Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Life (Basel) ; 13(12)2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38137939

ABSTRACT

Heart failure (HF) and chronic kidney disease (CKD) are associated with high mortality. In both disorders, impaired iron homeostasis, mostly in the form of a functional iron deficiency, is a frequent co-morbidity. In HF, functional iron deficiency and management by i.v. iron supplementation have been proven to affect both prognosis and functional capacity. In the same context, iron supplementation is routine for the adequate management of renal anemia in CKD. In numerous recent studies in HF and in CKD, sodium-glucose transporter 2 (SGLT2) inhibitor treatment has been proven to significantly reduce mortality. Furthermore, the same trials showed that these drugs alleviate iron deficiency and anemia. These effects of SGLT2 inhibitors may be due to an amelioration of inflammation with reduced interleukin-6 (IL-6) and to an enhancement of autophagy with increased sirtuin 1 (SIRT1), both associated with modified production of hepcidin and enhanced ferritinophagy. However, the exact pathogenic basis of the beneficial SGLT2 inhibitor action is not fully elucidated. Nevertheless, effects on iron homeostasis might be a potential explanatory mechanism for the powerful SGLT2 inhibitors' cardiovascular and renal outcome benefits. In addition, the interaction between iron supplementation and SGLT2 inhibitors and its potential impact on prognosis remains to be clarified by future studies. This review represents a significant effort to explore the complex relationships involved, seeking to elucidate the intricate mechanisms by which SGLT2 inhibitors influence iron homeostasis.

2.
Int J Mol Sci ; 24(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38003536

ABSTRACT

The interleukin-1 gene cluster encodes cytokines, which modulate mesangial cell proliferation and matrix expansion, both constituting central factors in the development and progression of immunoglobulin A nephropathy (IgAN). A candidate-gene study was performed to examine the association of polymorphisms of the interleukin-1 gene cluster with the risk of progressive IgAN. To gain deeper insights into the involvement of interleukin genes in IgAN, a meta-analysis of genetic association studies (GAS) that examine the association between interleukin variants and IgAN was conducted. Association study: The case-control study consisted of 121 unrelated Caucasians with sporadic, histologically diagnosed IgAN and of 246 age- and sex-matched healthy controls. Persistent proteinuria (>2 g/24 h) and/or impaired kidney function (serum creatinine > 1.5 mg/dL) defined progressive (n = 67) vs. non-progressive (n = 54) IgAN cases. Genotypes were assessed for two promoter-region single-nucleotide polymorphisms, C-899T (rs1800587) in IL1A and C-511T (rs16944) in IL1B, and for one penta-allelic variable-length tandem repeat polymorphism (VNTR 86 bp intron 2) in IL1RN. The association of these variants with the susceptibility of IgAN and the development of progressive IgAN (healthy status, IgAN, progressive IgAN) was tested using the generalized odds ratio (ORG) metric. Linkage disequilibrium and haplotype analysis were also performed. Meta-analysis: We included in the meta-analysis 15 studies investigating association between 14 interleukin variants harbored in eight different genes and IgAN. The ORG was used to evaluate the association between interleukin variants and IgAN using random effects models. The present case-control study revealed association of IL1B C-511T (rs16944) with the progression of IgAN (p = 0.041; ORG = 2.11 (1.09-4.07)). On haplotype analysis, significant results were derived for the haplotypes C-C-1 (p = 0.005; OR = 0.456 (0.261~0.797)) and C-T-2 (p = 0.003; OR = 4.208 (1.545-11.50)). Regarding association and meta-analysis results, variants in IL1B (rs1143627 and rs16944), IL1RN (rs928940, rs439154, and rs315951) and IL10 (rs1800871) were associated with IgAN based on either genotype or allele counts. Genetic variants and haplotypes in the IL1B, IL1RN, and IL10 genes might contribute to an increased risk for development and progression of IgAN.


