Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 264, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734696

ABSTRACT

BACKGROUND: Traditional biomarkers of chronic kidney disease (CKD) detect the disease in its late stages and hardly predict associated vascular damage. Integrin-linked kinase (ILK) is a scaffolding protein and a serine/threonine protein kinase that plays multiple roles in several pathophysiological processes during renal damage. However, the involvement of ILK as a biomarker of CKD and its associated vascular problems remains to be fully elucidated. METHODS: CKD was induced by an adenine-rich diet for 6 weeks in mice. We used an inducible ILK knockdown mice (cKD-ILK) model to decrease ILK expression. ILK content in mice's peripheral blood mononuclear cells (PBMCs) was determined and correlated with renal function parameters and with the expression of ILK and fibrosis and inflammation markers in renal and aortic tissues. Also, the expression of five miRNAs that target ILK was analyzed in whole blood of mice. RESULTS: The adenine diet increased ILK expression in PBMCs, renal cortex, and aortas, and creatinine and urea nitrogen concentrations in the plasma of WT mice, while these increases were not observed in cKD-ILK mice. Furthermore, ILK content in PBMCs directly correlated with renal function parameters and with the expression of renal and vascular ILK and fibrosis and inflammation markers. Finally, the expression of the five miRNAs increased in the whole blood of adenine-fed mice, although only four correlated with plasma urea nitrogen, and of those, three were downregulated in cKD-ILK mice. CONCLUSIONS: ILK, in circulating mononuclear cells, could be a potential biomarker of CKD and CKD-associated renal and vascular damage.


Subject(s)
Biomarkers , Kidney , Leukocytes, Mononuclear , Protein Serine-Threonine Kinases , RNA, Messenger , Renal Insufficiency, Chronic , Animals , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Leukocytes, Mononuclear/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Biomarkers/metabolism , Biomarkers/blood , Mice , Kidney/pathology , Kidney/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/blood , MicroRNAs/metabolism , Disease Models, Animal , Fibrosis
2.
Biomater Sci ; 11(14): 4916-4929, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37306667

ABSTRACT

White adipose tissue (WAT) hypertrophy is caused by the excessive storage of triglycerides (TGs) and is associated with obesity. We previously demonstrated that extracellular matrix mediator integrin beta1 (INTB1) and its downstream effector integrin linked kinase (ILK) are implicated in obesity establishment. We also considered in our previous works that ILK upregulation is a therapeutical strategy to reduce WAT hypertrophy. Carbon based nanomaterials (CNMs) have interesting potential to modify cell differentiation but have been never studied to change the properties of adipocytes. METHODS: GMC is a new graphene-based CNM that was tested for biocompatibility and functionality in cultured adipocytes. MTT, TG content, lipolysis quantification, and transcriptional changes were determined. Specific INTB1 blocking antibody and ILK depletion with specific siRNA were used to study the intracellular signalling. We complemented the study using subcutaneous WAT (scWAT) explants from transgenic ILK knockdown mice (cKD-ILK). GMC was topically administrated in the dorsal area of high fat diet-induced obese rats (HFD) for 5 consecutive days. The scWAT weights and some intracellular markers were analyzed after the treatment. RESULTS: graphene presence was characterized in GMC. It was non-toxic and effective in reducing TG content in vitro in a dose-dependent manner. GMC rapidly phosphorylated INTB1 and increased the expression and activity of hormone sensitive lipase (HSL), the lipolysis subproduct glycerol, and the expression of glycerol and fatty acid transporters. GMC also reduced the expression of adipogenesis markers. Pro-inflammatory cytokines were unaffected. ILK was overexpressed, and INTB1 or ILK blockade avoided functional GMC effects. Topical administration of GMC in HFD rats overexpressed ILK in scWAT, and their weight gains were reduced, while systemic (renal, hepatic) toxicity parameters were unaffected. CONCLUSIONS: GMC is safe and effective in reducing hypertrophied scWAT weight when topically applied and it can be considered of interest in anti-obesogenic strategies. GMC increases lipolysis and reduces adipogenesis inside adipocytes by mechanisms that imply the activation of INTB1, the overexpression of ILK, and changes in the expression and activity of several markers related to fat metabolism.


