Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Dis Model Mech ; 14(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-34308967

ABSTRACT

People of African ancestry living with the human immunodeficiency virus-1 (HIV-1) are at risk of developing HIV-associated nephropathy (HIVAN). Children with HIVAN frequently show high plasma fibroblast growth factor-2 (FGF-2) levels; however, the role of circulating FGF-2 in the pathogenesis of childhood HIVAN is unclear. Here, we explored how circulating FGF-2 affected the outcome of HIVAN in young HIV-Tg26 mice. Briefly, we demonstrated that FGF-2 was preferentially recruited in the kidneys of mice without pre-existing kidney disease, precipitating HIVAN by activating phosphorylated extracellular signal-regulated kinase (pERK) in renal epithelial cells, without inducing the expression of HIV-1 genes. Wild-type mice injected with recombinant adenoviral FGF-2 (rAd-FGF-2) vectors carrying a secreted form of human FGF-2 developed transient and reversible HIVAN-like lesions, including proteinuria and glomerular enlargement. HIV-Tg26 mice injected with rAd-FGF-2 vectors developed more-significant proliferative and pro-fibrotic inflammatory lesions, similar to those seen in childhood HIVAN. These lesions were partially reversed by treating mice with the FGF/VEGF receptor tyrosine kinase inhibitor PD173074. These findings suggest that high plasma FGF-2 levels may be an independent risk factor for precipitating HIVAN in young children.


Subject(s)
AIDS-Associated Nephropathy , HIV-1 , AIDS-Associated Nephropathy/genetics , Animals , Child, Preschool , Disease Models, Animal , Fibroblast Growth Factor 2 , HIV-1/genetics , Humans , Mice , Mice, Transgenic
2.
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
3.
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
4.
Med Res Arch ; 8(6)2020 Jun.
Article in English | MEDLINE | ID: mdl-33043139

ABSTRACT

Severe bleeding after cardiothoracic surgery with cardiopulmonary bypass (CPB) is associated with increased morbidity and mortality in adults and children. Fibroblast Growth Factor-2 (FGF-2) and Vascular Endothelial Growth Factor-A (VEGF-A) induce hemorrhage in murine models with heparin exposure. We aim to determine if plasma and urine levels of FGF-2 and VEGF-A in the immediate perioperative period can identify children with severe bleeding after CPB. We performed a prospective, observational biomarker study in 64 children undergoing CPB for congenital heart disease repair from June 2015 - January 2017 in a tertiary pediatric referral center. Primary outcome was severe bleeding defined as ≥ 20% estimated blood volume loss within 24-hours. Independent variables included perioperative plasma and urinary FGF-2 and VEGF-A levels. Analyses included comparative (Wilcoxon rank sum, Fisher's exact, and Student's t tests) and discriminative (receiver operator characteristic [ROC] curve) analyses. Forty-eight (75%) children developed severe bleeding. Median plasma and urinary FGF-2 and VEGF-A levels were elevated in children with severe bleeding compared to without bleeding (preoperative: plasma FGF-2 = 16[10-35] vs. 9[2-13] pg/ml; urine FGF-2= 28[15-76] vs. 14.5[1.5-22] pg/mg; postoperative: plasma VEGF-A = 146[34-379] vs. 53 [0-134] pg/ml; urine VEGF-A = 132 [52-257] vs. 45[0.1-144] pg/mg; all p < 0.05). ROC curve analyses of combined plasma and urinary FGF-2 and VEGF-A levels discriminated severe postoperative bleeding (AUC: 0.73-0.77) with mean sensitivity and specificity above 80%. We conclude that the perioperative plasma and urinary levels of FGF-2 and VEGF-A discriminate risk of severe bleeding after pediatric CPB.

