Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 13(13): e2302943, 2024 05.
Article in English | MEDLINE | ID: mdl-38266310

ABSTRACT

Decellularization of discarded whole livers and their recellularization with patient-specific induced pluripotent stem cells (iPSCs) to develop a functional organ is a promising approach to increasing the donor pool. The effect of extracellular matrix (ECM) of marginal livers on iPSC-hepatocyte differentiation and function has not been shown. To test the effect of donor liver ECM age and steatosis, young and old, as well as no, low, and high steatosis livers, are decellularized. All livers are decellularized successfully. High steatosis livers have fat remaining on the ECM after decellularization. Old donor liver ECM induces lower marker expression in early differentiation stages, compared to young liver ECM, while this difference is closed at later stages and do not affect iPSC-hepatocyte function significantly. High steatosis levels of liver ECM lead to higher albumin mRNA expression and secretion while at later stages of differentiation expression of major cytochrome (CYP) 450 enzymes is highest in low steatosis liver ECM. Both primary human hepatocytes and iPSC-hepatocytes show an increase in fat metabolism marker expression with increasing steatosis levels most likely induced by excess fat remaining on the ECM. Overall, removal of excess fat from liver ECM may be needed for inducing proper hepatic function after recellularization.


Subject(s)
Cell Differentiation , Extracellular Matrix , Fatty Liver , Hepatocytes , Induced Pluripotent Stem Cells , Liver , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Fatty Liver/metabolism , Fatty Liver/pathology , Extracellular Matrix/metabolism , Hepatocytes/metabolism , Hepatocytes/cytology , Liver/metabolism , Liver/pathology , Adult , Tissue Donors , Middle Aged , Cells, Cultured , Age Factors , Tissue Scaffolds/chemistry
2.
J Cardiovasc Pharmacol ; 82(6): 445-457, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37643020

ABSTRACT

ABSTRACT: The progression of chronic kidney disease results from the accumulation of extracellular matrix leading to end-stage renal disease. We previously demonstrated that a broad-spectrum matrix metalloproteinase (MMP) inhibitor reduced renal injury in rat models of hypertension and diabetes. However, the isoforms and mechanisms involved are unclear. This study examined the role of MMP2 during the development of proteinuria and renal injury after induction of hypertension or diabetes in Dahl salt-sensitive (SS) and MMP2 knockout (KO) rats. Mean arterial pressure rose from 115 ± 2 to 145 ± 2 mm Hg and 116 ± 1 to 152 ± 3 mm Hg in MMP2 KO and SS rats fed a high-salt (8% NaCl) diet for 3 weeks. The degree of proteinuria, glomerular injury, renal fibrosis, and podocyte loss was lower in MMP2 KO rats than in SS rats. Blood glucose and HbA1c levels, and mean arterial pressure rose to the same extent in streptozotocin-treated SS and MMP2 KO rats. However, the degree of proteinuria, glomerulosclerosis, renal fibrosis, renal hypertrophy, glomerular permeability to albumin, and the renal expression of MMP2 and TGFß1 were significantly reduced in MMP2 KO rats. Glomerular filtration rate fell by 33% after 12 weeks of diabetes in streptozotocin-treated SS rats compared with time-control rats, but glomerular filtration rate only fell by 12% in MMP2 KO rats. These results indicate that activation of MMP2 plays an essential role in the pathogenesis of hypertensive and diabetic nephropathy and suggests that an MMP2 inhibitor might slow the progression of chronic kidney disease.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Hypertension , Renal Insufficiency, Chronic , Rats , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Matrix Metalloproteinase 2/metabolism , Streptozocin/metabolism , Rats, Inbred Dahl , Hypertension/metabolism , Kidney , Proteinuria/genetics , Proteinuria/metabolism , Renal Insufficiency, Chronic/complications , Fibrosis , Blood Pressure , Sodium Chloride, Dietary , Diabetes Mellitus/metabolism
3.
Bioact Mater ; 25: 415-429, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37056249