Subject(s)
Glomerulonephritis, IGA , Humans , Glomerulonephritis, IGA/genetics , Glomerulonephritis, IGA/pathology , Case-Control Studies , Interleukin-10/genetics , Genetic Predisposition to Disease , Genotype , Interleukins/genetics , Polymorphism, Single Nucleotide , Interleukin-1/genetics , Interleukin-1beta/genetics
3.
Genes (Basel) ; 14(7)2023 07 21.
Article in English | MEDLINE | ID: mdl-37510392

ABSTRACT

Tumor necrosis factor-α (TNF-α) is a potent pro-inflammatory cytokine, involved in the pathogenesis and progression of immunoglobulin A nephropathy (IgAN). A bi-allelic polymorphism in the promoter region, at position -308 (G/A) of the TNF-α gene (rs1800629) is associated with an increased TNF-a production. However, several previous association studies of TNF-α G-308A polymorphism and IgAN rendered contradictory findings. The objective of the present study is to shed light on these inconclusive results and clarify the role of TNF-α and any possible contribution of this factor in the development and progression of sporadic IgAN. Therefore, a meta-analysis of all available genetic association studies relating the TNF-α G-308A polymorphism to the risk for development and/or progression of IgAN was conducted. Seven studies were included in the meta-analysis. Three of them included populations of European descent (Caucasians) and four involved Asians. The generalized odds ratio (ORG) was used to estimate the risk for the development and/or progression of the disease. Overall, the meta-analysis did not detect any significant association between the G-308A variant and both the risk of developing IgAN and the risk for progression of IgAN. In conclusion, these results suggest that TNF-α does not constitute a key component in the genetic architecture of sporadic IgAN. However, further evidence deciphering the influence of TNF-α on IgAN is still needed.


Subject(s)
Glomerulonephritis, IGA , Tumor Necrosis Factor-alpha , Humans , Genetic Association Studies , Glomerulonephritis, IGA/genetics , Polymorphism, Genetic , Tumor Necrosis Factor-alpha/genetics
4.
Int J Mol Sci ; 24(11)2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37298591

ABSTRACT

Besides being a marker of kidney disease severity, albuminuria exerts a toxic effect on renal proximal tubular epithelial cells (RPTECs). We evaluated whether an unfolded protein response (UPR) or DNA damage response (DDR) is elicited in RPTECs exposed to high albumin concentration. The deleterious outcomes of the above pathways, apoptosis, senescence, or epithelial-to-mesenchymal transition (EMT) were evaluated. Albumin caused reactive oxygen species (ROS) overproduction and protein modification, and a UPR assessed the level of crucial molecules involved in this pathway. ROS also induced a DDR evaluated by critical molecules involved in this pathway. Apoptosis ensued through the extrinsic pathway. Senescence also occurred, and the RPTECs acquired a senescence-associated secretory phenotype since they overproduced IL-1ß and TGF-ß1. The latter may contribute to the observed EMT. Agents against endoplasmic reticulum stress (ERS) only partially alleviated the above changes, while the inhibition of ROS upregulation prevented both UPR and DDR and all the subsequent harmful effects. Briefly, albumin overload causes cellular apoptosis, senescence, and EMT in RPTECs by triggering UPR and DDR. Promising anti-ERS factors are beneficial but cannot eliminate the albumin-induced deleterious effects because DDR also occurs. Factors that suppress ROS overproduction may be more effective since they could halt UPR and DDR.


Subject(s)
Kidney Tubules, Proximal , Signal Transduction , Albumins/metabolism , Albumins/toxicity , Cell Line , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Kidney Tubules, Proximal/metabolism , Reactive Oxygen Species/metabolism , Humans
5.
J Basic Clin Physiol Pharmacol ; 34(6): 745-754, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-35918786

ABSTRACT

OBJECTIVES: Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury (AKI). Experimental studies have shown that indoleamine 2,3-dioxygenase 1 (IDO) and the purinergic receptor P2X7 contribute to kidney I-R injury. We evaluated whether there is an interplay between IDO and P2X7 in the pathogenesis of I-R injury. METHODS: Primary renal proximal tubular epithelial cells (RPTECs) were subjected to anoxia or reoxygenation with or without specific inhibitors. Cell imaging, colorimetric assays, and Western blotting were used. RESULTS: Cell imaging revealed that inhibition of IDO, or all the purinergic receptors with an ATPase, or specific inhibition of P2X7 rescued the cells from anoxia or reoxygenation-induced cell death. This was confirmed with LDH release assay, which also detected the ferroptotic nature of cell death due to reoxygenation. On the contrary, activated cleaved caspase 3 increased during anoxia, showing that apoptosis prevails. All the aforementioned treatments prevented caspase increase. Both anoxia and reoxygenation increased extracellular ATP, IDO, and P2X7 expression. IDO remained unaffected by the above-mentioned treatments. On the contrary, treatment with apyrase or inhibition of P2X7decreased extracellular ATP and P2X7 expression, which are also decreased by inhibition of IDO. The first indicates a positive feedback loop regarding P2X7 activation, expression and function, while the latter implies that IDO controls P2X7 expression. CONCLUSIONS: In RPRECs subjected to anoxia or reoxygenation, IDO is upregulated, increasing P2X7 and contributing to anoxia or reoxygenation-induced cell death. Clarifying the molecular mechanisms implicated in kidney I-R injury is of particular interest since it may lead to new therapeutic strategies against AKI.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Apoptosis , Reperfusion Injury/metabolism , Acute Kidney Injury/prevention & control , Epithelial Cells , Hypoxia , Adenosine Triphosphate/pharmacology
6.
Int J Mol Sci ; 23(24)2022 Dec 17.
Article in English | MEDLINE | ID: mdl-36555751

