99 results on '"Pulakat L"'
Search Results
2. Mineralocorticoid Receptor Blockade Attenuates Hypertension and Myocardial Oxidative Stress without Reducing Left Ventricular Hypertrophy.
- Author
-
DeMarco, VG, primary, Whaley-Connell, AT, additional, Habibi, J, additional, Hayden, MR, additional, Ma, L, additional, Pulakat, L, additional, and Sowers, JR, additional
- Published
- 2010
- Full Text
- View/download PDF
3. Role of the NifM in maturation of the Fe-protein of nitrogenase.
- Author
-
Gavini, N., primary and Pulakat, L., additional
- Published
- 2002
- Full Text
- View/download PDF
4. Size-Dependent Effect of Nanoceria on Their Antibacterial Activity Towards Escherichia coli
- Author
-
Dar, M. A., primary, Gul, R., additional, Alfadda, A. A., additional, Karim, M. R., additional, Kim, D. W., additional, Cheung, C. L., additional, Almajid, A. A., additional, Alharthi, N. H., additional, and Pulakat, L., additional
- Published
- 2017
- Full Text
- View/download PDF
5. The β-blocker Nebivolol Is a GRK/β-arrestin Biased Agonist
- Author
-
Erickson, CE, Gul, R, Blessing, CP, Nguyen, J, Liu, T, Pulakat, L, Bastepe, M, Jackson, EK, Andresen, BT, Erickson, CE, Gul, R, Blessing, CP, Nguyen, J, Liu, T, Pulakat, L, Bastepe, M, Jackson, EK, and Andresen, BT
- Abstract
Nebivolol, a third generation β-adrenoceptor (β-AR) antagonist (β-blocker), causes vasodilation by inducing nitric oxide (NO) production. The mechanism via which nebivolol induces NO production remains unknown, resulting in the genesis of much of the controversy regarding the pharmacological action of nebivolol. Carvedilol is another β-blocker that induces NO production. A prominent pharmacological mechanism of carvedilol is biased agonism that is independent of Gαs and involves G protein-coupled receptor kinase (GRK)/β-arrestin signaling with downstream activation of the epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK). Due to the pharmacological similarities between nebivolol and carvedilol, we hypothesized that nebivolol is also a GRK/β-arrestin biased agonist. We tested this hypothesis utilizing mouse embryonic fibroblasts (MEFs) that solely express β2-ARs, and HL-1 cardiac myocytes that express β1- and β2-ARs and no detectable β3-ARs. We confirmed previous reports that nebivolol does not significantly alter cAMP levels and thus is not a classical agonist. Moreover, in both cell types, nebivolol induced rapid internalization of β-ARs indicating that nebivolol is also not a classical β-blocker. Furthermore, nebivolol treatment resulted in a time-dependent phosphorylation of ERK that was indistinguishable from carvedilol and similar in duration, but not amplitude, to isoproterenol. Nebivolol-mediated phosphorylation of ERK was sensitive to propranolol (non-selective β-AR-blocker), AG1478 (EGFR inhibitor), indicating that the signaling emanates from β-ARs and involves the EGFR. Furthermore, in MEFs, nebivolol-mediated phosphorylation of ERK was sensitive to pharmacological inhibition of GRK2 as well as siRNA knockdown of β-arrestin 1/2. Additionally, nebivolol induced redistribution of β-arrestin 2 from a diffuse staining pattern into more intense punctate spots. We conclude that nebivolol is a β2-AR, and likely β1-AR, GRK/β-a
- Published
- 2013
6. Properties of angiotensin II receptors in glial cells from the adult corpus callosum.
- Author
-
Matute, C., primary, Pulakat, L., additional, Rio, C., additional, Valcarcel, C., additional, and Miledi, R., additional
- Published
- 1994
- Full Text
- View/download PDF
7. The tRNA species for redundant genetic codons NNU and NNC. A thought on the absence of phenylalanine tRNA with AAA anticodon in Escherichia coli.
- Author
-
Gavini, N, primary and Pulakat, L, additional
- Published
- 1992
- Full Text
- View/download PDF
8. Role of translation of the pheA leader peptide coding region in attenuation regulation of the Escherichia coli pheA gene
- Author
-
Gavini, N, primary and Pulakat, L, additional
- Published
- 1991
- Full Text
- View/download PDF
9. Role of ribosome release in the basal level of expression of the Escherichia coli gene pheA
- Author
-
Gavini, N., primary and Pulakat, L., additional
- Published
- 1991
- Full Text
- View/download PDF
10. Role of Asp297 of the AT2 receptor in high-affinity binding to different peptide ligands
- Author
-
Knowle, D., Kurfis, J., Gavini, N., and Pulakat, L.
- Published
- 2001
- Full Text
- View/download PDF
11. Identification of an interaction between the angiotensin II receptor sub-type AT2 and the ErbB3 receptor, a member of the epidermal growth factor receptor family
- Author
-
Knowle, D., Ahmed, S., and Pulakat, L.
- Published
- 2000
- Full Text
- View/download PDF
12. Identification of a second site compensatory mutation in the Fe-protein that allows diazotrophic growth of Azotobacter vinelandii UW97
- Author
-
Lei, S., Pulakat, L., Suh, M. H., and Gavini, N.
- Published
- 2000
- Full Text
- View/download PDF
13. Activation of vanadium nitrogenase expression in Azotobacter vinelandii DJ54 revertant in the presence of molybdenum
- Author
-
Lei, S., Pulakat, L., and Gavini, N.
- Published
- 2000
- Full Text
- View/download PDF
14. Role of the third intracellular loop of the Angiotensin II receptor subtype AT2 in ligand-receptor interaction
- Author
-
Dittus, J., Cooper, S., Obermeir, G., and Pulakat, L.
- Published
- 1999
- Full Text
- View/download PDF
15. Role of Lys215 located in the fifth transmembrane domain of the AT2 receptor in ligand-receptor interaction
- Author
-
Pulakat, L., Tadessee, A. S., Dittus, J. J., and Gavini, Narasaiah
- Published
- 1997
- Full Text
- View/download PDF
16. Nif- phenotype of Azotobacter vinelandii UW97. Characterization and mutational analysis.
- Author
-
Pulakat, L, Hausman, B S, Lei, S, and Gavini, N
- Abstract
We have identified the molecular basis for the nitrogenase negative phenotype exhibited by Azotobacter vinelandii UW97. This strain was initially isolated following nitrosoguanidine mutagenesis. Recently, it was shown that this strain lacks the Fe protein activity, which results in the synthesis of a FeMo cofactor-deficient apodinitrogenase. Activation of this apodinitrogenase requires the addition of both MgATP and wild-type Fe protein to the crude extracts made by A. vinelandii UW97 (Allen, R.M., Homer, M.J., Chatterjee R., Ludden, P.W., Roberts, G.P., and Shah, V.K. (1993) J. Biol. Chem. 268 23670-23674). Earlier, we proposed the sequence of events in the MoFe protein assembly based on the biochemical and spectroscopic analysis of the purified apodinitrogenase from A. vinelandii DJ54 (Gavini, N., Ma, L., Watt, G., and Burgess, B.K. (1994) Biochemistry 33, 11842-11849). Taken together, these results imply that the assembly process of apodinitrogenase is arrested at the same step in both of these strains. Since A. vinelandii DJ54 is a delta nifH strain, this strain is not useful in identifying the features of the Fe protein involved in the MoFe protein assembly. Here, we report a systematic analysis of an A. vinelandii UW97 mutant and show that, unlike A. vinelandii DJ54, the nifH gene of A. vinelandii UW97 has no deletion in either coding sequence or the surrounding sequences. The specific mutation responsible for the Nif- phenotype of A. vinelandii UW97 is the substitution of a non-conserved serine at position 44 of the Fe protein by a phenylalanine as shown by DNA sequencing. Furthermore, oligonucleotide site-directed mutagenesis was employed to confirm that the Nif- phenotype in A. vinelandii UW97 is exclusively due to the substitution of the Fe protein residue serine 44 by phenylalanine. By contrast, replacing Ser-44 with alanine did not affect the Nif phenotype of A. vinelandii. Therefore, it seems that the Nif- phenotype of A. vinelandii UW97 is caused by a general structural disturbance of the Fe protein due to the presence of the bulky phenylalanine at position 44.
- Published
- 1996
17. Cardiovascular Protective Effects of NP-6A4, a Drug with the FDA Designation for Pediatric Cardiomyopathy, in Female Rats with Obesity and Pre-Diabetes.
- Author
-
Belenchia AM, Boukhalfa A, DeMarco VG, Mehm A, Mahmood A, Liu P, Tang Y, Gavini MP, Mooney B, Chen HH, and Pulakat L
- Subjects
- Female, Rats, Animals, Rats, Zucker, Obesity complications, Obesity drug therapy, Obesity metabolism, Prediabetic State, Cardiomyopathies drug therapy, Cardiomyopathies etiology, Heart Diseases, Hyperglycemia
- Abstract
Background: Obese and pre-diabetic women have a higher risk for cardiovascular death than age-matched men with the same symptoms, and there are no effective treatments. We reported that obese and pre-diabetic female Zucker Diabetic Fatty (ZDF-F) rats recapitulate metabolic and cardiac pathology of young obese and pre-diabetic women and exhibit suppression of cardio-reparative AT2R. Here, we investigated whether NP-6A4, a new AT2R agonist with the FDA designation for pediatric cardiomyopathy, mitigate heart disease in ZDF-F rats by restoring AT2R expression., Methods: ZDF-F rats on a high-fat diet (to induce hyperglycemia) were treated with saline, NP-6A4 (10 mg/kg/day), or NP-6A4 + PD123319 (AT2R-specific antagonist, 5 mg/kg/day) for 4 weeks (n = 21). Cardiac functions, structure, and signaling were assessed by echocardiography, histology, immunohistochemistry, immunoblotting, and cardiac proteome analysis., Results: NP-6A4 treatment attenuated cardiac dysfunction, microvascular damage (-625%) and cardiomyocyte hypertrophy (-263%), and increased capillary density (200%) and AT2R expression (240%) ( p < 0.05). NP-6A4 activated a new 8-protein autophagy network and increased autophagy marker LC3-II but suppressed autophagy receptor p62 and autophagy inhibitor Rubicon. Co-treatment with AT2R antagonist PD123319 suppressed NP-6A4's protective effects, confirming that NP-6A4 acts through AT2R. NP-6A4-AT2R-induced cardioprotection was independent of changes in body weight, hyperglycemia, hyperinsulinemia, or blood pressure., Conclusions: Cardiac autophagy impairment underlies heart disease induced by obesity and pre-diabetes, and there are no drugs to re-activate autophagy. We propose that NP-6A4 can be an effective drug to reactivate cardiac autophagy and treat obesity- and pre-diabetes-induced heart disease, particularly for young and obese women.
