
- Department of Pediatrics, General University Hospital of Patras, Medical School, University of Patras, 26504 Rion, Greece
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
- Centro di Salute Globale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Eric H. Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7500, South Africa
| | - Veshni Pillay-Fuentes Lorente
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7500, South Africa
| | - Kevin J. Downes
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Division of Infectious Diseases, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
- Department of Clinical Pharmacy, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
20
|
Yen CW, Yu MC, Lee J. Serum electrolyte abnormalities in pediatric patients presenting to an emergency department with various diseases: Age-related differences. Pediatr Neonatol 2022; 63:575-581. [PMID: 35987755 DOI: 10.1016/j.pedneo.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/14/2022] [Accepted: 04/28/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND This study evaluated the prevalence and frequency of serum electrolyte abnormalities (SEAs) in children presenting to a pediatric emergency department (PED) with various diseases. METHODS Pediatric patients (≤18 years) with blood electrolyte panels obtained in the PED of Lin-Kou Chang Gung Memorial Hospital, Taiwan, in the 5 years from January 1, 2016, to August 31, 2021, were enrolled in this retrospective observational study. Patients were divided into three age groups: Group A, < 4 years; Group B, 4-11 years; and Group C, 12-18 years. The associations between SEAs and clinical diseases in children and age-related differences were assessed. RESULTS This study included 182,058 pediatric patients visiting our PED over a 5-year period. A total of 250 (0.14%) patients with SEAs were included in the analysis. The study population consisted of 127 boys and 123 girls with a median (IQR) age of 9.0 (3.2-14.1) years. Hospital admission was required in 86.4% (n = 216) of the patients, and 32.4% (n = 81) of them were admitted to the pediatric intensive care unit (PICU). The median (IQR) hospital stay and PICU stay was 6.5 (4.0-11.0) and 4.0 (3.0-8.0) days, respectively. The PICU stay was longer in Group A (p < 0.05) and shorter in group C (p < 0.05). Hyponatremia was the most common SEA in group A (46.3%, n = 31), while hypokalemia was common in groups B (54.2%, n = 52) and C (32.2%, n = 28). Gastrointestinal, renal, and endocrine diseases were common clinical conditions associated with SEAs in pediatric patients in our PED. CONCLUSION The detection rate of SEAs in patients in the PED was 0.14%. Hyponatremia was a common SEA in pediatric patients aged <4 years, while the most common electrolyte disorder in those >4 years old was hypokalemia. In infants and young children, SEAs were associated with a longer PICU stay.
Collapse
Affiliation(s)
- Chen-Wei Yen
- Division of Pediatric General Medicine, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mei-Ching Yu
- Department of Pediatric Nephrology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jung Lee
- Division of Pediatric General Medicine, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
21
|
Fanous MS, Afolabi JM, Michael OS, Falayi OO, Iwhiwhu SA, Adebiyi A. Transdermal Measurement of Glomerular Filtration Rate in Mechanically Ventilated Piglets. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2022:10.3791/64413. [PMID: 36190295 PMCID: PMC9835146 DOI: 10.3791/64413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Transdermal measurement of glomerular filtration rate (GFR) has been used to evaluate kidney function in conscious animals. This technique is well established in rodents to study acute kidney injury and chronic kidney disease. However, GFR measurement using the transdermal system has not been validated in pigs, a species with a similar renal system to humans. Hence, we investigated the effect of sepsis on transdermal GFR in anesthetized and mechanically ventilated neonatal pigs. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP). The transdermal GFR measurement system consisting of a miniaturized fluorescence sensor was attached to the pig's shaved skin to determine the clearance of fluorescein-isothiocyanate (FITC) conjugated sinistrin, an intravenously injected GFR tracer. Our results show that at 12 h post-CLP, serum creatinine increased with a decrease in GFR. This study demonstrates, for the first time, the utility of the transdermal GFR approach in determining renal function in mechanically ventilated, neonatal pigs.
Collapse
Affiliation(s)
- Mina S. Fanous
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| | - Jeremiah M. Afolabi
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| | - Olugbenga S. Michael
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| | - Olufunke O. Falayi
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| | - Samson A. Iwhiwhu
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| | - Adebowale Adebiyi
- Department of Physiology, College of Medicine University of Tennessee Health Science Center
| |
Collapse
|
22
|
Sakamoto M, Haraguchi K, Tatsuta N, Marumoto M, Yamamoto M, Nakamura M. Breast milk contribution to tissue mercury levels in rat pups examined by cross-fostering at birth. ENVIRONMENTAL RESEARCH 2022; 208:112772. [PMID: 35063428 DOI: 10.1016/j.envres.2022.112772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
The developing perinatal brain is vulnerable to methylmercury (MeHg) exposure. The contribution of breast milk to tissue MeHg levels in offspring is a significant public health concern because breast milk contains a certain amount of MeHg. Here, the contribution of MeHg transferred via breast milk to the Hg levels in the tissues of pups (Wistar rats) was investigated. Mated maternal rats were fed a MeHg (2 ppm)-supplemented or a control diet during pregnancy. Following parturition, male neonates from each group were cross-fostered between exposed or control dams, and they were further raised by dams fed a MeHg-supplemented diet or a control diet during lactation. Consequently, we evaluated three pup groups, which were raised by dams exposed to MeHg during pregnancy (P pups), lactation (L pups), or pregnancy and lactation (PL pups). Total mercury (THg) concentrations in the tissues of the offspring were measured at birth (postnatal day 0 [PD0]), during lactation (PD6, PD12, and PD19), and after weaning (PD29 and PD36). Blood and brain THg levels in the P and PL pups declined dramatically during lactation, however, there were no considerable differences between the two groups at PD6 and PD12. In contrast, blood and brain THg levels in the L pups increased slightly during lactation. The increase in the THg levels in the blood and brain of L pups at PD12 were approximately 3.3% and 1.5%, respectively, compared to the corresponding THg levels in the neonates in the P and PL groups. Our results suggest that if the MeHg exposure level during pregnancy is not high enough to cause neuronal development defects in the fetus, the exposure via breast milk is not a significant concern.