ABSTRACT

Gliflozins are a new class of antidiabetic drugs with renoprotective properties. In cultures of primary human renal tubular epithelial cells (RPTECs) subjected to high-glucose conditions in the presence or absence of dapagliflozin, we evaluated cellular senescence pathways. High glucose increased sodium-glucose cotransporter-2 (SGLT-2) expression and glucose consumption, enhancing reactive oxygen species production. The latter induced DNA damage, ataxia telangiectasia mutated kinase (ATM), and p53 phosphorylation. Stabilized p53 increased the cell cycle inhibitor p21, resulting in cell cycle arrest and increasing the cellular senescence marker beta-galactosidase (GLB-1). RPTECs under high glucose acquired a senescence-associated secretory phenotype, which was detected by the production of IL-1ß, IL-8, and TGF-ß1. By decreasing SGLT-2 expression and glucose consumption, dapagliflozin inhibited the above pathway and prevented RPTEC senescence. In addition, dapagliflozin reduced the cell cycle inhibitor p16 independently of the glucose conditions. Neither glucose concentration nor dapagliflozin affected the epithelial-to-mesenchymal transition when assessed with α-smooth muscle actin (α-SMA). Thus, high glucose induces p21-dependent RPTEC senescence, whereas dapagliflozin prevents it. Since cellular senescence contributes to the pathogenesis of diabetic nephropathy, delineating the related molecular mechanisms and the effects of the widely used gliflozins on them is of particular interest and may lead to novel therapeutic approaches.


Subject(s)
Sodium-Glucose Transporter 2 Inhibitors , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/metabolism , Glucose/metabolism , Cellular Senescence/physiology , Epithelial Cells/metabolism
7.
Biomolecules ; 12(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36291624

ABSTRACT

Ischemia-reperfusion injury is the leading cause of acute kidney injury. Reactive oxygen species (ROS) production causes cell death or senescence. In cultures of primary human renal tubular epithelial cells (RPTECs) subjected to anoxia-reoxygenation, inhibition of the Krebs cycle at the level of malate dehydrogenase-2 (MDH-2) decreases hypoxia-inducible factor-1α and oxidative stress and protects from apoptotic or ferroptotic cell death. Inhibition of MDH-2 decreased reoxygenation-induced upregulation of p53 and p21, restored the levels of the proliferation marker Ki-67, and prevented the upregulation of the senescence marker beta-galactosidase and interleukin-1ß production. MDH-2 inhibition reduced the reoxygenation-induced upregulation of ATP, but the alterations of critical cell metabolism enzymes allowed enough ATP production to prevent cell energy collapse. Thus, inhibition of the Krebs cycle at the level of MDH-2 protects RPTECs from anoxia-reoxygenation-induced death or senescence. MDH-2 may be a promising pharmaceutical target against ischemia-reperfusion injury.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Malate Dehydrogenase , Reperfusion Injury , Humans , Adenosine Triphosphate/metabolism , Apoptosis , beta-Galactosidase/metabolism , Epithelial Cells/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Ki-67 Antigen/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Pharmaceutical Preparations/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Kidney Int ; 102(6): 1305-1319, 2022 12.
Article in English | MEDLINE | ID: mdl-35921911