ABSTRACT

Despite decades of efforts, state-of-the-art synthetic burn dressings to treat partial-thickness burns are still far from ideal. Current dressings adhere to the wound and necessitate debridement. This work describes the first "supramolecular hybrid hydrogel (SHH)" burn dressing that is biocompatible, self-healable, and on-demand dissoluble for easy and trauma-free removal, prepared by a simple, fast, and scalable method. These SHHs leverage the interactions of a custom-designed cationic copolymer via host-guest chemistry with cucurbit[7]uril and electrostatic interactions with clay nanosheets coated with an anionic polymer to achieve enhanced mechanical properties and fast on-demand dissolution. The SHHs show high mechanical strength (>50 kPa), self-heal rapidly in ∼1 min, and dissolve quickly (4-6 min) using an amantadine hydrochloride (AH) solution that breaks the supramolecular interactions in the SHHs. Neither the SHHs nor the AH solution has any adverse effects on human dermal fibroblasts or epidermal keratinocytes in vitro. The SHHs also do not elicit any significant cytokine response in vitro. Furthermore, in vivo murine experiments show no immune or inflammatory cell infiltration in the subcutaneous tissue and no change in circulatory cytokines compared to sham controls. Thus, these SHHs present excellent burn dressing candidates to reduce the time of pain and time associated with dressing changes.

4.
Cell Mol Gastroenterol Hepatol ; 16(2): 243-261, 2023.
Article in English | MEDLINE | ID: mdl-37085137

ABSTRACT

BACKGROUND & AIMS: Alterations in mitochondrial morphology and function and increased oxidative stresses in hepatocytes are well established in nonalcoholic fatty liver disease (NAFLD). Patients can undergo lifestyle changes, especially in earlier NAFLD stages, to reverse disease-induced phenotypes on a gross level. Yet, little is known about whether mitochondrial function and injuries recover upon reversal. Thus, we elucidated this question and interplays between the cytoskeletal network and mitochondria in the development and reversal of steatosis. METHODS: We cultured primary human hepatocytes stably for 2 weeks and used free fatty acid supplementation to induce steatosis over 7 days and reversed steatosis by free fatty acid withdrawal over the next 7 days. We assessed cytoskeletal and mitochondrial morphologies using immunocytochemistry and confocal microscopy. We evaluated mitochondrial respiration and function via the Seahorse analyzer, in which we fully optimized reagent dosing specifically for human hepatocytes. RESULTS: During early steatosis, intracellular lipid droplets displaced microtubules altering mitochondrial distribution, and disrupted the F-actin network, leading to loss of bile canaliculi in steatotic hepatocytes. Basal mitochondrial respiration, maximum respiratory capacity, and resistance to H2O2-induced cell death also increased as an adaptative response. Upon reversal of steatosis, F-actin and bile canaliculi were restored in hepatocytes. Nevertheless, we observed an increase in elongated mitochondrial branches accompanied by decreases in α-tubulin expression, mitochondrial proton leak, and susceptibility to H2O2-induced cell death. CONCLUSIONS: Despite the restoration of cytoskeletons morphologically upon reversal of steatosis, the mitochondria in hepatocytes were impaired owing to early adaptative respiratory increase. Hepatocytes thus were highly predisposed to H2O2-induced cell death. These results indicate the persistence of potential health risks for recovering NAFLD patients.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Fatty Acids, Nonesterified/metabolism , Actins/metabolism , Hydrogen Peroxide/metabolism , Hepatocytes/metabolism , Mitochondria/metabolism , Cytoskeleton/metabolism , Microtubules/metabolism
5.
Physiol Genomics ; 54(2): 58-70, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34859687

ABSTRACT

Hypertension is a leading risk factor for stroke, heart disease, chronic kidney disease, vascular cognitive impairment, and Alzheimer's disease. Previous genetic studies have nominated hundreds of genes linked to hypertension, and renal and cognitive diseases. Some have been advanced as candidate genes by showing that they can alter blood pressure or renal and cerebral vascular function in knockout animals; however, final validation of the causal variants and underlying mechanisms has remained elusive. This review chronicles 40 years of work, from the initial identification of adducin (ADD) as an ACTIN-binding protein suggested to increase blood pressure in Milan hypertensive rats, to the discovery of a mutation in ADD1 as a candidate gene for hypertension in rats that were subsequently linked to hypertension in man. More recently, a recessive K572Q mutation in ADD3 was identified in Fawn-Hooded Hypertensive (FHH) and Milan Normotensive (MNS) rats that develop renal disease, which is absent in resistant strains. ADD3 dimerizes with ADD1 to form functional ADD protein. The mutation in ADD3 disrupts a critical ACTIN-binding site necessary for its interactions with actin and spectrin to regulate the cytoskeleton. Studies using Add3 KO and transgenic strains, as well as a genetic complementation study in FHH and MNS rats, confirmed that the K572Q mutation in ADD3 plays a causal role in altering the myogenic response and autoregulation of renal and cerebral blood flow, resulting in increased susceptibility to hypertension-induced renal disease and cerebral vascular and cognitive dysfunction.