Collapse
Affiliation(s)
- Mineshi Sakamoto
- National Institute for Minamata Disease, Minamata, Kumamoto, 867-0008, Japan.
| | - Koichi Haraguchi
- National Institute for Minamata Disease, Minamata, Kumamoto, 867-0008, Japan
| | - Nozomi Tatsuta
- Tohoku University School of Medicine, Sendai, 880-2112, Japan
| | - Masumi Marumoto
- National Institute for Minamata Disease, Minamata, Kumamoto, 867-0008, Japan
| | - Megumi Yamamoto
- National Institute for Minamata Disease, Minamata, Kumamoto, 867-0008, Japan
| | - Masaaki Nakamura
- National Institute for Minamata Disease, Minamata, Kumamoto, 867-0008, Japan
| |
Collapse
|
23
|
Neumann U, van der Linde A, Krone RE, Krone NP, Güven A, Güran T, Elsedfy H, Poyrazoglu S, Darendeliler F, Bachega TASS, Balsamo A, Hannema SE, Birkebaek N, Vieites A, Thankamony A, Cools M, Milenkovic T, Bonfig W, Costa EC, Atapattu N, de Vries L, Guaragna-Filho G, Korbonits M, Mohnike K, Bryce J, Ahmed SF, Voet B, Blankenstein O, Claahsen-van der Grinten HL. Treatment of congenital adrenal hyperplasia in children aged 0-3 years: a retrospective multicenter analysis of salt supplementation, glucocorticoid and mineralocorticoid medication, growth and blood pressure. Eur J Endocrinol 2022; 186:587-596. [PMID: 35290211 PMCID: PMC9066592 DOI: 10.1530/eje-21-1085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 03/15/2022] [Indexed: 12/04/2022]
Abstract
Objectives International guidelines recommend additional salt supplementation during infancy in classic congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency. The influence of corticoid medication and growth has not been assessed. Aim To investigate the current use of salt supplementation, fludrocortisone (FC) and hydrocortisone (HC) dosage as well as weight, height, BMI and blood pressure (BP) in CAH children aged 0-3 years. Methods Retrospective multicentre analysis using data from the I-CAH registry. Salt-treated (ST) and non-salt-treated (NST) children were compared regarding FC and HC dosage, weight, height and BP at 0, 3, 6, 9, 12, 18, 24, 30, and 36 months. Results We analysed 2483 visits of 331 patients born after year 2000 in 13 countries (male, n = 145) with 203 ST patients (61%). NST children had significantly higher FC dosages at 1.5-4.5 months and higher HC dosages until 1.5 months of age. No differences in weight, length and BP between subgroups were observed. Children of the whole cohort showed increased BMI-SDS during the study period and about half of the reported BP readings were >P95. Conclusion In children treated with additional salt supplementation, FC and HC dosages are lower during the first months of life but without differences in weight, length and BP until 3 years of age compared to NST children. All children showed an increase in BMI-SDS and a high rate of BP readings >P95 until 3 years, indicating the start of weight gain and negative effects on blood pressure already in very early life.
Collapse
Affiliation(s)
- Uta Neumann
- Institute for Experimental Paediatric Endocrinology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Annelieke van der Linde
- Amalia Children’s Hospital, Radboud University Medical Centre, Nijmegen, Netherlands
- Amphia Hospital, Breda, The Netherlands
| | - Ruth E Krone
- Birmingham Women’s and Children’s Hospital, Birmingham, UK
| | - Nils P Krone
- University of Sheffield, Sheffield Children’s Hospital, Western Bank, Sheffield, UK
| | - Ayla Güven
- University of Health Science Zeynep Kamil Women and Children Hospital, Pediatric Endocrinology, Istanbul, Turkey
| | - Tülay Güran
- Marmara University Istanbul, Istanbul, Turkey
| | - Heba Elsedfy
- Pediatrics Department, Ain Shams University, Cairo, Egypt
| | - Sukran Poyrazoglu
- Pediatric Endocrinology Unit, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Feyza Darendeliler
- Pediatric Endocrinology Unit, Istanbul Faculty of Medicine, Istanbul, Turkey
| | | | | | - Sabine E Hannema
- Leiden University Medical Centre, Leiden, Netherlands
- Erasmus Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Niels Birkebaek
- Department of Pediatrics and Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Ana Vieites
- Centro de Investigaciones Endocrinológicas Buenos Aires, Buenos Aires, Argentina
| | - Ajay Thankamony
- University of Cambridge and Addenbrooke’s Hospital, Cambridge, UK
| | | | - Tatjana Milenkovic
- Institute for Mother and Child Healthcare of Serbia ‘Dr Vukan Čupić’, Belgrade, Serbia
| | - Walter Bonfig
- Technical University of Munich, Munich, Germany
- Klinikum Wels-Grieskirchen, Wels, Austria
| | | | | | - Liat de Vries
- Institute for Diabetes and Endocrinology, Schneider Children's Medical Center of Israel, Petah-Tikvah, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | - Klaus Mohnike
- Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | | | | | | | - Oliver Blankenstein
- Institute for Experimental Paediatric Endocrinology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
24
|
Voggel J, Mohr J, Nüsken KD, Dötsch J, Nüsken E, Alejandre Alcazar MA. Translational insights into mechanisms and preventive strategies after renal injury in neonates. Semin Fetal Neonatal Med 2022; 27:101245. [PMID: 33994314 DOI: 10.1016/j.siny.2021.101245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adverse perinatal circumstances can cause acute kidney injury (AKI) and contribute to chronic kidney disease (CKD). Accumulating evidence indicate that a wide spectrum of perinatal conditions interferes with normal kidney development and ultimately leads to aberrant kidney structure and function later in life. The present review addresses the lack of mechanistic knowledge with regard to perinatal origins of CKD and provides a comprehensive overview of pre- and peri-natal insults, including genetic predisposition, suboptimal nutritional supply, obesity and maternal metabolic disorders as well as placental insufficiency leading to intrauterine growth restriction (IUGR), prematurity, infections, inflammatory processes, and the need for life-saving treatments (e.g. oxygen supplementation, mechanical ventilation, medications) in neonates. Finally, we discuss future preventive, therapeutic, and regenerative directions. In summary, this review highlights the perinatal vulnerability of the kidney and the early origins of increased susceptibility toward AKI and CKD during postnatal life. Promotion of kidney health and prevention of disease require the understanding of perinatal injury in order to optimize perinatal micro- and macro-environments and enable normal kidney development.