5.
Dis Model Mech ; 13(10)2020 10 28.
Article in English | MEDLINE | ID: mdl-32917744

ABSTRACT

Modern antiretroviral therapies (ART) have decreased the prevalence of HIV-associated nephropathy (HIVAN). Nonetheless, we continue to see children and adolescents with HIVAN all over the world. Furthermore, once HIVAN is established in children, it is difficult to revert its long-term progression, and we need better animal models of childhood HIVAN to test new treatments. To define whether the HIV-1 trans-activator (Tat) gene precipitates HIVAN in young mice, and to develop an inducible mouse model of childhood HIVAN, an HIV-Tat gene cloned from a child with HIVAN was used to generate recombinant adenoviral vectors (rAd-Tat). rAd-Tat and LacZ control vectors (2×109) were expressed in the kidney of newborn wild-type and HIV-transgenic (Tg26) FVB/N mice without significant proteinuria (n=5; 8 per group). Mice were sacrificed 7 and 35 days later to assess their renal outcome, the expression of HIV-genes and growth factors, and markers of cell growth and differentiation by RT-qPCR, immunohistochemistry and/or western blots. HIV-Tat induced the expression of HIV-1 genes and heparin-binding growth factors in the kidney of HIV-Tg26 mice, and precipitated HIVAN in the first month of life. No significant renal changes were detected in wild-type mice infected with rAd-Tat vectors, suggesting that HIV-Tat alone does not induce renal disease. This new mouse model of childhood HIVAN highlights the critical role that HIV-Tat plays in the pathogenesis of HIVAN, and could be used to study the pathogenesis and treatment of HIVAN in children and adolescents.


Subject(s)
AIDS-Associated Nephropathy/pathology , tat Gene Products, Human Immunodeficiency Virus/metabolism , AIDS-Associated Nephropathy/genetics , Albuminuria/complications , Albuminuria/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Apoptosis , Cell Dedifferentiation , Cell Proliferation , Child , Disease Models, Animal , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , HIV-1/genetics , Humans , Kidney/injuries , Kidney/pathology , Mice, Transgenic , Podocytes/metabolism , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , beta-Galactosidase/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry
6.
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
7.
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
8.
PLoS One ; 11(4): e0153837, 2016.
Article in English | MEDLINE | ID: mdl-27097314

ABSTRACT

Renal endothelial cells (REc) are the first target of HIV-1 in the kidney. The integrity of REc is maintained at least partially by heparin binding growth factors that bind to heparan sulfate proteoglycans located on their cell surface. However, previous studies showed that the accumulation of two heparin-binding growth factors, Vascular Endothelial Cell Growth Factor-A (VEGF-A) and Fibroblast Growth Factor-2 (FGF-2), in combination with the viral protein Tat, can precipitate the progression of HIV-renal diseases. Nonetheless, very little is known about how these factors affect the behavior of REc in HIV+ children. We carried out this study to determine how VEGF-A, FGF-2, and HIV-Tat, modulate the cytoskeletal structure and permeability of cultured REc, identify key signaling pathways involved in this process, and develop a functional REc assay to detect HIV+ children affected by these changes. We found that VEGF-A and FGF-2, acting in synergy with HIV-Tat and heparin, affected the cytoskeletal structure and permeability of REc through changes in Rho-A, Src, and Rac-1 activity. Furthermore, urine samples from HIV+ children with renal diseases, showed high levels of VEGF-A and FGF-2, and induced similar changes in cultured REc and podocytes. These findings suggest that FGF-2, VEGF-A, and HIV-Tat, may affect the glomerular filtration barrier in HIV+ children through the induction of synergistic changes in Rho-A and Src activity. Further studies are needed to define the clinical value of the REc assay described in this study to identify HIV+ children exposed to circulating factors that may induce glomerular injury through similar mechanisms.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , HIV Infections/pathology , Kidney Diseases/complications , Vascular Endothelial Growth Factor A/pharmacology , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Activation/drug effects , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/urine , HIV-1/physiology , Heparin/pharmacology , Humans , Permeability/drug effects , Podocytes/pathology , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/pathology
9.
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
10.
Toxicol Rep ; 2: 838-849, 2015.
Article in English | MEDLINE | ID: mdl-26213685