- Published
- 2023
- Full Text
- View/download PDF
18. Using cultured canine cardiac slices to model the autophagic flux with doxorubicin.
- Author
-
Boukhalfa A, Robinson SR, Meola DM, Robinson NA, Ling LA, LaMastro JN, Upshaw JN, Pulakat L, Jaffe IZ, London CA, Chen HH, and Yang VK
- Subjects
- Animals, Dogs, Autophagy, Doxorubicin pharmacology, Doxorubicin metabolism, Sirolimus pharmacology, Myocytes, Cardiac metabolism, Cardiotoxicity metabolism
- Abstract
Chemotherapy-induced impairment of autophagy is implicated in cardiac toxicity induced by anti-cancer drugs. Imperfect translation from rodent models and lack of in vitro models of toxicity has limited investigation of autophagic flux dysregulation, preventing design of novel cardioprotective strategies based on autophagy control. Development of an adult heart tissue culture technique from a translational model will improve investigation of cardiac toxicity. We aimed to optimize a canine cardiac slice culture system for exploration of cancer therapy impact on intact cardiac tissue, creating a translatable model that maintains autophagy in culture and is amenable to autophagy modulation. Canine cardiac tissue slices (350 μm) were generated from left ventricular free wall collected from euthanized client-owned dogs (n = 7) free of cardiovascular disease at the Foster Hospital for Small Animals at Tufts University. Cell viability and apoptosis were quantified with MTT assay and TUNEL staining. Cardiac slices were challenged with doxorubicin and an autophagy activator (rapamycin) or inhibitor (chloroquine). Autophagic flux components (LC3, p62) were quantified by western blot. Cardiac slices retained high cell viability for >7 days in culture and basal levels of autophagic markers remained unchanged. Doxorubicin treatment resulted in perturbation of the autophagic flux and cell death, while rapamycin co-treatment restored normal autophagic flux and maintained cell survival. We developed an adult canine cardiac slice culture system appropriate for studying the effects of autophagic flux that may be applicable to drug toxicity evaluations., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Boukhalfa et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
19. A role for misaligned gene expression of fetal gene program in the loss of female-specific cardiovascular protection in young obese and diabetic females.
- Author
-
Pulakat L
- Subjects
- Adult, Pregnancy, Female, Humans, Male, Heart, Obesity, Prenatal Care, Gene Expression, Diabetes Mellitus, Type 2 metabolism
- Abstract
Healthy, premenopausal women have the advantage of female-specific cardiovascular protection compared to age-matched healthy men. However, pathologies such as obesity and Type 2 diabetes mellitus (T2DM) cause losing of this female-specific cardiovascular protection in young, obese and diabetic females. Molecular mechanisms underlying this loss of female-specific cardiovascular protection in young, obese and diabetic females are not clearly elucidated. This review takes a close look at the latest advances in our understanding of sex differences in adult cardiac gene expression patterns in health and disease. Based on the emerging data, this review proposes that female biased gene expression patterns in healthy adult hearts of human and pre-clinical models support the existence of active fetal gene program in healthy, premenopausal female heart compared to age-matched healthy male heart. However, the misalignment of gene expression pattern in this female-specific active cardiac fetal gene program caused by pathologies such as obesity and T2DM may contribute to the loss of female-specific cardiovascular protection in young, obese and diabetic females., Competing Interests: The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Pulakat.)
- Published
- 2023
- Full Text
- View/download PDF
20. Transdermal Delivery of High Molecular Weight Antibiotics to Deep Tissue Infections via Droplette Micromist Technology Device (DMTD).
- Author
-
Pulakat L, Chen HH, Gavini MP, Ling LA, Tang Y, Mehm A, Martin GL, Beale CN, Mooney BP, and Sun H
- Abstract
Wound infection by multidrug-resistant (MDR) bacteria is a major disease burden. Systemic administration of broad-spectrum antibiotics colistin methanesulfonate (CMS) and vancomycin are the last lines of defense against deep wound infections by MDR bacteria. However, systemic administration of CMS and vancomycin are linked to life-threatening vital organ damage. Currently there are no effective topical application strategies to deliver these high molecular weight antibiotics across the stratum corneum. To overcome this difficulty, we tested if high molecular weight antibiotics delivered by Droplette micromist technology device (DMTD), a transdermal delivery device that generates a micromist capable of packaging large molecules, could attenuate deep skin tissue infections. Using green fluorescent protein-tagged E. coli and live tissue imaging, we show that (1) the extent of attenuation of deep-skin E. coli infection was similar when treated with topical DMTD- or systemic IP (intraperitoneal)-delivered CMS; (2) DMTD-delivered micromist did not spread the infection deeper; (3) topical DMTD delivery and IP delivery resulted in similar levels of vancomycin in the skin after a 2 h washout period; and (4) IP-delivered vancomycin was about 1000-fold higher in kidney and plasma than DMTD-delivered vancomycin indicating systemic toxicity. Thus, topical DMTD delivery of these antibiotics is a safe treatment for the difficult-to-treat deep skin tissue infections by MDR bacteria.
- Published
- 2022
- Full Text
- View/download PDF
21. Suppression of Inflammatory Cardiac Cytokine Network in Rats with Untreated Obesity and Pre-Diabetes by AT2 Receptor Agonist NP-6A4.
- Author
-
Gavini MP, Mahmood A, Belenchia AM, Beauparlant P, Kumar SA, Ardhanari S, DeMarco VG, and Pulakat L
- Abstract
Obesity affects over 42% of the United States population and exacerbates heart disease, the leading cause of death in men and women. Obesity also increases pro-inflammatory cytokines that cause chronic tissue damage to vital organs. The standard-of-care does not sufficiently attenuate these inflammatory sequelae. Angiotensin II receptor AT2R is an anti-inflammatory and cardiovascular protective molecule; however, AT2R agonists are not used in the clinic to treat heart disease. NP-6A4 is a new AT2R peptide agonist with an FDA orphan drug designation for pediatric cardiomyopathy. NP-6A4 increases AT2R expression (mRNA and protein) and nitric oxide generation in human cardiovascular cells. AT2R-antagonist PD123319 and AT2RSiRNA suppress NP-6A4-effects indicating that NP-6A4 acts through AT2R. To determine whether NP-6A4 would mitigate cardiac damage from chronic inflammation induced by untreated obesity, we investigated the effects of 2-weeks NP-6A4 treatment (1.8 mg/kg delivered subcutaneously) on cardiac pathology of male Zucker obese (ZO) rats that display obesity, pre-diabetes and cardiac dysfunction. NP-6A4 attenuated cardiac diastolic and systolic dysfunction, cardiac fibrosis and cardiomyocyte hypertrophy, but increased myocardial capillary density. NP-6A4 treatment suppressed tubulointerstitial injury marker urinary β-NAG, and liver injury marker alkaline phosphatase in serum. These protective effects of NP-6A4 occurred in the presence of obesity, hyperinsulinemia, hyperglycemia, and hyperlipidemia, and without modulating blood pressure. NP-6A4 increased expression of AT2R (consistent with human cells) and cardioprotective erythropoietin (EPO) and Notch1 in ZO rat heart, but suppressed nineteen inflammatory cytokines. Cardiac miRNA profiling and in silico analysis showed that NP-6A4 activated a unique miRNA network that may regulate expression of AT2R, EPO, Notch1 and inflammatory cytokines, and mitigate cardiac pathology. Seventeen pro-inflammatory and pro-fibrotic cytokines that increase during lethal cytokine storms caused by infections such as COVID-19 were among the cytokines suppressed by NP-6A4 treatment in ZO rat heart. Thus, NP-6A4 activates a novel anti-inflammatory network comprised of 21 proteins in the heart that was not reported previously. Since NP-6A4's unique mode of action suppresses pro-inflammatory cytokine network and attenuates myocardial damage, it can be an ideal adjuvant drug with other anti-glycemic, anti-hypertensive, standard-of-care drugs to protect the heart tissues from pro-inflammatory and pro-fibrotic cytokine attack induced by obesity., Competing Interests: Author MG was employed by the company Novopyxis Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Gavini, Mahmood, Belenchia, Beauparlant, Kumar, Ardhanari, DeMarco and Pulakat.)
- Published
- 2021
- Full Text
- View/download PDF
22. Angiotensin Type 2 Receptors: Painful, or Not?
- Author
-
Pulakat L and Sumners C
- Abstract
Pain in response to various types of acute injury can be a protective stimulus to prevent the organism from using the injured part and allow tissue repair and healing. On the other hand, neuropathic pain, defined as 'pain caused by a lesion or disease of the somatosensory nervous system', is a debilitating pathology. The TRPA1 neurons in the Dorsal Root Ganglion (DRG) respond to reactive oxygen species (ROS) and induce pain. In acute nerve injury and inflammation, macrophages infiltrating the site of injury undergo an oxidative burst, and generate ROS that promote tissue repair and induce pain via TRPA1. The latter discourages using the injured limb, with a lack of movement helping wound healing. In chronic inflammation caused by diabetes, cancer etc., ROS levels increase systemically and modulate TRPA1 neuronal functions and cause debilitating neuropathic pain. It is important to distinguish between drug targets that elicit protective vs. debilitating pain when developing effective drugs for neuropathic pain. In this context, the connection of the Angiotensin type 2 receptor (AT
2 R) to neuropathic pain presents an interesting dilemma. Several lines of evidence show that AT2 R activation promotes anti-inflammatory and anti-nociceptive signaling, tissue repair, and suppresses ROS in chronic inflammatory models. Conversely, some studies suggest that AT2 R antagonists are anti-nociceptive and therefore AT2 R is a drug target for neuropathic pain. However, AT2 R expression in nociceptive neurons is lacking, indicating that neuronal AT2 R is not involved in neuropathic pain. It is also important to consider that Novartis terminated their phase II clinical trial (EMPHENE) to validate that AT2 R antagonist EMA401 mitigates post-herpetic neuralgia. This trial, conducted in Australia, United Kingdom, and a number of European and Asian countries in 2019, was discontinued due to pre-clinical drug toxicity data. Moreover, early data from the trial did not show statistically significant positive outcomes. These facts suggest that may AT2 R not be the proper drug target for neuropathic pain in humans and its inhibition can be harmful., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2020 Pulakat and Sumners.)- Published
- 2020
- Full Text
- View/download PDF
23. AT2R agonist NP-6A4 mitigates aortic stiffness and proteolytic activity in mouse model of aneurysm.
- Author
-
Sharma N, Belenchia AM, Toedebusch R, Pulakat L, and Hans CP
- Subjects
- Angiotensin II, Animals, Aorta metabolism, Aorta pathology, Apolipoproteins E deficiency, Apolipoproteins E metabolism, Collagen metabolism, Disease Models, Animal, Human Umbilical Vein Endothelial Cells metabolism, Humans, Male, Mice, Knockout, Osteopontin metabolism, Phenotype, Receptor, Angiotensin, Type 2 metabolism, Aortic Aneurysm, Abdominal metabolism, Aortic Aneurysm, Abdominal physiopathology, Proteolysis, Receptor, Angiotensin, Type 2 agonists, Vascular Stiffness
- Abstract
Clinical and experimental studies show that angiotensin II (AngII) promotes vascular pathology via activation of AngII type 1 receptors (AT1Rs). We recently reported that NP-6A4, a selective peptide agonist for AngII type 2 receptor (AT2R), exerts protective effects on human vascular cells subjected to serum starvation or doxorubicin exposure. In this study, we investigated whether NP-6A4-induced AT2R activation could mitigate AngII-induced abdominal aortic aneurism (AAA) using AngII-treated Apoe
-/- mice. Male Apoe-/- mice were infused with AngII (1 µg/kg/min) by implanting osmotic pumps subcutaneously for 28 days. A subset of mice was pre-treated subcutaneously with NP-6A4 (2.5 mg/kg/day) or vehicle for 14 days prior to AngII, and treatments were continued for 28 days. NP-6A4 significantly reduced aortic stiffness of the abdominal aorta induced by AngII as determined by ultrasound functional analyses and histochemical analyses. NP-6A4 also increased nitric oxide bioavailability in aortic tissues and suppressed AngII-induced increases in monocyte chemotactic protein-1, osteopontin and proteolytic activity of the aorta. However, NP-6A4 did not affect maximal intraluminal aortic diameter or AAA incidences significantly. These data suggest that the effects of AT2R agonist on vascular pathologies are selective, affecting the aortic stiffness and proteolytic activity without affecting the size of AAA., (© 2020 University of Missouri Columbia. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)- Published
- 2020
- Full Text
- View/download PDF
24. Pro-Senescence and Anti-Senescence Mechanisms of Cardiovascular Aging: Cardiac MicroRNA Regulation of Longevity Drug-Induced Autophagy.