Subject(s)
Calmodulin-Binding Proteins/genetics , Genetic Predisposition to Disease/genetics , Hypertension, Renal/genetics , Hypertension/genetics , Nephritis/genetics , Precision Medicine/methods , Animals , Blood Pressure/genetics , Cognitive Dysfunction/genetics , Disease Models, Animal , Homeostasis/genetics , Humans , Mutation , Precision Medicine/trends , Rats , Renal Circulation/genetics
6.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669830

ABSTRACT

Ischemic stroke is one of the most disabling diseases and a leading cause of death globally. Despite advances in medical care, the global burden of stroke continues to grow, as no effective treatments to limit or reverse ischemic injury to the brain are available. However, recent preclinical findings have revealed the potential role of transient receptor potential cation 6 (TRPC6) channels as endogenous protectors of neuronal tissue. Activating TRPC6 in various cerebral ischemia models has been found to prevent neuronal death, whereas blocking TRPC6 enhances sensitivity to ischemia. Evidence has shown that Ca2+ influx through TRPC6 activates the cAMP (adenosine 3',5'-cyclic monophosphate) response element-binding protein (CREB), an important transcription factor linked to neuronal survival. Additionally, TRPC6 activation may counter excitotoxic damage resulting from glutamate release by attenuating the activity of N-methyl-d-aspartate (NMDA) receptors of neurons by posttranslational means. Unresolved though, are the roles of TRPC6 channels in non-neuronal cells, such as astrocytes and endothelial cells. Moreover, TRPC6 channels may have detrimental effects on the blood-brain barrier, although their exact role in neurovascular coupling requires further investigation. This review discusses evidence-based cell-specific aspects of TRPC6 in the brain to assess the potential targets for ischemic stroke management.


Subject(s)
Ischemic Stroke/physiopathology , Ischemic Stroke/therapy , Neurovascular Coupling , TRPC6 Cation Channel/metabolism , Animals , Humans , Models, Biological , Neurons/metabolism , TRPC6 Cation Channel/chemistry
7.
J Pharmacol Exp Ther ; 377(1): 189-198, 2021 04.
Article in English | MEDLINE | ID: mdl-33414130

ABSTRACT

Previous studies identified a region on chromosome 1 associated with NG-nitro-L-arginine methyl ester (L-NAME) hypertension-induced renal disease in fawn-hooded hypertensive (FHH) rats. This region contains a mutant γ-adducin (Add3) gene that impairs renal blood flow (RBF) autoregulation, but its contribution to renal injury is unknown. The present study evaluated the hypothesis that knockout (KO) of Add3 impairs the renal vasoconstrictor response to the blockade of nitric oxide synthase and enhances hypertension-induced renal injury after chronic administration of L-NAME plus a high-salt diet. The acute hemodynamic effect of L-NAME and its chronic effects on hypertension and renal injury were compared in FHH 1Brown Norway (FHH 1BN) congenic rats (WT) expressing wild-type Add3 gene versus FHH 1BN Add3 KO rats. RBF was well autoregulated in WT rats but impaired in Add3 KO rats. Acute administration of L-NAME (10 mg/kg) raised mean arterial pressure (MAP) similarly in both strains, but RBF and glomerular filtration rate (GFR) fell by 38% in WT versus 15% in Add3 KO rats. MAP increased similarly in both strains after chronic administration of L-NAME and a high-salt diet; however, proteinuria and renal injury were greater in Add3 KO rats than in WT rats. Surprisingly, RBF, GFR, and glomerular capillary pressure were 41%, 82%, and 13% higher in L-NAME-treated Add3 KO rats than in WT rats. Hypertensive Add3 KO rats exhibited greater loss of podocytes and glomerular nephrin expression and increased interstitial fibrosis than in WT rats. These findings indicate that loss of ADD3 promotes L-NAME-induced renal injury by altering renal hemodynamics and enhancing the transmission of pressure to glomeruli. SIGNIFICANCE STATEMENT: A mutation in the γ-adducin (Add3) gene in fawn-hooded hypertensive rats that impairs autoregulation of renal blood flow is in a region of rat chromosome 1 homologous to a locus on human chromosome 10 associated with diabetic nephropathy. The present results indicate that loss of ADD3 enhanced NG-nitro-L-arginine methyl ester-induced hypertensive renal injury by altering the transmission of pressure to the glomerulus.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Hypertension, Renal/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Blood Pressure , Calmodulin-Binding Proteins/genetics , Enzyme Inhibitors/toxicity , Gene Deletion , Glomerular Filtration Rate , Homeostasis , Hypertension, Renal/etiology , Hypertension, Renal/physiopathology , Male , NG-Nitroarginine Methyl Ester/toxicity , Podocytes/drug effects , Podocytes/metabolism , Rats , Renal Circulation , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/physiopathology , Vasoconstriction
8.
Am J Physiol Renal Physiol ; 320(1): F97-F113, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33308016

