Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Clin Sci (Lond) ; 138(10): 573-597, 2024 May 22.
Article En | MEDLINE | ID: mdl-38718356

The three striatins (STRN, STRN3, STRN4) form the core of STRiatin-Interacting Phosphatase and Kinase (STRIPAK) complexes. These place protein phosphatase 2A (PP2A) in proximity to protein kinases thereby restraining kinase activity and regulating key cellular processes. Our aim was to establish if striatins play a significant role in cardiac remodelling associated with cardiac hypertrophy and heart failure. All striatins were expressed in control human hearts, with up-regulation of STRN and STRN3 in failing hearts. We used mice with global heterozygote gene deletion to assess the roles of STRN and STRN3 in cardiac remodelling induced by angiotensin II (AngII; 7 days). Using echocardiography, we detected no differences in baseline cardiac function or dimensions in STRN+/- or STRN3+/- male mice (8 weeks) compared with wild-type littermates. Heterozygous gene deletion did not affect cardiac function in mice treated with AngII, but the increase in left ventricle mass induced by AngII was inhibited in STRN+/- (but not STRN3+/-) mice. Histological staining indicated that cardiomyocyte hypertrophy was inhibited. To assess the role of STRN in cardiomyocytes, we converted the STRN knockout line for inducible cardiomyocyte-specific gene deletion. There was no effect of cardiomyocyte STRN knockout on cardiac function or dimensions, but the increase in left ventricle mass induced by AngII was inhibited. This resulted from inhibition of cardiomyocyte hypertrophy and cardiac fibrosis. The data indicate that cardiomyocyte striatin is required for early remodelling of the heart by AngII and identify the striatin-based STRIPAK system as a signalling paradigm in the development of pathological cardiac hypertrophy.


Angiotensin II , Cardiomegaly , Mice, Knockout , Myocytes, Cardiac , Animals , Angiotensin II/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Male , Humans , Muscle Proteins/metabolism , Muscle Proteins/genetics , Ventricular Remodeling , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Calmodulin-Binding Proteins , Nerve Tissue Proteins
2.
Kidney Int ; 103(6): 1105-1119, 2023 06.
Article En | MEDLINE | ID: mdl-37097268

Nuclear factor κB (NF-κB) activation is a deleterious molecular mechanism that drives acute kidney injury (AKI) and manifests in transplanted kidneys as delayed graft function. The TNFAIP3 gene encodes A20, a cytoplasmic ubiquitin ligase and a master negative regulator of the NF- κB signaling pathway. Common population-specific TNFAIP3 coding variants that reduce A20's enzyme function and increase NF- κB activation have been linked to heightened protective immunity and autoimmune disease, but have not been investigated in AKI. Here, we functionally identified a series of unique human TNFAIP3 coding variants linked to the autoimmune genome-wide association studies single nucleotide polymorphisms of F127C; namely F127C;R22Q, F127C;G281E, F127C;W448C and F127C;N449K that reduce A20's anti-inflammatory function in an NF- κB reporter assay. To investigate the impact of TNFAIP3 hypomorphic coding variants in AKI we tested a mouse Tnfaip3 hypomorph in a model of ischemia reperfusion injury (IRI). The mouse Tnfaip3 coding variant I325N increases NF- κB activation without overt inflammatory disease, providing an immune boost as I325N mice exhibit enhanced innate immunity to a bacterial challenge. Surprisingly, despite exhibiting increased intra-kidney NF- κB activation with inflammation in IRI, the kidney of I325N mice was protected. The I325N variant influenced the outcome of IRI by changing the dynamic expression of multiple cytoprotective mechanisms, particularly by increasing NF- κB-dependent anti-apoptotic factors BCL-2, BCL-XL, c-FLIP and A20, altering the active redox state of the kidney with a reduction of superoxide levels and the enzyme super oxide dismutase-1, and enhancing cellular protective mechanisms including increased Foxp3+ T cells. Thus, TNFAIP3 gene variants represent a kidney and population-specific molecular factor that can dictate the course of IRI.


Acute Kidney Injury , NF-kappa B , Humans , Mice , Animals , NF-kappa B/metabolism , Transcription Factors/genetics , Ubiquitin , Genome-Wide Association Study , Ligases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Acute Kidney Injury/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics
3.
Stroke ; 53(12): 3696-3705, 2022 12.
Article En | MEDLINE | ID: mdl-36205142

BACKGROUND: Cerebral small vessel disease (SVD) is common in older people and causes lacunar stroke and vascular cognitive impairment. Risk factors include old age, hypertension and variants in the genes COL4A1/COL4A2 encoding collagen alpha-1(IV) and alpha-2(IV), here termed collagen-IV, which are core components of the basement membrane. We tested the hypothesis that increased vascular collagen-IV associates with clinical hypertension and with SVD in older persons and with chronic hypertension in young and aged primates and genetically hypertensive rats. METHODS: We quantified vascular collagen-IV immunolabeling in small arteries in a cohort of older persons with minimal Alzheimer pathology (N=52; 21F/31M, age 82.8±6.95 years). We also studied archive tissue from young (age range 6.2-8.3 years) and older (17.0-22.7 years) primates (M mulatta) and compared chronically hypertensive animals (18 months aortic stenosis) with normotensives. We also compared genetically hypertensive and normotensive rats (aged 10-12 months). RESULTS: Collagen-IV immunolabeling in cerebral small arteries of older persons was negatively associated with radiological SVD severity (ρ: -0.427, P=0.005) but was not related to history of hypertension. General linear models confirmed the negative association of lower collagen-IV with radiological SVD (P<0.017), including age as a covariate and either clinical hypertension (P<0.030) or neuropathological SVD diagnosis (P<0.022) as fixed factors. Reduced vascular collagen-IV was accompanied by accumulation of fibrillar collagens (types I and III) as indicated by immunogold electron microscopy. In young and aged primates, brain collagen-IV was elevated in older normotensive relative to young normotensive animals (P=0.029) but was not associated with hypertension. Genetically hypertensive rats did not differ from normotensive rats in terms of arterial collagen-IV. CONCLUSIONS: Our cross-species data provide novel insight into sporadic SVD pathogenesis, supporting insufficient (rather than excessive) arterial collagen-IV in SVD, accompanied by matrix remodeling with elevated fibrillar collagen deposition. They also indicate that hypertension, a major risk factor for SVD, does not act by causing accumulation of brain vascular collagen-IV.