- Author
-
Pulakat L and Chen HH
- Abstract
Chronological aging as well as biological aging accelerated by various pathologies such as diabetes and obesity contribute to cardiovascular aging, and structural and functional tissue damage of the heart and vasculature. Cardiovascular aging in humans is characterized by structural pathologic remodeling including cardiac and vascular fibrosis, hypertrophy, stiffness, micro- and macro-circulatory impairment, left ventricular diastolic dysfunction precipitating heart failure with either reduced or preserved ejection fraction, and cardiovascular cell death. Cellular senescence, an important hallmark of aging, is a critical factor that impairs repair and regeneration of damaged cells in cardiovascular tissues whereas autophagy, an intracellular catabolic process is an essential inherent mechanism that removes senescent cells throughout life time in all tissues. Several recent reviews have highlighted the fact that all longevity treatment paradigms to mitigate progression of aging-related pathologies converge in induction of autophagy, activation of AMP kinase (AMPK) and Sirtuin pathway, and inhibition of mechanistic target of rapamycin (mTOR). These longevity treatments include health style changes such as caloric restriction, and drug treatments using rapamycin, the first FDA-approved longevity drug, as well as other experimental longevity drugs such as metformin, rapamycin, aspirin, and resveratrol. However, in the heart tissue, autophagy induction has to be tightly regulated since evidence show excessive autophagy results in cardiomyopathy and heart failure. Here we discuss emerging evidence for microRNA-mediated tight regulation of autophagy in the heart in response to treatment with rapamycin, and novel approaches to monitor autophagy progression in a temporal manner to diagnose and regulate autophagy induction by longevity treatments., (Copyright © 2020 Pulakat and Chen.)
- Published
- 2020
- Full Text
- View/download PDF
25. Challenges and New Therapeutic Approaches in the Management of Chronic Wounds.
- Author
-
Sun H, Pulakat L, and Anderson DW
- Subjects
- Animals, Biofilms, Drug Resistance, Bacterial, Humans, Wound Healing drug effects, Wounds and Injuries etiology, Wounds and Injuries metabolism, Wounds and Injuries microbiology, Chronic Disease therapy, Wounds and Injuries therapy
- Abstract
Chronic non-healing wounds are estimated to cost the US healthcare $28-$31 billion per year. Diabetic ulcers, arterial and venous ulcers, and pressure ulcers are some of the most common types of chronic wounds. The burden of chronic wounds continues to rise due to the current epidemic of obesity and diabetes and the increase in elderly adults in the population who are more vulnerable to chronic wounds than younger individuals. This patient population is also highly vulnerable to debilitating infections caused by opportunistic and multi-drug resistant pathogens. Reduced microcirculation, decreased availability of cytokines and growth factors that promote wound closure and healing, and infections by multi-drug resistant and biofilm forming microbes are some of the critical factors that contribute to the development of chronic non-healing wounds. This review discusses novel approaches to understand chronic wound pathology and methods to improve chronic wound care, particularly when chronic wounds are infected by multi-drug resistant, biofilm forming microbes., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2020
- Full Text
- View/download PDF
26. Deficiency of IL12p40 (Interleukin 12 p40) Promotes Ang II (Angiotensin II)-Induced Abdominal Aortic Aneurysm.
- Author
-
Sharma N, Dev R, Belenchia AM, Aroor AR, Whaley-Connell A, Pulakat L, and Hans CP
- Subjects
- Animals, Collagen metabolism, Interleukin-12 Subunit p40 deficiency, Macrophages physiology, Male, Matrix Metalloproteinase 2 metabolism, Mice, Mice, Inbred C57BL, Transforming Growth Factor beta2 physiology, Vascular Stiffness, Angiotensin II pharmacology, Aortic Aneurysm, Abdominal chemically induced, Interleukin-12 Subunit p40 physiology
- Abstract
Objective- Abdominal aortic aneurysm is caused by the accumulation of inflammatory cells in the aortic wall. Our recent studies demonstrated that inhibition of Notch signaling attenuates abdominal aortic aneurysm formation by shifting the macrophage balance towards anti-inflammatory (M2) phenotype. Using IL12p40
-/- (interleukin 12 p40) mice, we investigated the effects of M2-predominant macrophages on the development of abdominal aortic aneurysm. Approach and Results- Male (8-10 week-old) wild-type and IL12p40-/- mice (n=15) on C57BL/6 background were infused with Ang II (angiotensin II, 1000 ng/kg per minute) by implanting osmotic pumps subcutaneously for 28 days. In the IL12p40-/- mice, Ang II significantly increased the maximal intraluminal diameter (9/15) as determined by transabdominal ultrasound imaging. In addition, IL12p40-deletion significantly increased aortic stiffness in response to Ang II as measured by pulse wave velocity and atomic force microscopy. Histologically, IL12p40-/- mice exhibited increased maximal external diameter of aorta and aortic lesions associated with collagen deposition and increased elastin fragmentation compared with wild-type mice infused with Ang II. Mechanistically, IL12p40 deficiency by siRNA (small interfering RNA) augmented the Tgfβ2-mediated Mmp2 expression in wild-type bone marrow-derived macrophages without affecting the expression of Mmp9. No such effects of IL12p40 deficiency on MMP2/MMP9 was observed in human aortic smooth muscle cells or fibroblasts. Depletion of macrophages in IL12p40-/- mice by clodronate liposomes significantly decreased the maximal external diameter of aorta and aortic stiffness in response to Ang II as determined by imaging and atomic force microscopy. Conclusions- IL12p40 depletion promotes the development of abdominal aortic aneurysm, in part, by facilitating recruitment of M2-like macrophages and potentiating aortic stiffness and fibrosis mediated by Tgfβ2.- Published
- 2019
- Full Text
- View/download PDF
27. Comparison of Cardiac miRNA Transcriptomes Induced by Diabetes and Rapamycin Treatment and Identification of a Rapamycin-Associated Cardiac MicroRNA Signature.
- Author
-
Belenchia AM, Gavini MP, Toedebusch RG, DeMarco VG, and Pulakat L
- Subjects
- Animals, Diabetes Mellitus genetics, Disease Models, Animal, Humans, Male, Rats, Rats, Zucker, Diabetes Mellitus chemically induced, MicroRNAs metabolism, Sirolimus adverse effects, Transcriptome genetics
- Abstract
Rapamycin (Rap), an inhibitor of mTORC1, reduces obesity and improves lifespan in mice. However, hyperglycemia and lipid disorders are adverse side effects in patients receiving Rap treatment. We previously reported that diabetes induces pansuppression of cardiac cytokines in Zucker obese rats (ZO-C). Rap treatment (750 μ g/kg/day for 12 weeks) reduced their obesity and cardiac fibrosis significantly; however, it increased their hyperglycemia and did not improve their cardiac diastolic parameters. Moreover, Rap treatment of healthy Zucker lean rats (ZL-C) induced cardiac fibrosis. Rap-induced changes in ZL-C's cardiac cytokine profile shared similarities with that of diabetes-induced ZO-C. Therefore, we hypothesized that the cardiac microRNA transcriptome induced by diabetes and Rap treatment could share similarities. Here, we compared the cardiac miRNA transcriptome of ZL-C to ZO-C, Rap-treated ZL (ZL-Rap), and ZO (ZO-Rap). We report that 80% of diabetes-induced miRNA transcriptome (40 differentially expressed miRNAs by minimum 1.5-fold in ZO-C versus ZL-C; p ≤ 0.05) is similar to 47% of Rap-induced miRNA transcriptome in ZL (68 differentially expressed miRNAs by minimum 1.5-fold in ZL-Rap versus ZL-C; p ≤ 0.05). This remarkable similarity between diabetes-induced and Rap-induced cardiac microRNA transcriptome underscores the role of miRNAs in Rap-induced insulin resistance. We also show that Rap treatment altered the expression of the same 17 miRNAs in ZL and ZO hearts indicating that these 17 miRNAs comprise a unique Rap-induced cardiac miRNA signature. Interestingly, only four miRNAs were significantly differentially expressed between ZO-C and ZO-Rap, indicating that, unlike the nondiabetic heart, Rap did not substantially change the miRNA transcriptome in the diabetic heart. In silico analyses showed that (a) mRNA-miRNA interactions exist between differentially expressed cardiac cytokines and miRNAs, (b) human orthologs of rat miRNAs that are strongly correlated with cardiac fibrosis may modulate profibrotic TGF- β signaling, and (c) changes in miRNA transcriptome caused by diabetes or Rap treatment include cardioprotective miRNAs indicating a concurrent activation of an adaptive mechanism to protect the heart in conditions that exacerbate diabetes.
- Published
- 2018
- Full Text
- View/download PDF
28. Cell-Specific Protective Signaling Induced by the Novel AT2R-Agonist NP-6A4 on Human Endothelial and Smooth Muscle Cells.