ABSTRACT

We recently reported that the enhanced susceptibility to chronic kidney disease (CKD) in the fawn-hooded hypertensive (FHH) rat is caused, at least in part, by a mutation in γ-adducin (ADD3) that attenuates renal vascular function. The present study explored whether Add3 contributes to the modulation of podocyte structure and function using FHH and FHH.Add3 transgenic rats. The expression of ADD3 on the membrane of primary podocytes isolated from FHH was reduced compared with FHH.Add3 transgenic rats. We found that F-actin nets, which are typically localized in the lamellipodia, replaced unbranched stress fibers in conditionally immortalized mouse podocytes transfected with Add3 Dicer-substrate short interfering RNA (DsiRNA) and primary podocytes isolated from FHH rats. There were increased F/G-actin ratios and expression of the Arp2/3 complexes throughout FHH podocytes in association with reduced synaptopodin and RhoA but enhanced Rac1 and CDC42 expression in the renal cortex, glomeruli, and podocytes of FHH rats. The expression of nephrin at the slit diaphragm and the levels of focal adhesion proteins integrin-α3 and integrin-ß1 were decreased in the glomeruli of FHH rats. Cell migration was enhanced and adhesion was reduced in podocytes of FHH rats as well as in immortalized mouse podocytes transfected with Add3 DsiRNA. Mean arterial pressures were similar in FHH and FHH.Add3 transgenic rats at 16 wk of age; however, FHH rats exhibited enhanced proteinuria associated with podocyte foot process effacement. These results demonstrate that reduced ADD3 function in FHH rats alters baseline podocyte pathophysiology by rearrangement of the actin cytoskeleton at the onset of proteinuria in young animals.


Subject(s)
Actin Cytoskeleton/metabolism , Calmodulin-Binding Proteins/metabolism , Hypertension/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Renal Insufficiency, Chronic/metabolism , Actin Cytoskeleton/pathology , Animals , Arterial Pressure , Calmodulin-Binding Proteins/genetics , Cell Adhesion , Cell Line , Cell Movement , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Disease Progression , Focal Adhesions/metabolism , Focal Adhesions/pathology , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Integrins/metabolism , Male , Mice , Monomeric GTP-Binding Proteins/metabolism , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Rats, Inbred Strains , Rats, Transgenic , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Signal Transduction
9.
Am J Physiol Renal Physiol ; 319(4): F624-F635, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32830539