Cerebral Small Vessel Diseases , Hypertension , Stroke, Lacunar , Animals , Rats , Cerebral Small Vessel Diseases/complications , Stroke, Lacunar/complications , Hypertension/complications , Brain/pathology , Blood Pressure , Collagen Type IV/genetics
4.
Int J Mol Sci ; 23(14)2022 Jul 12.
Article En | MEDLINE | ID: mdl-35887055

Hypertension is a major public health concern and poses a significant risk for sudden cardiac death (SCD). However, the characterisation of human tissues tends to be macroscopic, with little appreciation for the quantification of the pathological remodelling responsible for the advancement of the disease. While the components of hypertensive remodelling are well established, the timeline and comparative quantification of pathological changes in hypertension have not been shown before. Here, we sought to identify the phasing of cardiac remodelling with hypertension using post-mortem tissue from SCD patients with early and advanced hypertensive heart disease (HHD). In order to study and quantify the progression of phenotypic changes, human specimens were contrasted to a well-described angiotensin-II-mediated hypertensive mouse model. While cardiomyocyte hypertrophy is an early adaptive response in the mouse that stabilises in established hypertension and declines as the disease progresses, this finding did not translate to the human setting. In contrast, optimising fibrosis quantification methods and applying them to each setting identified perivascular fibrosis as the prevailing possible cause for overall disease progression. Indeed, assessing myocardial inflammation highlights CD45+ inflammatory cell infiltration that precedes fibrosis and is an early-phase event in response to elevated arterial pressures that may underscore perivascular remodelling. Along with aetiology insight, we highlight cross-species comparison for quantification of cardiac remodelling in human hypertension. As such, this platform could assist with the development of therapies specific to the disease phase rather than targeting global components of hypertension, such as blood pressure lowering.


Hypertension , Ventricular Remodeling , Angiotensin II/physiology , Animals , Blood Pressure , Disease Models, Animal , Fibrosis , Heart , Humans , Mice , Myocardium/pathology , Myocytes, Cardiac/pathology
5.
Biochem J ; 479(13): 1467-1486, 2022 07 15.
Article En | MEDLINE | ID: mdl-35730579

The protein kinase PKN2 is required for embryonic development and PKN2 knockout mice die as a result of failure in the expansion of mesoderm, cardiac development and neural tube closure. In the adult, cardiomyocyte PKN2 and PKN1 (in combination) are required for cardiac adaptation to pressure-overload. The specific role of PKN2 in contractile cardiomyocytes during development and its role in the adult heart remain to be fully established. We used mice with cardiomyocyte-directed knockout of PKN2 or global PKN2 haploinsufficiency to assess cardiac development and function using high resolution episcopic microscopy, MRI, micro-CT and echocardiography. Biochemical and histological changes were also assessed. Cardiomyocyte-directed PKN2 knockout embryos displayed striking abnormalities in the compact myocardium, with frequent myocardial clefts and diverticula, ventricular septal defects and abnormal heart shape. The sub-Mendelian homozygous knockout survivors developed cardiac failure. RNASeq data showed up-regulation of PKN2 in patients with dilated cardiomyopathy, suggesting an involvement in adult heart disease. Given the rarity of homozygous survivors with cardiomyocyte-specific deletion of PKN2, the requirement for PKN2 in adult mice was explored using the constitutive heterozygous PKN2 knockout. Cardiac hypertrophy resulting from hypertension induced by angiotensin II was reduced in these haploinsufficient PKN2 mice relative to wild-type littermates, with suppression of cardiomyocyte hypertrophy and cardiac fibrosis. It is concluded that cardiomyocyte PKN2 is essential for heart development and the formation of compact myocardium and is also required for cardiac hypertrophy in hypertension. Thus, PKN signalling may offer therapeutic options for managing congenital and adult heart diseases.