Collapse
Affiliation(s)
- Jenny Voggel
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Jasmine Mohr
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Kai-Dietrich Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Jörg Dötsch
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Eva Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Miguel A Alejandre Alcazar
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany; Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine, University Hospital Cologne Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Gießen, Germany.
| |
Collapse
|
25
|
Claahsen - van der Grinten HL, Speiser PW, Ahmed SF, Arlt W, Auchus RJ, Falhammar H, Flück CE, Guasti L, Huebner A, Kortmann BBM, Krone N, Merke DP, Miller WL, Nordenström A, Reisch N, Sandberg DE, Stikkelbroeck NMML, Touraine P, Utari A, Wudy SA, White PC. Congenital Adrenal Hyperplasia-Current Insights in Pathophysiology, Diagnostics, and Management. Endocr Rev 2022; 43:91-159. [PMID: 33961029 PMCID: PMC8755999 DOI: 10.1210/endrev/bnab016] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Indexed: 11/19/2022]
Abstract
Congenital adrenal hyperplasia (CAH) is a group of autosomal recessive disorders affecting cortisol biosynthesis. Reduced activity of an enzyme required for cortisol production leads to chronic overstimulation of the adrenal cortex and accumulation of precursors proximal to the blocked enzymatic step. The most common form of CAH is caused by steroid 21-hydroxylase deficiency due to mutations in CYP21A2. Since the last publication summarizing CAH in Endocrine Reviews in 2000, there have been numerous new developments. These include more detailed understanding of steroidogenic pathways, refinements in neonatal screening, improved diagnostic measurements utilizing chromatography and mass spectrometry coupled with steroid profiling, and improved genotyping methods. Clinical trials of alternative medications and modes of delivery have been recently completed or are under way. Genetic and cell-based treatments are being explored. A large body of data concerning long-term outcomes in patients affected by CAH, including psychosexual well-being, has been enhanced by the establishment of disease registries. This review provides the reader with current insights in CAH with special attention to these new developments.
Collapse
Affiliation(s)
| | - Phyllis W Speiser
- Cohen Children’s Medical Center of NY, Feinstein Institute, Northwell Health, Zucker School of Medicine, New Hyde Park, NY 11040, USA
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine Dentistry & Nursing, University of Glasgow, Glasgow, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Endocrinology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Departments of Internal Medicine and Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Intitutet, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Angela Huebner
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Barbara B M Kortmann
- Radboud University Medical Centre, Amalia Childrens Hospital, Department of Pediatric Urology, Nijmegen, The Netherlands
| | - Nils Krone
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Deborah P Merke
- National Institutes of Health Clinical Center and the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Walter L Miller
- Department of Pediatrics, Center for Reproductive Sciences, and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| | - Anna Nordenström
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Nicole Reisch
- Medizinische Klinik IV, Klinikum der Universität München, Munich, Germany
| | - David E Sandberg
- Department of Pediatrics, Susan B. Meister Child Health Evaluation and Research Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, Center for Rare Endocrine Diseases of Growth and Development, Center for Rare Gynecological Diseases, Hôpital Pitié Salpêtrière, Sorbonne University Medicine, Paris, France
| | - Agustini Utari
- Division of Pediatric Endocrinology, Department of Pediatrics, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory of Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Justus Liebig University, Giessen, Germany
| | - Perrin C White
- Division of Pediatric Endocrinology, UT Southwestern Medical Center, Dallas TX 75390, USA
| |
Collapse
|
26
|
Hall AM, Trepiccione F, Unwin RJ. Drug toxicity in the proximal tubule: new models, methods and mechanisms. Pediatr Nephrol 2022; 37:973-982. [PMID: 34050397 PMCID: PMC9023418 DOI: 10.1007/s00467-021-05121-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/23/2021] [Accepted: 05/05/2021] [Indexed: 10/28/2022]
Abstract
The proximal tubule (PT) reabsorbs most of the glomerular filtrate and plays an important role in the uptake, metabolism and excretion of xenobiotics. Some therapeutic drugs are harmful to the PT, and resulting nephrotoxicity is thought to be responsible for approximately 1 in 6 of cases of children hospitalized with acute kidney injury (AKI). Clinically, PT dysfunction leads to urinary wasting of important solutes normally reabsorbed by this nephron segment, leading to systemic complications such as bone demineralization and a clinical scenario known as the renal Fanconi syndrome (RFS). While PT defects can be diagnosed using a combination of blood and urine markers, including urinary excretion of low molecular weight proteins (LMWP), standardized definitions of what constitutes clinically significant toxicity are lacking, and identifying which patients will go on to develop progressive loss of kidney function remains a major challenge. In addition, much of our understanding of cellular mechanisms of drug toxicity is still limited, partly due to the constraints of available cell and animal models. However, advances in new and more sophisticated in vitro models of the PT, along with the application of high-content analytical methods that can provide readouts more relevant to the clinical manifestations of nephrotoxicity, are beginning to extend our knowledge. Such technical progress should help in discovering new biomarkers that can better detect nephrotoxicity earlier and predict its long-term consequences, and herald a new era of more personalized medicine.
Collapse
Affiliation(s)
- Andrew M. Hall
- grid.7400.30000 0004 1937 0650Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland ,grid.412004.30000 0004 0478 9977Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- grid.9841.40000 0001 2200 8888Department of Translational Medical Science, University of Campania ‘Luigi Vanvitelli’, Naples, Italy ,grid.428067.f0000 0004 4674 1402Biogem Research Institute, Ariano Irpino, Italy
| | - Robert J. Unwin
- grid.83440.3b0000000121901201Department of Renal Medicine, University College London, London, UK
| |
Collapse
|
27
|
Guignard JP, Iacobelli S. Use of diuretics in the neonatal period. Pediatr Nephrol 2021; 36:2687-2695. [PMID: 33481099 DOI: 10.1007/s00467-021-04921-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/06/2020] [Accepted: 01/05/2021] [Indexed: 12/31/2022]
Abstract
The use of diuretics is extremely frequent in sick neonates, the more so in very premature newborn infants. The use of diuretics in patients whose kidney function is immature necessitates a thorough knowledge of renal developmental physiology and pathophysiology. This review presents the basic aspects of body fluid homeostasis in the neonate, discusses the development of kidney function, and describes the mechanisms involved in electrolyte and water reabsorption along the nephron. Diuretics are then classified according to the site of their action on sodium reabsorption. The use of diuretics in sodium-retaining states, in oliguric states, in electrolyte disorders, and in arterial hypertension, as well as in a few specific disorders, is presented. Common and specific adverse effects are discussed. Recommended dosages for the main diuretics used in the neonatal period are given. New developments in diuretic therapy are briefly mentioned.