ABSTRACT

Phosphorodiamidate morpholino oligonucleotides (PMO) are used as a promising exon-skipping gene therapy for Duchenne Muscular Dystrophy (DMD). One potential complication of high dose PMO therapy is its transient accumulation in the kidneys. Therefore new urinary biomarkers are needed to monitor this treatment. Here, we carried out a pilot proteomic profiling study using stable isotope labeling in mammals (SILAM) strategy to identify new biomarkers to monitor the effect of PMO on the kidneys of the dystrophin deficient mouse model for DMD (mdx-23). We first assessed the baseline renal status of the mdx-23 mouse compared to the wild type (C57BL10) mouse, and then followed the renal outcome of mdx-23 mouse treated with a single high dose intravenous PMO injection (800 mg/kg). Surprisingly, untreated mdx-23 mice showed evidence of renal injury at baseline, which was manifested by albuminuria, increased urine output, and changes in established urinary biomarker of acute kidney injury (AKI). The PMO treatment induced further transient renal injury, which peaked at 7 days, and returned to almost the baseline status at 30 days post-treatment. In the kidney, the SILAM approach followed by western blot validation identified changes in Meprin A subunit alpha at day 2, then returned to normal levels at day 7 and 30 after PMO injection. In the urine, SILAM approach identified an increase in Clusterin and γ-glutamyl transpeptidase 1 as potential candidates to monitor the transient renal accumulation of PMO. These results, which were confirmed by Western blots or ELISA, demonstrate the value of the SILAM approach to identify new candidate biomarkers of renal injury in mdx-23 mice treated with high dose PMO. Chemical compounds studied in this article: Phosphorodiamidate morpholino (PubChem CID: 22140692); isoflurane (PubChem CID: 3763); formic acid (PubChem CID: 284); acetonitrile (PubChem CID: 6342); acetone (PubChem CID: 180); methanol (PubChem CID: 887).

11.
Proteomics Clin Appl ; 9(5-6): 490-500, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25764519

ABSTRACT

A significant number of children infected with the human immunodeficiency virus 1 (HIV-1) virus all over the world are at risk of developing renal diseases that could have a significant impact on their treatment and quality of life. It is necessary to identify children undergoing the early stages of these renal diseases, as well as the potential renal toxicity that could be caused by antiretroviral drugs, in order to prevent the development of cardiovascular complications and chronic renal failure. This article describes the most common renal diseases seen in HIV-infected children, as well as the value and limitations of the clinical markers that are currently being used to monitor their renal function and histological damage in a noninvasive manner. In addition, we discuss the progress made during the last 10 years in the discovery and validation of new renal biomarkers for HIV-infected children and young adults. Although significant progress has been made during the early phases of the biomarkers discovery, more work remains to be done to validate the new biomarkers in a large number of patients. The future looks promising, however, the new knowledge needs to be integrated and validated in the context of the clinical environment where these children are living.


Subject(s)
HIV Infections/urine , Kidney Diseases/urine , Animals , Biomarkers/urine , Child, Preschool , HIV Infections/complications , Humans , Kidney Diseases/diagnosis , Kidney Diseases/virology
12.
J AIDS Clin Res ; 5(6)2014 Jun.
Article in English | MEDLINE | ID: mdl-25429350