Cardiomyopathies , Hypertension , Protein Kinase C/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Female , Hypertension/metabolism , Hypertension/pathology , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pregnancy
6.
Biochem J ; 479(3): 401-424, 2022 02 11.
Article En | MEDLINE | ID: mdl-35147166

The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade promotes cardiomyocyte hypertrophy and is cardioprotective, with the three RAF kinases forming a node for signal integration. Our aims were to determine if BRAF is relevant for human heart failure, whether BRAF promotes cardiomyocyte hypertrophy, and if Type 1 RAF inhibitors developed for cancer (that paradoxically activate ERK1/2 at low concentrations: the 'RAF paradox') may have the same effect. BRAF was up-regulated in heart samples from patients with heart failure compared with normal controls. We assessed the effects of activated BRAF in the heart using mice with tamoxifen-activated Cre for cardiomyocyte-specific knock-in of the activating V600E mutation into the endogenous gene. We used echocardiography to measure cardiac dimensions/function. Cardiomyocyte BRAFV600E induced cardiac hypertrophy within 10 d, resulting in increased ejection fraction and fractional shortening over 6 weeks. This was associated with increased cardiomyocyte size without significant fibrosis, consistent with compensated hypertrophy. The experimental Type 1 RAF inhibitor, SB590885, and/or encorafenib (a RAF inhibitor used clinically) increased ERK1/2 phosphorylation in cardiomyocytes, and promoted hypertrophy, consistent with a 'RAF paradox' effect. Both promoted cardiac hypertrophy in mouse hearts in vivo, with increased cardiomyocyte size and no overt fibrosis. In conclusion, BRAF potentially plays an important role in human failing hearts, activation of BRAF is sufficient to induce hypertrophy, and Type 1 RAF inhibitors promote hypertrophy via the 'RAF paradox'. Cardiac hypertrophy resulting from these interventions was not associated with pathological features, suggesting that Type 1 RAF inhibitors may be useful to boost cardiomyocyte function.


Cardiomegaly/pathology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins B-raf/physiology , Animals , Carbamates/pharmacology , Carbamates/toxicity , Cardiomegaly/metabolism , Cell Size/drug effects , Cells, Cultured , Dimerization , Gene Knock-In Techniques , Heart Failure/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Point Mutation , Protein Conformation/drug effects , Protein Interaction Mapping , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/biosynthesis , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Sulfonamides/toxicity
7.
Clin Sci (Lond) ; 135(14): 1631-1647, 2021 07 30.
Article En | MEDLINE | ID: mdl-34296750

Raf kinases signal via extracellular signal-regulated kinases 1/2 (ERK1/2) to drive cell division. Since activating mutations in BRAF (B-Raf proto-oncogene, serine/threonine kinase) are highly oncogenic, BRAF inhibitors including dabrafenib have been developed for cancer. Inhibitors of ERK1/2 signalling used for cancer are cardiotoxic in some patients, raising the question of whether dabrafenib is cardiotoxic. In the heart, ERK1/2 signalling promotes not only cardiomyocyte hypertrophy and is cardioprotective but also promotes fibrosis. Our hypothesis is that ERK1/2 signalling is not required in a non-stressed heart but is required for cardiac remodelling. Thus, dabrafenib may affect the heart in the context of, for example, hypertension. In experiments with cardiomyocytes, cardiac fibroblasts and perfused rat hearts, dabrafenib inhibited ERK1/2 signalling. We assessed the effects of dabrafenib (3 mg/kg/d) on male C57BL/6J mouse hearts in vivo. Dabrafenib alone had no overt effects on cardiac function/dimensions (assessed by echocardiography) or cardiac architecture. In mice treated with 0.8 mg/kg/d angiotensin II (AngII) to induce hypertension, dabrafenib inhibited ERK1/2 signalling and suppressed cardiac hypertrophy in both acute (up to 7 d) and chronic (28 d) settings, preserving ejection fraction. At the cellular level, dabrafenib inhibited AngII-induced cardiomyocyte hypertrophy, reduced expression of hypertrophic gene markers and almost completely eliminated the increase in cardiac fibrosis both in interstitial and perivascular regions. Dabrafenib is not overtly cardiotoxic. Moreover, it inhibits maladaptive hypertrophy resulting from AngII-induced hypertension. Thus, Raf is a potential therapeutic target for hypertensive heart disease and drugs such as dabrafenib, developed for cancer, may be used for this purpose.


Antineoplastic Agents/pharmacology , Fibrosis/drug therapy , Hypertension/drug therapy , Imidazoles/pharmacology , Oximes/pharmacology , Animals , Cardiomegaly/physiopathology , Disease Models, Animal , Hypertension/physiopathology , Mice, Inbred C57BL , Myocardium/pathology , Myocytes, Cardiac/metabolism , Rats , Signal Transduction/drug effects , Ventricular Remodeling/drug effects
8.
Biochem J ; 478(11): 2121-2143, 2021 06 11.
Article En | MEDLINE | ID: mdl-34032269

The Ser/Thr kinase MAP4K4, like other GCKIV kinases, has N-terminal kinase and C-terminal citron homology (CNH) domains. MAP4K4 can activate c-Jun N-terminal kinases (JNKs), and studies in the heart suggest it links oxidative stress to JNKs and heart failure. In other systems, MAP4K4 is regulated in striatin-interacting phosphatase and kinase (STRIPAK) complexes, in which one of three striatins tethers PP2A adjacent to a kinase to keep it dephosphorylated and inactive. Our aim was to understand how MAP4K4 is regulated in cardiomyocytes. The rat MAP4K4 gene was not properly defined. We identified the first coding exon of the rat gene using 5'-RACE, we cloned the full-length sequence and confirmed alternative-splicing of MAP4K4 in rat cardiomyocytes. We identified an additional α-helix C-terminal to the kinase domain important for kinase activity. In further studies, FLAG-MAP4K4 was expressed in HEK293 cells or cardiomyocytes. The Ser/Thr protein phosphatase inhibitor calyculin A (CalA) induced MAP4K4 hyperphosphorylation, with phosphorylation of the activation loop and extensive phosphorylation of the linker between the kinase and CNH domains. This required kinase activity. MAP4K4 associated with myosin in untreated cardiomyocytes, and this was lost with CalA-treatment. FLAG-MAP4K4 associated with all three striatins in cardiomyocytes, indicative of regulation within STRIPAK complexes and consistent with activation by CalA. Computational analysis suggested the interaction was direct and mediated via coiled-coil domains. Surprisingly, FLAG-MAP4K4 inhibited JNK activation by H2O2 in cardiomyocytes and increased myofibrillar organisation. Our data identify MAP4K4 as a STRIPAK-regulated kinase in cardiomyocytes, and suggest it regulates the cytoskeleton rather than activates JNKs.


