Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 14(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34494644

ABSTRACT

The current understanding of molecular mechanisms driving diabetic kidney disease (DKD) is limited, partly due to the complex structure of the kidney. To identify genes and signalling pathways involved in the progression of DKD, we compared kidney cortical versus glomerular transcriptome profiles in uninephrectomized (UNx) db/db mouse models of early-stage (UNx only) and advanced [UNxplus adeno-associated virus-mediated renin-1 overexpression (UNx-Renin)] DKD using RNAseq. Compared to normoglycemic db/m mice, db/db UNx and db/db UNx-Renin mice showed marked changes in their kidney cortical and glomerular gene expression profiles. UNx-Renin mice displayed more marked perturbations in gene components associated with the activation of the immune system and enhanced extracellular matrix remodelling, supporting histological hallmarks of progressive DKD in this model. Single-nucleus RNAseq enabled the linking of transcriptome profiles to specific kidney cell types. In conclusion, integration of RNAseq at the cortical, glomerular and single-nucleus level provides an enhanced resolution of molecular signalling pathways associated with disease progression in preclinical models of DKD, and may thus be advantageous for identifying novel therapeutic targets in DKD.


Subject(s)
Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Gene Expression Profiling , Hypertension/complications , Animals , Dependovirus/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice, Inbred C57BL , Renin/metabolism
2.
Physiol Rep ; 6(13): e13720, 2018 07.
Article in English | MEDLINE | ID: mdl-29984555

ABSTRACT

Chronic kidney disease (CKD) and uremia increase the risk of heart disease and sudden cardiac death. Coronary artery disease can only partly account for this. The remaining mechanistic links between CKD and sudden death are elusive, but may involve cardiac arrhythmias. For the present study, we hypothesized that a thorough electrophysiological study in mice with CKD would provide us valuable information that could aid in the identification of additional underlying causes of sudden cardiac death in patients with kidney disease. Partial (5/6) nephrectomy (NX) in mice induced mild CKD: plasma urea in NX was 24 ± 1 mmol/L (n = 23) versus 12 ± 1 mmol/L (n = 22) in sham-operated control mice (P < 0.05). Echocardiography did not identify structural or mechanical remodeling in NX mice. Baseline ECG parameters were comparable in conscious NX and control mice; however, the normal 24-h diurnal rhythm in QRS duration was lost in NX mice. Moreover, ß-adrenergic stimulation (isoprenaline, 200 µg/kg intraperitoneally) prolonged QRS duration in conscious NX mice (from 12 ± 1 to 15 ± 2 msec, P < 0.05), but not in sham-operated controls (from 13 ± 1 to 13 ± 2 msec, P > 0.05). No spontaneous arrhythmias were observed in conscious NX mice, and intracardiac pacing in anesthetized mice showed a comparable arrhythmia vulnerability in NX and sham-operated mice. Isoprenaline (2 mg/kg intraperitoneally) changed the duration of the QRS complex from 11.2 ± 0.4 to 11.9 ± 0.5 (P = 0.06) in NX mice and from 10.7 ± 0.6 to 10.6 ± 0.6 (P = 0.50) in sham-operated mice. Ex vivo measurements of cardiac ventricular conduction velocity were comparable in NX and sham mice. Transcriptional activity of Scn5a, Gja1 and several profibrotic genes was similar in NX and sham mice. We conclude that proper kidney function is necessary to maintain diurnal variation in QRS duration and that sympathetic regulation of the QRS duration is altered in kidney disease.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Heart Rate , Heart/drug effects , Isoproterenol/pharmacology , Renal Insufficiency, Chronic/physiopathology , Uremia/physiopathology , Animals , Heart/physiology , Male , Mice , Mice, Inbred C57BL
3.
Sci Rep ; 7(1): 6496, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747676