ABSTRACT

Chronic allograft dysfunction with progressive fibrosis of unknown cause remains a major issue after kidney transplantation, characterized by ischemia-reperfusion injury (IRI). One hypothesis to account for this is that spontaneous progressive tubulointerstitial fibrosis following IRI is driven by cellular senescence evolving from a prolonged, unresolved DNA damage response (DDR). Since cellular communication network factor 2 ((CCN2), formerly called connective tissue growth factor), an established mediator of kidney fibrosis, is also involved in senescence-associated pathways, we investigated the relation between CCN2 and cellular senescence following kidney transplantation. Tubular CCN2 overexpression was found to be associated with DDR, loss of kidney function and tubulointerstitial fibrosis in both the early and the late phase in human kidney allograft biopsies. Consistently, CCN2 deficient mice developed reduced senescence and tubulointerstitial fibrosis in the late phase; six weeks after experimental IRI. Moreover, tubular DDR markers and plasma urea were less elevated in CCN2 knockout than in wild-type mice. Finally, CCN2 administration or overexpression in epithelial cells induced upregulation of tubular senescence-associated genes including p21, while silencing of CCN2 alleviated DDR induced by anoxia-reoxygenation injury in cultured proximal tubule epithelial cells. Thus, our observations indicate that inhibition of CCN2 can mitigate IRI-induced acute kidney injury, DNA damage, and the subsequent DDR-senescence-fibrosis sequence. Hence, targeting CCN2 might help to protect the kidney from transplantation-associated post-IRI chronic kidney dysfunction.


Subject(s)
Acute Kidney Injury , Connective Tissue Growth Factor , DNA Damage , Reperfusion Injury , Animals , Humans , Mice , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Fibrosis , Kidney/pathology , Mice, Inbred C57BL , Reperfusion Injury/pathology
9.
Antioxidants (Basel) ; 10(12)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34943123

ABSTRACT

AKI, due to the fact of altered oxygen supply after kidney transplantation, is characterized by renal ischemia-reperfusion injury (IRI). Recent data suggest that AKI to CKD progression may be driven by cellular senescence evolving from prolonged DNA damage response (DDR) following oxidative stress. Cellular communication factor 2 (CCN2, formerly called CTGF) is a major contributor to CKD development and was found to aggravate DNA damage and the subsequent DDR-cellular senescence-fibrosis sequence following renal IRI. We therefore investigated the impact of CCN2 inhibition on oxidative stress and DDR in vivo and in vitro. Four hours after reperfusion, full transcriptome RNA sequencing of mouse IRI kidneys revealed CCN2-dependent enrichment of several signaling pathways, reflecting a different immediate stress response to IRI. Furthermore, decreased staining for γH2AX and p-p53 indicated reduced DNA damage and DDR in tubular epithelial cells of CCN2 knockout (KO) mice. Three days after IRI, DNA damage and DDR were still reduced in CCN2 KO, and this was associated with reduced oxidative stress, marked by lower lipid peroxidation, protein nitrosylation, and kidney expression levels of Nrf2 target genes (i.e., HMOX1 and NQO1). Finally, silencing of CCN2 alleviated DDR and lipid peroxidation induced by anoxia-reoxygenation injury in cultured PTECs. Together, our observations suggest that CCN2 inhibition might mitigate AKI by reducing oxidative stress-induced DNA damage and the subsequent DDR. Thus, targeting CCN2 might help to limit post-IRI AKI.

10.
Genes (Basel) ; 12(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34946835

ABSTRACT

BACKGROUND: Many lines of evidence highlight the genetic contribution on the development of diabetic nephropathy (DN). One of the studied genes is SERPINE1 whose the role in the risk of developing DN remains questionable. In order to elucidate the contribution of SERPINE1 in DN progression in the context of type 2 diabetes mellitus (T2DM), we conducted an association study and meta-analysis of SERPINE1 genetic variants. MATERIALS AND METHODS: A total of 190 patients with DN, 150 T2DM (type 2 diabetes mellitus) patients without DN and 238 healthy controls were recruited. We selected five tag single-nucleotide polymorphisms (SNPs) from the HapMap. The generalized odds ratio (ORG) was calculated to estimate the risk on DN development. Subgroup analyses based on ethnicity and type of diabetes were also performed. RESULTS: Both the present association study regarding SERPINE1 SNPs (rs2227667, rs2070682, rs1050813, rs2227690, rs2227692) did not found any significant association between SERPINE1 variants and DN and the meta-analysis of variant 4G>5G (rs1799889) did not also reveal a significant association between 4G>5G variant and DN in main and subgroup analyses. DISCUSSION: In conclusion, the present association study and meta-analysis provides strong evidence that SERPINE1 genetic variant 4G>5G is not implicated in the risk or development of DN in Caucasians. Further studies in other populations remain to further investigate the role of this variant in the course of DN.