Collapse
Affiliation(s)
| | - Silvia Iacobelli
- Néonatologie, Réanimation Néonatale et Pédiatrique, CHU La Réunion, Site Sud, Saint Pierre, France. .,Centre d'Etudes Périnatales de l'Océan Indien, CHU La Réunion, Saint Pierre et Université de la Réunion, Site Sud, EA 7388, Saint Pierre, France.
| |
Collapse
|
28
|
Lozic M, Minarik L, Racetin A, Filipovic N, Saraga Babic M, Vukojevic K. CRKL, AIFM3, AIF, BCL2, and UBASH3A during Human Kidney Development. Int J Mol Sci 2021; 22:ijms22179183. [PMID: 34502088 PMCID: PMC8431184 DOI: 10.3390/ijms22179183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
We aimed to investigate the spatio-temporal expression of possible CAKUT candidate genes CRKL, AIFM3, and UBASH3A, as well as AIF and BCL2 during human kidney development. Human fetal kidney tissue was stained with antibodies and analyzed by fluorescence microscopy and RT-PCR. Quantification of positive cells was assessed by calculation of area percentage and counting cells in nephron structures. Results showed statistically significant differences in the temporal expression patterns of the examined markers, depending on the investigated developmental stage. Limited but strong expression of CRKL was seen in developing kidneys, with increasing expression up to the period where the majority of nephrons are formed. Results also lead us to conclude that AIFM3 and AIF are important for promoting cell survival, but only AIFM3 is considered a CAKUT candidate gene due to the lack of AIF in nephron developmental structures. Our findings imply great importance of AIFM3 in energy production in nephrogenesis and tubular maturation. UBASH3A raw scores showed greater immunoreactivity in developing structures than mature ones which would point to a meaningful role in nephrogenesis. The fact that mRNA and proteins of CRKL, UBASH3A, and AIFM3 were detected in all phases of kidney development implies their role as renal development control genes.
Collapse
Affiliation(s)
- Mirela Lozic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
| | - Luka Minarik
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
- Department of Medical Genetics, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Natalija Filipovic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
| | - Mirna Saraga Babic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
| | - Katarina Vukojevic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia; (M.L.); (L.M.); (A.R.); (N.F.); (M.S.B.)
- Department of Medical Genetics, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Correspondence: ; Tel.: +385-21-557-807; Fax: +385-21-557-811
| |
Collapse
|
29
|
Thomson RB, Dynia DW, Burlein S, Thomson BR, Booth CJ, Knauf F, Wang T, Aronson PS. Deletion of Cdh16 Ksp-cadherin leads to a developmental delay in the ability to maximally concentrate urine in mouse. Am J Physiol Renal Physiol 2021; 320:F1106-F1122. [PMID: 33938239 DOI: 10.1152/ajprenal.00556.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system of the mammalian kidney. The principal aim of the present study was to determine if Ksp-cadherin played a critical role in the development and maintenance of the adult mammalian kidney by generating and evaluating a mouse line deficient in Ksp-cadherin. Ksp-null mutant animals were viable and fertile, and kidneys from both neonates and adults showed no evidence of structural abnormalities. Immunolocalization and Western blot analyses of Na+-K+-ATPase and E-cadherin indicated that Ksp-cadherin is not essential for either the genesis or maintenance of the polarized tubular epithelial phenotype. Moreover, E-cadherin expression was not altered to compensate for Ksp-cadherin loss. Plasma electrolytes, total CO2, blood urea nitrogen, and creatinine levels were also unaffected by Ksp-cadherin deficiency. However, a subtle but significant developmental delay in the ability to maximally concentrate urine was detected in Ksp-null mice. Expression analysis of the principal proteins involved in the generation of the corticomedullary osmotic gradient and the resultant movement of water identified misexpression of aquaporin-2 in the inner medullary collecting duct as the possible cause for the inability of young adult Ksp-cadherin-deficient animals to maximally concentrate their urine. In conclusion, Ksp-cadherin is not required for normal kidney development, but its absence leads to a developmental delay in maximal urinary concentrating ability.NEW & NOTEWORTHY Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system. Using knockout mice, we found that Ksp-cadherin is in fact not required for kidney development despite its high and specific expression along the nephron. However, its absence leads to a developmental delay in maximal urinary concentrating ability.
Collapse
Affiliation(s)
- R B Thomson
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - D W Dynia
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - S Burlein
- Department of Nephrology and Hypertension, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - B R Thomson
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - C J Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - F Knauf
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - T Wang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - P S Aronson
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
30
|
Mossoba ME, Mapa MST, Sprando J, Araujo M, Sprando RL. Evaluation of transporter expression in HK-2 cells after exposure to free and ester-bound 3-MCPD. Toxicol Rep 2021; 8:436-442. [PMID: 33717996 PMCID: PMC7932896 DOI: 10.1016/j.toxrep.2021.02.017] [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: 03/23/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/22/2022] Open
Abstract
3-Monochloropropane-1,2-diol (3-MCPD) and its fatty acid esters have the potential to induce nephrotoxicity. We used an in vitro cellular model of human proximal tubule cells to test the effects of 3-MCPD compound exposures on transporter gene expression. 3-MCPD-related nephrotoxicity could be associated with indirect modes of action relating to aquaporin homeostasis.
3-Monochloropropane-1,2-diol (3-MCPD) is a food processing contaminant in some infant formula products and other foods in the United States. Although rodent studies have demonstrated that 3-MCPD and its palmitic esters have the potential to induce nephrotoxicity, our recent human cell culture studies using the human renal proximal tubule cell line HK-2 have not strongly supported this finding. Considering this disparity, we sought to examine whether changes in transporter gene expression on proximal tubule cells could be modulated by these compounds and allow us to glean mechanistic information on a possible indirect path to proximal tubule injury in vivo. If fundamental processes like water and solute transport could be disrupted by 3-MCPD compounds, then a new avenue of toxicity could be further explored in both infant and adult models. In our current study, we used HK-2 cells as an in vitro cellular model of human proximal tubule cells to investigate the effects of low (10 μM) and high (100 μM) 3-MCPD compound exposures to these cells for 24 hours (h) on the expression of 20 transporter genes that are known to be relevant to proximal tubules. Although we detected consistent upregulation of AQP1 expression at the RNA transcript level following HK-2 treatment with both low and high doses of several ester-bound 3-MCPD compounds, these increases were not associated with statistically significant elevations in their protein expression levels. Moreover, we observed a lack of modulation of other members of the AQP protein family that are known to be expressed by human proximal tubule cells. Overall, our study suggests the possibility that 3-MCPD-related nephrotoxicity could be associated with indirect modes of action relating to aquaporin homeostasis, but additional studies with other human-derived models would be pertinent to further explore these findings and to better understand transporter expression differences under different stages of proximal tubule development.
