Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Brain ; 144(12): 3788-3807, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34972207

ABSTRACT

Pioglitazone, an FDA-approved compound, has been shown to target the novel mitochondrial protein mitoNEET and produce short-term neuroprotection and functional benefits following traumatic brain injury. To expand on these findings, we now investigate the dose- and time-dependent effects of pioglitazone administration on mitochondrial function after experimental traumatic brain injury. We then hypothesize that optimal pioglitazone dosing will lead to ongoing neuroprotection and cognitive benefits that are dependent on pioglitazone-mitoNEET signalling pathways. We show that delayed intervention is significantly more effective than early intervention at improving acute mitochondrial bioenergetics in the brain after traumatic brain injury. In corroboration, we demonstrate that mitoNEET is more heavily expressed, especially near the cortical contusion, in the 18 h following traumatic brain injury. To explore whether these findings relate to ongoing pathological and behavioural outcomes, mice received controlled cortical impact followed by initiation of pioglitazone treatment at either 3 or 18 h post-injury. Mice with treatment initiation at 18 h post-injury exhibited significantly improved behaviour and tissue sparing compared to mice with pioglitazone initiated at 3 h post-injury. Further using mitoNEET knockout mice, we show that this therapeutic effect is dependent on mitoNEET. Finally, we demonstrate that delayed pioglitazone treatment improves serial motor and cognitive performance in conjunction with attenuated brain atrophy after traumatic brain injury. This study illustrates that mitoNEET is the critical target for delayed pioglitazone intervention after traumatic brain injury, mitochondrial-targeting is highly time-dependent after injury and there is an extended therapeutic window to effectively treat mitochondrial dysfunction after brain injury.


Subject(s)
Brain Injuries, Traumatic , Iron-Binding Proteins/drug effects , Membrane Proteins/drug effects , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Pioglitazone/pharmacology , Animals , Mice , Mice, Inbred C57BL
2.
Am J Physiol Renal Physiol ; 320(5): F789-F798, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33615888

ABSTRACT

Calcineurin inhibitors (CNIs) are vital immunosuppressive therapies in the management of inflammatory conditions. A long-term consequence is nephrotoxicity. In the kidneys, the primary, catalytic calcineurin (CnA) isoforms are CnAα and CnAß. Although the renal phenotype of CnAα-/- mice substantially mirrors CNI-induced nephrotoxicity, the mechanisms downstream of CnAα are poorly understood. Since NADPH oxidase-2 (Nox2)-derived oxidative damage has been implicated in CNI-induced nephrotoxicity, we hypothesized that CnAα inhibition drives Nox2 upregulation and promotes oxidative stress. To test the hypothesis, Nox2 regulation was investigated in kidneys from CnAα-/-, CnAß-/-, and wild-type (WT) littermate mice. To identify the downstream mediator of CnAα, nuclear factor of activated T cells (NFAT) and NF-κB regulation was examined. To test if Nox2 is transcriptionally regulated via a NF-κB pathway, CnAα-/- and WT renal fibroblasts were treated with the NF-κB inhibitor caffeic acid phenethyl ester. Our findings showed that cyclosporine A treatment induced Nox2 upregulation and oxidative stress. Furthermore, Nox2 upregulation and elevated ROS generation occurred only in CnAα-/- mice. In these mice, NF-κB but not NFAT activity was increased. In CnAα-/- renal fibroblasts, NF-κB inhibition prevented Nox2 upregulation and reactive oxygen species (ROS) generation. In conclusion, these findings indicate that 1) CnAα loss stimulates Nox2 upregulation, 2) NF-κB is a novel CnAα-regulated transcription factor, and 3) NF-κB mediates CnAα-induced Nox2 and ROS regulation. Our results demonstrate that CnAα plays a key role in Nox2 and ROS generation. Furthermore, these novel findings provide evidence of divergent CnA isoform signaling pathways. Finally, this study advocates for CnAα-sparing CNIs, ultimately circumventing the CNI nephrotoxicity.NEW & NOTEWORTHY A long-term consequence of calcineurin inhibitors (CNIs) is oxidative damage and nephrotoxicity. This study indicates that NF-κB is a novel calcineurin-regulated transcription factor that is activated with calcineurin inhibition, thereby driving oxidative damage in CNI nephropathy. These findings provide additional evidence of divergent calcineurin signaling pathways and suggest that selective CNIs could improve the long-term outcomes of patients by mitigating renal side effects.


