Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pediatr Nephrol ; 36(12): 3933-3944, 2021 12.
Article in English | MEDLINE | ID: mdl-34125285

ABSTRACT

BACKGROUND: Children living with HIV frequently show high plasma levels of fibroblast growth factor-2 (FGF-2/bFGF). FGF-2 accelerates the progression of several experimental kidney diseases; however, the role of circulating FGF-2 in childhood HIV-chronic kidney diseases (HIV-CKDs) is unknown. We carried out this study to determine whether high plasma FGF-2 levels were associated with the development of HIV-CKDs in children. METHODS: The plasma and urine FGF-2 levels were measured in 84 children (< 12 years of age) living with HIV during the pre-modern antiretroviral era, and followed for at least 3 years to determine the prevalence of proteinuria and HIV-CKDs. We also assessed the distribution of the kidney FGF-2 binding sites by autoradiography and Alcian blue staining, and explored potential mechanisms by which circulating FGF-2 may precipitate HIV-CKDs in cultured kidney epithelial and mononuclear cells derived from children with HIV-CKDs. RESULTS: High plasma FGF-2 levels were associated with a high viral load. Thirteen children (~ 15%) developed HIV-CKDs and showed a large reservoir of FGF-2 low-affinity binding sites in the kidney, which can facilitate the recruitment of circulating FGF-2. Children with high plasma and urine FGF-2 levels had 73-fold increased odds (95% CI 9-791) of having HIV-CKDs relative to those with normal FGF-2 values. FGF-2 induced the proliferation and decreased the expression of APOL-1 mRNA in podocytes, and increased the attachment and survival of infected mononuclear cells cultured from children with HIV-CKDs. CONCLUSIONS: High plasma FGF-2 levels appear to be an additional risk factor for developing progressive childhood HIV-CKDs.


Subject(s)
Disease Progression , Fibroblast Growth Factor 2/blood , HIV Infections , Renal Insufficiency, Chronic , Child , HIV Infections/diagnosis , Humans , Kidney , Renal Insufficiency, Chronic/diagnosis
2.
Pediatr Nephrol ; 36(8): 2189-2201, 2021 08.
Article in English | MEDLINE | ID: mdl-33044676

ABSTRACT

HIV-associated nephropathy (HIVAN) predominantly affects people of African ancestry living with HIV who do not receive appropriate antiretroviral therapy (ART). Childhood HIVAN is characterized by heavy proteinuria and decreased kidney function. Kidney histology shows mesangial expansion, classic or collapsing glomerulosclerosis, and microcystic renal tubular dilatation leading to kidney enlargement. The pathogenesis of HIVAN involves the kidney recruitment of inflammatory cells and the infection of kidney epithelial cells. In addition, both viral and genetic factors play key roles in this disease. Modern ART has improved the outcome and decreased the prevalence of childhood HIVAN. However, physicians have had modest success providing chronic ART to children and adolescents, and we continue to see children with HIVAN all over the world. This article discusses the progress made during the last decade in our understanding of the pathogenesis and treatment of childhood HIVAN, placing particular emphasis on the mechanisms that mediate the infection of kidney epithelial cells, and the roles of cytokines, the HIV-Tat gene, and the Apolipoprotein-1 (APOL1) gene risk variants in this disease. In view of the large number of children living with HIV at risk of developing HIVAN, better prevention and treatment programs are needed to eradicate this disease.


Subject(s)
AIDS-Associated Nephropathy , HIV Infections , HIV-1 , AIDS-Associated Nephropathy/diagnosis , AIDS-Associated Nephropathy/epidemiology , AIDS-Associated Nephropathy/genetics , Adolescent , Apolipoprotein L1 , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/epidemiology , Humans , Kidney
3.
Hum Mol Genet ; 26(4): 768-780, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28164240