Subject(s)
Graphite , Lipolysis , Mice , Rats , Animals , Glycerol , Weight Gain , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Mice, Transgenic , Hypertrophy/complications , Integrins
3.
Exp Mol Med ; 54(3): 226-238, 2022 03.
Article in English | MEDLINE | ID: mdl-35246616

ABSTRACT

Cardiovascular disease is an important cause of death in patients with chronic kidney disease (CKD). Protein-bound uremic toxins, such as p-cresyl and indoxyl sulfate (IS), are poorly removed during hemodialysis, leading to vascular endothelial dysfunction and leukocyte extravasation. These processes can be related to dynamic adhesion structures called podosomes. Several studies have indicated the role of integrin-linked kinase (ILK) in the accumulation of integrin-associated proteins in podosomes. Here, we investigated the involvement of ILK and podosome formation in the adhesion and extravasation of monocytes under p-cresol (pc) and IS exposure. Incubation of THP-1 human monocyte cells with these toxins upregulated ILK kinase activity. Together, both toxins increased cell adhesion, podosome formation, extracellular matrix degradation, and migration of THP-1 cells, whereas ILK depletion with specific small interfering RNAs suppressed these processes. Interestingly, F-actin colocalized with cortactin in podosome cores, while ILK was colocalized in podosome rings under toxin stimulation. Podosome Wiskott-Aldrich syndrome protein (WASP)-interacting protein (WIP) and AKT protein depletion demonstrated that monocyte adhesion depends on podosome formation and that the ILK/AKT signaling pathway is involved in these processes. Ex vivo experiments showed that both toxins induced adhesion and podosome formation in leukocytes from wild-type mice, whereas these effects were not observed in leukocytes of conditional ILK-knockdown animals. In summary, under pc and IS stimulation, monocytes increase podosome formation and transmigratory capacity through an ILK/AKT signaling pathway-dependent mechanism, which could lead to vascular injury. Therefore, ILK could be a potential therapeutic target for the treatment of vascular damage associated with CKD.


Subject(s)
Podosomes , Protein Serine-Threonine Kinases , Animals , Cell Adhesion , Cresols , Cytoskeletal Proteins/metabolism , Humans , Indican/metabolism , Indican/pharmacology , Mice , Monocytes , Podosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , THP-1 Cells
4.
ACS Med Chem Lett ; 12(11): 1656-1662, 2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34790291

ABSTRACT

Integrin-linked kinase (ILK) has emerged as a controversial pseudokinase protein that plays a crucial role in the signaling process initiated by integrin-mediated signaling. However, ILK also exhibits a scaffolding protein function inside cells, controlling cytoskeletal dynamics, and has been related to non-neoplastic diseases such as chronic kidney disease (CKD). Although this protein always acts as a heterotrimeric complex bound to PINCH and parvin adaptor proteins, the role of parvin proteins is currently not well understood. Using in silico approaches for the design, we have generated and prepared a set of new tripeptides mimicking an α-parvin segment. These derivatives exhibit activity in phenotypic assays in an ILK-dependent manner without altering kinase activity, thus allowing the generation of new chemical probes and drug candidates with interesting ILK-modulating activities.

5.
Cell Physiol Biochem ; 54(1): 71-87, 2020 Jan 24.
Article in English | MEDLINE | ID: mdl-31972071

ABSTRACT

BACKGROUND/AIMS: Diabetes type 2, metabolic syndrome or non-alcoholic fatty liver disease are insulin resistance-related metabolic disorders, which lack a better prognosis before their full establishment. We studied the importance of the intracellular scaffold protein integrin linked kinaes (ILK) as a key modulator in the initial pathogenesis and the early progression of those insulin resistance- related disorders. METHODS: Adult mice with a global transgenic downregulation of ILK expression (cKD-ILK) and littermates without that depletion (CT) were fed with either standard (STD) or high fat (HFD) diets during 2 and 6 weeks. Weights, blood glucose and other systemic biochemical parameters were determined in animals under fasting conditions and after glucose or pyruvate intraperitoneal injections to test their tolerance. In RNA or proteins extracted from insulin-sensitive tissues, we determined by reverse transcription-quantitative PCR and western blot the expression of ILK, metabolites transporters and other metabolism and inflammatory markers. Glucose uptake capacity was studied in freshly isolated tissues. RESULTS: HFD feeding was able to early and progressively increase glycaemia, insulinemia, circulating glycerol, body weight gain, liver-mediated gluconeogenesis along this time lapse, but cKD-ILK have all these systemic misbalances exacerbated compared to CT in the same HFD time lapse. Interestingly, the tisular expression of ILK in HFD-fed CT was dramatically downregulated in white adipose tissue (WAT), skeletal muscle and liver at the same extent of the original ILK downregulation of cKD-ILK. We previously published that basal STD-fed cKD-ILK compared to basal STD-CT have different expression of glucose transporters GLUT4 in WAT and skeletal muscle. In the same STD-fed cKD-ILK, we observed here the increased expressions of hepatic GLUT2 and WAT pro-inflammatory cytokines TNF-α and MCP-1. The administration of HFD exacerbated the expression changes in cKD-ILK of these and other markers related to the imbalanced metabolism observed, such as WAT lipolysis (HSL), hepatic gluconeogenesis (PCK-1) and glycerol transport (AQP9). CONCLUSION: ILK expression may be taken as a predictive determinant of metabolic disorders establishment, because its downregulation seems to correlate with the early imbalance of glucose and glycerol transport and the subsequent loss of systemic homeostasis of these metabolites.