ABSTRACT

Recently, we reported a mutation in γ-adducin (ADD3) was associated with an impaired myogenic response of the afferent arteriole and hypertension-induced chronic kidney disease (CKD) in fawn hooded hypertensive (FHH) rats. However, the mechanisms by which altered renal blood flow (RBF) autoregulation promotes hypertension-induced renal injury remain to be determined. The present study compared the time course of changes in renal hemodynamics and the progression of CKD during the development of DOCA-salt hypertension in FHH 1BN congenic rats [wild-type (WT)] with an intact myogenic response versus FHH 1BNAdd3KO (Add3KO) rats, which have impaired myogenic response. RBF was well autoregulated in WT rats but not in Add3KO rats. Glomerular capillary pressure rose by 6 versus 14 mmHg in WT versus Add3KO rats when blood pressure increased from 100 to 150 mmHg. After 1 wk of hypertension, glomerular filtration rate increased by 38% and glomerular nephrin expression decreased by 20% in Add3KO rats. Neither were altered in WT rats. Proteinuria doubled in WT rats versus a sixfold increase in Add3KO rats. The degree of renal injury was greater in Add3KO than WT rats after 3 wk of hypertension. RBF, glomerular filtration rate, and glomerular capillary pressure were lower by 20%, 28%, and 19% in Add3KO rats than in WT rats, which was associated with glomerular matrix expansion and loss of capillary filtration area. The results indicated that impaired RBF autoregulation and eutrophic remodeling of preglomerular arterioles increase the transmission of pressure to glomeruli, which induces podocyte loss and accelerates the progression of CKD in hypertensive Add3KO rats.


Subject(s)
Blood Pressure , Glomerular Filtration Rate , Hypertension/complications , Kidney Glomerulus/blood supply , Proteinuria/etiology , Renal Circulation , Renal Insufficiency, Chronic/etiology , Animals , Arterioles/metabolism , Arterioles/physiopathology , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Desoxycorticosterone Acetate , Disease Models, Animal , Disease Progression , Homeostasis , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Male , Muscle Development , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/physiopathology , Rats, Transgenic , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Sodium Chloride, Dietary , Vascular Remodeling
10.
Geroscience ; 42(2): 547-561, 2020 04.
Article in English | MEDLINE | ID: mdl-32166556

ABSTRACT

Individuals with diabetes are more susceptible to cerebral vascular aging. However, the underlying mechanisms are not well elucidated. The present study examined whether the myogenic response of the middle cerebral artery (MCA) is impaired in diabetic rats due to high glucose (HG)-induced cerebral vascular smooth muscle cell (CVSMC) dysfunction, and whether this is associated with ATP depletion and changes in mitochondrial dynamics and membrane potential. The diameters of the MCA of diabetic rats increased to 135.3 ± 11.3% when perfusion pressure was increased from 40 to 180 mmHg, while it fell to 85.1 ± 3.1% in non-diabetic controls. The production of ROS and mitochondrial-derived superoxide were enhanced in cerebral arteries of diabetic rats. Levels of mitochondrial superoxide were significantly elevated in HG-treated primary CVSMCs, which was associated with decreased ATP production, mitochondrial respiration, and membrane potential. The expression of OPA1 was reduced, and MFF was elevated in HG-treated CVSMCs in association with fragmented mitochondria. Moreover, HG-treated CVSMCs displayed lower contractile and proliferation capabilities. These results demonstrate that imbalanced mitochondrial dynamics (increased fission and decreased fusion) and membrane depolarization contribute to ATP depletion in HG-treated CVSMCs, which promotes CVSMC dysfunction and may play an essential role in exacerbating the impaired myogenic response in the cerebral circulation in diabetes and accelerating vascular aging.


Subject(s)
Diabetes Mellitus, Experimental , Muscle, Smooth, Vascular , Vascular System Injuries , Animals , Cerebral Arteries , Diabetes Mellitus, Experimental/complications , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiopathology , Rats , Rats, Sprague-Dawley
11.
Int J Mol Sci ; 20(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514409

ABSTRACT

Hypertension is the most common modifiable risk factor for stroke, and understanding the underlying mechanisms of hypertension and hypertension-related stroke is crucial. 20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid (20-HETE), which plays an important role in vasoconstriction, autoregulation, endothelial dysfunction, angiogenesis, inflammation, and blood-brain barrier integrity, has been linked to hypertension and stroke. 20-HETE can promote hypertension by potentiating the vascular response to vasoconstrictors; it also can reduce blood pressure by inhibition of sodium transport in the kidney. The production of 20-HETE is elevated after the onset of both ischemic and hemorrhagic strokes; on the other hand, subjects with genetic variants in CYP4F2 and CYP4A11 that reduce 20-HETE production are more susceptible to stroke. This review summarizes recent genetic variants in CYP4F2, and CYP4A11 influencing 20-HETE production and discusses the role of 20-HETE in hypertension and the susceptibility to the onset, progression, and prognosis of ischemic and hemorrhagic strokes.