ABSTRACT

Many genetic mutations have been identified as monogenic causes of nephrotic syndrome (NS), but important knowledge gaps exist in the roles of these genes in kidney cell biology and renal diseases. More animal models are needed to assess the functions of these genes in vivo, and to determine how they cause NS in a timely manner. Drosophila nephrocytes and human podocytes share striking similarities, but to what degree these known NS genes play conserved roles in nephrocytes remains unknown. Here we systematically studied 40 genes associated with NS, including 7 that have not previously been analysed for renal function in an animal model. We found that 85% of these genes are required for nephrocyte functions, suggesting that a majority of human genes known to be associated with NS play conserved roles in renal function from flies to humans. To investigate functional conservation in more detail, we focused on Cindr, the fly homolog of the human NS gene CD2AP. Silencing Cindr in nephrocytes led to dramatic nephrocyte functional impairment and shortened life span, as well as collapse of nephrocyte lacunar channels and effacement of nephrocyte slit diaphragms. These phenotypes could be rescued by expression of a wild-type human CD2AP gene, but not a mutant allele derived from a patient with CD2AP-associated NS. We conclude that the Drosophila nephrocyte can be used to elucidate clinically relevant molecular mechanisms underlying the pathogenesis of most monogenic forms of NS, and to efficiently generate personalized in vivo models of genetic renal diseases bearing patient-specific mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Kidney/physiopathology , Microfilament Proteins/genetics , Nephrotic Syndrome/genetics , Animals , Disease Models, Animal , Drosophila melanogaster/genetics , Gene Silencing , Humans , Mutation , Nephrotic Syndrome/physiopathology , Phenotype , Podocytes/metabolism , Podocytes/pathology
5.
Pediatr Res ; 85(3): 329-338, 2019 02.
Article in English | MEDLINE | ID: mdl-30643188

ABSTRACT

BACKGROUND: Neonates with serum creatinine (SCr) rise ≥0.3 mg/dL and/or ≥50% SCr rise are more likely to die, even when controlling for confounders. These thresholds have not been tested in newborns. We hypothesized that different gestational age (GA) groups require different SCr thresholds. METHODS: Neonates in Assessment of Worldwide Acute Kidney Epidemiology in Neonates (AWAKEN) with ≥1 SCr on postnatal days 1-2 and ≥1 SCr on postnatal days 3-8 were assessed. We compared the mortality predictability of SCr absolute (≥0.3 mg/dL) vs percent (≥50%) rise. Next, we determine usefulness of combining absolute with percent rise. Finally, we determined the optimal absolute, percent, and maximum SCr thresholds that provide the highest mortality area under curve (AUC) and specificity for different GA groups. RESULTS: The ≥0.3 mg/dL rise outperformed ≥50% SCr rise. Addition of percent rise did not improve mortality predictability. The optimal SCr thresholds to predict AUC and specificity were ≥0.3 and ≥0.6 mg/dL for ≤29 weeks GA, and ≥0.1 and ≥0.3 mg/dL for >29 week GA. The maximum SCr value provides great specificity. CONCLUSION: Unique SCr rise cutoffs for different GA improves outcome prediction. Percent SCr rise does not add value to the neonatal AKI definition.


Subject(s)
Acute Kidney Injury/diagnosis , Acute Kidney Injury/epidemiology , Creatinine/blood , Acute Kidney Injury/blood , Biomarkers/blood , Female , Gestational Age , Hospital Mortality , Humans , Infant, Newborn , Intensive Care, Neonatal , Male , Predictive Value of Tests , Prognosis , Registries , Reproducibility of Results , Retrospective Studies , Sample Size
7.
J Am Soc Nephrol ; 28(9): 2607-2617, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28428331

ABSTRACT

Clinical studies have identified patients with nephrotic syndrome caused by mutations in genes involved in the biosynthesis of coenzyme Q10 (CoQ10), a lipid component of the mitochondrial electron transport chain and an important antioxidant. However, the cellular mechanisms through which these mutations induce podocyte injury remain obscure. Here, we exploited the striking similarities between Drosophila nephrocytes and human podocytes to develop a Drosophila model of these renal diseases, and performed a systematic in vivo analysis assessing the role of CoQ10 pathway genes in renal function. Nephrocyte-specific silencing of Coq2, Coq6, and Coq8, which are genes involved in the CoQ10 pathway that have been associated with genetic nephrotic syndrome in humans, induced dramatic adverse changes in these cells. In particular, silencing of Coq2 led to an abnormal localization of slit diaphragms, collapse of lacunar channels, and more dysmorphic mitochondria. In addition, Coq2-deficient nephrocytes showed elevated levels of autophagy and mitophagy, increased levels of reactive oxygen species, and increased sensitivity to oxidative stress. Dietary supplementation with CoQ10 at least partially rescued these defects. Furthermore, expressing the wild-type human COQ2 gene specifically in nephrocytes rescued the defective protein uptake, but expressing the mutant allele derived from a patient with COQ2 nephropathy did not. We conclude that transgenic Drosophila lines carrying mutations in the CoQ10 pathway genes are clinically relevant models with which to explore the pathogenesis of podocyte injury and could serve as a new platform to test novel therapeutic approaches.