ABSTRACT

Atherosclerotic cardiovascular disease is a major complication of chronic kidney disease (CKD). CKD leads to uremia, which modulates the phenotype of aortic smooth muscle cells (SMCs). Phenotypic modulation of SMCs plays a key role in accelerating atherosclerosis. We investigated the hypothesis that uremia potentiates neointima formation in response to vascular injury in mice. Carotid wire injury was performed on C57BL/6 wt and apolipoprotein E knockout (Apoe -/-) mice two weeks after induction of uremia by 5/6 nephrectomy. Wire injury led to neointima formation and downregulation of genes encoding classical SMC markers (i.e., myocardin, α-smooth muscle actin, SM22-alpha, and smooth muscle myosin heavy chain) in both wt and Apoe -/- mice. Contrary to our expectations, uremia did not potentiate neointima formation, nor did it affect intimal lesion composition as judged from magnetic resonance imaging and histological analyses. Also, there was no effect of uremia on SMC marker gene expression in the injured carotid arteries, suggesting that there may be different effects of uremia on SMCs in different vascular beds. In conclusion, uremia does not accelerate neointima formation in response to wire injury of the carotid artery in mice.


Subject(s)
Atherosclerosis/pathology , Neointima , Uremia/complications , Animals , Apolipoproteins E/deficiency , Carotid Artery Injuries/pathology , Histocytochemistry , Magnetic Resonance Imaging , Mice, Inbred C57BL , Mice, Knockout , Muscle Cells , Renal Insufficiency/complications
4.
Eur J Pharmacol ; 816: 154-160, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-28351665

ABSTRACT

Chronic kidney disease affects as much as 13% of the population, and is associated with a markedly increased risk of developing cardiovascular disease. One of the underlying reasons is accelerated development of atherosclerosis. This can be ascribed both to increased occurrence of traditional cardiovascular risk factors, and to risk factors that may be unique to patients with chronic kidney disease. The latter is reflected in the observation that the current treatment modalities, mainly directed against traditional risk factors, are insufficient to prevent cardiovascular disease in the patient with chronic kidney disease. This review discusses mechanisms accelerating uremic atherosclerosis with a specific focus on the putative roles of apolipoprotein(apo)s B and M that may be particularly important in patients with chronic kidney disease.


Subject(s)
Apolipoproteins B/metabolism , Apolipoproteins M/metabolism , Atherosclerosis/etiology , Atherosclerosis/metabolism , Uremia/complications , Animals , Atherosclerosis/drug therapy , Humans , Molecular Targeted Therapy
5.
Atherosclerosis ; 257: 64-70, 2017 02.
Article in English | MEDLINE | ID: mdl-28107707

ABSTRACT

BACKGROUND AND AIMS: Chronic kidney disease leads to uremia and markedly accelerates atherosclerosis. Phenotypic modulation of smooth muscle cells (SMCs) in the arterial media plays a key role in accelerating atherogenesis. The aim of this study was to investigate whether uremia per se modulates the phenotype of aortic SMCs in vivo. METHODS: Moderate uremia was induced by 5/6 nephrectomy in apolipoprotein E knockout (ApoE-/-) and wildtype C57Bl/6 mice. Plasma analysis, gene expression, histology, and myography were used to determine uremia-mediated changes in the arterial wall. RESULTS: Induction of moderate uremia in ApoE-/- mice increased atherosclerosis in the aortic arch en face 1.6 fold (p = 0.04) and induced systemic inflammation. Based on histological analyses of aortic root sections, uremia increased the medial area, while there was no difference in the content of elastic fibers or collagen in the aortic media. In the aortic arch, mRNA and miRNA expression patterns were consistent with a uremia-mediated phenotypic modulation of SMCs; e.g. downregulation of myocardin, α-smooth muscle actin, and transgelin; and upregulation of miR146a. Notably, these expression patterns were observed after acute (2 weeks) and chronic (19 and 30 weeks) uremia, both under normo- and hypercholesterolemic settings. Functionally, aortic constriction was decreased in uremic as compared to non-uremic aorta segments, as measured by myography. CONCLUSIONS: Uremia modulates the phenotype of aortic SMCs as determined by mRNA/miRNA expression, an increased medial area, and decreased aortic contractility. We propose that this phenotypic modulation of SMCs precedes the acceleration of atherosclerosis observed in uremic mice.