Subject(s)
Diet, High-Fat/adverse effects , Down-Regulation , Metabolic Diseases/etiology , Protein Serine-Threonine Kinases/genetics , Animals , Female , Gluconeogenesis , Inflammation/etiology , Inflammation/genetics , Insulin Resistance , Lipolysis , Male , Metabolic Diseases/genetics , Mice , Mice, Inbred BALB C
6.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1284-1297, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30726718

ABSTRACT

Kidney fibrosis is one of the main pathological findings of progressive chronic kidney disease (CKD) although the pathogenesis of renal scar formation remains incompletely explained. Integrin-linked kinase (ILK), a major scaffold protein between the extracellular matrix (ECM) and intracellular signaling pathways, is involved in several pathophysiological processes during renal damage. However, ILK contribution in the CKD progress remains to be fully elucidated. In the present work, we studied 1) the renal functional and structural consequences of CKD genesis and progression when ILK is depleted and 2) the potential of ILK depletion as a therapeutic approach to delay CKD progression. We induced an experimental CKD model, based on an adenine-supplemented diet on adult wild-type (WT) and ILK-depleted mice, with a tubulointerstitial damage profile resembling that is observed in human CKD. The adenine diet induced in WT mice a progressive increase in plasma creatinine and urea concentrations. In the renal cortex it was also observed tubular damage, interstitial fibrosis and progressive increased ECM components, pro-inflammatory and chemo-attractant cytokines, EMT markers and TGF-ß1 expressions. These observations were highly correlated to a simultaneous increase of ILK expression and activity. In adenine-fed transgenic ILK-depleted mice, all these changes were prevented. Additionally, we evaluated the potential role of ILK depletion to be applied after the disease induction, as an effective approach to interventions in human CKD subjects. In this scenario, two weeks after the establishment of adenine-induced CKD, ILK was abrogated in WT mice and stabilized renal damage, avoiding CKD progression. We propose ILK to be a potential target to delay renal disease progression.


Subject(s)
Adenine/administration & dosage , Gene Knockdown Techniques , Kidney Tubules/metabolism , Protein Serine-Threonine Kinases/genetics , Renal Insufficiency, Chronic/genetics , Actins/genetics , Actins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Creatinine/blood , Diet , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Gene Expression Regulation , Humans , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Urea/blood
7.
Nefrologia (Engl Ed) ; 38(6): 639-646, 2018.
Article in English, Spanish | MEDLINE | ID: mdl-30337107

ABSTRACT

BACKGROUND: Patients with chronic kidney disease present with an accumulation of uraemic toxins, which have been identified as pathogenic agents associated with cardiovascular mortality, which is very high is this patient group. A phenomenon common to the progressive renal dysfunction and associated vascular damage, is the abnormal accumulation of extracellular matrix (ECM) proteins in the renal or vascular structures. OBJECTIVE: To determine the contribution of uraemia or the uraemic toxins to the production of cytokinins and ECM in aortas of uraemic animals or human aortic smooth muscle cells (HASMCs). MATERIALS AND METHODS: Mice were used with uraemia induced by a diet rich in adenine (0.2%) for 2, 4 or 6 weeks. Kidney function was evaluated by means of urine volume, plasma levels of creatinine, urea, fractional excretion of sodium, and vascular damage using histology, as well as protein expression using RT-qPCR. The HASMCs were incubated in vitro with uraemic toxins: p-cresol 10-100 (µg/ml) and indoxyl-sulphate25-100 (µg/ml) alone or simultaneously. The protein expression was evaluated using Western blot and confocal microscopy. RESULTS: The administration of adenine produced progressive kidney damage in the mice, thickening of the aortic wall, and increasing the expression of TGF-ß1 and ECM proteins. The toxins at high doses and combined also induced the expression of TGF-ß1 and ECM proteins by the HASMCs. CONCLUSIONS: The uraemia produced by an adenine rich diet or high doses of uraemic toxins induced the abnormal deposit of ECM proteins in the vascular wall or its production by HASMCs. The understanding of the mechanisms that underlie this pathophysiological process may be useful in the prevention of cardiovascular damage associated with the progress of chronic kidney disease, a disease, at the moment that is irreversible and occasional silent until its diagnosis in advanced stages.