Subject(s)
Alkyl and Aryl Transferases/genetics , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Ubiquinone/analogs & derivatives , Vitamins/pharmacology , Alkyl and Aryl Transferases/deficiency , Alleles , Animals , Autophagy/drug effects , Cell Line , Cells, Cultured , Disease Models, Animal , Gene Silencing , Humans , Mitochondria/ultrastructure , Mitophagy/drug effects , Organisms, Genetically Modified , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Ubiquinone/biosynthesis , Ubiquinone/genetics , Ubiquinone/pharmacology , Vitamins/biosynthesis
8.
J Am Soc Nephrol ; 28(3): 862-875, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27811066

ABSTRACT

Studies have shown that podocytes and renal tubular epithelial cells from patients with HIV-associated nephropathy (HIVAN) express HIV-1 transcripts, suggesting that productive infection of renal epithelial cells precipitates development of HIVAN. However, podocytes and renal tubular epithelial cells do not express CD4 receptors, and it is unclear how these cells become productively infected in vivo We investigated the mechanisms underlying the infection by HIV-1 of podocytes cultured from the urine of children with HIVAN. We observed low-level productive infection on exposure of these cells to primary cell-free HIV-1 supernatants. However, envelope-defective recombinant HIV-1 did not infect the renal epithelial cell lines. Moreover, treatment of podocytes to inhibit endocytic transport or dynamin activity or remove cell surface heparan sulfate proteoglycans reduced infection efficiency. Transfection of CD4- 293T cells with a cDNA expression library developed from a podocyte cell line derived from a child with HIVAN led to the identification of TNF-α as a possible mediator of HIV-1 infection. Overexpression of transmembrane TNF-α in cultured CD4- renal tubular epithelial cells, 293T cells, and HeLa cells enabled the infection of these cells; exposure to soluble TNF-α did not. Immunohistochemistry showed TNF-α expression in podocytes of renal sections from children with HIVAN. Furthermore, we found that TNF-α enhanced NF-κB activation and integration of HIV-1 into the podocyte DNA. Finally, inhibition of dynamin activity blocked TNF-α-mediated infection. These data establish a role for transmembrane TNF-α in facilitating the viral entry and integration of HIV-1 into the DNA of renal epithelial cells.


Subject(s)
AIDS-Associated Nephropathy/virology , HIV-1/physiology , Podocytes/virology , Tumor Necrosis Factor-alpha/physiology , Cells, Cultured , Child , Humans , Membrane Proteins
9.
J Am Soc Nephrol ; 28(4): 1106-1116, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27864430

ABSTRACT

People of African ancestry carrying certain APOL1 mutant alleles are at elevated risk of developing renal diseases. However, the mechanisms underlying APOL1-associated renal diseases are unknown. Because the APOL1 gene is unique to humans and some primates, new animal models are needed to understand the function of APOL1 in vivo We generated transgenic Drosophila fly lines expressing the human APOL1 wild type allele (G0) or the predominant APOL1 risk allele (G1) in different tissues. Ubiquitous expression of APOL1 G0 or G1 in Drosophila induced lethal phenotypes, and G1 was more toxic than was G0. Selective expression of the APOL1 G0 or G1 transgene in nephrocytes, fly cells homologous to mammalian podocytes, induced increased endocytic activity and accumulation of hemolymph proteins, dextran particles, and silver nitrate. As transgenic flies with either allele aged, nephrocyte function declined, cell size increased, and nephrocytes died prematurely. Compared with G0-expressing cells, however, G1-expressing cells showed more dramatic phenotypes, resembling those observed in cultured mammalian podocytes overexpressing APOL1-G1. Expressing the G0 or G1 APOL1 transgene in nephrocytes also impaired the acidification of organelles. We conclude that expression of an APOL1 transgene initially enhances nephrocyte function, causing hypertrophy and subsequent cell death. This new Drosophila model uncovers a novel mechanism by which upregulated expression of APOL1-G1 could precipitate renal disease in humans. Furthermore, this model may facilitate the identification of APOL1-interacting molecules that could serve as new drug targets to treat APOL1-associated renal diseases.