Collapse
Key Words
- 1-Li, 1-Linoleoyl-3-chloropropanediol
- 1-Ol, 1-Oleoyl-3-chloropropanediol
- 1-Pa, 1-Palmitoyl-3-chloropropanediol
- 3-MCPD, 3-Monochloropropane-1,2-diol
- 3-Monochloropropane-1,2-diol
- HK-2
- HK-2, Human Kidney-2
- Kidney
- Li, Linoleic Acid
- Li-Li, 1,2-Di-linoleoyl-3-chloropropanediol
- Ol, Oleic Acid
- Ol-Li, 1-Oleoyl-2-linoleoyl-3-chloropropanediol
- Ol-Ol, 1,2-Di-oleoyl-3-chloropropanediol
- PMA, Phenylmercuric Acetate
- Pa, Palmitic Acid
- Pa-Li, 1-Palmitoyl-2-linoleoyl-3-chloropropanediol
- Pa-Ol, 1-Palmitoyl-2-oleoyl-3-chloropropanediol
- Pa-Pa, 1,2-Di-palmitoyl-3-chloropropanediol
- VAL, Valproic Acid
Collapse
Affiliation(s)
- Miriam E Mossoba
- U.S. Food and Drug Administration (U.S. FDA), Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), Division of Toxicology (DT), Laurel, MD, 20817, United States
| | - Mapa S T Mapa
- U.S. Food and Drug Administration (U.S. FDA), Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), Division of Toxicology (DT), Laurel, MD, 20817, United States
| | - Jessica Sprando
- Virginia-Maryland College of Veterinary Medicine, 205 Duck Pond Road, Blacksburg, VA, 24061, United States
| | - Magali Araujo
- U.S. Food and Drug Administration (U.S. FDA), Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), Division of Toxicology (DT), Laurel, MD, 20817, United States
| | - Robert L Sprando
- U.S. Food and Drug Administration (U.S. FDA), Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), Division of Toxicology (DT), Laurel, MD, 20817, United States
| |
Collapse
|
31
|
Liu L, Gu M, Liu J, Liu Q, Xu X, Fan R, Peng F, Jiang Y. The Effect on the Kidney in Patients With Anti-N-methyl D-aspartate Receptor Antibody Encephalitis. Front Neurol 2021; 12:601495. [PMID: 33643189 PMCID: PMC7907499 DOI: 10.3389/fneur.2021.601495] [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: 09/01/2020] [Accepted: 01/15/2021] [Indexed: 11/27/2022] Open
Abstract
Objectives: The function of the N-methyl-D-aspartate receptor (NMDAR) in the kidney has been studied. However, the effect on the kidney from anti-NAMDAR antibody encephalitis has not been investigated thus far. Methods: Case data were collected from 82 patients with anti-NMDAR antibody encephalitis and 166 age- and sex-matched healthy controls (HCs). Clinical characteristics, urinalysis [including urine pH and urine specific gravity (SG)], serum creatinine (Scr), and estimated glomerular filtration rate (eGFR) based on Cr levels were evaluated. Results: At initial admission, urine pH levels and urine SG levels in anti-NMDAR antibody encephalitis patients were significantly higher and lower, respectively, than HCs (both p < 0.001). There were no significant differences in Scr and eGFR between anti-NMDAR antibody encephalitis patients and HCs. Urine pH levels in patients with anti-NMDAR antibody <1:32 were significantly lower than those in patients with anti-NMDAR antibody ≥1:32 (p = 0.029). Urine pH levels were significantly lower (p = 0.004) and urine SG levels were significantly higher (p = 0.027) in a follow-up evaluation 3 months after treatment. Conclusions: The changes in urinalysis occur in patients with anti-NMDAR antibody encephalitis. The pathophysiological changes in anti-NMDAR antibody encephalitis were not limited to the CNS.
Collapse
Affiliation(s)
- Lizhi Liu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Neurology, Huizhou Hospital of Traditional Chinese Medicine, Huizhou, China
| | - Meifeng Gu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jia Liu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Department of Neurology, Huizhou Hospital of Traditional Chinese Medicine, Huizhou, China
| | - Xiaofeng Xu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Fan
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Jiang
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Chen ZG, Huang J, Fan R, Weng RH, Shinohara RT, Landis JR, Chen Y, Jiang Y. Urinalysis in patients with neuromyelitis optica spectrum disorder. Eur J Neurol 2019; 27:619-625. [PMID: 31729806 DOI: 10.1111/ene.14128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/09/2019] [Accepted: 11/04/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE Increasing evidence has demonstrated that aquaporin-4 (AQP4) immunoglobulin G causes damage to the kidney in neuromyelitis optica spectrum disorder (NMOSD). However, changes in urinalysis in NMOSD have not been investigated thus far. Our objective was to evaluate the changes in urinalysis in NMOSD patients. METHODS Case data were collected from 44 patients with AQP4 antibody-positive NMOSD, 53 patients with multiple sclerosis (MS) and 79 age- and sex-matched healthy controls. Analyses of early morning urine and 24-h urine samples comparing NMOSD with MS patients were conducted. RESULTS In the acute phase, urine pH levels (P < 0.001) and urine specific gravity levels (P < 0.001) from NMOSD patients were significantly higher and lower, respectively, than for MS patients. 24-h urine sodium and 24-h urine volume from NMOSD patients were significantly higher than for MS patients (both P = 0.001). A 24-h urine volume higher than 2500 ml (odds ratio 11.7, 95% confidence interval 1.863-73.066) and a 24-h urine sodium higher than 200 mmol (odds ratio 16.0, 95% confidence interval 2.122-120.648) are more likely to occur in NMOSD patients in the acute phase than in MS patients. CONCLUSIONS The urinalysis results were significantly different between NMOSD patients and MS patients. The pathophysiological changes in AQP4 antibody-positive NMOSD patients were not limited to the central nervous system.
Collapse
Affiliation(s)
- Z G Chen
- Department of Neurology and Multiple Sclerosis Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Neurology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - J Huang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - R Fan
- Department of Neurology and Multiple Sclerosis Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - R H Weng
- Department of Neurology and Multiple Sclerosis Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Neurology, Second People's Hospital of Foshan, Foshan, China
| | - R T Shinohara
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - J R Landis
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Jiang
- Department of Neurology and Multiple Sclerosis Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Protein Abundance of Clinically Relevant Drug Transporters in The Human Kidneys. Int J Mol Sci 2019; 20:ijms20215303. [PMID: 31653114 PMCID: PMC6862022 DOI: 10.3390/ijms20215303] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Renal drug transporters such as the organic cation transporters (OCTs), organic anion transporters (OATs) and multidrug resistance proteins (MRPs) play an important role in the tubular secretion of many drugs influencing their efficacy and safety. However, only little is known about the distinct protein abundance of these transporters in human kidneys, and about the impact of age and gender as potential factors of inter-subject variability in their expression and function. The aim of this study was to determine the protein abundance of MDR1, MRP1-4, BCRP, OAT1-3, OCT2-3, MATE1, PEPT1/2, and ORCTL2 by liquid chromatography-tandem mass spectrometry-based targeted proteomics in a set of 36 human cortex kidney samples (20 males, 16 females; median age 53 and 55 years, respectively). OAT1 and 3, OCT2 and ORCTL2 were found to be most abundant renal SLC transporters while MDR1, MRP1 and MRP4 were the dominating ABC transporters. Only the expression levels of MDR1 and ORCTL2 were significantly higher abundant in older donors. Moreover, we found several significant correlations between different transporters, which may indicate their functional interplay in renal vectorial transport processes. Our data may contribute to a better understanding of the molecular processes determining renal excretion of drugs.