Subject(s)
Calcineurin Inhibitors/toxicity , Calcineurin/metabolism , Cyclosporine/toxicity , Immunosuppressive Agents/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , NADPH Oxidase 2/metabolism , NF-kappa B/metabolism , Animals , Calcineurin/deficiency , Calcineurin/genetics , Cell Line , Fibrosis , Kidney/enzymology , Kidney/pathology , Kidney Diseases/enzymology , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Up-Regulation
3.
Neurobiol Dis ; 140: 104866, 2020 07.
Article in English | MEDLINE | ID: mdl-32289370

ABSTRACT

Traumatic brain injury (TBI) leads to acute necrosis at the site of injury followed by a sequence of secondary events lasting from hours to weeks and often years. Targeting mitochondrial impairment following TBI has shown improvements in brain mitochondrial bioenergetics and neuronal function. Recently formoterol, a highly selective ß2-adrenoreceptor agonist, was found to induce mitochondrial biogenesis (MB) via Gßγ-Akt-eNOS-sGC pathway. Activation of MB is a novel approach that has been shown to restore mitochondrial function in several disease and injury models. We hypothesized that activation of MB as a target of formoterol after TBI would mitigate mitochondrial dysfunction, enhance neuronal function and improve behavioral outcomes. TBI-injured C57BL/6 male mice were injected (i.p.) with vehicle (normal saline) or formoterol (0.3 mg/kg) at 15 min, 8 h, 16 h, 24 h and then daily after controlled cortical impact (CCI) until euthanasia. After CCI, mitochondrial copy number and bioenergetic function were decreased in the ipsilateral cortex of the CCI-vehicle group. Compared to CCI-vehicle, cortical and hippocampal mitochondrial respiration rates as well as cortical mitochondrial DNA copy number were increased in the CCI-formoterol group. Mitochondrial Ca2+ buffering capacity in the hippocampus was higher in the CCI-formoterol group compared to CCI-vehicle group. Both assessments of cognitive performance, novel object recognition (NOR) and Morris water maze (MWM), decreased following CCI and were restored in the CCI-formoterol group. Although no changes were seen in the amount of cortical tissue spared between CCI-formoterol and CCI-vehicle groups, elevated levels of hippocampal neurons and improved white matter sparing in the corpus callosum were observed in CCI-formoterol group. Collectively, these results indicate that formoterol-mediated MB activation may be a potential therapeutic target to restore mitochondrial bioenergetics and promote functional recovery after TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Cognition/drug effects , Formoterol Fumarate/pharmacology , Mitochondria/drug effects , Recovery of Function/drug effects , Animals , Disease Models, Animal , Formoterol Fumarate/therapeutic use , Hippocampus/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Organelle Biogenesis , White Matter/drug effects
4.
Anesth Analg ; 129(1): 192-203, 2019 07.
Article in English | MEDLINE | ID: mdl-31082969

ABSTRACT

BACKGROUND: Clinical studies implicate the perioperative period in cognitive complications, and increasing experimental evidence shows that the anesthetic agents can affect neuronal processes that underpin learning and memory. Calcineurin, a Ca-dependent phosphatase critically involved in synaptic plasticity, is activated after isoflurane exposure, but its role in the neurological response to anesthesia is unclear. METHODS: We investigated the effect of chronic calcineurin inhibition on postanesthetic cognitive function. Mice were treated with 30 minutes of isoflurane anesthesia during a chronic cyclosporine A regimen. Behavioral end points during the perianesthesia period were quantified. Visuospatial learning was assessed with the water radial arm maze. Total and biotinylated surface protein expression of the α5ß3γ2 γ-aminobutyric acid (GABA) type A receptors was measured. Expression of the GABA synthesis enzyme glutamate decarboxylase (GAD)-67 was also measured. RESULTS: Mice treated with cyclosporine A before anesthesia showed significant deficits in visuospatial learning compared to sham and cyclosporine A-treated mice (n = 10 per group, P = .0152, Tukey post hoc test). Induction and emergence were unaltered by cyclosporine A. Analysis of hippocampal protein expression revealed an increased surface expression of the α5 GABA type A receptor subunit after isoflurane treatment (P = .019, Dunnett post hoc testing), as well as a decrease in GAD-67 expression. Cyclosporine A did not rescue either effect. CONCLUSIONS: Our results confirm the work of others that isoflurane induces changes to inhibitory network function and exclude calcineurin inhibition via cyclosporine A as an intervention. Further, our studies suggest that calcineurin mediates a protective role in the neurological response to anesthesia, and patients receiving cyclosporine A may be an at-risk group for memory problems related to anesthesia.


Subject(s)
Anesthetics, Inhalation/toxicity , Behavior, Animal/drug effects , Calcineurin Inhibitors/toxicity , Cyclosporine/toxicity , Hippocampus/drug effects , Isoflurane/toxicity , Memory/drug effects , Spatial Behavior/drug effects , Visual Perception/drug effects , Animals , Cognition/drug effects , Glutamate Decarboxylase/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice, Inbred C57BL , Receptors, GABA-A/metabolism , Time Factors
5.
Am J Physiol Renal Physiol ; 316(4): F646-F653, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30649891