Subject(s)
Aortic Diseases/etiology , Atherosclerosis/etiology , Inflammation/etiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Uremia/complications , Vasoconstriction , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Genetic Predisposition to Disease , Inflammation/blood , Inflammation/genetics , Inflammation/physiopathology , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uremia/blood
6.
Arterioscler Thromb Vasc Biol ; 36(9): 1782-90, 2016 09.
Article in English | MEDLINE | ID: mdl-27444197

ABSTRACT

OBJECTIVE: Atherosclerotic lesions contain hypoxic areas, but the pathophysiological importance of hypoxia is unknown. Hypoxia-inducible factor-1α (HIF-1α) is a key transcription factor in cellular responses to hypoxia. We investigated the hypothesis that HIF-1α has effects on macrophage biology that promotes atherogenesis in mice. APPROACH AND RESULTS: Studies with molecular probes, immunostaining, and laser microdissection of aortas revealed abundant hypoxic, HIF-1α-expressing macrophages in murine atherosclerotic lesions. To investigate the significance of macrophage HIF-1α, Ldlr(-/-) mice were transplanted with bone marrow from mice with HIF-1α deficiency in the myeloid cells or control bone marrow. The HIF-1α deficiency in myeloid cells reduced atherosclerosis in aorta of the Ldlr(-/-) recipient mice by ≈72% (P=0.006).In vitro, HIF-1α-deficient macrophages displayed decreased differentiation to proinflammatory M1 macrophages and reduced expression of inflammatory genes. HIF-1α deficiency also affected glucose uptake, apoptosis, and migratory abilities of the macrophages. CONCLUSIONS: HIF-1α expression in macrophages affects their intrinsic inflammatory profile and promotes development of atherosclerosis.


Subject(s)
Aorta, Thoracic/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Foam Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Transplantation , Cell Differentiation , Cell Hypoxia , Cell Movement , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Disease Progression , Foam Cells/pathology , Genetic Predisposition to Disease , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction
7.
BMC Dermatol ; 16(1): 9, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27401543

ABSTRACT

BACKGROUND: Risk of cardiovascular disease is increased in patients with psoriasis, but molecular mechanisms linking the two conditions have not been clearly established. Lack of appropriate animal models has hampered generation of new knowledge in this area of research and we therefore sought to develop an animal model with combined atherosclerosis and psoriasis-like skin inflammation. METHODS: Topical 12-O-tetradecanoylphorbol-13-acetate (TPA) was applied to the ears twice per week for 8 weeks in atherosclerosis-prone apolipoprotein E deficient (ApoE(-/-)) mice. RESULTS: TPA led to localized skin inflammation with increased epidermal thickness, infiltration of inflammatory-like cells and augmented tissue interleukin-17F levels. Systemic effects of the topical application of TPA were demonstrated by increased plasma concentration of serum amyloid A and splenic immune modulation, respectively. However, atherosclerotic plaque area and composition, and mRNA levels of several inflammatory genes in the aortic wall were not significantly affected by TPA-induced skin inflammation. CONCLUSIONS: TPA-induced psoriasis-like skin inflammation in atherosclerosis-prone ApoE(-/-) mice evoked systemic immune-inflammatory effects, but did not affect atherogenesis. The results may question the role of psoriasis-induced inflammation in the pathogenesis of atherosclerosis in psoriasis patients.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Carcinogens/pharmacology , Hypercholesterolemia , Inflammation , Psoriasis/chemically induced , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cytokines/metabolism , Disease Models, Animal , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Serum Amyloid A Protein/metabolism , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...