- Author
-
Toedebusch R, Belenchia A, and Pulakat L
- Abstract
Cardiovascular disease incidence continues to rise and new treatment paradigms are warranted. We reported previously that activation of Angiotensin II receptor (encoded by the X-linked Agtr2 gene) by a new peptide agonist, NP-6A4, was more effective in protecting mouse cardiomyocyte HL-1 cells and human coronary artery vascular smooth muscle cells (hCAVSMCs) from acute nutrient deficiency than other drugs tested. To elucidate further the protective effects of NP-6A4 in human cells, we studied the effects of NP-6A4 treatment on functions of human coronary artery endothelial cells (hCAECs), and hCAVSMCs. In hCAVSMCs, NP-6A4 (1 μM) increased Agtr2 mRNA (sixfold, p < 0.05) after 12-h exposure, whereas in hCAECs, significant increase in Agtr2 mRNA (hCAECs: eightfold) was observed after prolonged exposure. Interestingly, NP-6A4 treatment (1 μM, 12 h) increased AT2R protein levels in all human cells tested. Pre-treatment with AT2R-antagonist PD123319 (20 μM) and anti-AT2R siRNA (1 μM) suppressed this effect. Thus, NP-6A4 activates a positive feedback loop for AT2R expression and signaling in hCAVSMCs and hCAECs. NP-6A4 (1-20 μM) increased cell index (CI) of hCAVSMCs as determined by real time cell analyzer (RTCA), indicating that high concentrations of NP-6A4 were not cytotoxic for hCAVSMCs, rather promoting better cell attachment and growth. Seahorse Extracellular Flux Assay revealed that NP-6A4 (1 μM) treatment for 7 days increased whole cell-based mitochondrial parameters of hCAVSMCs, specifically maximal respiration ( p < 0.05), spare respiratory capacity ( p < 0.05) and ATP production ( p < 0.05). NP-6A4 (1 μM; 7 days) also suppressed Reactive Oxygen Species (ROS) in hCAVSMCs. Exposure to Doxorubicin (DOXO) (1 μM) increased ROS in hCAVSMCs and this effect was suppressed by NP-6A4 (1 μM). In hCAECs grown in complete medium, NP-6A4 (1 μM) and Ang II (1 μM) exerted similar changes in CI. Additionally, NP-6A4 (5 μM: 12 h) increased expression of eNOS (sixfold, p < 0.05) and generation of nitric oxide (1.3-fold, p < 0.05) in hCAECs and pre-treatment with PD123319 (20 μM) suppressed this effect partially (65%). Finally, NP-6A4 decreased phosphorylation of Jun-N-terminal kinase, implicated in apoptosis of ECs in atherosclerotic sites. Taken together, NP-6A4, through its ability to increase AT2R expression and signaling, exerts different cell-specific protective effects in human VSMCs and ECs.
- Published
- 2018
- Full Text
- View/download PDF
29. Diabetic Cardiomyopathy: Impact of Biological Sex on Disease Development and Molecular Signatures.
- Author
-
Toedebusch R, Belenchia A, and Pulakat L
- Abstract
Diabetic cardiomyopathy refers to a unique set of heart-specific pathological variables induced by hyperglycemia and insulin resistance. Given that cardiovascular disease (CVD) is the leading cause of death in the world, and type 2 diabetes incidence continues to rise, understanding the complex interplay between these two morbidities and developing novel therapeutic strategies is vital. Two hallmark characteristics specific to diabetic cardiomyopathy are diastolic dysfunction and cardiac structural mal-adaptations, arising from cardiac cellular responses to the complex toxicity induced by hyperglycemia with or without hyperinsulinemia. While type 2 diabetes is more prevalent in men compared to women, cardiovascular risk is higher in diabetic women than in diabetic men, suggesting that diabetic women take a steeper path to cardiomyopathy and heart failure. Accumulating evidence from randomized clinical trials indicate that although pre-menopausal women have lower risk of CVDs, compared to age-matched men, this advantage is lost in diabetic pre-menopausal women, which suggests estrogen availability does not protect from increased cardiovascular risk. Notably, few human studies have assessed molecular and cellular mechanisms regarding similarities and differences in the progression of diabetic cardiomyopathy in men versus women. Additionally, most pre-clinical rodent studies fail to include female animals, leaving a void in available data to truly understand the impact of biological sex differences in diabetes-induced dysfunction of cardiovascular cells. Elegant reviews in the past have discussed in detail the roles of estrogen-mediated signaling in cardiovascular protection, sex differences associated with telomerase activity in the heart, and cardiac responses to exercise. In this review, we focus on the emerging cellular and molecular markers that define sex differences in diabetic cardiomyopathy based on the recent clinical and pre-clinical evidence. We also discuss miR-208a, MED13, and AT2R, which may provide new therapeutic targets with hopes to develop novel treatment paradigms to treat diabetic cardiomyopathy uniquely between men and women.
- Published
- 2018
- Full Text
- View/download PDF
30. Cardiovascular disease progression in female Zucker Diabetic Fatty rats occurs via unique mechanisms compared to males.
- Author
-
Lum-Naihe K, Toedebusch R, Mahmood A, Bajwa J, Carmack T, Kumar SA, Ardhanari S, DeMarco VG, Emter CA, and Pulakat L
- Subjects
- Animals, Biomarkers metabolism, Cardiomegaly metabolism, Cardiomegaly pathology, Cardiovascular Diseases metabolism, Cytokines metabolism, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 2 metabolism, Diabetic Cardiomyopathies metabolism, Diabetic Cardiomyopathies pathology, Disease Progression, Female, Fibrosis metabolism, Fibrosis pathology, Male, MicroRNAs metabolism, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Neuropilin-1 metabolism, Rats, Rats, Zucker, Receptor, Angiotensin, Type 2 metabolism, Cardiovascular Diseases pathology, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Type 2 pathology
- Abstract
Population studies have shown that compared to diabetic men, diabetic women are at a higher risk of cardiovascular disease. However, the mechanisms underlying this gender disparity are unclear. Our studies in young murine models of type 2 diabetes mellitus (T2DM) and cardiovascular disease show that diabetic male rats develop increased cardiac fibrosis and suppression of intracardiac anti-fibrotic cytokines, while premenopausal diabetic female rats do not. This protection from cardiac fibrosis in female rats can be an estrogen-related effect. However, diabetic female rats develop early subclinical myocardial deformation, cardiac hypertrophy via elevated expression of pro-hypertrophic miR-208a, myocardial damage, and suppression of cardio-reparative Angiotensin II receptor 2 (Agtr2). Diabetic rats of both sexes exhibit a reduction in cardiac capillary density. However, diabetic female rats have reduced expression of neuropilin 1 that attenuates cardiomyopathy compared to diabetic male rats. A combination of cardiac hypertrophy and reduced capillary density likely contributed to increased myocardial structural damage in diabetic female rats. We propose expansion of existing cardiac assessments in diabetic female patients to detect myocardial deformation, cardiac hypertrophy and capillary density via non-invasive imaging, as well as suggest miR-208a, AT2R and neuropilin 1 as potential therapeutic targets and mechanistic biomarkers for cardiac disease in females.
- Published
- 2017
- Full Text
- View/download PDF
31. mTORC1 inhibitors rapamycin and metformin affect cardiovascular markers differentially in ZDF rats.
- Author
-
Nistala R, Raja A, and Pulakat L
- Subjects
- Animals, Biomarkers blood, Biomarkers urine, Cardiovascular Diseases blood, Cardiovascular Diseases enzymology, Cardiovascular Diseases etiology, Diabetes Mellitus, Type 2 blood, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 enzymology, Diabetic Nephropathies enzymology, Diabetic Nephropathies etiology, Diabetic Nephropathies prevention & control, Disease Models, Animal, Disease Progression, Liver drug effects, Liver enzymology, Male, Mechanistic Target of Rapamycin Complex 1, Multiprotein Complexes metabolism, Proteinuria enzymology, Proteinuria etiology, Proteinuria prevention & control, Rats, Zucker, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism, Time Factors, Cardiovascular Diseases prevention & control, Diabetes Mellitus, Type 2 drug therapy, Hypoglycemic Agents pharmacology, Insulin Resistance, Metformin pharmacology, Multiprotein Complexes antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Sirolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Mammalian target for rapamycin complex 1 (mTORC1) is a common target for the action of immunosuppressant macrolide rapamycin and glucose-lowering metformin. Inhibition of mTORC1 can exert both beneficial and detrimental effects in different pathologies. Here, we investigated the differential effects of rapamycin (1.2 mg/kg per day delivered subcutaneously for 6 weeks) and metformin (300 mg/kg per day delivered orally for 11 weeks) treatments on male Zucker diabetic fatty (ZDF) rats that mimic the cardiorenal pathology of type 2 diabetic patients and progress to insulin insufficiency. Rapamycin and metformin improved proteinuria, and rapamycin also reduced urinary gamma glutamyl transferase (GGT) indicating improvement of tubular health. Metformin reduced food and water intake, and urinary sodium and potassium, whereas rapamycin increased urinary sodium. Metformin reduced plasma alkaline phosphatase, but induced transaminitis as evidenced by significant increases in plasma AST and ALT. Metformin also induced hyperinsulinemia, but did not suppress fasting plasma glucose after ZDF rats reached 17 weeks of age, and worsened lipid profile. Rapamycin also induced mild transaminitis. Additionally, both rapamycin and metformin increased plasma uric acid and creatinine, biomarkers for cardiovascular and renal disease. These observations define how rapamycin and metformin differentially modulate metabolic profiles that regulate cardiorenal pathology in conditions of severe type 2 diabetes.
- Published
- 2017
- Full Text
- View/download PDF
32. Regulation of the cardioprotective adiponectin and its receptor AdipoR1 by salt.
- Author
-
Arnold N, Mahmood A, Ramdas M, Ehlinger PP, and Pulakat L
- Subjects
- Adiponectin genetics, Angiotensin II pharmacology, Animals, Cell Line, Dose-Response Relationship, Drug, Down-Regulation, Mice, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptors, Adiponectin genetics, Signal Transduction drug effects, Adiponectin metabolism, Myocytes, Cardiac drug effects, Receptors, Adiponectin metabolism, Sodium Chloride pharmacology
- Abstract
Both circulating adiponectin (APN) and cardiac APN exert cardioprotective effects and improve insulin sensitivity and mitochondrial function. Low circulating APN serves as a biomarker for cardiovascular risk. Ablation of adiponectin receptor 1 (AdipoR1) causes myocardial mitochondrial dysfunction. Although high salt intake is a contributor to cardiovascular disease, how it modulates the expression of APN or AdipoR1 in cardiomyocytes is not known. We report that APN mRNA expression was attenuated in a dose-dependent manner in mouse cardiomyocyte cell line HL-1 exposed to salt concentrations ranging from 0.75% to 1.5% for 12 h. High-salt exposure (0.88% and 1.25% for 12 h) also suppressed APN and AdipoR1 protein expression significantly in rat cardiac muscle H9c2 cells. Co-immunostaining for AdipoR1 and mitochondrial complex 1 indicated that AdipoR1 may be co-localized with mitochondria. These data show for the first time that high salt is an important suppressor of cardiovascular protective APN and AdipoR1.