ABSTRACT

Zn2+ deficiency (ZnD) is a common comorbidity of many chronic diseases. In these settings, ZnD exacerbates hypertension. Whether ZnD alone is sufficient to alter blood pressure (BP) is unknown. To explore the role of Zn2+ in BP regulation, adult mice were fed a Zn2+-adequate (ZnA) or a Zn2+-deficient (ZnD) diet. A subset of ZnD mice were either returned to the ZnA diet or treated with hydrochlorothiazide (HCTZ), a Na+-Cl- cotransporter (NCC) inhibitor. To reduce intracellular Zn2+ in vitro, mouse distal convoluted tubule cells were cultured in N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN, a Zn2+ chelator)- or vehicle (DMSO)-containing medium. To replete intracellular Zn2+, TPEN-exposed cells were then cultured in Zn2+-supplemented medium. ZnD promoted a biphasic BP response, characterized by episodes of high BP. BP increases were accompanied by reduced renal Na+ excretion and NCC upregulation. These effects were reversed in Zn2+-replete mice. Likewise, HCTZ stimulated natriuresis and reversed BP increases. In vitro, Zn2+ depletion increased NCC expression. Furthermore, TPEN promoted NCC surface localization and Na+ uptake activity. Zn2+ repletion reversed TPEN effects on NCC. These data indicate that 1) Zn2+ contributes to BP regulation via modulation of renal Na+ transport, 2) renal NCC mediates ZnD-induced hypertension, and 3) NCC is a Zn2+-regulated transporter that is upregulated with ZnD. This study links dysregulated renal Na+ handling to ZnD-induced hypertension. Furthermore, NCC is identified as a novel mechanism by which Zn2+ regulates BP. Understanding the mechanisms of ZnD-induced BP dysregulation may have an important therapeutic impact on hypertension.


Subject(s)
Hypertension/metabolism , Kidney/metabolism , Sodium/metabolism , Zinc/deficiency , Animals , Blood Pressure/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Diet , Ethylenediamines/pharmacology , Hydrochlorothiazide/pharmacology , Hypertension/etiology , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Mice , Mice, Inbred C57BL , Natriuresis/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology
6.
PLoS One ; 12(4): e0175242, 2017.
Article in English | MEDLINE | ID: mdl-28414804

ABSTRACT

Calcineurin inhibitors are powerful immunosuppressants that revolutionized organ transplantation. However, non-immune effects of the calcineurin inhibitor, such as cyclosporine A (CsA), have significantly hindered their use. Specifically, nephrotoxicity, which is associated with tubulointerstitial fibrosis, inflammation, and podocyte damage, affects up to half of all transplant patients. Calcineurin is involved in many aspects of kidney development and function; therefore, mechanisms of CsA-induced nephrotoxicity are complex and not yet fully understood. MicroRNAs are short non-coding RNAs that regulate protein-coding RNA expression through post-translational repression of target messenger RNAs. MicroRNA dysregulation is known to be involved in kidney diseases including fibrosis. In this study, we compared the renal microRNA expression profiles between mice that received CsA (20 mg/kg) or vehicle daily for six weeks. The results demonstrate that CsA induces significant changes in renal microRNA expression profile. We used combined criteria of False Discovery Rate (≤0.1), fold change (≥2) and median signal strength (≥50) and identified 76 differencially expressed microRNAs. This approach identified microRNAs previously linked to renal fibrosis that includes let-7d, miR-21, miR-29, miR-30, miR-130, miR-192, and miR-200 as well as microRNAs that have not been reported to be related to nephrotoxicity or immunosuppression. Pathway analysis of microRNA/mRNA changes highlights the Wnt, TGF-ß, mTOR, and VEGF pathways. The mRNA expression profiles were compared in the same samples. The change of mRNA and microRNA profiles showed close correlations. To validate that the observed microRNA and mRNA expression level changes in mice kidney tissue were directly related to CsA treatment, the expression change induced by CsA treatment of three microRNAs (miR-21, miR-186, and miR-709) and three mRNAs (BMPR1a, SMURF1 and SMAD7) were compared in HEK293 cell line. A similar trend of expression level change was induced by CsA treatment in all selected microRNAs and mRNAs in the in vitro cell model. These data provide a roadmap for future work to study the role of the known and novel candidate microRNAs in the mechanism of nephrotoxicity and their further therapeutic potential.


Subject(s)
Calcineurin Inhibitors/toxicity , Cyclosporine/toxicity , Kidney/drug effects , Kidney/metabolism , MicroRNAs/genetics , Animals , Gene Regulatory Networks/drug effects , HEK293 Cells , Humans , Immunosuppressive Agents/toxicity , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Transcriptome
7.
BMC Health Serv Res ; 17(1): 51, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28103930

ABSTRACT

BACKGROUND: Depression is a prevalent mental health disorder and the fourth leading cause of disability in the world as per the World Health Organization. Use of antidepressants can lead to adverse drug events (ADEs), defined as any injury resulting from medication use. This study aimed to examine changes in hospital admissions due to antidepressant-related ADEs (ArADEs) among different socio-demographic groups and changes in lengths of stay (LOS) and hospital charges in ArADE admissions from 2001 to 2011. METHODS: The Healthcare Cost and Utilization Project database was used. ArADE admissions in different socio-demographic groups were examined including characteristics such as age, gender, rural/urban, and income. LOS and hospital charges for ArADE cases were compared between 2001 and 2011. Chi-square test and t test were used for statistical analyses. RESULTS: There were 17,375 and 20,588 ArADE related admissions in 2001 and 2011, respectively. There was a 17.6% increase among the group of 18 to 64 years old and a 64.8% increase among the group of 65 years or older while the other age groups experienced decreased admission rates. Males and females had similar increases. Patients from the lower income areas experienced a two-fold increase while those from the higher income areas experienced a decrease. The mean LOS for all ArADE related admissions increased from 2.18 to 2.81 days and mean hospital charges increased from $8,456.2 to $21,572.5. CONCLUSIONS: There was an increase in ArADE hospital admissions. The greater increase in ArADE admissions among elderly, urban or low-income patients should be noted and addressed by practitioners and policy makers. The large increase in hospital charges needs further research.