Collapse
|
34
|
Sakamoto M, Tatsuta N, Chan HM, Domingo JL, Murata K, Nakai K. Brain methylmercury uptake in fetal, neonate, weanling, and adult rats. ENVIRONMENTAL RESEARCH 2018; 167:15-20. [PMID: 30005196 DOI: 10.1016/j.envres.2018.06.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 06/08/2023]
Abstract
Fetuses and neonates are known to be highly susceptible to methylmercury (MeHg) toxicity, but little is known about the relative uptake of MeHg from blood to the developing brain. We measured time-course changes in mercury (Hg) concentrations in the brain of fetal, neonate, weanling, and adult rats after an injection of 0.08 μg (0.4 nmol) Hg/g MeHg. In the prenatal experiment, MeHg was subcutaneously injected to pregnant dams on embryonic days 17, 18, 18.5, 19, 19.5, or 20, and Hg concentrations in tissues were measured in both mothers and fetuses on embryonic day 21 (1 day before parturition). Brain Hg levels in fetuses peaked 2 days after injection and were approximately 1.5 times higher than in mothers. In the postnatal experiment, the same MeHg dose was injected subcutaneously to male rats on postnatal days 1 (neonates), 35 (weanlings), or 56 (adults). Mercury concentrations in tissues were measured 1, 2, 3, 4, 5, or 6 days after the injection. Brain Hg levels peaked most rapidly in neonates, and were approximately 1.5 times higher than levels in weanlings or adults. Throughout the examined period, peak Hg levels in the brain and the Hg brain/blood ratio 24 h after injection were highest in fetuses, followed by the levels in neonates, and decreased with life stage. These findings suggest that relatively higher brain MeHg uptake is an important factor in the vulnerability of fetuses and neonates to MeHg exposure.
Collapse
Affiliation(s)
- Mineshi Sakamoto
- Department of Environment and Public Health, National Institute for Minamata Disease, 4058-18 Hama, Kumamoto 867-0008, Japan; Development and Environmental Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Nozomi Tatsuta
- Development and Environmental Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hing Man Chan
- Center for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - José L Domingo
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Universitat "Rovira I Virgili", Reus, Spain
| | - Katsuyuki Murata
- Department of Environmental Health Sciences, Akita University School of Medicine, Akita, Japan
| | - Kunihiko Nakai
- Development and Environmental Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Antenatal Bartter's syndrome (aBS) is the most severe form of Bartter's syndrome, requiring close follow-up, in particular during the neonatal period, primarily because of prematurity. The recent identification of a novel and very severe form of aBS merits an update on this topic. RECENT FINDING Despite the identification of several genes involved in Bartter's syndrome, about 20% of patients clinically diagnosed with aBS remained without genetic explanation for decades. We recently identified mutations in MAGED2 as a cause of an X-linked form of aBS characterized by a very early onset of severe polyhydramnios and extreme prematurity leading to high mortality. Remarkably, all symptoms in surviving patients with MAGE-D2 mutations resolve spontaneously, within weeks after preterm birth. Interestingly, MAGE-D2 affects the expression of the sodium chloride cotransporters NKCC2 and NCC, explaining thereby the severity of the disease. Importantly, a more recent analysis of MAGED2 in a large French cohort of patients with aBS confirmed our data and showed that females can also be affected. SUMMARY MAGE-D2 is critical for renal salt reabsorption in the fetus, amniotic fluid volume regulation, and maintenance of pregnancy. Most importantly, MAGED2 must be included in the genetic screening of every form of aBS.
Collapse
Affiliation(s)
- Martin Kömhoff
- University Children's Hospital, Philipps-University, Marburg, Germany
| | - Kamel Laghmani
- INSERM/UPMC/CNRS-U1138, ERL8228, Team 3, Paris Cedex 06, France
| |
Collapse
|
36
|
Abstract
Kidneys are physiologically hypoxic due to huge oxygen consumption for tubular reabsorption. The physiological hypoxia makes the kidney an appropriate organ for sensitively detecting oxygen levels and producing erythropoietin (EPO). In preterm neonates, immature kidneys cannot produce sufficient EPO, which results in anemia of prematurity (AOP). The cause of EPO insufficiency in AOP has been unclear, therefore current therapeutic options are transfusion and injection of recombinant human EPO. This report shows that the cause of insufficient EPO production in AOP is elevated renal oxygen levels due to poor oxygen consumption by immature tubules. Neonatal mice with AOP showed low tubular transporter expression and elevated renal oxygen levels compared with those without AOP. Enhancing transporter expression in AOP mice induced renal hypoxia and EPO production. In preterm neonates, red blood cell counts, hemoglobin levels, and hematocrit levels correlated with tubular function, but not with serum creatinine, gestational age, or birth weight. Furthermore, pharmacological upregulation of hypoxia signaling ameliorated AOP in mice. These data suggest that tubular maturation with increased oxygen consumption is required for renal EPO production.