Subject(s)
Apolipoproteins/genetics , Cell Death/physiology , Kidney Diseases/genetics , Kidney/pathology , Lipoproteins, HDL/genetics , Alleles , Animals , Animals, Genetically Modified , Apolipoprotein L1 , Cells, Cultured , Disease Models, Animal , Disease Progression , Drosophila , Gene Expression Regulation , Humans , Hypertrophy/genetics , Kidney Diseases/pathology
10.
Pediatr Nephrol ; 31(7): 1167-78, 2016 07.
Article in English | MEDLINE | ID: mdl-26857710

ABSTRACT

BACKGROUND: Current definitions of acute kidney injury (AKI) are not sufficiently sensitive to identify all newborns with AKI during the first week of life. METHODS: To determine whether the rate of decline of serum creatinine (SCr) during the first week of life can be used to identify newborns with AKI, we reviewed the medical records of 106 term neonates at risk of AKI who were treated with hypothermia for hypoxic ischemic encephalopathy (HIE). RESULTS: Of the newborns enrolled in the study, 69 % showed a normal rate of decline of SCr to ≥50 % and/or reached SCr levels of ≤0.6 mg/dl before the 7th day of life, and therefore had an excellent clinical outcome (control group). Thirteen newborns with HIE (12 %) developed AKI according to an established neonatal definition (AKI-KIDGO group), and an additional 20 newborns (19 %) showed a rate of decline of SCr of <33, <40, and <46 % from birth to days 3, 5, or 7 of life, respectively (delayed rise in estimated SCr clearance group). Compared to the control group, newborns in the other two groups required more days of mechanical ventilation and vasopressor drugs and had higher gentamicin levels, more fluid overload, lower urinary epidermal growth factor levels, and a prolonged length of stay. CONCLUSIONS: The rate of decline of SCr provides a sensitive approach to identify term newborns with AKI during the first week of life.


Subject(s)
Acute Kidney Injury/diagnosis , Creatinine/blood , Hypoxia-Ischemia, Brain/complications , Acute Kidney Injury/etiology , Biomarkers/blood , Female , Humans , Infant, Newborn , Kidney Function Tests/methods , Male , Retrospective Studies
11.
Am J Physiol Heart Circ Physiol ; 309(8): H1314-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26276817

ABSTRACT

Critically ill children can develop bleeding complications when treated with heparin-like drugs. These events are usually attributed to the anticoagulant activity of these drugs. However, previous studies showed that fibroblast growth factor-2 (FGF-2), a heparin-binding growth factor released in the circulation of these patients, could precipitate intestinal hemorrhages in mice treated with the heparin-like drug pentosan polysulfate (PPS). Yet very little is known about how FGF-2 induces bleeding complications in combination with heparin-like drugs. Here, we examined the mechanisms by which circulating FGF-2 induces intestinal hemorrhages in mice treated with PPS. We used a well-characterized mouse model of intestinal hemorrhages induced by FGF-2 plus PPS. Adult FVB/N mice were infected with adenovirus carrying Lac-Z or a secreted form of recombinant human FGF-2, and injected with PPS, at doses that do not induce bleeding complications per se. Mice treated with FGF-2 in combination with PPS developed an intestinal inflammatory reaction that increased the permeability and disrupted the integrity of submucosal intestinal vessels. These changes, together with the anticoagulant activity of PPS, induced lethal hemorrhages. Moreover, a genetically modified form of the endothelial ligand angiopoietin-1 (Ang-1*), which has powerful antipermeability and anti-inflammatory activity, prevented the lethal bleeding complications without correcting the anticoagulant status of these mice. These findings define new mechanisms through which FGF-2 and Ang-1* modulate the outcome of intestinal bleeding complications induced by PPS in mice and may have wider clinical implications for critically ill children treated with heparin-like drugs.