Subject(s)
Antidepressive Agents/administration & dosage , Depressive Disorder/drug therapy , Hospitalization/trends , Adolescent , Adult , Aged , Antidepressive Agents/economics , Depressive Disorder/economics , Depressive Disorder/epidemiology , Drug-Related Side Effects and Adverse Reactions/economics , Drug-Related Side Effects and Adverse Reactions/epidemiology , Female , Health Care Costs , Hospital Charges/statistics & numerical data , Hospitalization/statistics & numerical data , Hospitals/statistics & numerical data , Humans , Length of Stay/statistics & numerical data , Male , Medication Errors , Middle Aged , Poverty , Retrospective Studies , Young Adult
8.
Am J Physiol Cell Physiol ; 312(1): C47-C55, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27806940

ABSTRACT

Zn2+ deficiency (ZnD) is comorbid with chronic kidney disease and worsens kidney complications. Oxidative stress is implicated in the detrimental effects of ZnD. However, the sources of oxidative stress continue to be identified. Since NADPH oxidases (Nox) are the primary enzymes that contribute to renal reactive oxygen species generation, this study's objective was to determine the role of these enzymes in ZnD-induced oxidative stress. We hypothesized that ZnD promotes NADPH oxidase upregulation, resulting in oxidative stress and kidney damage. To test this hypothesis, wild-type mice were pair-fed a ZnD or Zn2+-adequate diet. To further investigate the effects of Zn2+ bioavailability on NADPH oxidase regulation, mouse tubular epithelial cells were exposed to the Zn2+ chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) or vehicle followed by Zn2+ supplementation. We found that ZnD diet-fed mice develop microalbuminuria, electrolyte imbalance, and whole kidney hypertrophy. These markers of kidney damage are accompanied by elevated Nox2 expression and H2O2 levels. In mouse tubular epithelial cells, TPEN-induced ZnD stimulates H2O2 generation. In this in vitro model of ZnD, enhanced H2O2 generation is prevented by NADPH oxidase inhibition with diphenyleneiodonium. Specifically, TPEN promotes Nox2 expression and activation, which are reversed when intracellular Zn2+ levels are restored following Zn2+ supplementation. Finally, Nox2 knockdown by siRNA prevents TPEN-induced H2O2 generation and cellular hypertrophy in vitro. Together, these findings reveal that Nox2 is a Zn2+-regulated enzyme that mediates ZnD-induced oxidative stress and kidney hypertrophy. Understanding the specific mechanisms by which ZnD contributes to kidney damage may have an important impact on the treatment of chronic kidney disease.


Subject(s)
Kidney/enzymology , NADPH Oxidases/metabolism , Oxidative Stress , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/pathology , Zinc/deficiency , Animals , Female , Kidney/pathology , Male , Mice , Zinc/metabolism
9.
Neuroscience ; 341: 42-51, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27890830

ABSTRACT

Vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia behind Alzheimer's disease (AD) and is a frequent co-morbidity with AD. Despite its prevalence, little is known about the molecular mechanisms underlying the cognitive dysfunction resulting from cerebrovascular disease. Astrocytic end-feet almost completely surround intraparenchymal blood vessels in the brain and express a variety of channels and markers indicative of their specialized functions in the maintenance of ionic and osmotic homeostasis and gliovascular signaling. These functions are mediated by end-foot enrichment of the aquaporin 4 water channel (AQP4), the inward rectifying potassium channel Kir4.1 and the calcium-dependent potassium channel MaxiK. Using our hyperhomocysteinemia (HHcy) model of VCID we examined the time-course of astrocytic end-foot changes along with cognitive and neuroinflammatory outcomes. We found that there were significant astrocytic end-foot disruptions in the HHcy model. AQP4 becomes dislocalized from the end-feet, there is a loss of Kir4.1 and MaxiK protein expression, as well as a loss of the Dp71 protein known to anchor the Kir4.1, MaxiK and AQP4 channels to the end-foot membrane. Neuroinflammation occurs prior to the astrocytic changes, while cognitive impairment continues to decline with the exacerbation of the astrocytic changes. We have previously reported similar astrocytic changes in models of cerebral amyloid angiopathy (CAA) and therefore, we believe astrocytic end-foot disruption could represent a common cellular mechanism of VCID and may be a target for therapeutic development.