Collapse
|
37
|
Hardy A, Benford D, Halldorsson T, Jeger MJ, Knutsen HK, More S, Naegeli H, Noteborn H, Ockleford C, Ricci A, Rychen G, Schlatter JR, Silano V, Solecki R, Turck D, Bresson JL, Dusemund B, Gundert-Remy U, Kersting M, Lambré C, Penninks A, Tritscher A, Waalkens-Berendsen I, Woutersen R, Arcella D, Court Marques D, Dorne JL, Kass GE, Mortensen A. Guidance on the risk assessment of substances present in food intended for infants below 16 weeks of age. EFSA J 2017; 15:e04849. [PMID: 32625502 PMCID: PMC7010120 DOI: 10.2903/j.efsa.2017.4849] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Following a request from the European Commission to EFSA, the EFSA Scientific Committee (SC) prepared a guidance for the risk assessment of substances present in food intended for infants below 16 weeks of age. In its approach to develop this guidance, the EFSA SC took into account, among others, (i) an exposure assessment based on infant formula as the only source of nutrition; (ii) knowledge of organ development in human infants, including the development of the gut, metabolic and excretory capacities, the brain and brain barriers, the immune system, the endocrine and reproductive systems; (iii) the overall toxicological profile of the substance identified through the standard toxicological tests, including critical effects; (iv) the relevance for the human infant of the neonatal experimental animal models used. The EFSA SC notes that during the period from birth up to 16 weeks, infants are expected to be exclusively fed on breast milk and/or infant formula. The EFSA SC views this period as the time where health-based guidance values for the general population do not apply without further considerations. High infant formula consumption per body weight is derived from 95th percentile consumption. The first weeks of life is the time of the highest relative consumption on a body weight basis. Therefore, when performing an exposure assessment, the EFSA SC proposes to use the high consumption value of 260 mL/kg bw per day. A decision tree approach is proposed that enables a risk assessment of substances present in food intended for infants below 16 weeks of age. The additional information needed when testing substances present in food for infants below 16 weeks of age and the approach to be taken for the risk assessment are on a case-by-case basis, depending on whether the substance is added intentionally to food and is systemically available.
Collapse
|
38
|
Segar JL. Renal adaptive changes and sodium handling in the fetal-to-newborn transition. Semin Fetal Neonatal Med 2017; 22:76-82. [PMID: 27881286 DOI: 10.1016/j.siny.2016.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Appropriate fluid and electrolyte management is critical for optimal care of very low birth weight or sick infants. Delivery of such care requires an understanding of developmental changes in renal water and salt handling that occur with advancing gestational age as well as postnatal age. This review focuses on the principles of sodium homeostasis during fetal and postnatal life. The physiology of renal tubular transport mechanisms, as well as neurohumoral factors impacting renal tubular transport are highlighted. Clinical implications and guidelines to the provision of sodium to this vulnerable population are also discussed.
Collapse
Affiliation(s)
- Jeffrey L Segar
- Department of Pediatrics, University of Iowa Carver College of Medicine, University of Iowa Children's Hospital, Iowa City, IA, USA.
| |
Collapse
|
39
|
Tang L, Zheng S, Ren H, He D, Zeng C, Wang WE. Activation of angiotensin II type 1 receptors increases D 4 dopamine receptor expression in rat renal proximal tubule cells. Hypertens Res 2017; 40:652-657. [PMID: 28230199 DOI: 10.1038/hr.2017.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/22/2022]
Abstract
Both the dopaminergic and renin-angiotensin systems play important roles in the regulation of blood pressure. Our previous study showed that the stimulation of dopaminergic D4 receptors reduced angiotensin II type 1 (AT1) receptor expression in renal proximal tubule (RPT) cells. In this study, we tested whether AT1 receptors, in return, would regulate D4 receptor expression and function in RPT cells. Expression of the D4 receptor from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) RPT cells and renal cortex tissues were determined by western blot, and Na+-K+ ATPase activity was determined using an enzyme assay. Urine volume and urine sodium of WKY rats and SHRs treated with or without D4 receptor stimulation were measured. Thus, activation of AT1 receptors with angiotensin II (Ang II) increased D4 receptor protein expression in RPT cells, and this increase was blocked by nicardipine, a calcium influx blocker. The D4 receptor agonist PD168077 inhibited Na+-K+ ATPase activity in WKY RPT cells but not in SHR RPT cells. Ang II pre-treatment promoted D4 receptor-mediated inhibition of Na+-K+ ATPase in RPT cells in WKY rats but not in SHRs. Meanwhile, Ang II pre-treatment augmented the natriuretic effect of PD168077 in WKY rats but not in SHRs. In conclusion, AT1 stimulation can regulate the expression and natriuretic function of dopaminergic D4 receptors in RPT cells and might be involved in the pathogenesis of essential hypertension.
Collapse
Affiliation(s)
- Luxun Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Duofen He
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Wei Eric Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
40
|
Bischoff AR, Dornelles AD, Carvalho CG. Treatment of Hypernatremia in Breastfeeding Neonates: A Systematic Review. Biomed Hub 2017; 2:1-10. [PMID: 31988896 PMCID: PMC6945909 DOI: 10.1159/000454980] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/06/2016] [Indexed: 11/23/2022] Open
Abstract
Background/Aims Hypernatremic dehydration in term neonates is associated with inadequate fluid intake, usually related to insufficient lactation. The use of hypotonic fluids is appropriate to dilute serum sodium (SNa), but cerebral edema may develop when it happens abruptly. Our objective was to clarify how to correct hypernatremic dehydration properly. Methods The following databases were searched, limited to studies published until January 31st, 2016: Clinical Trials, MEDLINE/PubMed, EMBASE, LILACS, and the Cochrane Library. We included open-label trials, nonrandomized controlled trials, or prospective and retrospective case series evaluating relevant outcomes. Information regarding the way of administering the treatment, type of fluid used, rates of complications and outcomes, as well as the rate of SNa reduction were collected. Results Searches yielded 771 articles: 64 had the full text reviewed and 9 were included. No randomized clinical trials or systematic reviews focusing on treatment of hypernatremic dehydration and its outcomes were found. We found a scarcity of high quality studies and great methodology heterogeneity. Conclusions More severe hypernatremia is at greater risk of causing severe adverse effects of treatment. There is no consensus about the optimal rate of SNa drop in this population, but a slower correction appears to be safer. Questions as when parenteral fluids are indicated remain unanswered.
Collapse
Affiliation(s)
- Adrianne Rahde Bischoff
- Departmental Clinical Fellowship, Division of Neonatology, Department of Pediatrics, University of Toronto and the Hospital for Sick Children, Toronto, ON, Canada
| | | | - Clarissa Gutierrez Carvalho
- Departamento de Pediatria, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Pediatria, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
41
|
Abstract
It is widely accepted that sodium is an essential nutritional electrolyte and its deficiency is associated with neurological sequelae and poor growth. The provision of an adequate sodium intake to preterm neonates is hampered by the technical difficulty in clinically assessing total body sodium content. As addressed in this review, there is a lack of consensus on the definition of hyponatremia early in life, but there is no evidence that it should deviate from the widely accepted normative data for adult subjects. A low urinary sodium content is accepted by many as reflecting total body sodium deficiency, yet spot urinary sodium measurements are of questionable clinical value. The hormonal regulation of sodium homeostasis is here reviewed and the mechanism accounting for sodium deficiency-induced growth impairment in preterm infants addressed. Lastly, we provide evidence-based gestational and postnatal age-dependent recommendations for the provision of adequate sodium intake to preterm neonates.