Subject(s)
Angiopoietin-1/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Gastrointestinal Hemorrhage/prevention & control , Genetic Therapy/methods , Intestine, Small/metabolism , Adenoviridae/genetics , Angiopoietin-1/genetics , Animals , Blood Coagulation , Capillary Permeability , Disease Models, Animal , Fibroblast Growth Factor 2/genetics , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/genetics , Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/pathology , Gene Transfer Techniques , Genetic Vectors , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/prevention & control , Intestine, Small/blood supply , Intestine, Small/pathology , Macrophages/metabolism , Male , Matrix Metalloproteinases/metabolism , Mice , Pentosan Sulfuric Polyester
12.
J Am Soc Nephrol ; 25(8): 1800-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24578133

ABSTRACT

Podocyte injury has a critical role in the pathogenesis of HIV-associated nephropathy (HIVAN). The HIV-1 transactivator of transcription (Tat), combined with fibroblast growth factor-2 (FGF-2), can induce the dedifferentiation and proliferation of cultured human podocytes. Cellular internalization of Tat requires interactions with heparan sulfate proteoglycans and cholesterol-enriched lipid rafts (LRs). However, the specific distribution of Tat in human podocytes and its ability to associate with LRs have not been documented. Here, we found that Tat is preferentially recruited to LRs in podocytes isolated from children with HIVAN. Furthermore, we identified arginines in the basic domain (RKKRRQRRR) of Tat as essential for (1) targeting Tat to LRs, (2) Tat-mediated increases in the expression of Rho-A and matrix metalloproteinase-9 in LRs, and (3) Tat-mediated enhancement of FGF-2 signaling in human podocytes and HIV-transgenic mouse kidneys and the exacerbation of renal lesions in these mice. Tat carrying alanine substitutions in the basic domain (AKKAAQAAA) remained localized in the cytosol and did not associate with LRs or enhance FGF-2 signaling in cultured podocytes. These results show the specific association of Tat with LRs in podocytes isolated from children with HIVAN, confirm Tat as a regulator of FGF-2 signaling in LRs, and identify the key domain of Tat responsible for promoting these effects and aggravating renal injury in HIV-transgenic mice. Moreover, these results provide a molecular framework for developing novel therapies to improve the clinical outcome of children with HIVAN.


Subject(s)
AIDS-Associated Nephropathy/metabolism , Fibroblast Growth Factor 2/metabolism , HIV-1 , Membrane Microdomains/physiology , Podocytes/physiology , Signal Transduction/physiology , tat Gene Products, Human Immunodeficiency Virus/physiology , AIDS-Associated Nephropathy/pathology , Animals , Arginine/metabolism , Cell Culture Techniques , Child , Humans , Matrix Metalloproteinase 9/metabolism , Mice, Transgenic , rhoA GTP-Binding Protein/metabolism
13.
Clin Infect Dis ; 59(9): e96-138, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25234519

ABSTRACT

It is important to realize that guidelines cannot always account for individual variation among patients. They are not intended to supplant physician judgment with respect to particular patients or special clinical situations. IDSA considers adherence to these guidelines to be voluntary, with the ultimate determination regarding their application to be made by the physician in the light of each patient's individual circumstances.