- Published
- 2017
- Full Text
- View/download PDF
33. Differential Regulation of Cardiac Function and Intracardiac Cytokines by Rapamycin in Healthy and Diabetic Rats.
- Author
-
Luck C, DeMarco VG, Mahmood A, Gavini MP, and Pulakat L
- Subjects
- Animals, Blood Glucose analysis, Diabetes Mellitus, Experimental, Echocardiography, Enzyme-Linked Immunosorbent Assay, Granulocyte-Macrophage Colony-Stimulating Factor analysis, Heart diagnostic imaging, Insulin blood, Interferon-gamma, Interleukin-10 analysis, Male, Myocardium pathology, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Rats, Rats, Zucker, Time Factors, Triglycerides blood, Uric Acid blood, Cytokines analysis, Heart drug effects, Myocardium metabolism, Sirolimus pharmacology
- Abstract
Diabetes is comorbid with cardiovascular disease and impaired immunity. Rapamycin improves cardiac functions and extends lifespan by inhibiting the mechanistic target of rapamycin complex 1 (mTORC1). However, in diabetic murine models, Rapamycin elevates hyperglycemia and reduces longevity. Since Rapamycin is an immunosuppressant, we examined whether Rapamycin (750 μ g/kg/day) modulates intracardiac cytokines, which affect the cardiac immune response, and cardiac function in male lean (ZL) and diabetic obese Zucker (ZO) rats. Rapamycin suppressed levels of fasting triglycerides, insulin, and uric acid in ZO but increased glucose. Although Rapamycin improved multiple diastolic parameters ( E / E ', E '/ A ', E / Vp ) initially, these improvements were reversed or absent in ZO at the end of treatment, despite suppression of cardiac fibrosis and phosphoSer473Akt. Intracardiac cytokine protein profiling and Ingenuity® Pathway Analysis indicated suppression of intracardiac immune defense in ZO, in response to Rapamycin treatment in both ZO and ZL. Rapamycin increased fibrosis in ZL without increasing phosphoSer473Akt and differentially modulated anti-fibrotic IL-10, IFN γ , and GM-CSF in ZL and ZO. Therefore, fundamental difference in intracardiac host defense between diabetic ZO and healthy ZL, combined with differential regulation of intracardiac cytokines by Rapamycin in ZO and ZL hearts, underlies differential cardiac outcomes of Rapamycin treatment in health and diabetes.
- Published
- 2017
- Full Text
- View/download PDF
34. Loss of Nlrp3 Does Not Protect Mice from Western Diet-Induced Adipose Tissue Inflammation and Glucose Intolerance.
- Author
-
Ringling RE, Gastecki ML, Woodford ML, Lum-Naihe KJ, Grant RW, Pulakat L, Vieira-Potter VJ, and Padilla J
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Adipose Tissue pathology, Diet, Western, Glucose Intolerance, Inflammation pathology, NLR Family, Pyrin Domain-Containing 3 Protein genetics
- Abstract
We tested the hypothesis that loss of Nlrp3 would protect mice from Western diet-induced adipose tissue (AT) inflammation and associated glucose intolerance and cardiovascular complications. Five-week old C57BL6J wild-type (WT) and Nlrp3 knockout (Nlrp3-/-) mice were randomized to either a control diet (10% kcal from fat) or Western diet (45% kcal from fat and 1% cholesterol) for 24 weeks (n = 8/group). Contrary to our hypothesis that obesity-mediated white AT inflammation is Nlrp3-dependent, we found that Western diet-induced expression of AT inflammatory markers (i.e., Cd68, Cd11c, Emr1, Itgam, Lgals, Il18, Mcp1, Tnf, Ccr2, Ccl5 mRNAs, and Mac-2 protein) were not accompanied by increased caspase-1 cleavage, a hallmark feature of NLRP3 inflammasome activation. Furthermore, Nlrp3 null mice were not protected from Western diet-induced white or brown AT inflammation. Although Western diet promoted glucose intolerance in both WT and Nlrp3-/- mice, Nlrp3-/- mice were protected from Western diet-induced aortic stiffening. Additionally, Nlrp3-/- mice exhibited smaller cardiomyocytes and reduced cardiac fibrosis, independent of diet. Collectively, these findings suggest that presence of the Nlrp3 gene is not required for Western diet-induced AT inflammation and/or glucose intolerance; yet Nlrp3 appears to play a role in potentiating arterial stiffening, cardiac hypertrophy and fibrosis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
35. Differential Effects of β-Blockers, Angiotensin II Receptor Blockers, and a Novel AT2R Agonist NP-6A4 on Stress Response of Nutrient-Starved Cardiovascular Cells.
- Author
-
Mahmood A and Pulakat L
- Subjects
- Animals, Cell Line, Humans, Mice, Muscle, Smooth, Vascular cytology, Myocytes, Smooth Muscle cytology, Receptor, Angiotensin, Type 2 metabolism, Adrenergic beta-Antagonists pharmacology, Angiotensin Receptor Antagonists pharmacology, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Receptor, Angiotensin, Type 2 agonists, Stress, Physiological immunology
- Abstract
In order to determine differences in cardiovascular cell response during nutrient stress to different cardiovascular protective drugs, we investigated cell responses of serum starved mouse cardiomyocyte HL-1 cells and primary cultures of human coronary artery vascular smooth muscles (hCAVSMCs) to treatment with β-blockers (atenolol, metoprolol, carvedilol, nebivolol, 3 μM each), AT1R blocker losartan (1 μM) and AT2R agonists (CGP42112A and novel agonist NP-6A4, 300 nM each). Treatment with nebivolol, carvedilol, metoprolol and atenolol suppressed Cell Index (CI) of serum-starved HL-1 cells (≤17%, ≤8%, ≤15% and ≤15% respectively) as measured by the Xcelligence Real-Time Cell Analyzer (RTCA). Conversely, CI was increased by Ang II (≥9.6%), CGP42112A (≥14%), and NP-6A4 (≥25%) respectively and this effect was blocked by AT2R antagonist PD123319, but not by AT1R antagonist losartan. Thus, the CI signature for each drug could be unique. MTS cell proliferation assay showed that NP-6A4, but not other drugs, increased viability (≥20%) of HL-1 and hCAVSMCs. Wheat Germ Agglutinin (WGA) staining showed that nebivolol was most effective in reducing cell sizes of HL-1 and hCAVSMCs. Myeloid Cell Leukemia 1 (MCL-1) is a protein critical for cardiovascular cell survival and implicated in cell adhesion. β-blockers significantly suppressed and NP-6A4 increased MCL-1 expression in HL-1 and hCAVSMCs as determined by immunofluorescence. Thus, reduction in cell size and/or MCL-1 expression might underlie β-blocker-induced reduction in CI of HL-1. Conversely, increase in cell viability and MCL-1 expression by NP-6A4 through AT2R could have resulted in NP-6A4 mediated increase in CI of HL-1. These data show for the first time that activation of the AT2R-MCL-1 axis by NP-6A4 in nutrient-stressed mouse and human cardiovascular cells (mouse HL-1 cells and primary cultures of hCAVSMCs) might underlie improved survival of cells treated by NP-6A4 compared to other drugs tested in this study.
- Published
- 2015
- Full Text
- View/download PDF
36. Regulation of cardiac miR-208a, an inducer of obesity, by rapamycin and nebivolol.
- Author
-
Gul R, Mahmood A, Luck C, Lum-Naihe K, Alfadda AA, Speth RC, and Pulakat L
- Subjects
- Angiotensin II pharmacology, Animals, Cells, Cultured, Gene Expression Regulation drug effects, Male, Mediator Complex physiology, Mice, MicroRNAs metabolism, Myocytes, Cardiac drug effects, Obesity metabolism, Obesity pathology, Rats, Rats, Zucker, Signal Transduction drug effects, Signal Transduction genetics, Weight Gain drug effects, Weight Gain genetics, MicroRNAs genetics, Myocytes, Cardiac metabolism, Nebivolol pharmacology, Obesity genetics, Sirolimus pharmacology
- Abstract
Objective: Resistance to obesity is observed in rodents and humans treated with rapamycin (Rap) or nebivolol (Neb). Because cardiac miR-208a promotes obesity, this study tested whether the modes of actions of Rap and Neb involve inhibition of miR-208a., Methods: Mouse cardiomyocyte HL-1 cells and Zucker obese (ZO) rats were used to investigate regulation of cardiac miR-208a., Results: Angiotensin II (Ang II) increased miR-208a expression in HL-1 cells. Pretreatment with an AT1 receptor (AT1R) antagonist, losartan (1 μM), antagonized this effect, whereas a phospholipase C inhibitor, U73122 (10 μM), and an NADPH oxidase inhibitor, apocynin (0.5 mM), did not. Ang II-induced increase in miR-208a was suppressed by Rap (10 nM), an inhibitor of nutrient sensor kinase mTORC1, and Neb (1 μM), a 3rd generation β-blocker that suppressed bioavailable AT1R binding of (125) I-Ang II. Thus, suppression of AT1R expression by Neb, inhibition of AT1R activation by losartan, and inhibition of AT1R-induced activation of mTORC1 by Rap attenuated the Ang II-induced increase in miR-208a. In ZO rats, Rap treatment (750 μg kg(-1) day(-1) ; 12 weeks) reduced obesity despite similar food intake, suppressed cardiac miR-208a, and increased cardiac MED13, a suppresser of obesity., Conclusions: Rap and Neb suppressed cardiac miR-208a. Suppression of miR-208a and increase in MED13 correlated with attenuated weight gain despite leptin resistance., (© 2015 The Obesity Society.)
- Published
- 2015
- Full Text
- View/download PDF
37. The two faces of miR-29.
- Author
-
Ślusarz A and Pulakat L
- Subjects
- Animals, Diabetes Complications genetics, Diabetic Angiopathies genetics, Gene Expression Regulation drug effects, Genetic Predisposition to Disease, Humans, Immunosuppressive Agents adverse effects, Myeloid Cell Leukemia Sequence 1 Protein genetics, Neoplasms genetics, Sirolimus adverse effects, Cardiovascular Diseases genetics, Diabetes Mellitus genetics, MicroRNAs genetics
- Abstract
Diabetes mellitus is a metabolic homeostasis disease that contributes to additional comorbidities such as cardiovascular disease (CVD) and cancer. It has a long undiagnosed latent period during which there can be irreparable damage to the pancreas and cardiovascular tissues. Recent studies have highlighted the roles of several microRNAs in CVD. Determining the microRNAs that link diabetes mellitus and CVD is an important topic to be explored. In the present review, we discuss the microRNAs that contribute to the progression of diabetes mellitus and CVD and focus on the miR-29 family microRNAs whose expression is upregulated by hyperglycemia and proinflammatory cytokines, the hallmarks of diabetes mellitus. Upregulation of miR-29 expression is a key factor in the loss of pancreatic β cells and development of the first stage of type 1 diabetes mellitus (T1DM). Additionally, miR-29-mediated suppression of myeloid cell leukemia 1 (MCL-1), an important prosurvival protein, underlies Marfan's syndrome, abdominal aortic aneurysm, and diabetes mellitus-associated cardiomyocyte disorganization. Suppression of miR-29 expression and subsequent increase in the prosurvival MCL-1, however, promotes tumor development. Therefore, miR-29 mimics that suppress MCL-1 are hailed as tumor suppressors. The critical question is whether an increase in miR-29 levels is well tolerated in conditions of comorbidities in which insulin resistance is an underlying disease. In light of increasing awareness of the interconnection of diabetes mellitus, CVD, and cancer, it is of utmost importance to understand the mechanism of action of current treatment options on all of the comorbidities and careful evaluation of cardiovascular toxicity must accompany any treatment paradigm that increases miR-29 levels.