ABSTRACT

OBJECTIVE: Kaposi's sarcoma (KS) is an angioproliferative disease frequently seen in patients with the acquired immunodeficiency syndrome (AIDS). Previous studies suggest that the HIV-1 protein Tat and Fibroblast Growth Factor 2 (FGF-2) have synergistic angiogenic effects in AIDS-KS tumors. However, the mechanisms by which FGF-2 is released and activated in KS tumors are not clearly defined. We carried out this study to determine whether an FGF-binding protein (FGFBP1 or BP1) that enhances the angiogenic activity of FGF-2 is expressed in AIDS-KS tumors, and to define whether BP1, FGF-2, and HIV-Tat protein-protein interactions could play a potential clinically role in the pathogenesis of AIDS-KS. METHODS: BP1 was localized in AIDS-KS lesions by immunohistochemistry and in situ hybridization studies. The binding of radiolabeled FGF-2 to His-tagged BP1 or the FGF-receptor 1 was assessed in the presence and absence of HIV-Tat and other viral proteins. Mice carrying tetracycline-regulated BP1 transgene mice were used to determine whether activation of BP1 during wound healing induces KS-like lesions. RESULTS: BP1 expression was detected in AIDS-KS tumor keratinocytes, spindle cells, and infiltrating mononuclear cells. In addition, HIV-Tat competed for the binding of FGF-2 to immobilized BP1, but does not affect the interactions of FGF-2 with its high affinity receptor (FGFR-1). In contrast, two other HIV-proteins, Nef and gp120, did not affect the binding of FGF-2 to BP1 or to FGFR-1. Finally, up-regulation of BP1 expression in tetracycline-regulated -conditional BP1 transgenic mice subjected to skin wounds, induced KS-like skin lesions. CONCLUSION: Taking into consideration the results of previous studies showing that both HIV-Tat and BP1 enhance the mitogenic and angiogenic activity of locally-stored FGF-2, both in vitro and in vivo, our findings suggest a novel mechanism by which the release and activity of FGFs can be modulated in AIDS-KS tumors by HIV-Tat as well as BP1.

13.
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
14.
Pediatr Nephrol ; 27(3): 469-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21959768

ABSTRACT

Fibroblast growth factor-2 (FGF-2) is an angiogenic growth factor involved in renal growth and regeneration. Previous studies in rodents revealed that single intrarenal injections of FGF-2 improved the outcome of acute kidney injury (AKI). Septic children usually show elevated plasma levels of FGF-2, and are at risk of developing AKI. However, the role of circulating FGF-2 in the pathogenesis of AKI is not well understood. We have developed a mouse model to determine how FGF-2 released into the circulation modulates the outcome of AKI induced by lipopolysaccharide (LPS). Young FVB/N mice were infected with adenoviruses carrying a secreted form of human FGF-2 or control LacZ vectors. Subsequently, when the circulating levels of FGF-2 were similar to those seen in septic children, mice were injected with a non-lethal dose of LPS or control buffer. All mice injected with LPS developed hypotension and AKI, from which they recovered after 5 days. FGF-2 did not improve the outcome of AKI, and induced more significant renal proliferative and apoptotic changes during the recovery phase. These findings suggest that circulating FGF-2 may not necessarily prevent the development or improve the outcome of AKI. Moreover, the renal accumulation of FGF-2 might cause further renal damage.


Subject(s)
Acute Kidney Injury/etiology , Fibroblast Growth Factor 2/physiology , Lipopolysaccharides/toxicity , Actins/analysis , Acute Kidney Injury/blood , Acute Kidney Injury/physiopathology , Acute-Phase Proteins/urine , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Fibroblast Growth Factor 2/blood , Kidney/drug effects , Kidney/pathology , Lipocalin-2 , Lipocalins/urine , Male , Mice , Oncogene Proteins/urine , Proliferating Cell Nuclear Antigen/analysis , Systole/drug effects
15.
Anticancer Res ; 30(2): 519-27, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20332464