Subject(s)
HIV Infections/complications , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Humans , Kidney Transplantation , United States
14.
bioRxiv ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38496548

ABSTRACT

Background: People carrying two APOL1 risk alleles (RA) G1 or G2 are at greater risk of developing HIV-associated nephropathy (HIVAN). Studies in transgenic mice showed that the expression of HIV-1 genes in podocytes, and nef in particular, led to HIVAN. However, it remains unclear whether APOL1-RA and HIV-1 Nef interact to induce podocyte cell death. Method: We generated transgenic (Tg) flies that express APOL1-G1 (derived from a child with HIVAN) and HIV-1 nef specifically in the nephrocytes, the fly equivalent of mammalian podocytes, and assessed their individual and combined effects on the nephrocyte filtration structure and function. Results: We found that HIV-1 Nef acts in synergy with APOL1-G1 resulting in nephrocyte structural and functional defects. Specifically, HIV-1 Nef itself can induce endoplasmic reticulum (ER) stress without affecting autophagy. Furthermore, Nef exacerbates the organelle acidification defects and autophagy reduction induced by APOL1-G1. The synergy between HIV-1 Nef and APOL1-G1 is built on their joint effects on elevating ER stress, triggering nephrocyte dysfunction and ultimately cell death. Conclusions: Using a new Drosophila model of HIV-1-related kidney diseases, we identified ER stress as the converging point for the synergy between HIV-1 Nef and APOL1-G1 in inducing nephrocyte cell death. Given the high relevance between Drosophila nephrocytes and human podocytes, this finding suggests ER stress as a new therapeutic target for HIV-1 and APOL1-associated nephropathies.

15.
Pediatr Res ; 73(2): 194-200, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23174703

ABSTRACT

BACKGROUND: Nephrocalcinosis (NC) is an important clinical problem seen in critically ill preterm neonates treated with loop diuretics. No reliable animal models are available to study the pathogenesis of NC in preterm infants. The purpose of this study was to develop a reproducible and clinically relevant animal model of NC for these patients and to explore the impact of extracellular fluid (ECF) volume contraction induced by sodium and chloride depletion in this process. METHODS: Three-week-old weanling Sprague-Dawley rats were fed diets deficient in either chloride or sodium or both. A subgroup of rats from each dietary group was injected daily with furosemide (40 mg/kg i.p.). RESULTS: Rats fed a control diet, with or without furosemide, or a chloride-depleted diet alone, did not develop NC. By contrast, 50% of the rats injected with furosemide and fed the chloride-depleted diet developed NC. Moreover, 94% of the rats fed the combined sodium- and chloride-depleted diet developed NC, independently of furosemide use. NC was associated with the development of severe ECF volume contraction; hypochloremic, hypokalemic, metabolic alkalosis; increased phosphaturia; and growth retardation. CONCLUSION: Severe ECF volume contraction induced by chronic sodium and chloride depletion appears to play an important role in the pathogenesis of NC.


Subject(s)
Chlorides/metabolism , Kidney/metabolism , Nephrocalcinosis/etiology , Sodium Chloride, Dietary/metabolism , Sodium/deficiency , Water-Electrolyte Balance , Animals , Blood Pressure , Calcium/urine , Disease Models, Animal , Furosemide , Kidney/physiopathology , Male , Nephrocalcinosis/chemically induced , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , Phosphorus/urine , Rats , Rats, Sprague-Dawley , Time Factors , Weight Gain
16.
Pediatr Nephrol ; 28(11): 2179-88, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23783654

ABSTRACT

BACKGROUND: The goal of this study was to assess the value of a urinary biomarker profile comprised of neutrophil gelatinase-associated lipocalin (NGAL), fibroblast growth factor-2 (FGF-2), and epidermal growth factor (EGF), to detect acute kidney injury (AKI) in critically ill neonates. METHODS: We conducted a prospective cohort pilot study of at-risk neonates treated in a level IIIC neonatal intensive care unit (NICU) with therapeutic hypothermia (HT) (n = 25) or extracorporeal membrane oxygenation (ECMO) (n = 10). Urine was collected at baseline, 48 h of illness, and > 24 h post-recovery of their corresponding treatments. Control samples were collected from 27 healthy newborns. The data were expressed as urinary concentrations and values normalized for urinary creatinine. AKI was defined as the presence of oliguria >24 h and/or elevated serum creatinine (SCr), or the failure to improve the estimated creatinine clearance (eCCL) by >50% post-recovery. Non-parametric statistical tests and ROC analyses were used to interpret the data. RESULTS: Fifteen at-risk newborns had AKI. In the first 48 h of illness, the urinary levels of NGAL and FGF-2 had high sensitivity but poor specificity to identify neonates with AKI. At recovery, low urinary EGF levels identified neonates with AKI with a sensitivity of 74% and specificity of 84%. Overall, in the early stages of a critical illness, the urinary levels of NGAL and FGF-2 were sensitive, but not specific, to identify neonates at risk of AKI. Low EGF levels post-recovery identified critically ill neonates with AKI. CONCLUSIONS: These findings require validation in larger prospective studies.