Alternative Splicing , Calmodulin-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/metabolism , Mutation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Calmodulin-Binding Proteins/genetics , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Isoforms , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Sequence Homology
9.
Biochem J ; 478(11): 2059-2079, 2021 06 11.
Article En | MEDLINE | ID: mdl-34002209

Insulin and insulin-like growth factor stimulate protein synthesis and cardioprotection in the heart, acting through their receptors (INSRs, IGF1Rs) and signalling via protein kinase B (PKB, also known as Akt). Protein synthesis is increased in hearts perfused at alkaline pHo to the same extent as with insulin. Moreover, α1-adrenergic receptor (α1-AR) agonists (e.g. phenylephrine) increase protein synthesis in cardiomyocytes, activating PKB/Akt. In both cases, the mechanisms are not understood. Our aim was to determine if insulin receptor-related receptors (INSRRs, activated in kidney by alkaline pH) may account for the effects of alkaline pHo on cardiac protein synthesis, and establish if α1-ARs signal through the insulin receptor family. Alkaline pHo activated PKB/Akt signalling to the same degree as insulin in perfused adult male rat hearts. INSRRs were expressed in rat hearts and, by immunoblotting for phosphorylation (activation) of INSRRs/INSRs/IGF1Rs, we established that INSRRs, together with INSRs/IGF1Rs, are activated by alkaline pHo. The INSRR/INSR/IGF1R kinase inhibitor, linsitinib, prevented PKB/Akt activation by alkaline pHo, indicating that INSRRs/INSRs/IGF1Rs are required. Activation of PKB/Akt in cardiomyocytes by α1-AR agonists was also inhibited by linsitinib. Furthermore, linsitinib inhibited cardiomyocyte hypertrophy induced by α1-ARs in cultured cells, reduced the initial cardiac adaptation (24 h) to phenylephrine in vivo (assessed by echocardiography) and increased cardiac fibrosis over 4 days. We conclude that INSRRs are expressed in the heart and, together with INSRs/IGF1Rs, the insulin receptor family provide a potent system for promoting protein synthesis and cardioprotection. Moreover, this system is required for adaptive hypertrophy induced by α1-ARs.


Alkalies/pharmacology , Fibrosis/pathology , Hypertrophy/pathology , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Animals , Animals, Newborn , Female , Fibrosis/chemically induced , Fibrosis/metabolism , Humans , Hydrogen-Ion Concentration , Hypertrophy/chemically induced , Hypertrophy/metabolism , Imidazoles/pharmacology , Insulin/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Pyrazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Insulin/genetics , Receptors, Adrenergic, alpha-1/genetics
10.
Sci Rep ; 10(1): 21968, 2020 12 15.
Article En | MEDLINE | ID: mdl-33319836

Acute kidney injury (AKI) is a major health problem affecting millions of patients globally. There is no effective treatment for AKI and new therapies are urgently needed. Novel drug development, testing and progression to clinical trials is overwhelmingly expensive. Drug repurposing is a more cost-effective measure. We identified 2 commonly used drugs (colchicine and metformin) that alter inflammatory cell function and signalling pathways characteristic of AKI, and tested them in models of acute and chronic kidney injury to assess therapeutic benefit. We assessed the renoprotective effects of colchicine or metformin in C57BL/6 mice challenged with renal ischemia reperfusion injury (IRI), treated before or after injury. All animals underwent analysis of renal function and biomolecular phenotyping at 24 h, 48 h and 4 weeks after injury. Murine renal tubular epithelial cells were studied in response to in vitro mimics of IRI. Pre-emptive treatment with colchicine or metformin protected against AKI, with lower serum creatinine, improved histological changes and decreased TUNEL staining. Pro-inflammatory cytokine profile and multiple markers of oxidative stress were not substantially different between groups. Metformin augmented expression of multiple autophagic proteins which was reversed by the addition of hydroxychloroquine. Colchicine led to an increase in inflammatory cells within the renal parenchyma. Chronic exposure after acute injury to either therapeutic agent in the context of reduced renal mass did not mitigate the development of fibrosis, with colchicine significantly worsening an ischemic phenotype. These data indicate that colchicine and metformin affect acute and chronic kidney injury differently. This has significant implications for potential drug repurposing, as baseline renal disease must be considered when selecting medication.