ABSTRACT

BACKGROUND: Worldwide among men, prostate cancer ranks third in cancer occurrence and sixth in cancer mortality. A number of 1, 4-naphthoquinone derivatives have been identified that possess significant pharmacological effects associated with antitumor activities. In this study, the in vitro effects of N-(3-chloro-1,4-dioxo 1,4-dihydro-naphthalen-2-yl)-benzamide (NCDDNB) were evaluated on androgen-dependent (CWR-22) and androgen-independent (PC-3, DU-145) human prostate cancer cell lines, and on a normal bone marrow cell line (HS-5). Specifically, the in vitro activity of this compound on cell cycle regulation and apoptosis was evaluated. MATERIALS AND METHODS: Established methods of cell viability, cell cycle, Western blot and apoptosis were used. RESULTS: The effect of NCDDNB on CWR-22, PC-3, DU-145 and HS-5 cells revealed significant anti-tumor activities with IC(50)s, of 2.5, 2.5, 6.5, and 25 muM respectively. The results of cell cycle analysis showed that NCDDNB arrested PC-3, DU-145, and CWR-22 cells in the G(1)-phase of the cell cycle. The compound showed no effect on the cell cycle progression in the HS-5 bone marrow cell line. These findings were further validated using Western blot analysis. NCDDNB showed the greatest amount of apoptosis in the androgen-independent PC-3 cells in a time-dependent manner with the apoptotic apex at day 5 of treatment. Furthermore, NCDDNB induced-apoptosis in DU-145 and CWR-22 cells peaked at day 3 of treatment. CONCLUSION: Although the mechanism of action of this compound has not been completely elucidated, the effect on the cell cycle and the induction of apoptosis in different prostate cancer cell lines prompted us to carry out a more in-depth preclinical evaluation. This study suggests that NCDDNB may have an impact on treatment of prostate cancer while protecting the bone marrow.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/toxicity , Cell Proliferation/drug effects , Naphthoquinones/toxicity , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Benzamides/chemical synthesis , Blotting, Western , Cell Cycle/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques , Male , Naphthoquinones/chemical synthesis , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
Prostate ; 70(7): 755-64, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20058239

ABSTRACT

BACKGROUND: Class IA PI 3-kinases produce phosphatidylinositol (3,4,5)-trisphosphate (PIP3). PIP3 is bound by AKT which facilities its activation by PDK1. Activated AKT promotes cell survival and stimulates cell proliferation. Class IA PI 3-kinases are heterodimers consisting of a regulatory subunit p85 and a catalytic subunit p110. The p110alpha isoform has been shown to be mutated in a number of tumor types. A number of recent studies suggest that the p110beta isoform may be functionally relevant in prostate cancer. In this study we extend this work to include the examination of the expression and functional properties of p110alpha and p110beta in three different prostate cancer cell lines, DU145, LNCaP, PC3, as well as the non-tumorigenic but immortalized RWPE1 prostate epithelial cell line. METHODS: Western blot analysis was used to measure protein expression and quantitative real-time PCR was used to measure mRNA levels. After targeted knockdown using isoform-specific siRNAs to reduce PI 3-kinase p110alpha or p110beta isoform expression, we measured downstream signally events such as phosphorylation of AKT, ERK 1/2, PDK, and FOXO, as well as biological consequences such as changes in apoptosis, and alterations in cell cycle progression. RESULTS: In all three prostate cancer cell lines examined, targeted knockdown of p110beta, and not p110alpha, resulted in significantly reduced AKT, PDK, and FOXO phosphorylation. While knockdown of either p110 isoform resulted in an increase in apoptosis and a cell cycle arrest in G1 in the remaining non-apoptotic cells, these effects were much more pronounced with knockdown of p110beta. CONCLUSIONS: Our results support the concept that p110beta appears to be the predominant functional class I PI 3-kinase isoform in prostate cancer cells.


Subject(s)
Cell Proliferation , Cell Survival/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Analysis of Variance , Blotting, Western , Cell Cycle/physiology , Cell Line, Tumor , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Humans , Male , Microscopy, Fluorescence , Phosphorylation , Prostate/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Neurotox Res ; 16(3): 194-204, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19526284