Subject(s)
Diabetic Nephropathies , Plasminogen Activator Inhibitor 1 , Polymorphism, Single Nucleotide , Aged , Female , Humans , Male , Alleles , Case-Control Studies , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Genetic Predisposition to Disease/genetics , Genotype , Odds Ratio , Plasminogen Activator Inhibitor 1/genetics , Polymorphism, Single Nucleotide/genetics
11.
Biomolecules ; 11(10)2021 10 15.
Article in English | MEDLINE | ID: mdl-34680153

ABSTRACT

Ischemia-reperfusion injury is the commonest form of acute kidney injury (AKI). Tubular epithelial cell senescence contributes to incomplete recovery from AKI and predisposes to subsequent chronic kidney disease. In cultures of primary proximal renal tubular epithelial cells (RPTECs) subjected to anoxia or reoxygenation, we evaluated the role of indoleamine 2,3-dioxygenase 1 (IDO) in cellular senescence. Proteins of interest were assessed with Western blotting or enzyme-linked immunosorbent assay or histochemically. Under anoxia or reoxygenation, IDO expression and activity were increased. Moreover, the two IDO-derived pathways, the general control nonderepressible 2 kinase (GCN2K) pathway and the aryl-hydrocarbon receptor (AhR) pathway, were also activated. A DNA damage response (DDR) took place and led to increased levels of the cell-cycle inhibitors p21 and p16, and senescence-associated ß-galactosidase (SA-ß-Gal) activity. Cell proliferation was inhibited, and more IL-6 was produced. The IDO inhibitor 1-DL-methyl-tryptophan ameliorated the DDR; decreased p21, p16, and SA-ß-Gal activity; restored cell proliferation; and decreased IL-6 production. The AhR inhibitor CH223191 did not affect the above parameters. In conclusion, anoxia and the subsequent reoxygenation upregulate IDO. IDO depletes tryptophan and activates GCN2K. The latter enhances the anoxia- or reoxygenation-induced DDR, resulting in increased p21 and p16 expression and eventually leading to RPTEC senescence. Since cellular senescence affects AKI outcome, the role of IDO in cellular senescence and the possible therapeutic role of IDO inhibitors deserve further investigation.


Subject(s)
Cellular Senescence/genetics , Hypoxia/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-6/genetics , Protein Serine-Threonine Kinases/genetics , Tryptophan/analogs & derivatives , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Azo Compounds/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Humans , Hypoxia/drug therapy , Hypoxia/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Oxygen/metabolism , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Tryptophan/pharmacology , rho GTP-Binding Proteins/genetics
12.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33899121

ABSTRACT

The present study evaluated indoleamine 2,3­dioxygenase 1 (IDO) kinetics and how it affects cell survival during the two distinct phases of ischemia­reperfusion (I­R) injury. Primary renal proximal tubular epithelial cells (RPTECs) were cultured under anoxia or reoxygenation with or without the IDO inhibitor 1­DL­methyltryptophan, the aryl­hydrocarbon receptor (AhR) inhibitor CH223191 or the ferroptosis inhibitor α­tocopherol. Using cell imaging, colorimetric assays, PCR and western blotting, it was demonstrated that IDO was upregulated and induced apoptosis during anoxia. The related molecular pathway entails tryptophan degradation, general control non­derepressible­2 kinase (GCN2K) activation, increased level of phosphorylated eukaryotic translation initiation factor 2α, activating transcription factor (ATF)4, ATF3, C/EBP homologous protein, phosphorylated p53, p53, Bax, death receptor­5 and eventually activated cleaved caspase­3. Reoxygenation also upregulated IDO, which, in this case, induced ferroptosis. The related molecular pathway encompasses kynurenine production, AhR activation, cytochrome p450 enzymes increase, reactive oxygen species generation and eventually ferroptosis. In conclusion, in RPTECs, both anoxia and reoxygenation upregulated IDO, which in turn induced GCN2K­mediated apoptosis and AhR­mediated ferroptosis. Since both phases of I­R injury share IDO upregulation as a common point, its inhibition may prove a useful therapeutic strategy for preventing or attenuating I­R injury.