Subject(s)
Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/metabolism , Stroke/metabolism , Animals , Disease Progression , Humans , Models, Biological , Polymorphism, Genetic
12.
Physiol Rep ; 7(8): e14065, 2019 04.
Article in English | MEDLINE | ID: mdl-31008571

ABSTRACT

This study describes a modified technique to fill the renal vasculature with a silicon rubber (Microfil) compound and obtain morphologic information about the intrarenal distribution of capillary blood flow under a variety of conditions. Kidneys and cremaster muscles of rats were perfused in vivo with Microfil using a perfusion pressure equal to the animal's mean arterial pressure at body temperature. Microfil did not alter arteriolar diameter or the pattern of flow in the microcirculation of the cremaster muscle. The modified protocol reproducibly filled the renal vasculature, including; glomerular, peritubular, and vasa recta capillaries. We compared the filling of the renal circulation in control rats with that seen in animals subjected to maneuvers reported to alter the intrarenal distribution of blood flow. Infusion of angiotensin II, hypotension, volume expansion, and mannitol- or furosemide-induced diuresis redistributed flow between renal cortical and medullary capillaries. The advantage of the current technique is that it provides anatomical information regarding the number, diameter, and branching patterns of capillaries in the postglomerular circulation critical in determining the intrarenal distribution of cortical and medullary blood flow.


Subject(s)
Capillaries/diagnostic imaging , Kidney/diagnostic imaging , Microcirculation , Perfusion Imaging/methods , Renal Circulation , Abdominal Muscles/blood supply , Angiotensin II/pharmacology , Animals , Capillaries/physiology , Diuretics/pharmacology , Female , Furosemide/pharmacology , Kidney/blood supply , Kidney/drug effects , Mannitol/pharmacology , Rats , Rats, Wistar , Silicone Elastomers/pharmacokinetics , Vasoconstrictor Agents/pharmacology
13.
Am J Physiol Renal Physiol ; 315(6): F1843-F1854, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30207168

ABSTRACT

The incidence and severity of acute kidney injury is increased in patients with diabetes and with aging. However, the mechanisms involved have not been clearly established. The present study examined the effects of aging and diabetes on the severity of renal ischemia-reperfusion (IR) injury in Sprague-Dawley (SD) and type 2 diabetic (T2DN) rats. T2DN rats develop diabetes at 3 mo of age and progressive proteinuria and diabetic nephropathy as they age from 6 to 18 mo. Plasma creatinine levels after bilateral IR were significantly higher (3.4 ± 0.1 mg/dl) in 18-mo-old elderly T2DN rats than in middle-aged (12 mo) T2DN rats with less severe diabetic nephropathy or young (3 mo) and elderly (18 mo) control SD rats (1.5 ± 0.2, 1.8 ± 0.1, and 1.7 ± 0.1 mg/dl, respectively). Elderly T2DN rats exhibited a greater fall in medullary blood flow 2 h following renal IR and a more severe and prolonged decline in glomerular filtration rate than middle-aged T2DN and young or elderly SD rats. The basal expression of the adhesion molecules ICAM-1 and E-selectin and the number of infiltrating immune cells was higher in the kidney of elderly T2DN than age-matched SD rats or young and middle-aged T2DN rats before renal IR. These results indicate that elderly T2DN rats with diabetic nephropathy are more susceptible to renal IR injury than diabetic animals with mild injury or age-matched control animals. This is associated with increased expression of ICAM-1, E-selectin and immune cell infiltration, renal medullary vasocongestion, and more prolonged renal medullary ischemia.


Subject(s)
Acute Kidney Injury/etiology , Aging , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Kidney/blood supply , Renal Circulation , Reperfusion Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Age Factors , Aging/metabolism , Aging/pathology , Animals , Blood Glucose/metabolism , Blood Pressure , Creatinine/blood , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Glomerular Filtration Rate , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , Male , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...