Subject(s)
Astrocytes/immunology , Astrocytes/pathology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Dementia, Vascular/immunology , Dementia, Vascular/pathology , Animals , Brain/blood supply , Brain/immunology , Brain/pathology , CD11b Antigen/metabolism , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Disease Progression , Dystrophin/metabolism , Gliosis/immunology , Gliosis/pathology , Hyperhomocysteinemia/immunology , Hyperhomocysteinemia/pathology , Interleukin-12 Subunit p35/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Maze Learning/physiology , Mice, Inbred C57BL , Potassium Channels, Inwardly Rectifying/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
10.
J Cell Mol Med ; 18(12): 2361-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25287476

ABSTRACT

Calcineurin is a calcium-dependent phosphatase that is involved in many cellular processes including hypertrophy. Inhibition or genetic loss of calcineurin blocks pathological cardiac hypertrophy and diabetic renal hypertrophy. However, calcineurin does not appear to be involved in physiological cardiac hypertrophy induced by exercise. The role of calcineurin in a compensatory, non-pathological model of renal hypertrophy has not been tested. Therefore, in this study, we examined activation of calcineurin and the effect of calcineurin inhibition or knockout on compensatory hypertrophy following uninephrectomy (UNX). UNX induces ~15% increase in the size of the remaining kidney; the data show no change in the generation of reactive oxygen species (ROS), Nox4 or transforming growth factor-ß expression confirming the model as one of compensatory hypertrophy. Next, analyses of the remaining kidney reveal that total calcineurin activity is increased, and, to a lesser extent, transcriptional activity of the calcineurin substrate nuclear factor of activated T cell is up-regulated following UNX. However, inhibition of calcineurin with cyclosporine failed to prevent compensatory renal hypertrophy. Likewise, hypertrophy was comparable to WT in mice lacking either isoform of the catalytic subunit of calcineurin (CnAα-/- or CnAß-/-). In conclusion, similar to its role in the heart, calcineurin is required for pathological but not compensatory renal hypertrophy. This separation of signalling pathways could therefore help further define key factors necessary for pathological hypertrophy including diabetic nephropathy.


Subject(s)
Calcineurin/metabolism , Kidney/metabolism , Kidney/surgery , Nephrectomy/methods , Animals , Blotting, Western , Calcineurin/genetics , Gene Expression , Hypertrophy/etiology , Kidney/pathology , Mice, Knockout , Nephrectomy/adverse effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Curr Opin Nephrol Hypertens ; 23(5): 473-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25036747

ABSTRACT

PURPOSE OF REVIEW: Research over the past decade has significantly deepened our understanding of mechanisms that drive the development of hypertension. In particular, a novel paradigm of inflammation as a common mediator of cardiovascular and kidney disease has emerged. This review will summarize the role of the immune system in cardiovascular disease, explore some of the most promising new therapeutic directions and consider their potential as new treatments for hypertension. RECENT FINDINGS: Recent data continue to demonstrate that targeting the immune system can prevent hypertension in a variety of experimental models. Tempering the enthusiasm for a long-awaited new approach to treating hypertension is decades of clinical data, showing that classic immunosuppression regimens are associated with significant side-effects - including cardiovascular disease - that effectively preclude their use in the setting of chronic hypertension. New, more specific therapies are being developed that target cytokines including IL-17, IL-6 and TNFα. SUMMARY: Preclinical data convincingly demonstrate a key role for the immune system and specific cytokine mediators. Several biotherapeutics targeting these pathways are on the market and more are in development. Side-effects, however, continue to resemble those of classic immunosuppressants, highlighting the challenge of translating these research advances into new therapies for hypertension. VIDEO ABSTRACT: http://links.lww.com/CONH/A9.


Subject(s)
Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Immune System/drug effects , Immunosuppressive Agents/therapeutic use , Animals , Antihypertensive Agents/adverse effects , Cytokines/metabolism , Drug Design , Humans , Hypertension/immunology , Hypertension/metabolism , Hypertension/physiopathology , Immune System/immunology , Immune System/metabolism , Immune System/physiopathology , Immunosuppressive Agents/adverse effects , Inflammation Mediators/metabolism , Signal Transduction/drug effects , Treatment Outcome
12.
Am J Physiol Cell Physiol ; 306(6): C551-8, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24336651