Subject(s)
Blood Vessels/pathology , Cytokines/physiology , Extracellular Matrix Proteins/physiology , Renal Insufficiency, Chronic/complications , Uremia/complications , Adenine/administration & dosage , Animals , Fibrosis/etiology , Male , Mice , Mice, Inbred C57BL , Toxins, Biological/physiology , Transforming Growth Factor beta1/physiology
8.
FASEB J ; 32(2): 920-934, 2018 02.
Article in English | MEDLINE | ID: mdl-29054855

ABSTRACT

Ras proteins regulate cell survival, growth, differentiation, blood pressure, and fibrosis in some organs. We have demonstrated that H- ras gene deletion produces mice hypotension via a soluble guanylate cyclase-protein kinase G (PKG)-dependent mechanism. In this study, we analyzed the consequences of H- ras deletion on cardiac remodeling induced by continuous angiotensin II (AngII) infusion and the molecular mechanisms implied. Left ventricular posterior wall thickness and mass and cardiomyocyte cross-sectional area were similar between AngII-treated H-Ras knockout (H -ras-/-) and control wild-type (H -ras+/+) mice, as were extracellular matrix protein expression. Increased cardiac PKG-Iß protein expression in H -ras-/- mice suggests the involvement of this protein in heart protection. Ex vivo experiments on cardiac explants could support this mechanism, as PKG blockade blunted protection against AngII-induced cardiac hypertrophy and fibrosis markers in H -ras-/- mice. Genetic modulation studies in cardiomyocytes and cardiac and embryonic fibroblasts revealed that the lack of H-Ras down-regulates the B-RAF/MEK/ERK pathway, which induces the glycogen synthase kinase-3ß-dependent activation of the transcription factor, cAMP response element-binding protein, which is responsible for PKG-Iß overexpression in H -ras-/- mouse embryonic fibroblasts. This study demonstrates that H- ras deletion protects against AngII-induced cardiac remodeling, possibly via a mechanism in which PKG-Iß overexpression could play a partial role, and points to H-Ras and/or downstream proteins as potential therapeutic targets in cardiovascular disease.-Martín-Sánchez, P., Luengo, A., Griera, M., Orea, M. J., López-Olañeta, M., Chiloeches, A., Lara-Pezzi, E., de Frutos, S., Rodríguez-Puyol, M., Calleros, L., Rodríguez-Puyol, D. H- ras deletion protects against angiotensin II-induced arterial hypertension and cardiac remodeling through protein kinase G-Iß pathway activation.


Subject(s)
Angiotensin II/adverse effects , Cardiomegaly/enzymology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Hypertension/enzymology , MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/deficiency , Angiotensin II/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fibroblasts/enzymology , Fibroblasts/pathology , Gene Deletion , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Hypertension/chemically induced , Hypertension/pathology , Mice , Mice, Knockout
9.
Hum Gene Ther Methods ; 28(6): 340-351, 2017 12.
Article in English | MEDLINE | ID: mdl-29160106

ABSTRACT

Vesicular stomatitis virus G glycoprotein (VSVg) is extensively used for retroviral and lentiviral vector (LV) pseudotyping. However, VSVg pseudotyped vectors are serum inactivated, blocking the in vivo gene delivery. Several strategies have been employed to prevent complement inactivation, including chemical and genetic envelope modifications. This study employed the streptococcal albumin-binding domain (ABD) to generate a construct to express ABD as a glycosylphosphatidylinositol-anchored protein. LV particles bearing ABD are able to bind bovine and human serum albumin in vitro. Neither the lentiviral vector production titer nor the in vitro transduction was affected by the ABD display. The study demonstrated that ABD-bearing LVs are protected from human complement inactivation. More importantly, intravenous administration demonstrated that the presence of ABD significantly reduces lentivector sequestration in liver and bone-marrow cells. Therefore, the use of ABD represents an improvement for in vivo gene therapy applications. The results strongly point to ABD display as a universal strategy to increase the in vivo efficacy of different viral vectors.


Subject(s)
Bacterial Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Glycoproteins/genetics , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Viral Load , Albumins/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bone Marrow/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , HEK293 Cells , Humans , Jurkat Cells , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Domains , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vesiculovirus/metabolism , Vesiculovirus/physiology , Viral Envelope Proteins/metabolism
10.
Biochim Biophys Acta Gene Regul Mech ; 1860(9): 922-935, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28736155