Acute Kidney Injury/prevention & control , Colchicine/administration & dosage , Drug Repositioning , Kidney Failure, Chronic/prevention & control , Metformin/administration & dosage , Animals , Humans , Mice , Mice, Inbred C57BL
11.
Hypertension ; 76(4): 1208-1218, 2020 10.
Article En | MEDLINE | ID: mdl-32903101

Systemic hypertension increases cardiac workload causing cardiomyocyte hypertrophy and increased cardiac fibrosis. An underlying feature is increased production of reactive oxygen species. Redox-sensitive ASK1 (apoptosis signal-regulating kinase 1) activates stress-regulated protein kinases (p38-MAPK [mitogen-activated protein kinases] and JNKs [c-Jun N-terminal kinases]) and promotes fibrosis in various tissues. Here, we determined the specificity of ASK1 signaling in the heart, with the hypothesis that ASK1 inhibitors may be used to manage fibrosis in hypertensive heart disease. Using immunoblotting, we established that moderate levels of H2O2 activate ASK1 in neonatal rat cardiomyocytes and perfused rat hearts. ASK1 was activated during ischemia in adult rat hearts, but not on reperfusion, consistent with activation by moderate (not high) reactive oxygen species levels. In contrast, IL (interleukin)-1ß activated an alternative kinase, TAK1 (transforming growth factor-activated kinase 1). ASK1 was not activated by IL1ß in cardiomyocytes and activation in perfused hearts was due to increased reactive oxygen species. Selonsertib (ASK1 inhibitor) prevented activation of p38-MAPKs (but not JNKs) by oxidative stresses in cultured cardiomyocytes and perfused hearts. In vivo (C57Bl/6J mice with osmotic minipumps for drug delivery), selonsertib (4 mg/[kg·d]) alone did not affect cardiac function/dimensions (assessed by echocardiography). However, it suppressed hypertension-induced cardiac hypertrophy resulting from angiotensin II (0.8 mg/[kg·d], 7d), with inhibition of Nppa/Nppb mRNA upregulation, reduced cardiomyocyte hypertrophy and, notably, significant reductions in interstitial and perivascular fibrosis. Our data identify a specific reactive oxygen species→ASK1→p38-MAPK pathway in the heart and establish that ASK1 inhibitors protect the heart from hypertension-induced cardiac remodeling. Thus, targeting the ASK1→p38-MAPK nexus has potential therapeutic viability as a treatment for hypertensive heart disease.


Hypertension/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Myocardium/metabolism , Ventricular Remodeling/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Benzamides/pharmacology , Heart/drug effects , Hydrogen Peroxide/pharmacology , Imidazoles/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Ventricular Remodeling/drug effects
12.
Sci Rep ; 10(1): 6930, 2020 04 24.
Article En | MEDLINE | ID: mdl-32332767

Normothermic machine perfusion (NMP) is an emerging modality for kidney preservation prior to transplantation. NMP may allow directed pharmacomodulation of renal ischemia-reperfusion injury (IRI) without the need for systemic donor/recipient therapies. Three proven anti-IRI agents not in widespread clinical use, CD47-blocking antibody (αCD47Ab), soluble complement receptor 1 (sCR1), and recombinant thrombomodulin (rTM), were compared in a murine model of kidney IRI. The most effective agent was then utilized in a custom NMP circuit for the treatment of isolated porcine kidneys, ascertaining the impact of the drug on perfusion and IRI-related parameters. αCD47Ab conferred the greatest protection against IRI in mice after 24 hours. αCD47Ab was therefore chosen as the candidate agent for addition to the NMP circuit. CD47 receptor binding was demonstrated by immunofluorescence. Renal perfusion/flow improved with CD47 blockade, with a corresponding reduction in oxidative stress and histologic damage compared to untreated NMP kidneys. Tubular and glomerular functional parameters were not significantly impacted by αCD47Ab treatment during NMP. In a murine renal IRI model, αCD47Ab was confirmed as a superior anti-IRI agent compared to therapies targeting other pathways. NMP enabled effective, direct delivery of this drug to porcine kidneys, although further efficacy needs to be proven in the transplantation setting.


Antibodies/pharmacology , Kidney/pathology , Perfusion , Reperfusion Injury/pathology , Temperature , Animals , Blood Urea Nitrogen , CD47 Antigen/immunology , Chemokines/genetics , Chemokines/metabolism , Complement C3/metabolism , Complement C9/metabolism , Creatinine/blood , Drug Delivery Systems , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis A Virus Cellular Receptor 1/metabolism , Hydrogen Peroxide/metabolism , Inflammation Mediators/metabolism , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Complement/metabolism , Reperfusion Injury/blood , Swine
13.
FASEB J ; 33(10): 11528-11540, 2019 10.
Article En | MEDLINE | ID: mdl-31370677

Acute kidney injury (AKI) remains an important source of progressive chronic kidney injury. Loss of renal blood flow with subsequent restoration, termed ischemia reperfusion (IR), is a common cause of AKI. The cell surface receptor signal regulatory protein α (SIRP-α) is expressed on macrophages and limits inflammation and phagocytosis. SIRP-α has recently been found to have wider cell-based expression and play a role in renal IR. We have explored this in a genetic model of deficient SIRP-α signaling. Mice lacking SIRP-α cytoplasmic signaling (SIRP-αmut) and wild-type (WT) littermate controls underwent renal ischemia and reperfusion. Chimeric mice transplanted with WT or SIRP-αmut bone marrow were similarly challenged following engraftment. Molecular and immunohistochemical analysis of renal function, tissue damage, and key molecular targets was performed. SIRP-αmut mice were protected from renal IR compared with WT animals, demonstrating improved serum creatinine, less histologic damage, reduced proinflammatory cytokine production, and diminished production of reactive oxygen species (ROS). Resistance to renal IR in SIRP-αmut occurred alongside down-regulation of CD47 and thrombospondin-1, which are known to exert SIRP-α crosstalk and also promote IR. In chimeric mice, lack of SIRP-α signaling conferred protection to IR regardless of the genotype of circulating cells. Renal tubular epithelial cells from SIRP-αmut mice produced fewer ROS and proinflammatory cytokines in vitro. These results identify parenchymal SIRP-α as an independent driver of IR-mediated AKI and a potential therapeutic target.-Ghimire, K., Chiba, T., Minhas, N., Meijles, D. N., Lu, B., O'Connell, P., Rogers, N. M. Deficiency in SIRP-α cytoplasmic recruitment confers protection from acute kidney injury.