ABSTRACT

Skeletal muscle atrophy is prevalent in chronic diseases, and microRNAs (miRs) may play a key role in the wasting process. miR-23a was previously shown to inhibit the expression of atrogin-1 and muscle RING-finger protein-1 (MuRF1) in muscle. It also was reported to be regulated by cytoplasmic nuclear factor of activated T cells 3 (NFATc3) in cardiomyocytes. The objective of this study was to determine if miR-23a is regulated during muscle atrophy and to evaluate the relationship between calcineurin (Cn)/NFAT signaling and miR-23a expression in skeletal muscle cells during atrophy. miR-23a was decreased in the gastrocnemius of rats with acute streptozotocin-induced diabetes, a condition known to increase atrogin-1 and MuRF1 expression and cause atrophy. Treatment of C2C12 myotubes with dexamethasone (Dex) for 48 h also reduced miR-23a as well as RCAN1.4 mRNA, which is transcriptionally regulated by NFAT. NFATc3 nuclear localization and the amount of miR-23a decreased rapidly within 1 h of Dex administration, suggesting a link between Cn signaling and miR-23a. The level of miR-23a was lower in primary myotubes from mice lacking the α- or ß-isoform of the CnA catalytic subunit than wild-type mice. Dex did not further suppress miR-23a in myotubes from Cn-deficient mice. Overexpression of CnAß in C2C12 myotubes prevented Dex-induced suppression of miR-23a. Finally, miR-23a was present in exosomes isolated from the media of C2C12 myotubes, and Dex increased its exosomal abundance. Dex did not alter the number of exosomes released into the media. We conclude that atrophy-inducing conditions downregulate miR-23a in muscle by mechanisms involving attenuated Cn/NFAT signaling and selective packaging into exosomes.


Subject(s)
Calcineurin/metabolism , Diabetes Mellitus, Experimental/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , MicroRNAs/metabolism , Muscular Atrophy/metabolism , Animals , Biological Transport , Calcium-Binding Proteins , Cells, Cultured , Dexamethasone , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscular Atrophy/genetics , NFATC Transcription Factors/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction , Streptozocin
13.
Int J Neurosci ; 124(4): 236-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23931049

ABSTRACT

Although aging itself is not a disease, there are many comorbidities that become more common with aging. Heart disease, cancer, and other chronic illnesses are either more common or more severe in aging patients. Approximately 5.5 million people in the United States have Alzheimer's disease (AD), with the principal risk factor being age. It is estimated that the incidence of AD diagnosis doubles every 5 years after the age of 65. Therefore, as the population ages, the impact of AD on the healthcare landscape will increase. Understanding how to manage patients with AD is critical as we begin to care for more elderly patients in the perioperative period. In addition to their other health considerations, aging surgical patients are increasingly more likely to have pre-existing AD or be at risk for developing AD. There is growing interest to determine how anesthesia affects the development or progression of AD. Similarly, a best practice for the anesthetic management of patients with AD is not yet defined. Finally, the relationship between AD and susceptibility to or exacerbation of postoperative cognitive dysfunction (POCD) is not well understood. In this review, we will discuss both the clinical and the preclinical data related to anesthesia and AD, describe the overlapping pathophysiology of neurodegeneration and provide some insight into the anesthetic care of patients with AD.


Subject(s)
Alzheimer Disease/psychology , Anesthesia/adverse effects , Cognition Disorders/chemically induced , Cognition Disorders/physiopathology , Postoperative Complications/chemically induced , Postoperative Complications/psychology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cognition Disorders/complications , Cognition Disorders/pathology , Humans , Nerve Degeneration/chemically induced , Nerve Degeneration/complications , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Perioperative Period , Postoperative Complications/pathology , Postoperative Complications/physiopathology
14.
J Biol Chem ; 289(8): 4896-905, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24371139

ABSTRACT

Hypertrophy is an adaptive response that enables organs to appropriately meet increased functional demands. Previously, we reported that calcineurin (Cn) is required for glomerular and whole kidney hypertrophy in diabetic rodents (Gooch, J. L., Barnes, J. L., Garcia, S., and Abboud, H. E. (2003). Calcineurin is activated in diabetes and is required for glomerular hypertrophy and ECM accumulation. Am. J. Physiol. Renal Physiol. 284, F144-F154; Reddy, R. N., Knotts, T. L., Roberts, B. R., Molkentin, J. D., Price, S. R., and Gooch, J. L. (2011). Calcineurin Aß is required for hypertrophy but not matrix expansion in the diabetic kidney. J. Cell Mol. Med. 15, 414-422). Because studies have also implicated the reactive oxygen species-generating enzymes NADPH oxidases (Nox) in diabetic kidney responses, we tested the hypothesis that Nox and Cn cooperate in a common signaling pathway. First, we examined the role of the two main isoforms of Cn in hypertrophic signaling. Using primary kidney cells lacking a catalytic subunit of Cn (CnAα(-/-) or CnAß(-/-)), we found that high glucose selectively activates CnAß, whereas CnAα is constitutively active. Furthermore, CnAß but not CnAα mediates hypertrophy. Next, we found that chronic reactive oxygen species generation in response to high glucose is attenuated in CnAß(-/-) cells, suggesting that Cn is upstream of Nox. Consistent with this, loss of CnAß reduces basal expression and blocks high glucose induction of Nox2 and Nox4. Inhibition of nuclear factor of activated T cells (NFAT), a CnAß-regulated transcription factor, decreases Nox2 and Nox4 expression, whereas NFAT overexpression increases Nox2 and Nox4, indicating that the CnAß/NFAT pathway modulates Nox. These data reveal that the CnAß/NFAT pathway regulates Nox and plays an important role in high glucose-mediated hypertrophic responses in the kidney.