ABSTRACT

Two processes are associated with progressive loss of renal function: 1) decreased aquaporin-2 (AQP2) expression and urinary concentrating capacity (Nephrogenic Diabetes Insipidus, NDI); and 2) changes in extracellular matrix (ECM) composition, e.g. increased collagen I (Col I) deposition, characteristic of tubule-interstitial fibrosis. AQP2 expression is regulated by both the ECM-to-intracellular scaffold protein integrin-linked kinase (ILK) by NFATc/AP1 and other transcription factors. In the present work, we used in vivo and in vitro approaches to examine ILK participation in NFATc3/AP-1-mediated increases in AQP2 gene expression. Both NFATc3 knock-out mice and ILK conditional-knockdown mice (cKD-ILK) display symptoms of NDI (polyuria and reduced AQP2 expression). NFATc3 is upregulated in the renal medulla tubular cells of cKD-ILK mice but with reduced nuclear localization. Inner medullary collecting duct mIMCD3 cells were subjected to ILK depletion and transfected with reporter plasmids. Pharmacological activators or inhibitors determined the effect of ILK activity on NFATc/AP-1-dependent increases in transcription of AQP2. Finally, mIMCD3 cultured on Col I showed reduced activity of the ILK/GSK3ß/NFATc/AQP2 axis, suggesting this pathway is a potential target for therapeutic treatment of NDI.


Subject(s)
Aquaporin 2/genetics , NFATC Transcription Factors/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic/genetics , Animals , Cell Line , Diabetes Insipidus, Nephrogenic/genetics , Diabetes Insipidus, Nephrogenic/metabolism , Integrins/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Polyuria/genetics , Polyuria/metabolism , Transcription Factor AP-1/metabolism
11.
J Endocrinol ; 234(2): 115-128, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28490443

ABSTRACT

The development of insulin resistance is characterized by the impairment of glucose uptake mediated by glucose transporter 4 (GLUT4). Extracellular matrix changes are induced when the metabolic dysregulation is sustained. The present work was devoted to analyze the possible link between the extracellular-to-intracellular mediator integrin-linked kinase (ILK) and the peripheral tissue modification that leads to glucose homeostasis impairment. Mice with general depletion of ILK in adulthood (cKD-ILK) maintained in a chow diet exhibited increased glycemia and insulinemia concurrently with a reduction of the expression and membrane presence of GLUT4 in the insulin-sensitive peripheral tissues compared with their wild-type littermates (WT). Tolerance tests and insulin sensitivity indexes confirmed the insulin resistance in cKD-ILK, suggesting a similar stage to prediabetes in humans. Under randomly fed conditions, no differences between cKD-ILK and WT were observed in the expression of insulin receptor (IR-B) and its substrate IRS-1 expressions. The IR-B isoform phosphorylated at tyrosines 1150/1151 was increased, but the AKT phosphorylation in serine 473 was reduced in cKD-ILK tissues. Similarly, ILK-blocked myotubes reduced their GLUT4 promoter activity and GLUT4 expression levels. On the other hand, the glucose uptake capacity in response to exogenous insulin was impaired when ILK was blocked in vivo and in vitro, although IR/IRS/AKT phosphorylation states were increased but not different between groups. We conclude that ILK depletion modifies the transcription of GLUT4, which results in reduced peripheral insulin sensitivity and glucose uptake, suggesting ILK as a molecular target and a prognostic biomarker of insulin resistance.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin Resistance/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Glucose/metabolism , Glucose Transporter Type 4/genetics , Homeostasis/physiology , Hyperglycemia , Hyperinsulinism , Insulin/blood , Male , Mice , Muscle Fibers, Skeletal/metabolism , Protein Serine-Threonine Kinases/genetics
12.
Am J Physiol Renal Physiol ; 311(6): F1346-F1357, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27760768

ABSTRACT

Within the past decade tremendous efforts have been made to understand the mechanism behind aquaporin-2 (AQP2) water channel trafficking and recycling, to open a path toward effective diabetes insipidus therapeutics. A recent study has shown that integrin-linked kinase (ILK) conditional-knockdown mice developed polyuria along with decreased AQP2 expression. To understand whether ILK also regulates AQP2 trafficking in kidney tubular cells, we performed in vitro analysis using LLCPK1 cells stably expressing rat AQP2 (LLC-AQP2 cells). Upon treatment of LLC-AQP2 cells with ILK inhibitor cpd22 and ILK-siRNA, we observed increased accumulation of AQP2 in the perinuclear region, without any significant increase in the rate of endocytosis. This perinuclear accumulation did not occur in cells expressing a serine-256-aspartic acid mutation that retains AQP2 in the plasma membrane. We then examined clathrin-mediated endocytosis after ILK inhibition using rhodamine-conjugated transferrin. Despite no differences in overall transferrin endocytosis, the endocytosed transferrin also accumulated in the perinuclear region where it colocalized with AQP2. These accumulated vesicles also contained the recycling endosome marker Rab11. In parallel, the usual vasopressin-induced AQP2 membrane accumulation was prevented after ILK inhibition; however, ILK inhibition did not measurably affect AQP2 phosphorylation at serine-256 or its dephosphorylation at serine-261. Instead, we found that inhibition of ILK increased F-actin polymerization. When F-actin was depolymerized with latrunculin, the perinuclear located AQP2 dispersed. We conclude that ILK is important in orchestrating dynamic cytoskeletal architecture during recycling of AQP2, which is necessary for its subsequent entry into the exocytotic pathway.