ABSTRACT

Although the etiology of Parkinson's disease (PD) remains elusive, a number of toxins including elevated salsolinol, an endogenous metabolite of dopamine may contribute to its pathology. It was reported recently that nicotine may have protective effects against salsolinol-induced toxicity in human neuroblastoma derived SH-SY5Y cells and that these effects of nicotine are mediated by nicotinic receptors. Donepezil (Aricept) is a reversible non-competitive acetylcholinesterase inhibitor that is approved for use in mild to moderate Alzheimer's disease. The increase in acetylcholine concentrations is believed to be the major contributory factor in donepezil's therapeutic efficacy. However, cholinesterase inhibitors may also directly interact with nicotinic receptors and possess neuroprotective properties. In this study, we sought to determine whether donepezil may have protective effects against salsolinol-induced toxicity in SH-SY5Y cells and whether the combination of donepezil and nicotine may result in additive protection. Moreover, it was of interest to elucidate the role of nicotinic receptors as well as cell cycle and apoptosis in mechanism of action of these compounds. SH-SY5Y cells were exposed to 0.6 mM salsolinol with and without various drug pretreatments for 48 h. Nicotine (50 muM) resulted in approximately 54% protection and donepezil (5 muM) resulted in approximately 40% protection, and the combination of the two resulted in an additive (approximately 93%) protection against salsolinol-induced toxicity. Salsolinol caused an arrest of the cells in G(1)-phase of cell cycle and an increase in apoptotic indices that were blocked by the combination of donepezil and nicotine. Mecamylamine, a non-selective nicotinic receptor antagonist completely blocked the effects of nicotine and partially attenuated the effects of donepezil. A combination of atropine, a muscarinic receptor antagonist and mecamylamine completely blocked the effects of donepezil, indicating involvement of both nicotinic and muscarinic receptors in donepezil's actions. The findings suggest a therapeutic potential for the combination of donepezil and nicotine in PD.


Subject(s)
Apoptosis/drug effects , Cholinesterase Inhibitors/pharmacology , Indans/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Piperidines/pharmacology , Salsoline Alkaloids/toxicity , Analysis of Variance , Annexin A5/metabolism , Atropine/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Donepezil , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Flow Cytometry/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mecamylamine/pharmacology , Neuroblastoma/pathology , Nicotinic Antagonists/pharmacology , Propidium
18.
Anticancer Res ; 29(1): 191-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19331150

ABSTRACT

BACKGROUND: Breast cancer is the most frequent cancer and the second leading cause of cancer deaths in women today. A number of 1,4-naphthoquinone derivatives have been found to possess significant pharmacological effects associated with marked antimicrobial and antitumor activities. In the present study, the in vitro effect of 2,3-dichloro-5,8-dimethoxy-1,4-naphthoquinone (DCDMNQ) was evaluated on estrogen-positive MCF-7 and estrogen-negative MDA-MB-436 and Hs-578T human breast cancer cell lines. Moreover, the in vitro activity of this compound on cell cycle regulation and apoptosis were evaluated. MATERIALS AND METHODS: Established methods of cell viability, cell cycle, Western blot and apoptosis were used. RESULTS: The effect of DCDMNQ on MCF-7, MDA-MB-436 and Hs-578T cells revealed significant antitumor activities with IC(50)s, of 0.6 +/- 0.02, 1.4 +/- 0.25 and 3.1 +/- 0.4 microM respectively. Cell cycle analysis showed that DCDMNQ inhibited progression through the cell cycle in MCF-7 and MDA-MB-436 cell lines in a time-dependent manner. DCDMNQ arrested cells in the S-phase of the cell cycle with the greatest proportion of cells in the S-phase by day 5. This cell-cycle arrest was corroborated by inhibition of topoisomerase I induced by DCDMNQ. These findings were further validated using Western blot analysis of retinoblastoma protein time-dependent phosphorylation. Furthermore, DCDMNQ induced apoptosis in both estrogen-positive and -negative cell lines in a time-dependent manner. However, the highest percentages of apoptotic cells were observed in the MDA-MB-436 cell line. CONCLUSION: Although the mechanism of action of DCDMNQ has not been completely elucidated, it appears that this compound can inhibit topoisomerase I in a concentration-dependent manner. These promising results to explore novel naphthoquinone analogues as potential breast cancer agents. This study suggests that DCDMNQ may have an impact on treatment of estrogen-positive and -negative breast cancer while protecting the bone marrow.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Naphthoquinones/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Topoisomerases, Type I/metabolism , Flow Cytometry , Humans , Topoisomerase I Inhibitors
19.
Anticancer Res ; 27(3B): 1393-9, 2007.
Article in English | MEDLINE | ID: mdl-17595753