- Published
- 2015
- Full Text
- View/download PDF
38. Mineralocorticoid receptor antagonism treats obesity-associated cardiac diastolic dysfunction.
- Author
-
Bender SB, DeMarco VG, Padilla J, Jenkins NT, Habibi J, Garro M, Pulakat L, Aroor AR, Jaffe IZ, and Sowers JR
- Subjects
- Animals, Diastole, Disease Models, Animal, Echocardiography, Heart Ventricles diagnostic imaging, Heart Ventricles physiopathology, Rats, Rats, Zucker, Receptors, Mineralocorticoid metabolism, Ventricular Dysfunction, Left etiology, Ventricular Dysfunction, Left physiopathology, Mineralocorticoid Receptor Antagonists pharmacology, Obesity complications, Spironolactone pharmacology, Stroke Volume drug effects, Ventricular Dysfunction, Left drug therapy
- Abstract
Patients with obesity and diabetes mellitus exhibit a high prevalence of cardiac diastolic dysfunction (DD), an independent predictor of cardiovascular events for which no evidence-based treatment exists. In light of renin-angiotensin-aldosterone system activation in obesity and the cardioprotective action of mineralocorticoid receptor (MR) antagonists in systolic heart failure, we examined the hypothesis that MR blockade with a blood pressure-independent low-dose spironolactone (LSp) would treat obesity-associated DD in the Zucker obese (ZO) rat. Treatment of ZO rats exhibiting established DD with LSp normalized cardiac diastolic function, assessed by echocardiography. This was associated with reduced cardiac fibrosis, but not reduced hypertrophy, and restoration of endothelium-dependent vasodilation of isolated coronary arterioles via a nitric oxide-independent mechanism. Further mechanistic studies revealed that LSp reduced cardiac oxidative stress and improved endothelial insulin signaling, with no change in arteriolar stiffness. Infusion of Sprague-Dawley rats with the MR agonist aldosterone reproduced the DD noted in ZO rats. In addition, improved cardiac function in ZO-LSp rats was associated with attenuated systemic and adipose inflammation and an anti-inflammatory shift in cardiac immune cell mRNAs. Specifically, LSp increased cardiac markers of alternatively activated macrophages and regulatory T cells. ZO-LSp rats had unchanged blood pressure, serum potassium, systemic insulin sensitivity, or obesity-associated kidney injury, assessed by proteinuria. Taken together, these data demonstrate that MR antagonism effectively treats established obesity-related DD via blood pressure-independent mechanisms. These findings help identify a particular population with DD that might benefit from MR antagonist therapy, specifically patients with obesity and insulin resistance., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
39. Regulation of cardiac expression of the diabetic marker microRNA miR-29.
- Author
-
Arnold N, Koppula PR, Gul R, Luck C, and Pulakat L
- Subjects
- Animals, Blood Glucose drug effects, Diabetes Complications pathology, Gene Expression Regulation drug effects, Heart Diseases etiology, Heart Diseases pathology, Humans, Insulin blood, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells pathology, Mechanistic Target of Rapamycin Complex 1, Mice, MicroRNAs genetics, Multiprotein Complexes antagonists & inhibitors, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Rats, Sirolimus administration & dosage, TOR Serine-Threonine Kinases antagonists & inhibitors, Diabetes Complications genetics, Heart Diseases genetics, MicroRNAs biosynthesis, Multiprotein Complexes metabolism, Myeloid Cell Leukemia Sequence 1 Protein biosynthesis, TOR Serine-Threonine Kinases metabolism
- Abstract
Diabetes mellitus (DM) is an independent risk factor for heart disease and its underlying mechanisms are unclear. Increased expression of diabetic marker miR-29 family miRNAs (miR-29a, b and c) that suppress the pro-survival protein Myeloid Cell Leukemia 1(MCL-1) is reported in pancreatic β-cells in Type 1 DM. Whether an up-regulation of miR-29 family miRNAs and suppression of MCL-1 (dysregulation of miR-29-MCL-1 axis) occurs in diabetic heart is not known. This study tested the hypothesis that insulin regulates cardiac miR-29-MCL-1 axis and its dysregulation correlates with DM progression. In vitro studies with mouse cardiomyocyte HL-1 cells showed that insulin suppressed the expression of miR-29a, b and c and increased MCL-1 mRNA. Conversely, Rapamycin (Rap), a drug implicated in the new onset DM, increased the expression of miR-29a, b and c and suppressed MCL-1 and this effect was reversed by transfection with miR-29 inhibitors. Rap inhibited mammalian target of rapamycin complex 1 (mTORC1) signaling in HL-1 cells. Moreover, inhibition of either mTORC1 substrate S6K1 by PF-4708671, or eIF4E-induced translation by 4E1RCat suppressed MCL-1. We used Zucker diabetic fatty (ZDF) rat, a rodent model for DM, to test whether dysregulation of cardiac miR-29-MCL-1 axis correlates with DM progression. 11-week old ZDF rats exhibited significantly increased body weight, plasma glucose, insulin, cholesterol, triglycerides, body fat, heart weight, and decreased lean muscle mass compared to age-matched lean rats. Rap treatment (1.2 mg/kg/day, from 9-weeks to 15-weeks) significantly reduced plasma insulin, body weight and heart weight, and severely dysregulated cardiac miR-29-MCL1 axis in ZDF rats. Importantly, dysregulation of cardiac miR-29-MCL-1 axis in ZDF rat heart correlated with cardiac structural damage (disorganization or loss of myofibril bundles). We conclude that insulin and mTORC1 regulate cardiac miR-29-MCL-1 axis and its dysregulation caused by reduced insulin and mTORC1 inhibition increases the vulnerability of a diabetic heart to structural damage.
- Published
- 2014
- Full Text
- View/download PDF
40. The β-blocker Nebivolol Is a GRK/β-arrestin biased agonist.
- Author
-
Erickson CE, Gul R, Blessing CP, Nguyen J, Liu T, Pulakat L, Bastepe M, Jackson EK, and Andresen BT
- Subjects
- Animals, Arrestins genetics, Carbazoles pharmacology, Carvedilol, Cell Line, ErbB Receptors metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Fibroblasts drug effects, Fibroblasts metabolism, MAP Kinase Signaling System, Mice, Nebivolol, Nitric Oxide metabolism, Phosphorylation, Propanolamines pharmacology, Protein Processing, Post-Translational, Protein Transport, Receptors, Adrenergic, beta-2 metabolism, beta-Arrestin 1, beta-Arrestin 2, beta-Arrestins, Adrenergic beta-1 Receptor Antagonists pharmacology, Arrestins metabolism, Benzopyrans pharmacology, Ethanolamines pharmacology, G-Protein-Coupled Receptor Kinase 2 metabolism
- Abstract
Nebivolol, a third generation β-adrenoceptor (β-AR) antagonist (β-blocker), causes vasodilation by inducing nitric oxide (NO) production. The mechanism via which nebivolol induces NO production remains unknown, resulting in the genesis of much of the controversy regarding the pharmacological action of nebivolol. Carvedilol is another β-blocker that induces NO production. A prominent pharmacological mechanism of carvedilol is biased agonism that is independent of Gαs and involves G protein-coupled receptor kinase (GRK)/β-arrestin signaling with downstream activation of the epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK). Due to the pharmacological similarities between nebivolol and carvedilol, we hypothesized that nebivolol is also a GRK/β-arrestin biased agonist. We tested this hypothesis utilizing mouse embryonic fibroblasts (MEFs) that solely express β2-ARs, and HL-1 cardiac myocytes that express β1- and β2-ARs and no detectable β3-ARs. We confirmed previous reports that nebivolol does not significantly alter cAMP levels and thus is not a classical agonist. Moreover, in both cell types, nebivolol induced rapid internalization of β-ARs indicating that nebivolol is also not a classical β-blocker. Furthermore, nebivolol treatment resulted in a time-dependent phosphorylation of ERK that was indistinguishable from carvedilol and similar in duration, but not amplitude, to isoproterenol. Nebivolol-mediated phosphorylation of ERK was sensitive to propranolol (non-selective β-AR-blocker), AG1478 (EGFR inhibitor), indicating that the signaling emanates from β-ARs and involves the EGFR. Furthermore, in MEFs, nebivolol-mediated phosphorylation of ERK was sensitive to pharmacological inhibition of GRK2 as well as siRNA knockdown of β-arrestin 1/2. Additionally, nebivolol induced redistribution of β-arrestin 2 from a diffuse staining pattern into more intense punctate spots. We conclude that nebivolol is a β2-AR, and likely β1-AR, GRK/β-arrestin biased agonist, which suggests that some of the unique clinically beneficial effects of nebivolol may be due to biased agonism at β1- and/or β2-ARs.
- Published
- 2013
- Full Text
- View/download PDF
41. Renin inhibition and AT(1)R blockade improve metabolic signaling, oxidant stress and myocardial tissue remodeling.
- Author
-
Whaley-Connell A, Habibi J, Rehmer N, Ardhanari S, Hayden MR, Pulakat L, Krueger C, Ferrario CM, DeMarco VG, and Sowers JR
- Subjects
- Aldosterone blood, Amides pharmacology, Animals, Blood Pressure, Cell Size, Drug Interactions, Fibrosis, Fumarates pharmacology, Mice, Myocardium ultrastructure, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Rats, Rats, Sprague-Dawley, Rats, Transgenic, Renin genetics, Signal Transduction, Tetrazoles pharmacology, Valine analogs & derivatives, Valine pharmacology, Valsartan, Ventricular Remodeling, Angiotensin II Type 1 Receptor Blockers pharmacology, Myocardium pathology, Oxidative Stress, Receptor, Angiotensin, Type 1 metabolism, Renin antagonists & inhibitors
- Abstract
Objective: Strategies that block angiotensin II actions on its angiotensin type 1 receptor or inhibit actions of aldosterone have been shown to reduce myocardial hypertrophy and interstitial fibrosis in states of insulin resistance. Thereby, we sought to determine if combination of direct renin inhibition with angiotensin type 1 receptor blockade in vivo, through greater reductions in systolic blood pressure (SBP) and aldosterone would attenuate left ventricular hypertrophy and interstitial fibrosis to a greater extent than either intervention alone., Materials/methods: We utilized the transgenic Ren2 rat which manifests increased tissue expression of murine renin which, in turn, results in increased renin-angiotensin system activity, aldosterone secretion and insulin resistance. Ren2 rats were treated with aliskiren, valsartan, the combination (aliskiren+valsartan), or vehicle for 21 days., Results: Compared to Sprague-Dawley controls, Ren2 rats displayed increased systolic blood pressure, elevated serum aldosterone levels, cardiac tissue hypertrophy, interstitial fibrosis and ultrastructural remodeling. These biochemical and functional alterations were accompanied by increases in the NADPH oxidase subunit Nox2 and 3-nitrotyrosine content along with increases in mammalian target of rapamycin and reductions in protein kinase B phosphorylation. Combination therapy contributed to greater reductions in systolic blood pressure and serum aldosterone but did not result in greater improvement in metabolic signaling or markers of oxidative stress, fibrosis or hypertrophy beyond either intervention alone., Conclusions: Thereby, our data suggest that the greater impact of combination therapy on reductions in aldosterone does not translate into greater reductions in myocardial fibrosis or hypertrophy in this transgenic model of tissue renin overexpression., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
42. Angiotensin type 1 receptor resistance to blockade in the opossum proximal tubule cell due to variations in the binding pocket.