Subject(s)
Cell Hypoxia , Epithelial Cells/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kidney Tubules, Proximal/cytology , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Azo Compounds/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Ferroptosis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism
13.
Int J Mol Sci ; 22(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33923087

ABSTRACT

Chronic kidney disease (CKD) is an important global public health problem due to its high prevalence and morbidity. Although the treatment of nephrology patients has changed considerably, ineffectiveness and side effects of medications represent a major issue. In an effort to elucidate the contribution of genetic variants located in several genes in the response to treatment of patients with CKD, we performed a systematic review and meta-analysis of all available pharmacogenetics studies. The association between genotype distribution and response to medication was examined using the dominant, recessive, and additive inheritance models. Subgroup analysis based on ethnicity was also performed. In total, 29 studies were included in the meta-analysis, which examined the association of 11 genes (16 polymorphisms) with the response to treatment regarding CKD. Among the 29 studies, 18 studies included patients with renal transplantation, 8 involved patients with nephrotic syndrome, and 3 studies included patients with lupus nephritis. The present meta-analysis provides strong evidence for the contribution of variants harbored in the ABCB1, IL-10, ITPA, MIF, and TNF genes that creates some genetic predisposition that reduces effectiveness or is associated with adverse events of medications used in CKD.


Subject(s)
Pharmacogenomic Testing , Pharmacogenomic Variants , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/genetics , Azathioprine/pharmacokinetics , Cyclosporine/pharmacokinetics , Humans , Polymorphism, Genetic , Prednisolone/pharmacokinetics , Tacrolimus/pharmacokinetics , Treatment Outcome
15.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33760196

ABSTRACT

In antibody­mediated rejection (ABMR), the graft endothelium is at the forefront of the kidney transplant against the assault from the recipient's humoral immune system, and is a target of the latter. The present study investigated the effect of antibodies against human leukocyte antigen (HLA) class I (anti­HLAI) on the immunological properties of human glomerular endothelial cells. Additionally, the effect of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) inhibitor (everolimus), or the general control nonderepressible 2 kinase (GCN2K) activator (halofuginone) on anti­HLAI antibody­mediated alterations was assessed. Cell integrity was examined, an lactate dehydrogenase (LDH) release assay was performed and cleaved caspase­3 levels were determined. Furthermore, cell proliferation was analyzed by performing a bromodeoxyuridine assay and the cellular proteins involved in signal transduction or immune effector mechanisms were assessed via western blotting. IL­8, monocyte chemoattractive protein­1 (MCP­1), von Willebrand factor (vWF) and transforming growth factor­beta 1 (TGF­ß1) were assayed via ELISA. The results revealed that anti­HLAI triggered integrin signaling, activated mTOR and GCN2K, preserved cell integrity and promoted cell proliferation. Additionally, by increasing intercellular adhesion molecule 1 (ICAM­1), HLA­DR, IL­8 and MCP­1 levels, anti­HLAI enhanced the ability of immune cells to interact with endothelial cells thus facilitating graft rejection. Contrarily, by upregulating CD46 and CD59, anti­HLAI rendered the endothelium less vulnerable to complement­mediated injury. Finally, by enhancing vWF and TGF­ß1, anti­HLAI may render the endothelium prothrombotic and facilitate fibrosis and graft failure, respectively. According to our results, mTORC1 inhibition and GCN2K activation may prove useful pharmaceutical targets, as they prevent cell proliferation and downregulate ICAM­1, IL­8, MCP­1 and TGF­ß1. mTORC1 inhibition also decreases vWF.


Subject(s)
Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Protein Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/genetics , Antibodies, Anti-Idiotypic/immunology , CD59 Antigens/genetics , CD59 Antigens/immunology , Cell Proliferation/drug effects , Endothelial Cells/immunology , Everolimus/pharmacology , Graft Rejection/genetics , Graft Rejection/pathology , Histocompatibility Antigens Class I/genetics , Humans , Immunity, Humoral/genetics , Immunity, Humoral/immunology , Kidney Transplantation/adverse effects , Mechanistic Target of Rapamycin Complex 1/immunology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/immunology , Signal Transduction , TOR Serine-Threonine Kinases/immunology , von Willebrand Factor/genetics
16.
J Immunother ; 44(4): 164-166, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33721881

ABSTRACT

Immune checkpoint inhibitors by blocking specific inhibitory pathways induce T-cell-mediated tumor lysis. However, many remain to be elucidated about their effect on human humoral immunity. We evaluated the effect of the PD1 inhibitor nivolumab on humoral immunity by following the levels of antibodies against hepatitis B surface antigen (anti-HBs) in a hemodialysis patient successfully vaccinated against hepatitis B virus 5 years ago and now starting nivolumab for renal cell carcinoma lung metastases. Anti-HBs kinetics after administration of an extra vaccine dose were also evaluated. Nivolumab increased anti-HBs and facilitated a further increase following an additional vaccine dose. The observed time frame indicates that nivolumab boosts humoral immune response by affecting long-lived plasma cells and at least memory B cells. This may protect cancer patients from pathogens encountered in the past or against which vaccination has been performed and provide information for the emerging immune checkpoint inhibitors administration concept against chronic infectious diseases.