Acute Kidney Injury/metabolism , Cytoplasm/metabolism , Receptors, Immunologic/metabolism , Animals , CD47 Antigen/metabolism , Cytokines/metabolism , Down-Regulation/physiology , Inflammation/metabolism , Kidney/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Thrombospondin 1/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1150-L1164, 2019 06 01.
Article En | MEDLINE | ID: mdl-30892078

Pulmonary hypertension (PH) is a leading cause of death in sickle cell disease (SCD) patients. Hemolysis and oxidative stress contribute to SCD-associated PH. We have reported that the protein thrombospondin-1 (TSP1) is elevated in the plasma of patients with SCD and, by interacting with its receptor CD47, limits vasodilation of distal pulmonary arteries ex vivo. We hypothesized that the TSP1-CD47 interaction may promote PH in SCD. We found that TSP1 and CD47 are upregulated in the lungs of Berkeley (BERK) sickling (Sickle) mice and patients with SCD-associated PH. We then generated chimeric animals by transplanting BERK bone marrow into C57BL/6J (n = 24) and CD47 knockout (CD47KO, n = 27) mice. Right ventricular (RV) pressure was lower in fully engrafted Sickle-to-CD47KO than Sickle-to-C57BL/6J chimeras, as shown by the reduced maximum RV pressure (P = 0.013) and mean pulmonary artery pressure (P = 0.020). The afterload of the sickle-to-CD47KO chimeras was also lower, as shown by the diminished pulmonary vascular resistance (P = 0.024) and RV effective arterial elastance (P = 0.052). On myography, aortic segments from Sickle-to-CD47KO chimeras showed improved relaxation to acetylcholine. We hypothesized that, in SCD, TSP1-CD47 signaling promotes PH, in part, by increasing reactive oxygen species (ROS) generation. In human pulmonary artery endothelial cells, treatment with TSP1 stimulated ROS generation, which was abrogated by CD47 blockade. Explanted lungs of CD47KO chimeras had less vascular congestion and a smaller oxidative footprint. Our results show that genetic absence of CD47 ameliorates SCD-associated PH, which may be due to decreased ROS levels. Modulation of TSP1-CD47 may provide a new molecular approach to the treatment of SCD-associated PH.


Anemia, Sickle Cell/pathology , CD47 Antigen/metabolism , Hypertension, Pulmonary/pathology , Pulmonary Artery/pathology , Thrombospondin 1/metabolism , Anemia, Sickle Cell/genetics , Animals , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/genetics , Cells, Cultured , Endothelial Cells/pathology , Humans , Hypertension, Pulmonary/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Artery/cytology , Reactive Oxygen Species/metabolism , Ventricular Function, Right/physiology
15.
J Mol Cell Cardiol ; 129: 118-129, 2019 04.
Article En | MEDLINE | ID: mdl-30771309

Reactive oxygen species (ROS) play a key role in development of heart failure but, at a cellular level, their effects range from cytoprotection to induction of cell death. Understanding how this is regulated is crucial to develop novel strategies to ameliorate only the detrimental effects. Here, we revisited the fundamental hypothesis that the level of ROS per se is a key factor in the cellular response by applying different concentrations of H2O2 to cardiomyocytes. High concentrations rapidly reduced intracellular ATP and inhibited protein synthesis. This was associated with activation of AMPK which phosphorylated and inhibited Raptor, a crucial component of mTOR complex-1 that regulates protein synthesis. Inhibition of protein synthesis by high concentrations of H2O2 prevents synthesis of immediate early gene products required for downstream gene expression, and such mRNAs (many encoding proteins required to deal with oxidant stress) were only induced by lower concentrations. Lower concentrations of H2O2 promoted mTOR phosphorylation, associated with differential recruitment of some mRNAs to the polysomes for translation. Some of the upregulated genes induced by low H2O2 levels are cytoprotective. We identified p21Cip1/WAF1 as one such protein, and preventing its upregulation enhanced the rate of cardiomyocyte apoptosis. The data support the concept of a "redox rheostat" in which different degrees of ROS influence cell energetics and intracellular signalling pathways to regulate mRNA and protein expression. This sliding scale determines cell fate, modulating survival vs death.