- Author
-
Nistala R, Andresen BT, Pulakat L, Meuth A, Sinak C, Mandavia C, Thekkumkara T, Speth RC, Whaley-Connell A, and Sowers JR
- Subjects
- Angiotensin II metabolism, Angiotensin II pharmacology, Animals, Binding Sites, Cell Line, Drug Resistance genetics, Humans, Iodine Radioisotopes, Janus Kinase 2 metabolism, Kidney Tubules, Proximal cytology, Phylogeny, Polymorphism, Genetic genetics, Protein Structure, Secondary, Protein Structure, Tertiary, RNA, Messenger genetics, Rats, Receptor, Angiotensin, Type 1 metabolism, Species Specificity, Vasoconstrictor Agents metabolism, Vasoconstrictor Agents pharmacology, Angiotensin II Type 1 Receptor Blockers pharmacology, Imidazoles pharmacology, Kidney Tubules, Proximal drug effects, Opossums genetics, Receptor, Angiotensin, Type 1 chemistry, Receptor, Angiotensin, Type 1 genetics, Tetrazoles pharmacology
- Abstract
Blockade of the angiotensin (ANG) II receptor type 1 (AT(1)R) with angiotensin receptor blockers (ARBs) is widely used in the treatment of hypertension. However, ARBs are variably effective in reducing blood pressure, likely due, in part, to polymorphisms in the ARB binding pocket of the AT(1)R. Therefore, we need a better understanding of variations/polymorphisms that alter binding of ARBs in heterogeneous patient populations. The opossum proximal tubule cell (OKP) line is commonly used in research to evaluate renal sodium handling and therefore blood pressure. Investigating this issue, we found natural sequence variations in the opossum AT(1)R paralleling those observed in the human AT(1)R. Therefore, we posited that these sequence variations may explain ARB resistance. We demonstrate that OKP cells express AT(1)R mRNA, bind (125)I-labeled ANG II, and exhibit ANG II-induced phosphorylation of Jak2. However, Jak2 phosphorylation is not inhibited by five different ARBs commonly used to treat hypertension. Additionally, nonradioactive ANG II competes (125)I-ANG II efficiently, whereas a 10-fold molar excess of olmesartan and the ANG II receptor type 2 blocker PD-123319 is unable to block (125)I-ANG II binding. In contrast, ANG II binding to OKP cells stably expressing rat AT(1A)Rs, which have a conserved AT(1)R-binding pocket with human AT(1)R, is efficiently inhibited by olmesartan. A novel observation was that resistance to ARB binding to opossum AT(1)Rs correlates with variations from the human receptor at positions 108, 163, 192, and 198 within the ARB-binding pocket. These observations highlight the potential utility of evaluating AT(1)R polymorphisms within the ARB-binding pocket in various hypertensive populations.
- Published
- 2013
- Full Text
- View/download PDF
43. RAS-Mediated Adaptive Mechanisms in Cardiovascular Tissues: Confounding Factors of RAS Blockade Therapy and Alternative Approaches.
- Author
-
Gul R, Ramdas M, Mandavia CH, Sowers JR, and Pulakat L
- Abstract
Since the classic experiments by Tigerstedt and Bergman that established the role of renin in hypertension a century ago, aggressive efforts have been launched to effectively block the renin-angiotensin system (RAS). Blockade of RAS is advocated at multiple levels by direct renin inhibitor, angiotensin-converting enzyme inhibitor and/or angiotensin II type 1 receptor blocker, or aldosterone inhibitor (spironolactone), and has now become part of the standard of care to control hypertension and related metabolic diseases including diabetes. However, recent lessons learned from randomized clinical trials question the wisdom of blocking RAS at multiple levels. In this context, it is highly pertinent that components of RAS are evolutionarily conserved, and novel physiological/adaptive/protective roles for renin and angiotensin-converting enzyme are currently emerging. Angiotensin II, the classical RAS effector peptide responsible for hypertension, hypertrophy, fluid retention and fibrosis, manifests its cardiovascular protective effect when it activates the angiotensin II type 2 receptor. Additionally, angiotensin-converting enzyme 2 and the angiotensin II metabolite Ang-(1-7) that acts through the Mas proto-oncogene constitute the cardiovascular and renal protective branch of RAS. It is conceivable that modulating this vasodilative/anti-inflammatory branch of RAS by activation of the RAS components that constitute this branch may offer a safer long-term treatment strategy to balance RAS activity and achieve homeostasis compared to chronic multilevel RAS inhibition.
- Published
- 2012
- Full Text
- View/download PDF
44. Over-nutrition and metabolic cardiomyopathy.
- Author
-
Mandavia CH, Pulakat L, DeMarco V, and Sowers JR
- Subjects
- Animals, Cardio-Renal Syndrome etiology, Cardio-Renal Syndrome metabolism, Endoplasmic Reticulum metabolism, Fatty Acids metabolism, Fibrosis etiology, Fibrosis metabolism, Glucose metabolism, Heart Failure, Systolic etiology, Humans, Insulin metabolism, Insulin Resistance, Magnetic Resonance Imaging, Cine, Myocardium pathology, Obesity complications, Obesity etiology, Obesity metabolism, Signal Transduction, Cardiomyopathy, Hypertrophic etiology, Cardiomyopathy, Hypertrophic metabolism, Heart Failure, Systolic metabolism, Myocardium metabolism, Overnutrition complications, Overnutrition metabolism, Oxidative Stress
- Abstract
Cardiovascular disease, which accounts for the highest morbidity and mortality in the United States, has several major risk factors, including aging and diabetes. Overweight and obesity, especially abdominal obesity, have been increasingly implicated as independent risk factors in the development of cardiovascular disease. Metabolic and/or diabetic cardiomyopathy has been especially associated with excess body weight caused by chronic over-nutrition and high-fat feeding. In the initial stages, obesity is now understood to cause significant dysregulation of cardiac fatty acid and glucose metabolism. These abnormalities are due, in part, to increased oxidative stress, which in turn can cause deleterious effects on intracellular signaling pathways that control cellular growth and proliferation. This increase in oxidative stress is coupled with reduced anti-oxidant species and dysregulation of metabolic signaling pathways. The cardiomyopathy seen with obesity is associated with increased interstitial fibrosis and diastolic dysfunction. Over time, evolving abnormalities include hypertrophy and systolic dysfunction, eventually leading to heart failure., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
45. Regulation of Overnutrition-Induced Cardiac Inflammatory Mechanisms.
- Author
-
Gul R, Demarco VG, Sowers JR, Whaley-Connell A, and Pulakat L
- Abstract
BACKGROUND: Unlike conventional β-blockers, nebivolol, a third-generation β-adrenergic receptor blocker with vasodilator properties, promotes insulin sensitivity. Objective: The objective of this study was to determine whether nebivolol regulates overnutrition-induced activation of cardiac nutrient sensor kinases and inflammatory signaling. METHODS: Young Zucker obese (ZO) rats, a rodent model for overnutrition, and age-matched Zucker lean rats were treated with nebivolol (10 mg/kg/day; 21 days) and cardiac function was monitored by echocardiography and pressure volume loop analysis. Activation status of nutrient sensor serine/threonine kinases mammalian target for rapamycin (mTOR), and p70 S6kinase (S6K1) and S6K1-substrate RPS6, inflammatory marker Janus kinase 2 (Jak2) and its substrate STAT1, and energy sensor AMP-dependent kinase (AMPK) were monitored by determining phosphorylation status of pSer(2448) of mTOR, pThr(389) of S6K1, pSer(235/236) of RPS6, pTyr(1007/1008) of Jak2, pTyr(701) of STAT1, and pThr(172) of AMPK, respectively. RESULTS: Nebivolol reduced weight and improved cardiac function of ZO rats as shown by improvements in the myocardial performance index and a decrease in the diastolic parameter tau (τ), the time constant of isovolumic relaxation. Nebivolol also attenuated excessive activation of the nutrient sensor kinases mTOR and S6K1 and their substrate RPS6 as well as the inflammatory marker Jak2 and substrate STAT1 in ZO myocardium (p < 0.05). Moreover, nebivolol reversed suppression of the energy sensor kinase AMPK in ZO hearts (p < 0.05). CONCLUSION: We report for the first time that nebivolol regulates overnutrition-induced activation of cardiac mTOR and Jak/STAT signaling and reverses suppression of AMPK. Since it also suppresses weight gain, nebivolol appears effective in the treatment of overnutrition-related cardiac inflammation and diastolic dysfunction.
- Published
- 2012
- Full Text
- View/download PDF
46. Combination of direct renin inhibition with angiotensin type 1 receptor blockade improves aldosterone but does not improve kidney injury in the transgenic Ren2 rat.
- Author
-
Whaley-Connell A, Habibi J, Nistala R, Hayden MR, Pulakat L, Sinak C, Locher B, Ferrario CM, and Sowers JR
- Subjects
- Acute Kidney Injury drug therapy, Aldosterone biosynthesis, Amides pharmacology, Animals, Fumarates pharmacology, Rats, Rats, Sprague-Dawley, Rats, Transgenic, Renin genetics, Renin metabolism, Tetrazoles pharmacology, Valine analogs & derivatives, Valine pharmacology, Valsartan, Acute Kidney Injury metabolism, Aldosterone metabolism, Angiotensin II Type 1 Receptor Blockers pharmacology, Receptor, Angiotensin, Type 1 metabolism, Renin antagonists & inhibitors
- Abstract
Enhanced renin-angiotensin-aldosterone system (RAAS) activation contributes to proteinuria and chronic kidney disease by increasing glomerular and tubulointerstitial oxidative stress, promotion of fibrosis. Renin activation is the rate limiting step in angiotensin (Ang II) and aldosterone generation, and recent work suggests direct renin inhibition improves proteinuria comparable to that seen with Ang type 1 receptor (AT(1)R) blockade. This is important as, even with contemporary use of AT(1)R blockade, the burden of kidney disease remains high. Thereby, we sought to determine if combination of direct renin inhibition with AT(1)R blockade in vivo, via greater attenuation of kidney oxidative stress, would attenuate glomerular and proximal tubule injury to a greater extent than either intervention alone. We utilized the transgenic Ren2 rat with increased tissue RAS activity and higher serum levels of aldosterone, which manifests hypertension and proteinuria. Ren2 rats were treated with renin inhibition (aliskiren), AT(1)R blockade (valsartan), the combination (aliskiren+valsartan), or vehicle for 21days. Compared to Sprague-Dawley controls, Ren2 rats displayed increased systolic pressure (SBP), circulating aldosterone, proteinuria and greater urine levels of the proximal tubule protein excretory marker beta-N-acetylglucosaminidase (β-NAG). These functional and biochemical alterations were accompanied by increases in kidney tissue NADPH oxidase subunit Rac1 and 3-nitrotyrosine (3-NT) content as well as fibronectin and collagen type III. These findings occurred in conjunction with reductions in the podocyte-specific protein podocin as well as the proximal tubule-specific megalin. Further, in transgenic animals there was increased tubulointerstitial fibrosis on light microscopy as well as ultrastructural findings of glomerular podocyte foot-process effacement and reduced tubular apical endosomal/lysosomal activity. Combination therapy led to greater reductions in SBP and serum aldosterone, but did not result in greater improvement in markers of glomerular and tubular injury (i.e. β-NAG) compared to either intervention alone. Further, combination therapy did not improve markers of oxidative stress and podocyte and proximal tubule integrity in this transgenic model of RAAS-mediated kidney damage despite greater reductions in serum aldosterone and BP levels., (Published by Elsevier B.V.)