Subject(s)
Aquaporin 2/metabolism , Cytoskeleton/metabolism , Exocytosis/physiology , Kidney/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cytoskeleton/drug effects , Endocytosis/drug effects , Endocytosis/physiology , Exocytosis/drug effects , Kidney/drug effects , Male , Mice , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics
14.
FASEB J ; 28(8): 3645-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24784577

ABSTRACT

One of the clinical alterations observed in chronic renal disease (CRD) is the impaired urine concentration, known as diabetes insipidus (DI). Tubulointerstitial fibrosis of the kidney is also a pathological finding observed in CRD and involves composition of extracellular matrix (ECM). However, an association between these two events has not been elucidated. In this study, we showed that the extracellular-to-intracellular scaffold protein integrin-linked kinase (ILK) regulates expression of tubular water channel aquaporin-2 (AQP2) and its apical membrane presence in the renal tubule. Basally, polyuria and decreased urine osmolality were present in ILK conditional-knockdown (cKD-ILK) adult mice compared with nondepleted ILK littermates. No changes were observed in arginine-vasopressin (AVP) blood levels, renal receptor (V2R), or AQP3 expression. However, tubular AQP2 was decreased in expression and apical membrane presence in cKD-ILK mice, where the canonical V2R/cAMP axis activation is still functional, but independent of the absence of ILK. Thus, cKD-ILK constitutes a nephrogenic diabetes insipidus (NDI) model. AQP2 and ILK colocalize in cultured inner medullary collecting duct (mIMCD3) cells. Specific ILK siRNAs and collagen I (Col) decrease ILK and AQP2 levels and AQP2 presence on the membrane of tubular mIMCD3 cells, which impairs the capacity of the cells to transport water under hypotonic stress. The present work points to ILK as a therapeutic target in NDI.


Subject(s)
Aquaporin 2/physiology , Body Water/metabolism , Extracellular Matrix Proteins/physiology , Kidney Concentrating Ability/physiology , Kidney Tubules, Collecting/metabolism , Polyuria/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Aquaporin 2/biosynthesis , Aquaporin 2/genetics , Aquaporin 3/biosynthesis , Aquaporin 3/genetics , Arginine Vasopressin/blood , Biological Transport, Active , Cell Membrane/chemistry , Cell Polarity , Cells, Cultured , Collagen Type I/pharmacology , Deamino Arginine Vasopressin/pharmacology , Diabetes Insipidus, Nephrogenic/metabolism , Disease Models, Animal , Kidney Tubules, Collecting/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , Osmotic Pressure/physiology , Phosphorylation , Polyuria/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Vasopressin/biosynthesis , Receptors, Vasopressin/genetics
15.
Cardiovasc Res ; 99(3): 535-44, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23715557

ABSTRACT

AIMS: Integrin-linked kinase (ILK) regulates proliferation, differentiation, cell adhesion, and motility in many cell types and has been related to cancer progression, fibrosis, and vascular diseases. We designed the present study to directly explore the effect of ILK deletion on the regulation of vascular tone through the soluble guanylate cyclase (sGC) /protein kinase G (PKG) pathway in healthy adult mice. METHODS AND RESULTS: Experiments were carried out using a tamoxifen-inducible CRE-LOX system to conditionally delete the ILK gene in adult mice. Mice lacking ILK expression (cKO) presented increased vascular content and increased activity of sGC and PKG, resulting in a more intense vasodilatory response to a single dose of a nitric oxide (NO) donor [sodium nitroprusside (SNP)] or PKG agonist [8-bromoguanosine 3',5'-cyclic monophosphate sodium salt (8-Br)]. Five minutes after SNP or 8-Br administration the reduction in the systolic arterial pressure was enhanced in cKO mice (SNP WT: -7.4 ± 1.2 mmHG; SNP cKO: -14.0 ± 2.5; 8-Br WT: -2.9 ± 1.5 mmHG; 8-Br cKO: -10.0 ± 3.4 mmHG). ILK deletion restored the vascular response to SNP after chronic oral nitrite administration. In addition, ILK deletion also increased hypotensive SNP effect in angiotensin II-treated animals, suggesting a role for ILK in basal and pathological states. CONCLUSION: Deletion of ILK in adult animals increased the vascular response to NO. These findings show, for the first time, a requirement for ILK in regulating sGC-PKG expression in vivo.