AMP-Activated Protein Kinases/metabolism , Apoptosis , Gene Expression Regulation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoprotection/drug effects , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Genes, Immediate-Early , Hydrogen Peroxide/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Phosphorylation/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Physiological/drug effects
16.
Sci Signal ; 10(501)2017 Oct 17.
Article En | MEDLINE | ID: mdl-29042481

Senescent cells withdraw from the cell cycle and do not proliferate. The prevalence of senescent compared to normally functioning parenchymal cells increases with age, impairing tissue and organ homeostasis. A contentious principle governing this process has been the redox theory of aging. We linked matricellular protein thrombospondin 1 (TSP1) and its receptor CD47 to the activation of NADPH oxidase 1 (Nox1), but not of the other closely related Nox isoforms, and associated oxidative stress, and to senescence in human cells and aged tissue. In human endothelial cells, TSP1 promoted senescence and attenuated cell cycle progression and proliferation. At the molecular level, TSP1 increased Nox1-dependent generation of reactive oxygen species (ROS), leading to the increased abundance of the transcription factor p53. p53 mediated a DNA damage response that led to senescence through Rb and p21cip, both of which inhibit cell cycle progression. Nox1 inhibition blocked the ability of TSP1 to increase p53 nuclear localization and p21cip abundance and its ability to promote senescence. Mice lacking TSP1 showed decreases in ROS production, p21cip expression, p53 activity, and aging-induced senescence. Conversely, lung tissue from aging humans displayed increases in the abundance of vascular TSP1, Nox1, p53, and p21cip Finally, genetic ablation or pharmacological blockade of Nox1 in human endothelial cells attenuated TSP1-mediated ROS generation, restored cell cycle progression, and protected against senescence. Together, our results provide insights into the functional interplay between TSP1 and Nox1 in the regulation of endothelial senescence and suggest potential targets for controlling the aging process at the molecular level.


CD47 Antigen/genetics , Cellular Senescence/genetics , Endothelial Cells/metabolism , NADPH Oxidase 1/genetics , Thrombospondin 1/genetics , Adult , Aged , Aging/genetics , Animals , CD47 Antigen/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation , Humans , Lung/blood supply , Lung/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NADPH Oxidase 1/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Thrombospondin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Clin Sci (Lond) ; 131(15): 2019-2035, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28522681

Pulmonary arterial hypertension (PAH) is a rapidly degenerating and devastating disease of increased pulmonary vessel resistance leading to right heart failure. Palliative modalities remain limited despite recent endeavors to investigate the mechanisms underlying increased pulmonary vascular resistance (PVR), i.e. aberrant vascular remodeling and occlusion. However, little is known of the molecular mechanisms responsible for endothelial proliferation, a root cause of PAH-associated vascular remodeling. Lung tissue specimens from PAH and non-PAH patients and hypoxia-exposed human pulmonary artery endothelial cells (ECs) (HPAEC) were assessed for mRNA and protein expression. Reactive oxygen species (ROS) were measured using cytochrome c and Amplex Red assays. Findings demonstrate for the first time an up-regulation of NADPH oxidase 1 (Nox1) at the transcript and protein level in resistance vessels from PAH compared with non-PAH patients. This coincided with an increase in ROS production and expression of bone morphogenetic protein (BMP) antagonist Gremlin1 (Grem1). In HPAEC, hypoxia induced Nox1 subunit expression, assembly, and oxidase activity leading to elevation in sonic hedgehog (SHH) and Grem1 expression. Nox1 gene silencing abrogated this cascade. Moreover, loss of either Nox1, SHH or Grem1 attenuated hypoxia-induced EC proliferation. Together, these data support a Nox1-SHH-Grem1 signaling axis in pulmonary vascular endothelium that is likely to contribute to pathophysiological endothelial proliferation and the progression of PAH. These findings also support targeting of Nox1 as a viable therapeutic option to combat PAH.


Cell Proliferation , Hypertension, Pulmonary/enzymology , Intercellular Signaling Peptides and Proteins/metabolism , NADPH Oxidases/metabolism , Adult , Aged , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , NADPH Oxidase 1 , NADPH Oxidases/genetics , Pulmonary Artery/enzymology , Reactive Oxygen Species/metabolism , Signal Transduction
18.
Antioxid Redox Signal ; 26(16): 886-901, 2017 06 01.
Article En | MEDLINE | ID: mdl-27958762

AIMS: Macropinocytosis has been implicated in cardiovascular and other disorders, yet physiological factors that initiate fluid-phase internalization and the signaling mechanisms involved remain poorly identified. The present study was designed to examine whether matrix protein thrombospondin-1 (TSP1) stimulates macrophage macropinocytosis and, if so, to investigate the potential signaling mechanism involved. RESULTS: TSP1 treatment of human and murine macrophages stimulated membrane ruffle formation and pericellular solute internalization by macropinocytosis. Blockade of TSP1 cognate receptor CD47 and NADPH oxidase 1 (Nox1) signaling, inhibition of phosphoinositide 3-kinase, and transcriptional knockdown of myotubularin-related protein 6 abolished TSP1-induced macropinocytosis. Our results demonstrate that Nox1 signaling leads to dephosphorylation of actin-binding protein cofilin at Ser-3, actin remodeling, and macropinocytotic uptake of unmodified native low-density lipoprotein (nLDL), leading to foam cell formation. Finally, peritoneal chimera studies suggest the role of CD47 in macrophage lipid macropinocytosis in hypercholesterolemic ApoE-/- mice in vivo. INNOVATION: Activation of a previously unidentified TSP1-CD47 signaling pathway in macrophages stimulates direct receptor-independent internalization of nLDL, leading to significant lipid accumulation and foam cell formation. These findings reveal a new paradigm in which delimited Nox1-mediated redox signaling, independent of classical lipid oxidation, contributes to early propagation of vascular inflammatory disease. CONCLUSIONS: The findings of the present study demonstrate a new mechanism of solute uptake with implications for a wide array of cell types, including macrophages, dendritic cells, and cancer cells, and multiple pathological conditions in which matrix proteins are upregulated. Antioxid. Redox Signal. 26, 886-901.