Collapse
|
42
|
Na(+), K(+), Cl(-), acid-base or H2O homeostasis in children with urinary tract infections: a narrative review. Pediatr Nephrol 2016; 31:1403-9. [PMID: 26701834 DOI: 10.1007/s00467-015-3273-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
Guidelines on the diagnosis and management of urinary tract infections in childhood do not address the issue of abnormalities in Na(+), K(+), Cl(-) and acid-base balance. We have conducted a narrative review of the literature with the aim to describe the underlying mechanisms of these abnormalities and to suggest therapeutic maneuvers. Abnormalities in Na(+), K(+), Cl(-) and acid-base balance are common in newborns and infants and uncommon in children of more than 3 years of age. Such abnormalities may result from factitious laboratory results, from signs and symptoms (such as excessive sweating, poor fluid intake, vomiting and passage of loose stools) of the infection itself, from a renal dysfunction, from improper parenteral fluid management or from the prescribed antimicrobials. In addition, two transient renal tubular dysfunctions may occur in infants with infectious renal parenchymal involvement: a reduced capacity to concentrate urine and pseudohypoaldosteronism secondary to renal tubular unresponsiveness to aldosterone that presents with hyponatremia, hyperkalemia and acidosis. In addition to antimicrobials, volume resuscitation with an isotonic solution is required in these children. In secondary pseudohypoaldosteronism, isotonic solutions (such as 0.9 % saline or lactated Ringer) correct not only the volume depletion but also the hyperkalemia and acidosis. In conclusion, our review suggests that in infants with infectious renal parenchymal involvement, non-renal and renal causes concur to cause fluid volume depletion and abnormalities in electrolyte and acid-base balance, most frequently hyponatremia.
Collapse
|
43
|
Lava SAG, Bianchetti MG, Simonetti GD. Salt intake in children and its consequences on blood pressure. Pediatr Nephrol 2015; 30:1389-96. [PMID: 25127918 DOI: 10.1007/s00467-014-2931-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 02/07/2023]
Abstract
Sodium is the most abundant extracellular cation and therefore pivotal in determining fluid balance. At the beginning of life, a positive sodium balance is needed to grow. Newborns and preterm infants tend to lose sodium via their kidneys and therefore need adequate sodium intake. Among older children and adults, however, excessive salt intake leads to volume expansion and arterial hypertension. Children who are overweight, born preterm, or small for gestational age and African American children are at increased risk of developing high blood pressure due to a high salt intake because they are more likely to be salt sensitive. In the developed world, salt intake is generally above the recommended intake also among children. Although a positive sodium balance is needed for growth during the first year of life, in older children, a sodium-poor diet seems to have the same cardiovascular protective effects as among adults. This is relevant, since: (1) a blood pressure tracking phenomenon was recognized; (2) the development of taste preferences is important during childhood; and (3) salt intake is often associated with the consumption of sugar-sweetened beverages (predisposing children to weight gain).
Collapse
Affiliation(s)
- Sebastiano A G Lava
- Division of Pediatric Nephrology, University Children's Hospital Bern, Bern, Switzerland
| | | | | |
Collapse
|
44
|
Shabala S, Bose J, Hedrich R. Salt bladders: do they matter? TRENDS IN PLANT SCIENCE 2014; 19:687-91. [PMID: 25361704 DOI: 10.1016/j.tplants.2014.09.001] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/27/2014] [Accepted: 09/02/2014] [Indexed: 05/02/2023]
Abstract
Soil salinity is claiming about three hectares of arable land from conventional crop farming every minute. At the same time, the challenge of feeding 9.3 billion people by 2050 is forcing agricultural production into marginal areas, and providing sufficient food for this growing population cannot be achieved without a major breakthrough in crop breeding for salinity tolerance. In this Opinion article, we argue that the current trend of targeting Na(+) exclusion mechanisms in breeding programmes for salinity tolerance in crops needs revising. We propose that progress in this area will be achieved by learning from halophytes, naturally salt-loving plants capable of surviving in harsh saline environments, by targeting the mechanisms conferring Na(+) sequestration in external storage organs.
Collapse
Affiliation(s)
- Sergey Shabala
- School of Land and Food, University of Tasmania, Hobart, Australia.
| | - Jayakumar Bose
- School of Land and Food, University of Tasmania, Hobart, Australia
| | - Rainer Hedrich
- Institute for Molecular Plant Physiology and Biophysics, Würzburg, Germany.
| |
Collapse
|
45
|
Soleimani M. The multiple roles of pendrin in the kidney. Nephrol Dial Transplant 2014; 30:1257-66. [PMID: 25281699 DOI: 10.1093/ndt/gfu307] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/25/2014] [Indexed: 12/30/2022] Open
Abstract
The [Formula: see text] exchanger pendrin (SLC26A4, PDS) is located on the apical membrane of B-intercalated cells in the kidney cortical collecting duct and the connecting tubules and mediates the secretion of bicarbonate and the reabsorption of chloride. Given its dual function of bicarbonate secretion and chloride reabsorption in the distal tubules, it was thought that pendrin plays important roles in systemic acid-base balance and electrolyte and vascular volume homeostasis under basal conditions. Mice with the genetic deletion of pendrin or humans with inactivating mutations in PDS gene, however, do not display excessive salt and fluid wasting or altered blood pressure under baseline conditions. Very recent reports have unmasked the basis of incongruity between the mild phenotype in mutant mice and the role of pendrin as an important player in salt reabsorption in the distal tubule. These studies demonstrate that pendrin and the Na-Cl cotransporter (NCC; SLC12A3) cross compensate for the loss of each other, therefore masking the role that each transporter plays in salt reabsorption under baseline conditions. In addition, pendrin regulates calcium reabsorption in the distal tubules. Furthermore, combined deletion of pendrin and NCC not only causes severe volume depletion but also results in profound calcium wasting and luminal calcification in medullary collecting ducts. Based on studies in pathophysiological states and the examination of genetically engineered mouse models, the evolving picture points to important roles for pendrin (SLC26A4) in kidney physiology and in disease states. This review summarizes recent advances in the characterization of pendrin and the multiple roles it plays in the kidney, with emphasis on its essential roles in several diverse physiological processes, including chloride homeostasis, vascular volume and blood pressure regulation, calcium excretion and kidney stone formation.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, OH, USA Research Services, Veterans Affairs Medical Center, Cincinnati, OH, USA Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|