Subject(s)
Cyclic GMP/metabolism , Protein Serine-Threonine Kinases/deficiency , Vasodilation/physiology , Animals , Aorta/drug effects , Aorta/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Guanylate Cyclase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacology , Nitroprusside/administration & dosage , Nitroprusside/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Vasodilation/drug effects
16.
Int J Biochem Cell Biol ; 45(2): 232-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23084979

ABSTRACT

The circulating levels of heat shock proteins (HSP) are increased in cardiovascular diseases; however, the implication of this for the fibrotic process typical of such diseases remains unclear. HSP70 can interact with the vascular smooth muscle cells (SMC), the major producer of extracellular matrix (ECM) proteins, through the Toll-like receptors 4 (TLR4). The transforming growth factor type-ß1 (TGF-ß1) is a well known vascular pro-fibrotic cytokine that is regulated in part by AP-1-dependent transcriptional mechanisms. We hypothesized that extracellular HSP70 could interact with SMCs, inducing TGF-ß1 synthesis and subsequent changes in the vascular ECM. We demonstrate that extracellular HSP70 binds to human aorta SMC TLR4, which up-regulates the AP-1-dependent transcriptional activity of the TGF-ß1 promoter. This is achieved through the mitogen activated protein kinases JNK and ERK, as demonstrated by the use of specific blockers and the knockdown of TLR4 with specific small interfering RNAs. The TGF-ß1 upregulation increase the expression of the ECM proteins type I collagen and fibronectin. This novel observation may elucidate the mechanisms by which HSP70 contributes in the inflammation and fibrosis present in atherosclerosis and other fibrosis-related diseases.


Subject(s)
Extracellular Matrix/metabolism , HSP70 Heat-Shock Proteins/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta1/metabolism , Cells, Cultured , Collagen Type I/metabolism , Fibronectins/metabolism , Gene Expression , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Toll-Like Receptor 4/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/genetics , Up-Regulation
17.
Blood ; 119(19): 4565-76, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22446484

ABSTRACT

Membrane-anchored ephrinB2 and its receptor EphB4 are involved in the formation of blood and lymphatic vessels in normal and pathologic conditions. Eph/ephrin activation requires cell-cell interactions and leads to bidirectional signaling pathways in both ligand- and receptor-expressing cells. To investigate the functional consequences of blocking ephrinB2 activity, 2 highly specific human single-chain Fv (scFv) Ab fragments against ephrinB2 were generated and characterized. Both Ab fragments suppressed endothelial cell migration and tube formation in vitro in response to VEGF and provoked abnormal cell motility and actin cytoskeleton alterations in isolated endothelial cells. As only one of them (B11) competed for binding of ephrinB2 to EphB4, these data suggest an EphB-receptor-independent blocking mechanism. Anti-ephrinB2 therapy reduced VEGF-induced neovascularization in a mouse Matrigel plug assay. Moreover, systemic administration of ephrinB2-blocking Abs caused a drastic reduction in the number of blood and lymphatic vessels in xenografted mice and a concomitant reduction in tumor growth. Our results show for the first time that specific Ab-based ephrinB2 targeting may represent an effective therapeutic strategy to be used as an alternative or in combination with existing antiangiogenic drugs for treating patients with cancer and other angiogenesis-related diseases.


Subject(s)
Antibodies/pharmacology , Ephrin-B2/antagonists & inhibitors , Lymphangiogenesis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/metabolism , Antibodies/therapeutic use , Antibody Specificity , Down-Regulation/drug effects , Ephrin-B2/immunology , Ephrin-B2/metabolism , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunotherapy/methods , Lymphangiogenesis/physiology , Mice , Mice, Nude , Mice, SCID , Molecular Targeted Therapy , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Int J Biochem Cell Biol ; 44(6): 963-71, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22429882

ABSTRACT

The mechanisms involved in the continuous expression of constitutive genes are unclear. We hypothesize that steady state intracellular reactive oxygen species (ROS), which their levels are tightly maintained, could be regulating the expression of these constitutive genes in resting cells. We analyzed the regulation of an important constitutive gene, TGF-ß1, after decreasing intracellular ROS concentration in human mesangial cells. Decreased intracellular hydrogen peroxide by catalase addition reduced TGF-ß1 protein, mRNA expression and promoter activity. Furthermore, catalase decreased the basal activity of Activated Protein-1 (AP-1) that regulates TGF-ß1 promoter activity. This effect disappeared when AP-1 binding site was removed. Similar results were observed with another protein containing AP-1 binding sites in its promoter, such as eNOS, but it was not the case in other constitutive genes without any AP-1 binding site, as COX1 or PKG1. The pharmacological inhibition of the different ROS synthesis sources by blocking NADPH oxidase, the mitochondrial respiratory chain or xanthine oxidase, or the use of human fibroblasts with genetically deficient mitochondrial activity, induced a similar, significant reduction of steady state ROS concentration as the one observed with catalase. Moreover, there was decreased TGF-ß1 expression in all the cases excepting the xanthine oxidase blockade. These findings suggest a novel role for the steady state intracellular ROS concentration, where the compartmentalized, different systems involved in the intracellular ROS production, could be essential for the expression of constitutive AP1-dependent genes, as TGF-ß1.