Subject(s)
Immune Checkpoint Inhibitors/immunology , Immunity, Humoral/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Hepatitis B/immunology , Hepatitis B Antibodies/immunology , Hepatitis B Surface Antigens/metabolism , Hepatitis B Vaccines/immunology , Humans , Immunotherapy/methods , Male , Memory B Cells/immunology , Middle Aged , Nivolumab/immunology , Renal Dialysis/methods
17.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572206

ABSTRACT

Direct allorecognition is the earliest and most potent immune response against a kidney allograft. Currently, it is thought that passenger donor professional antigen-presenting cells (APCs) are responsible. Further, many studies support that graft ischemia-reperfusion injury increases the probability of acute rejection. We evaluated the possible role of primary human proximal renal tubular epithelial cells (RPTECs) in direct allorecognition by CD4+ T-cells and the effect of anoxia-reoxygenation. In cell culture, we detected that RPTECs express all the required molecules for CD4+ T-cell activation (HLA-DR, CD80, and ICAM-1). Anoxia-reoxygenation decreased HLA-DR and CD80 but increased ICAM-1. Following this, RPTECs were co-cultured with alloreactive CD4+ T-cells. In T-cells, zeta chain phosphorylation and c-Myc increased, indicating activation of T-cell receptor and co-stimulation signal transduction pathways, respectively. T-cell proliferation assessed with bromodeoxyuridine assay and with the marker Ki-67 increased. Previous culture of RPTECs under anoxia raised all the above parameters in T-cells. FOXP3 remained unaffected in all cases, signifying that proliferating T-cells were not differentiated towards a regulatory phenotype. Our results support that direct allorecognition may be mediated by RPTECs even in the absence of donor-derived professional APCs. Also, ischemia-reperfusion injury of the graft may enhance the above capacity of RPTECs, increasing the possibility of acute rejection.


Subject(s)
Epithelial Cells/immunology , Graft Rejection/immunology , Kidney Transplantation/adverse effects , Kidney Tubules, Proximal/immunology , Reperfusion Injury/immunology , Allografts/cytology , Allografts/immunology , Allografts/pathology , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Graft Rejection/pathology , Humans , Isoantigens/immunology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Lymphocyte Activation , Primary Cell Culture , Reperfusion Injury/pathology , Transplantation, Homologous/adverse effects
18.
Mol Med Rep ; 23(1)2021 01.
Article in English | MEDLINE | ID: mdl-33179104

ABSTRACT

During the reperfusion phase of ischemia­reperfusion injury, reactive oxygen species (ROS) production aggravates the course of many diseases, including acute kidney injury. Among the various enzymes implicated in ROS production are the enzymes of the cytochromes P450 superfamily (CYPs). Since arylhydrocarbon receptor (AhR) controls the expression of certain CYPs, the involvement of this pathway was evaluated in reperfusion injury. Because AhR may interact with the nuclear factor erythroid 2­related factor 2 (Nrf2) and the hypoxia­inducible factor­1α (HIF­1α), whether such an interaction takes place and affects reperfusion injury was also assessed. Proximal renal proximal tubular epithelial cells were subjected to anoxia and subsequent reoxygenation. At the onset of reoxygenation, the AhR inhibitor CH223191, the HIF­1α activator roxadustat, or the ferroptosis inhibitor α­tocopherol were used. The activity of AhR, Nrf2, HIF­1α, and their transcriptional targets were assessed with western blotting. ROS production, lipid peroxidation and cell death were measured with colorimetric assays or cell imaging. Reoxygenation induced ROS production, lipid peroxidation and cell ferroptosis, whereas CH223191 prevented all. Roxadustat did not affect the above parameters. Reoxygenation activated AhR and increased CYP1A1, while CH223191 prevented both. Reoxygenation with or without CH223191 did not alter Nrf2 or HIF­1α activity. Thus, AhR is activated during reoxygenation and induces ROS production, lipid peroxidation and ferroptotic cell death. These detrimental effects may be mediated by AhR­induced CYP overexpression, while the Nrf2 or the HIF­1α pathways remain unaffected. Accordingly, the AhR pathway may represent a promising therapeutic target for the prevention of reperfusion injury.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Kidney Tubules, Proximal/cytology , Oxygen/adverse effects , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Reperfusion Injury/metabolism , Animals , Azo Compounds/pharmacology , Cell Hypoxia , Cells, Cultured , Cytochrome P-450 CYP1A1/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Ferroptosis , Glycine/analogs & derivatives , Glycine/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoquinolines/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Lipid Peroxidation , Mice , Models, Biological , NF-E2-Related Factor 2/metabolism , Oxygen/pharmacology , Pyrazoles/pharmacology , Reperfusion Injury/chemically induced , alpha-Tocopherol/pharmacology
19.
Arch Immunol Ther Exp (Warsz) ; 68(6): 31, 2020 Oct 04.
Article in English | MEDLINE | ID: mdl-33011826