- Published
- 2012
- Full Text
- View/download PDF
47. Nebivolol improves diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the transgenic (mRen2) rat.
- Author
-
Ma L, Gul R, Habibi J, Yang M, Pulakat L, Whaley-Connell A, Ferrario CM, and Sowers JR
- Subjects
- AMP-Activated Protein Kinase Kinases, Animals, Diastole physiology, Disease Models, Animal, Fibrosis, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular physiopathology, Male, Myocardium metabolism, Myocardium pathology, NADPH Oxidases metabolism, Nebivolol, Nitric Oxide metabolism, Oxidative Stress physiology, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Rats, Rats, Sprague-Dawley, Rats, Transgenic, Ventricular Dysfunction, Left pathology, Ventricular Dysfunction, Left physiopathology, Ventricular Remodeling physiology, Adrenergic beta-1 Receptor Antagonists pharmacology, Benzopyrans pharmacology, Diastole drug effects, Ethanolamines pharmacology, Oxidative Stress drug effects, Renin genetics, Renin physiology, Ventricular Remodeling drug effects
- Abstract
Angiotensin II contributes to myocardial tissue remodeling and interstitial fibrosis through NADPH oxidase-mediated generation of oxidative stress in the progression of heart failure. Recent data have suggested that nebivolol, a third-generation β-blocker, improves diastolic dysfunction by targeting nitric oxide (NO) and metabolic pathways that decrease interstitial fibrosis. We sought to determine if targeting NO would improve diastolic function in a model of tissue renin-angiotensin system overactivation. We used the transgenic (TG) (mRen2)27 rat, which overexpresses the murine renin transgene and manifests insulin resistance and left ventricular dysfunction. We treated 6- to 7-wk-old TG (mRen2)27 rats and age-matched Sprague-Dawley control rats with nebivolol (10 mg·kg(-1)·day(-1)) or placebo via osmotic minipumps for a period of 21 days. Compared with Sprague-Dawley control rats, TG (mRen2)27 rats displayed a prolonged diastolic relaxation time and reduced initial filling rate associated with increased interstitial fibrosis and left ventricular hypertrophy. These findings were temporally related to increased NADPH oxidase activity and subunits p47(phox) and Rac1 and increased total ROS and peroxynitrite formation in parallel with reductions in the antioxidant heme oxygenase as well as the phosphorylation/activation of endothelial NO synthase and PKB/Akt. Treatment with nebivolol restored diastolic function and interstitial fibrosis through increases in the phosphorylation of 5'-AMP-activated protein kinase, Akt, and endothelial NO synthase and reductions in oxidant stress. These results support that targeting NO with nebivolol treatment improves diastolic dysfunction through reducing myocardial oxidative stress by enhancing 5'-AMP-activated protein kinase and Akt activation of NO biosynthesis.
- Published
- 2012
- Full Text
- View/download PDF
48. Mitochondria and Oxidative Stress in the Cardiorenal Metabolic Syndrome.
- Author
-
Aroor AR, Mandavia C, Ren J, Sowers JR, and Pulakat L
- Abstract
Mitochondria play a fundamental role in the maintenance of normal structure, function, and survival of tissues. There is considerable evidence for mitochondrial dysfunction in association with metabolic diseases including insulin resistance, obesity, diabetes, and the cardiorenal metabolic syndrome. The phenomenon of reactive oxygen species (ROS)-induced ROS release through interactions between cytosolic and mitochondrial oxidative stress contributes to a vicious cycle of enhanced oxidative stress and mitochondrial dysfunction. Activation of the cytosolic and mitochondrial NADPH oxidase system, impairment of the mitochondrial electron transport, activation of p66shc pathway-targeting mitochondria, endoplasmic reticular stress, and activation of the mammalian target of the rapamycin-S6 kinase pathway underlie dysregulation of mitochondrial dynamics and promote mitochondrial oxidative stress. These processes are further modulated by acetyltransferases including sirtuin 1 and sirtuin 3, the former regulating nuclear acetylation and the latter regulating mitochondrial acetylation. The regulation of mitochondrial functions by microRNAs forms an additional layer of molecular control of mitochondrial oxidative stress. Alcohol further exacerbates mitochondrial oxidative stress induced by overnutrition and promotes the development of metabolic diseases.
- Published
- 2012
- Full Text
- View/download PDF
49. Overweight female rats selectively breed for low aerobic capacity exhibit increased myocardial fibrosis and diastolic dysfunction.
- Author
-
DeMarco VG, Johnson MS, Ma L, Pulakat L, Mugerfeld I, Hayden MR, Garro M, Knight W, Britton SL, Koch LG, and Sowers JR
- Subjects
- AMP-Activated Protein Kinases metabolism, Aerobiosis genetics, Aerobiosis physiology, Animals, Baroreflex genetics, Baroreflex physiology, Blood Pressure physiology, Blotting, Western, Cardiac Catheterization, Citrate (si)-Synthase metabolism, Diastole physiology, Female, Fibrosis, Heart Function Tests, Hemodynamics physiology, Hydroxymethylglutaryl CoA Reductases metabolism, Immunohistochemistry, Magnetic Resonance Imaging, Male, Microscopy, Electron, Transmission, Myocardium ultrastructure, Overweight genetics, Physical Conditioning, Animal physiology, Physical Endurance physiology, Rats, Running physiology, Telemetry, Ventricular Remodeling physiology, Myocardium pathology, Overweight physiopathology, Oxygen Consumption genetics
- Abstract
The statistical association between endurance exercise capacity and cardiovascular disease suggests that impaired aerobic metabolism underlies the cardiovascular disease risk in men and women. To explore this connection, we applied divergent artificial selection in rats to develop low-capacity runner (LCR) and high-capacity runner (HCR) rats and found that disease risks segregated strongly with low running capacity. Here, we tested if inborn low aerobic capacity promotes differential sex-related cardiovascular effects. Compared with HCR males (HCR-M), LCR males (LCR-M) were overweight by 34% and had heavier retroperitoneal, epididymal, and omental fat pads; LCR females (LCR-F) were 20% heavier than HCR females (HCR-F), and their retroperitoneal, but not perireproductive or omental, fat pads were heavier as well. Unlike HCR-M, blood pressure was elevated in LCR-M, and this was accompanied by left ventricular (LV) hypertrophy. Like HCR-F, LCR-F exhibited normal blood pressure and LV weight as well as increased spontaneous cage activity compared with males. Despite normal blood pressures, LCR-F exhibited increased myocardial interstitial fibrosis and diastolic dysfunction, as indicated by increased LV stiffness, a decrease in the initial filling rate, and an increase in diastolic relaxation time. Although females exhibited increased arterial stiffness, ejection fraction was normal. Increased interstitial fibrosis and diastolic dysfunction in LCR-F was accompanied by the lowest protein levels of phosphorylated AMP-actived protein kinase [phospho-AMPK (Thr(172))] and silent information regulator 1. Thus, the combination of risk factors, including female sex, intrinsic low aerobic capacity, and overweightness, promote myocardial stiffness/fibrosis sufficient to induce diastolic dysfunction in the absence of hypertension and LV hypertrophy.
- Published
- 2012
- Full Text
- View/download PDF
50. Mineralocorticoid receptor-dependent proximal tubule injury is mediated by a redox-sensitive mTOR/S6K1 pathway.
- Author
-
Whaley-Connell AT, Habibi J, Nistala R, DeMarco VG, Pulakat L, Hayden MR, Joginpally T, Ferrario CM, Parrish AR, and Sowers JR
- Subjects
- Albuminuria metabolism, Albuminuria pathology, Animals, Animals, Genetically Modified, Blood Pressure, Immunohistochemistry methods, Male, Microscopy, Electron, Transmission methods, Oxidation-Reduction, Oxidative Stress, Rats, Rats, Sprague-Dawley, Ribosomal Protein S6 Kinases metabolism, Spironolactone metabolism, Kidney Tubules drug effects, Receptors, Mineralocorticoid metabolism, TOR Serine-Threonine Kinases metabolism
- Abstract
Background/aims: The mammalian target of rapamycin (mTOR) is a serine kinase that regulates phosphorylation (p) of its target ribosomal S6 kinase (S6K1), whose activation can lead to glomerular and proximal tubular cell (PTC) injury and associated proteinuria. Increased mTOR/S6K1 signaling regulates signaling pathways that target fibrosis through adherens junctions. Recent data indicate aldosterone signaling through the mineralocorticoid receptor (MR) can activate the mTOR pathway. Further, antagonism of the MR has beneficial effects on proteinuria that occur independent of hemodynamics., Methods: Accordingly, hypertensive transgenic TG(mRen2)27 (Ren2) rats, with elevated serum aldosterone and proteinuria, and age-matched Sprague-Dawley rats were treated with either a low dose (1 mg/kg/day) or a conventional dose (30 mg/kg/day) of spironolactone (MR antagonist) or placebo for 3 weeks., Results: Ren2 rats displayed increases in urine levels of the PTC brush border lysosomal enzyme N-acetyl-β-aminoglycosidase (β-NAG) in conjunction with reductions in PTC megalin, the apical membrane adherens protein T-cadherin and basolateral α-(E)-catenin, and fibrosis. In concert with these abnormalities, Ren2 renal cortical tissue also displayed increased Ser2448 (p)/activation of mTOR and Thr389 (p)-S6K1 and increased 3-nitrotyrosine (3-NT) content, a marker for peroxynitrite. Low-dose spironolactone had no effect on blood pressure but decreased proteinuria and β-NAG comparable to a conventional dose of this MR antagonist. Both doses of spironolactone attenuated ultrastructural maladaptive alterations and led to comparable reductions in (p)-mTOR/(p)-S6K1, 3-NT, fibrosis, and increased expression of α-(E)-catenin, T- and N-cadherin., Conclusions: Thereby, MR antagonism improves proximal tubule integrity by targeting mTOR/S6K1 signaling and redox status independent of changes in blood pressure., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.