Subject(s)
Acute Kidney Injury/urine , Biomarkers/urine , Acute Kidney Injury/therapy , Acute-Phase Proteins/urine , Blood Urea Nitrogen , Creatinine/blood , Critical Care , Critical Illness , Epidermal Growth Factor/urine , Extracorporeal Membrane Oxygenation , Female , Fibroblast Growth Factor 2/urine , Humans , Hypothermia, Induced , Infant, Newborn , Intensive Care Units, Neonatal , Lipocalin-2 , Lipocalins/urine , Male , Pilot Projects , Prospective Studies , Proto-Oncogene Proteins/urine , ROC Curve , Water-Electrolyte Balance/physiology
17.
Pediatr Nephrol ; 28(11): 2189-98, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23872928

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) increases the morbidity of critically ill children. Thus, it is necessary to identify better renal biomarkers to follow the outcome of these patients. This prospective case-control study explored the clinical value of a urinary biomarker profile comprised of neutrophil gelatinase lipocalin (uNGAL), fibroblast growth factor-2 (uFGF-2), and epidermal growth factor (uEGF) to follow these patients. METHODS: Urine samples were collected from 21 healthy children, and 39 critically ill children (mean age 7.5 years ± 6.97 SD) admitted to a pediatric intensive care unit with sepsis or requiring extra corporeal membrane oxygenation (ECMO). uNGAL, uFGF-2, and uEGF levels were measured using ELISA kits during the first 24 h of admission to PICU, at peak of illness, and upon resolution of the critical illness. RESULTS: On admission, the uNGAL and uFGF-2 levels were increased, and the uEGF levels were decreased, in critically ill children with AKI (n = 19) compared to those without AKI (n = 20), and healthy controls. A biomarker score using the combined cut-off values of uNGAL, uFGF-2, and uEGF (AUC = 0.90) showed the highest specificity to identify children with AKI, relative to each biomarker alone. uNGAL and uFGF-2 on admission showed high sensitivity and specificity to predict mortality (AUC = 0.82). CONCLUSIONS: The biomarker profile comprised of uNGAL, uFGF-2, and uEGF increased the specificity to detect AKI in critically ill children, when compared to each biomarker used alone. uNGAL and uFGF-2 may also predict the risk of death. Further validation of these findings in a large sample size is warranted.


Subject(s)
Acute Kidney Injury/urine , Epidermal Growth Factor/urine , Fibroblast Growth Factor 2/urine , Acute Kidney Injury/mortality , Biomarkers , Case-Control Studies , Child , Child, Preschool , Creatinine/blood , Critical Care , Critical Illness , Extracorporeal Membrane Oxygenation , Female , Gelatinases/blood , Humans , Infant , Length of Stay , Lipocalins/blood , Male , Neutrophils/enzymology , Pilot Projects , Predictive Value of Tests , Prognosis , Prospective Studies , ROC Curve , Sepsis/complications , Sepsis/urine , Survival Analysis
18.
Dis Model Mech ; 16(12)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37969018

ABSTRACT

People of African ancestry who carry the APOL1 risk alleles G1 or G2 are at high risk of developing kidney diseases through not fully understood mechanisms that impair the function of podocytes. It is also not clear whether the APOL1-G1 and APOL1-G2 risk alleles affect these cells through similar mechanisms. Previously, we have developed transgenic Drosophila melanogaster lines expressing either the human APOL1 reference allele (G0) or APOL1-G1 specifically in nephrocytes, the cells homologous to mammalian podocytes. We have found that nephrocytes that expressed the APOL1-G1 risk allele display accelerated cell death, in a manner similar to that of cultured human podocytes and APOL1 transgenic mouse models. Here, to compare how the APOL1-G1 and APOL1-G2 risk alleles affect the structure and function of nephrocytes in vivo, we generated nephrocyte-specific transgenic flies that either expressed the APOL1-G2 or both G1 and G2 (G1G2) risk alleles on the same allele. We found that APOL1-G2- and APOL1-G1G2-expressing nephrocytes developed more severe changes in autophagic pathways, acidification of organelles and the structure of the slit diaphragm, compared to G1-expressing nephrocytes, leading to their premature death. We conclude that both risk alleles affect similar key cell trafficking pathways, leading to reduced autophagy and suggesting new therapeutic targets to prevent APOL1 kidney diseases.