Subject(s)
Gene Expression , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/physiology , Binding Sites , Catalase/metabolism , Cells, Cultured , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Promoter Regions, Genetic , Transforming Growth Factor beta1/genetics
19.
Am J Physiol Cell Physiol ; 301(2): C441-50, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21525433

ABSTRACT

Chronic hypoxia (CH) activates the Ca(2+)-dependent transcription factor nuclear factor of activated T cells isoform c3 (NFATc3) in mouse pulmonary arteries. However, the mechanism of this response has not been explored. Since we have demonstrated that NFATc3 is required for CH-induced pulmonary arterial remodeling, establishing how CH activates NFATc3 is physiologically significant. The goal of this study was to test the hypothesis that endothelin-1 (ET-1) contributes to CH-induced NFATc3 activation. We propose that this mechanism requires increased pulmonary arterial smooth muscle cell (PASMC) intracellular Ca(2+) concentration ([Ca(2+)](i)) and stimulation of RhoA/Rho kinase (ROK), leading to calcineurin activation and actin cytoskeleton polymerization, respectively. We found that: 1) CH increases pulmonary arterial pre-pro-ET-1 mRNA expression and lung RhoA activity; 2) inhibition of ET receptors, calcineurin, L-type Ca(2+) channels, and ROK blunts CH-induced NFATc3 activation in isolated intrapulmonary arteries from NFAT-luciferase reporter mice; and 3) both ET-1-induced NFATc3 activation in isolated mouse pulmonary arteries ex vivo and ET-1-induced NFATc3-green fluorescence protein nuclear import in human PASMC depend on ROK and actin polymerization. This study suggests that CH increases ET-1 expression, thereby elevating PASMC [Ca(2+)](i) and RhoA/ROK activity. As previously demonstrated, elevated [Ca(2+)](i) is required to activate calcineurin, which dephosphorylates NFATc3, allowing its nuclear import. Here, we demonstrate that ROK increases actin polymerization, thus providing structural support for NFATc3 nuclear transport.


Subject(s)
Endothelin-1/metabolism , Hypoxia/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NFATC Transcription Factors/metabolism , Pulmonary Artery/metabolism , Actins/metabolism , Active Transport, Cell Nucleus , Animals , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling , Cells, Cultured , Chronic Disease , Cytoskeleton/metabolism , Disease Models, Animal , Endothelin Receptor Antagonists , Endothelin-1/antagonists & inhibitors , Endothelin-1/genetics , Genes, Reporter , Humans , Hypoxia/genetics , Male , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NFATC Transcription Factors/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/drug effects , RNA, Messenger/metabolism , Receptors, Endothelin/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Transcriptional Activation , Transfection , Up-Regulation , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
20.
Nephron Extra ; 1(1): 124-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22470386

ABSTRACT

BACKGROUND/AIMS: We have previously shown that aquaporin-2 (AQP2) is down-regulated in the renal medulla of rats made hypertensive by chronic inhibition of nitric oxide synthase. It has been shown that AQP2 expression is regulated by the calcineurin/nuclear factor of activated T cells (NFATc). Nitric oxide (NO) regulates the activity of NFATc via c-Jun-N-terminal kinase 2 (JNK2). Therefore, we hypothesized that increases in NO enhance NFATc-mediated up-regulation of AQP2 promoter activity. METHODS: AQP2 mRNA and protein expression were detected in mouse renal papilla. AQP2 promoter luciferase reporter- and NFAT luciferase reporter-transfected MDCK cells were used to determine AQP2 promoter activity and NFATc activity, respectively. Cells were incubated with classic activators and inhibitors of NFATc and the NO pathway. RESULTS: Our results demonstrate that both Ca(2+) and NO have a synergistic effect resulting in an increase in AQP2 mRNA and protein in mouse papilla and activation of the AQP2 promoter in kidney-derived cells. In addition, NO enhances Ca(2+)-induced NFATc activation. The underlying mechanism involves increased NFATc nuclear import and decreased export via protein kinase G-mediated inhibition of JNK1/2. CONCLUSIONS: This is the first study defining novel regulatory roles for NO and NFATc in the control of AQP2, which is an important renal protein.

SELECTION OF CITATIONS
SEARCH DETAIL
...