Actin Depolymerizing Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD47 Antigen/metabolism , Hypercholesterolemia/metabolism , Macrophages/cytology , NADPH Oxidase 1/metabolism , Animals , Disease Models, Animal , Humans , Macrophages/metabolism , Mice , Phosphorylation , Pinocytosis , Protein Interaction Maps , RAW 264.7 Cells , Signal Transduction , THP-1 Cells
19.
Proc Natl Acad Sci U S A ; 113(36): E5308-17, 2016 09 06.
Article En | MEDLINE | ID: mdl-27540115

Despite numerous reports implicating NADPH oxidases (Nox) in the pathogenesis of many diseases, precise regulation of this family of professional reactive oxygen species (ROS) producers remains unclear. A unique member of this family, Nox1 oxidase, functions as either a canonical or hybrid system using Nox organizing subunit 1 (NoxO1) or p47(phox), respectively, the latter of which is functional in vascular smooth muscle cells (VSMC). In this manuscript, we identify critical requirement of ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50; aka NHERF1) for Nox1 activation and downstream responses. Superoxide (O2 (•-)) production induced by angiotensin II (AngII) was absent in mouse EBP50 KO VSMC vs. WT. Moreover, ex vivo incubation of aortas with AngII showed a significant increase in O2 (•-) in WT but not EBP50 or Nox1 nulls. Similarly, lipopolysaccharide (LPS)-induced oxidative stress was attenuated in femoral arteries from EBP50 KO vs. WT. In silico analyses confirmed by confocal microscopy, immunoprecipitation, proximity ligation assay, FRET, and gain-/loss-of-function mutagenesis revealed binding of EBP50, via its PDZ domains, to a specific motif in p47(phox) Functional studies revealed AngII-induced hypertrophy was absent in EBP50 KOs, and in VSMC overexpressing EBP50, Nox1 gene silencing abolished VSMC hypertrophy. Finally, ex vivo measurement of lumen diameter in mouse resistance arteries exhibited attenuated AngII-induced vasoconstriction in EBP50 KO vs. WT. Taken together, our data identify EBP50 as a previously unidentified regulator of Nox1 and support that it promotes Nox1 activity by binding p47(phox) This interaction is pivotal for agonist-induced smooth muscle ROS, hypertrophy, and vasoconstriction and has implications for ROS-mediated physiological and pathophysiological processes.


ATPases Associated with Diverse Cellular Activities/metabolism , DNA Helicases/metabolism , Hypertrophy/metabolism , NADPH Oxidase 1/genetics , Phosphoproteins/metabolism , Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Adaptor Proteins, Signal Transducing , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , DNA Helicases/genetics , Femoral Artery/drug effects , Femoral Artery/metabolism , Femoral Artery/pathology , Humans , Hypertrophy/chemically induced , Hypertrophy/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , NADPH Oxidase 1/metabolism , Oxidative Stress/drug effects , Phosphoproteins/genetics , Proteins/genetics , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchangers/genetics , Superoxides/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics
20.
Circulation ; 133(24): 2391-403, 2016 Jun 14.
Article En | MEDLINE | ID: mdl-27162237

BACKGROUND: The NADPH oxidase, by generating reactive oxygen species, is involved in the pathophysiology of many cardiovascular diseases and represents a therapeutic target for the development of novel drugs. A single-nucleotide polymorphism, C242T of the p22(phox) subunit of NADPH oxidase, has been reported to be negatively associated with coronary heart disease and may predict disease prevalence. However, the underlying mechanisms remain unknown. METHODS AND RESULTS: With the use of computer molecular modeling, we discovered that C242T single-nucleotide polymorphism causes significant structural changes in the extracellular loop of p22(phox) and reduces its interaction stability with Nox2 subunit. Gene transfection of human pulmonary microvascular endothelial cells showed that C242T p22(phox) significantly reduced Nox2 expression but had no significant effect on basal endothelial O2 (.-) production or the expression of Nox1 and Nox4. When cells were stimulated with tumor necrosis factor-α (or high glucose), C242T p22(phox) significantly inhibited tumor necrosis factor-α-induced Nox2 maturation, O2 (.-) production, mitogen-activated protein kinases and nuclear factor κB activation, and inflammation (all P<0.05). These C242T effects were further confirmed using p22(phox) short-hairpin RNA-engineered HeLa cells and Nox2(-/-) coronary microvascular endothelial cells. Clinical significance was investigated by using saphenous vein segments from non-coronary heart disease subjects after phlebotomies. TT (C242T) allele was common (prevalence of ≈22%) and, in comparison with CC, veins bearing TT allele had significantly lower levels of Nox2 expression and O2 (.-) generation in response to high-glucose challenge. CONCLUSIONS: C242T single-nucleotide polymorphism causes p22(phox) structural changes that inhibit endothelial Nox2 activation and oxidative response to tumor necrosis factor-α or high-glucose stimulation. C242T single-nucleotide polymorphism may represent a natural protective mechanism against inflammatory cardiovascular diseases.


Endothelial Cells/enzymology , NADPH Oxidases/genetics , Vascular Diseases/enzymology , Animals , Endothelial Cells/pathology , HeLa Cells , Humans , Inflammation/enzymology , Inflammation/metabolism , Inflammation/pathology , Membrane Glycoproteins/metabolism , Mice , Models, Molecular , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Oxidative Stress/physiology , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism , Vascular Diseases/metabolism , Vascular Diseases/pathology
...