Subject(s)
Drosophila melanogaster , Kidney Diseases , Animals , Mice , Humans , Drosophila melanogaster/metabolism , Apolipoprotein L1/genetics , Apolipoprotein L1/metabolism , Cell Death , Mice, Transgenic , Autophagy/genetics , Mammals/metabolism
19.
Cell Biosci ; 13(1): 199, 2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37925499

ABSTRACT

BACKGROUND: People of Sub-Saharan African ancestry are at higher risk of developing chronic kidney disease (CKD), attributed to the Apolipoprotein L1 (APOL1) gene risk alleles (RA) G1 and G2. The underlying mechanisms by which the APOL1-RA precipitate CKD remain elusive, hindering the development of potential treatments. RESULTS: Using a Drosophila genetic modifier screen, we found that SNARE proteins (Syx7, Ykt6, and Syb) play an important role in preventing APOL1 cytotoxicity. Reducing the expression of these SNARE proteins significantly increased APOL1 cytotoxicity in fly nephrocytes, the equivalent of mammalian podocytes, whereas overexpression of Syx7, Ykt6, or Syb attenuated their toxicity in nephrocytes. These SNARE proteins bound to APOL1-G0 with higher affinity than APOL1-G1/G2, and attenuated APOL1-G0 cytotoxicity to a greater extent than either APOL1-RA. CONCLUSIONS: Using a Drosophila screen, we identified SNARE proteins (Syx7, Ykt6, and Syb) as antagonists of APOL1-induced cytotoxicity by directly binding APOL1. These data uncovered a new potential protective role for certain SNARE proteins in the pathogenesis of APOL1-CKD and provide novel therapeutic targets for APOL1-associated nephropathies.

20.
bioRxiv ; 2023 Sep 13.
Article in English | MEDLINE | ID: mdl-37745500

ABSTRACT

Antiretroviral therapy (ART) has profoundly decreased HIV-1 associated morbidity. However, despite ART, immune cells remain latently infected and slowly release viral proteins, leading to chronic inflammation and HIV associated comorbidities. Thus, new strategies are needed to reduce the inflammatory effects of HIV-1. In previous studies we found that gamma secretase inhibitor (GSIXX) ameliorated renal lesions of HIV-Tg26 mice carrying replication defective HIV-1 PNL4-3 by inhibiting Notch activation. Since gamma secretase inhibition is not a safe strategy in humans, here we examined the specific role of the Notch3 pathway in the pathogenesis of the renal lesions and outcome of HIV-Tg26 mice. We found that Notch3 is activated in podocytes and other renal cells in HIV-Tg26 mice and human biopsies with HIV-1 associated Nephropathy (HIVAN). Knockdown of Notch3 in HIV-Tg26 mice revealed a marked reduction in the mortality rate, improvement in renal injury and function. RNA sequencing and immunolabeling data revealed that Notch3 deletion drastically reduced infiltrating renal macrophages in HIV-Tg-N3KO mice in association with renal reduction of HIV-nef mRNA expression levels. In fact, bone marrow derived macrophages from HIV-Tg26 mice showed a significant activation of Notch3 signaling. Further, systemic levels of TNF-alpha and MCP-1 and other inflammatory chemokines and cytokines were reduced in Tg-N3KO mice as compared to HIV-Tg26 mice and this translated to a marked reduction of HIV-induced skin lesions. Taken together, these studies strongly point to a dual inhibitory/therapeutic effect of Notch3 inhibition on HIV-induced systemic, skin and renal lesions independently of ART.

SELECTION OF CITATIONS
SEARCH DETAIL