ABSTRACT

BACKGROUND: Breast cancer patients are at increased risk of osteoporosis. Contributing factors include age and/or chemotherapy. The selective estrogen modulator, raloxifene (RAL), effective in the prevention of breast cancer and approved for the treatment and prevention of osteoporosis, may prove beneficial in current breast cancer treatment modules. The purpose of this study was to evaluate RAL in combination with 5-fluorouracil (5-FU) and trimetrexate (TMX) to determine the most effective sequence in which to administer these cell cycle specific agents while taking into consideration the cellular mechanism of action. The goal was to maintain cytotoxicity to breast cancer cells and capitalize on the selective estrogen receptor modulatory effects of RAL. MATERIALS AND METHODS: MCF-7 cells were exposed to (i) TMX, 5-FU or RAL alone, or (ii) RAL 24 h prior to 5-FU followed 2 h later by TMX, or (iii) 5-FU 2 h prior to TMX followed 24 h later by RAL. The cell viability was determined using the Quick Cell Proliferation Assay. RESULTS: The growth rate of MCF- 7 cells exposed to early RAL was 68.25 +/- 4.11% that of the control, however, late RAL exposure produced a growth of 34.75 +/- 4.79% that of the control. Late RAL maintained the cytotoxicity of the regimen. The findings were further supported by cell flow cytometry and Western blot analysis data. CONCLUSION: RAL given prior to 5-FU/TMX significantly compromised cytotoxicity to breast cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Density Conservation Agents/administration & dosage , Breast Neoplasms/drug therapy , Raloxifene Hydrochloride/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorouracil/administration & dosage , Humans , Trimetrexate/administration & dosage
20.
Anticancer Res ; 27(3B): 1537-46, 2007.
Article in English | MEDLINE | ID: mdl-17595773

ABSTRACT

BACKGROUND: Prostate cancer ranks third worldwide in cancer incidence and sixth in cancer mortality among men. A number of 1,4-naphthoquinone derivatives have been found to possess significant pharmacological effects associated with marked antimicrobial and antitumor activities. In the present study the in vitro effect of 2,3-dichloro-5,8-dimethoxy-1,4-naphthoquinone (DCDMNQ) was evaluated on androgen-dependent (LNCaP, CWR-22) and androgen-independent (PC-3. DU-145) human prostate cancer cell lines, and/or a normal bone marrow cell line (HS-5). Moreover, the in vitro activity of this compound on cell cycle regulation and apoptosis was evaluated. MATERIALS AND METHODS: Established methods of cell viability, cell cycle, Western blot and apoptosis were used. RESULTS: The effect of DCDMNQ on LNCaP, CWR-22, PC-3, DU-145 and HS-5 cells revealed significant anti-tumor activities with IC50s, of 1, 3. 1.5, 3 and 10 microM respectively. Cell cycle analysis showed that DCDMNQ inhibited progression through the cell cycle in PC-3 and DU-145 cell lines in a time-dependent manner. The result for the CWR-22 cell line showed that DCDMNQ arrested cells in the G -phase of the cell cycle with the greatest proportion of cells in the G1-phase by day 5; however, the LNCaP cell line was inconsistent. The compound showed no effect on the cell cycle progression in the bone marrow HS-5 cell line. These findings were further validated using Western blot analysis. Furthermore, DCDMNQ induced apoptosis in the androgen-independent cells, preferentially over that of the androgen-dependent cell lines, in a time-dependent manner. CONCLUSION: Although the mechanism of action of this compound has not been completely elucidated, the effect on the cell cycle and the induction of apoptosis in different prostate cancer cell lines prompted us to carry out a more in-depth preclinical evaluation of it. This study suggests that DCDMNQ may have an impact on treatment of prostate cancer while protecting the bone marrow.


Subject(s)
Androgens/metabolism , Antineoplastic Agents/pharmacology , Naphthoquinones/pharmacology , Prostatic Neoplasms/metabolism , Apoptosis , Blotting, Western , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Humans , Inhibitory Concentration 50 , Male , Retinoblastoma Protein/analysis , Retinoblastoma Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...