Subject(s)
Calcineurin/metabolism , Glucose/pharmacology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , NFATC Transcription Factors/metabolism , Animals , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Hypertrophy/pathology , Kidney/pathology , Mice , Models, Biological , NADPH Oxidase 2 , NADPH Oxidase 4 , NFATC Transcription Factors/genetics , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
15.
Transplantation ; 96(3): 239-44, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23823650

ABSTRACT

BACKGROUND: Calcineurin is a ubiquitously expressed calcium-dependent phosphatase that is inhibited by the immunosuppressant drugs cyclosporine and tacrolimus. Measuring calcineurin activity in transplant patients has been complicated by a lack of consistent correlation between drug level and enzyme activity, particularly with chronic use. Data from mice lacking the CnAα or CnAß isoform of the catalytic subunit of calcineurin demonstrate that loss of CnAß results in immunosuppression, whereas loss of CnAα does not. As such, methods to examine activity of the CnAß isoform may be more clinically relevant than nonspecific assays. METHODS: Because the current enzyme assays are nonisoform specific, we examined association of the CnAα or CnAß isoform with calmodulin (CaM), a shared binding protein that is only associated with the catalytic subunit in the presence of calcium. We then compared semiquantitated data with total calcineurin activity and immune status in a cohort of pretransplantation controls and postrenal transplantation patients. RESULTS: We found no difference in calcineurin activity between the groups and no difference in the amount of non-isoform-specific catalytic subunit bound to CaM. However, association of CnAα-CaM is increased in transplant patients, whereas CnAß-CaM is decreased. In addition, the amount of CnAß-CaM corresponds positively with T-cell activity and negatively with tacrolimus level. Finally, CnAß-CaM is lower in stable transplant patients compared with those with acute rejection. CONCLUSIONS: These data suggest that monitoring specifically the ß isoform may be more informative than non-isoform-specific assay methods.


Subject(s)
Calcineurin/physiology , Kidney Transplantation , Animals , Calcineurin Inhibitors , Female , Humans , Immunosuppressive Agents/pharmacology , Male , Mice , Middle Aged , Protein Isoforms
16.
PLoS One ; 8(4): e62503, 2013.
Article in English | MEDLINE | ID: mdl-23638102

ABSTRACT

Mice lacking the α isoform of the catalytic subunit of calcineurin (CnAα) were first reported in 1996 and have been an important model to understand the role of calcineurin in the brain, immune system, bones, muscle, and kidney. Research using the mice has been limited, however, by failure to thrive and early lethality of most null pups. Work in our laboratory led to the rescue of CnAα-/- mice by supplemental feeding to compensate for a defect in salivary enzyme secretion. The data revealed that, without intervention, knockout mice suffer from severe caloric restriction. Since nutritional deprivation is known to significantly alter development, it is imperative that previous conclusions based on CnAα-/- mice are revisited to determine which aspects of the phenotype were attributable to caloric restriction versus a direct role for CnAα. In this study, we find that defects in renal development and function persist in adult CnAα-/- mice including a significant decrease in glomerular filtration rate and an increase in blood urea nitrogen levels. These data indicate that impaired renal development we previously reported was not due to caloric restriction but rather a specific role for CnAα in renal development and function. In contrast, we find that rather than being hypoglycemic, rescued mice are mildly hyperglycemic and insulin resistant. Examination of muscle fiber types shows that previously reported reductions in type I muscle fibers are no longer evident in rescued null mice. Rather, loss of CnAα likely alters insulin response due to a reduction in insulin receptor substrate-2 (IRS2) expression and signaling in muscle. This study illustrates the importance of re-examining the phenotypes of CnAα-/- mice and the advances that are now possible with the use of adult, rescued knockout animals.


Subject(s)
Calcineurin/genetics , Caloric Restriction , Gene Deletion , Kidney/physiology , Muscle, Skeletal/metabolism , Animals , Calcineurin/analysis , Insulin/metabolism , Insulin Resistance , Kidney/metabolism , Kidney/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/ultrastructure , Muscle, Skeletal/ultrastructure , Phenotype , Signal Transduction
17.
Expert Rev Mol Med ; 14: e14, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22805659

ABSTRACT

Organ transplantation is the state of the art for treating end-stage organ failure. Over 25000 organ transplants are performed in the USA each year. Survival rates following transplantation are now approaching 90% for 1 year and 75% for 5 years. Central to this success was the introduction of drugs that suppress the immune system and prevent rejection. The most commonly used class of immunosuppressing drugs are calcineurin inhibitors (CNIs). Calcineurin is a ubiquitous enzyme that is important for T-cell function. With more people taking CNIs for longer and longer periods of time the consequences of calcineurin inhibition on other organ systems - particularly the kidney - have become a growing concern. Virtually all people who take a CNI will develop some degree of kidney toxicity and up to 10% will progress to kidney failure. In the past 15 years, research into calcineurin action has identified distinct actions of the two main isoforms of the catalytic subunit of the enzyme. The α-isoform is required for kidney function whereas the ß-isoform has a predominant role in the immune system. This review will discuss the current state of knowledge about calcineurin isoforms and how these new insights may reshape post-transplant immunosuppression.