ABSTRACT

Hypoxia-inducible factor (HIF) prolyl-hydroxylase inhibitors are currently used for the treatment of renal anemia. Since HIF affects immune cells, we evaluated the effect of such a drug, the roxadustat, on adaptive immunity. Cell proliferation was assessed in a two-way mixed lymphocyte reaction (MLR) with BrdU assay. In CD4+ T cells isolated from the two-way MLRs, western blotting was performed to detect the impact of roxadustat on HIF-1α and HIF-2α, the apoptotic marker cleaved caspase-3, and the master transcription factors of CD4+ T cells differentiation towards Th1, Th2, Th17, Treg and Tfh subsets. The signature cytokines of the above CD4+ T-cell subsets IFN-γ, IL-4, IL-17, IL-10, and IL-21 were measured in the supernatants. For assessing humoral immunity, we developed a suitable antibody-mediated complement-dependent cytotoxicity assay. Roxadustat stabilized HIF-1α and HIF-2α, suppressed cell proliferation, inhibited CD4+ T-cell differentiation into Th1 and Th17 subsets, while it favored differentiation towards Th2, Treg and Tfh. Roxadustat suppressed humoral immunity too. These immunosuppressive properties of roxadustat indicate that the recently introduced HIF prolyl-hydroxylase inhibitors in medical therapeutics may render the patients vulnerable to infections. This possibility should be further evaluated in clinical trials.


Subject(s)
Glycine/analogs & derivatives , Immunosuppressive Agents/pharmacology , Isoantibodies/metabolism , Isoquinolines/pharmacology , Th1 Cells/immunology , Th17 Cells/metabolism , Adult , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Female , Glycine/pharmacology , Healthy Volunteers , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Isoantigens/immunology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Male
20.
Biomed Rep ; 13(2): 3, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32509306

ABSTRACT

Ischemia-reperfusion (I-R) injury is involved in the pathogenesis of several human diseases. In the present study, the kinetics of the H2S producing enzymes-nuclear factor erythroid 2-like 2 (Nrf2)-antioxidant proteins axis under anoxia or reoxygenation was evaluated, as well as its effects on survival of mouse renal proximal tubular epithelial cells (RPTECs). In RPTECs subjected to anoxia and subsequent reoxygenation, reactive oxygen species (ROS) production, lipid peroxidation, ferroptotic cell death, the levels of the H2S producing enzymes and H2S, the expression of Nrf2 and its transcriptional targets superoxide dismutase-3, glutathione reductase, ferritin H and cystine-glutamate antiporter, as well as apoptosis, and the levels of p53, Bax and phosphorylated p53 were assessed. When needed, the H2S producing enzyme inhibitor aminooxyacetate, or the ferroptosis inhibitor α-tocopherol, were used. Reoxygenation induced ferroptosis, whereas anoxia activated the p53-Bax pathway and induced apoptosis. The H2S producing enzymes-Nrf2-antioxidant proteins axis was activated only during anoxia and not during reoxygenation, when cellular viability is threatened by ROS overproduction and the ensuing ferroptosis. The activation of the above axis during anoxia ameliorated the effects of the apoptotic p53-Bax pathway, but did not adequately protect against apoptosis. In conclusion, the H2S-Nrf2 axis is activated by anoxia, and although it reduces apoptosis, it does not completely prevent apoptotic cell death. Additionally, following reoxygenation, the above axis was not activated. This mistimed activation of the H2S producing enzymes-Nrf2-antioxidant proteins axis contributes to reoxygenation-induced cell death. Determining the exact molecular mechanisms involved in reoxygenation-induced cell death may assist in the development of clinically relevant interventions for preventing I-R injury.

SELECTION OF CITATIONS
SEARCH DETAIL
...