Subject(s)
Calcineurin Inhibitors , Enzyme Inhibitors/adverse effects , Immunosuppressive Agents/adverse effects , Animals , Calcineurin/immunology , Calcineurin/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/immunology , Isoenzymes/metabolism , Kidney Transplantation/adverse effects
18.
Am J Pathol ; 178(4): 1605-13, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435446

ABSTRACT

Calcineurin is an important signal transduction mediator in T cells, neurons, the heart, and kidneys. Recent evidence points to unique actions of the two main isoforms of the catalytic subunit. Although the ß isoform is required for T-cell development, α is important in the brain and kidney. In addition, mice lacking α but not ß suffer from failure to thrive and early mortality. The purpose of this study was to identify the cause of postnatal death of calcineurin α null (CnAα(-/-)) mice and to determine the mechanism of α activity that contributes to the phenotype. CnAα(-/-) mice and wild-type littermate controls were fed a modified diet and then salivary gland function and histology were examined. In vitro studies were performed to identify the mechanism of α action. Data show that calcineurin is required for normal submandibular gland function and secretion of digestive enzymes. Loss of α does not impair nuclear factor of activated T-cell activity or expression but results in impaired protein trafficking downstream of the inositol trisphosphate receptor. These findings show a novel function of calcineurin in digestion and protein trafficking. Significantly, these data also provide a mechanism to rescue to adulthood a valuable animal model of calcineurin inhibitor-mediated neuronal and renal toxicities.


Subject(s)
Calcineurin/genetics , Calcineurin/physiology , Salivary Glands/metabolism , Animals , Brain/metabolism , Calcineurin/metabolism , Female , Immunohistochemistry/methods , Kidney/metabolism , Luciferases/metabolism , Male , Mice , Mice, Transgenic , Neurons/metabolism , Protein Isoforms , Protein Transport , Signal Transduction , Subcellular Fractions/metabolism
19.
J Cell Mol Med ; 15(2): 414-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19778355

ABSTRACT

Calcineurin is an important signalling protein that regulates a number of molecular and cellular processes. Previously, we found that inhibition of calcineurin with cyclosporine reduced renal hypertrophy and blocked glomerular matrix expansion in the diabetic kidney. Isoforms of the catalytic subunit of calcineurin are reported to have tissue specific expression and functions. In particular, the ß isoform has been implicated in cardiac and skeletal muscle hypertrophy. Therefore, we examined the role of calcineurin ß in diabetic renal hypertrophy and glomerular matrix expansion. Type I diabetes was induced in wild-type and ß(-/-) mice and then renal function, extracellular matrix expansion and hypertrophy were evaluated. The absence of ß produced a significant decrease in total calcineurin activity in the inner medulla (IM) and reduced nuclear factor of activated T-cells (NFATc) activity. Loss of ß did not alter diabetic renal dysfunction assessed by glomerular filtration rate, urine albumin excretion and blood urea nitrogen. Similarly, matrix expansion in the whole kidney and glomerulus was not different between diabetic wild-type and ß(-/-) mice. In contrast, whole kidney and glomerular hypertrophy were significantly reduced in diabetic ß(-/-) mice. Moreover, ß(-/-) renal fibroblasts demonstrated impaired phosphorylation of Erk1/Erk2, c-Jun N-terminal kinases (JNK) and mammalian target of rapamycin (mTOR) following stimulation with transforming growth factor-ß and did not undergo hypertrophy with 48 hrs culture in high glucose. In conclusion, loss of the ß isoform of calcineurin is sufficient to reproduce beneficial aspects of cyclosporine on diabetic renal hypertrophy but not matrix expansion. Therefore, while multiple signals appear to regulate matrix, calcineurin ß appears to be a central mechanism involved in organ hypertrophy.


Subject(s)
Calcineurin/metabolism , Diabetes Mellitus, Experimental/metabolism , Extracellular Matrix/metabolism , Kidney/metabolism , Kidney/pathology , NFATC Transcription Factors/metabolism , Albumins , Animals , Blood Urea Nitrogen , Cyclosporine/pharmacology , Diabetes Mellitus, Experimental/pathology , Glomerular Filtration Rate , Glucose/pharmacology , Hypertrophy , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , NFATC Transcription Factors/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/pharmacology
20.
J Ren Nutr ; 20(5 Suppl): S24-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20797566

ABSTRACT

Muscle atrophy is a significant consequence of chronic kidney disease that increases a patient's risk of mortality and decreases their quality of life. The loss of lean body mass results, in part, from an increase in the rate of muscle protein degradation. In this review, the proteolytic systems that are activated during chronic kidney disease and the key insulin signaling pathways that regulate the protein degradative processes are described.


Subject(s)
Insulin/physiology , Kidney Diseases/complications , Muscular Atrophy/etiology , Signal Transduction , Acidosis/complications , Animals , Chronic Disease , Glucocorticoids/biosynthesis , Humans , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Muscle Proteins/metabolism , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...