98 results on '"Tallant EA"'
Search Results
2. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2.
- Author
-
Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, Diz DI, Gallagher PE, Ferrario, Carlos M, Jessup, Jewell, Chappell, Mark C, Averill, David B, Brosnihan, K Bridget, Tallant, E Ann, Diz, Debra I, and Gallagher, Patricia E
- Published
- 2005
- Full Text
- View/download PDF
3. Calmodulin antagonists elevate the levels of 32P-labeled polyphosphoinositides in human platelets
- Author
-
Tallant Ea and Robert W. Wallace
- Subjects
Blood Platelets ,Phosphatidylinositol 4,5-Diphosphate ,medicine.medical_specialty ,Perphenazine ,Chlorpromazine ,Biophysics ,Trifluoperazine ,Phosphatidylinositols ,Biochemistry ,chemistry.chemical_compound ,Calmodulin ,Phosphatidylinositol Phosphates ,Internal medicine ,medicine ,Humans ,Platelet ,Phosphatidylinositol ,Molecular Biology ,Promazine ,Sulfonamides ,Chemistry ,Cell Biology ,Metabolism ,Kinetics ,Tamoxifen ,Endocrinology ,Phosphatidylinositol 4,5-bisphosphate ,lipids (amino acids, peptides, and proteins) ,medicine.drug - Abstract
The calmodulin antagonists trifluoperazine, chlorpromazine, perphenazine, promazine, tamoxifen and the naphthalene sulfonamide derivatives W7 and W13 increased the level of 32P-incorporation into human platelet PIP and PIP2. Various drugs with poor anti-calmodulin activity were ineffective. The increase in 32P-PIP and 32P-PIP2 required micromolar concentrations of trifluoperazine and was time-dependent, reaching half-maximal within two minutes of the addition of the drug. These results indicate that the calmodulin antagonists perturb polyphosphoinositide metabolism, probably at the level of the PI- and PIP-kinases and/or the PIP2- and PIP-phosphomonoesterases.
- Published
- 1985
4. High levels of a heat-labile calmodulin-binding protein (CaM-BP80) in bovine neostriatum
- Author
-
Wai Yiu Cheung, Wallace Rw, and Tallant Ea
- Subjects
Hot Temperature ,Calmodulin ,Macromolecular Substances ,Radioimmunoassay ,Biochemistry ,Cyclase ,Drug Stability ,Postsynaptic potential ,Protein A/G ,Animals ,Tissue Distribution ,Brain Chemistry ,Kidney Medulla ,biology ,Chemistry ,Phosphoric Diester Hydrolases ,Binding protein ,Myocardium ,Phosphodiesterase ,Corpus Striatum ,Molecular Weight ,biology.protein ,Calmodulin-Binding Proteins ,Cattle ,Protein G ,Carrier Proteins ,Subcellular Fractions - Abstract
Bovine brain contains a heat-labile, 80,000-dalton calmodulin-binding protein (CaM-BP80) which inhibits the calmodulin-dependent activities of cyclic 3',5'-nucleotide phosphodiesterase, adenylate cyclase, and Ca2+-ATPase in vitro. CaM-BP80 is composed of two polypeptides (60,000 and 18,500 daltons) present in a 1:1 ratio. An antibody directed against CaM-BP80 was raised in rabbits, and a radioimmunoassay was developed, having a sensitivity of 60 fmol of CaM-BP80. Using the radioimmunoassay, we determined the levels of CaM-BP80 in various bovine tissues. The protein was found primarily in the brain, present in particularly high levels in the neostriatum. These results, together with immunohistochemical localization of CaM-BP80 at the postsynaptic densities and the microtubules of postsynaptic dendrites [Wood, J.G., Wallace, R., Whitaker, J., & Cheung, W.Y. (1980) J. Cell Biol. 84, 66-76], suggest that the protein may have a role in the cerebrum at the site of neurotransmitter action and at the level of microtubular function.
- Published
- 1980
5. Calmodulin-dependent protein phosphatase: a developmental study
- Author
-
Tallant Ea and Cheung Wy
- Subjects
Aging ,Calmodulin ,Ranidae ,Swine ,Phosphatase ,Synaptogenesis ,DUSP6 ,Chick Embryo ,Biochemistry ,Retina ,Mice ,Species Specificity ,Cerebellum ,medicine ,Phosphoprotein Phosphatases ,Animals ,biology ,Cerebrum ,Chemistry ,Fishes ,Brain ,Protein phosphatase 2 ,Regucalcin ,Rats ,Cytosol ,medicine.anatomical_structure ,biology.protein ,Calmodulin-Binding Proteins ,Cattle ,Chickens - Abstract
Calmodulin-dependent protein phosphatase, one of the major calmodulin-binding proteins in bovine brain, dephosphorylates casein with a specific activity of 15 nmol mg-1 min-1 at 30 degrees C. The stimulation of phosphatase activity by calmodulin is reversed by ethylene glycol bis (beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid or trifluoperazine, a calmodulin antagonist. Antibodies raised in rabbit against the phosphatase inhibit the enzyme activity. The levels of the protein in brain extracts from various animals, determined by a radioimmunoassay, range from 20 micrograms/g of tissue in chick and fish brains to 143 micrograms in rat cerebrum. The ontogeny of the phosphatase was studied in nervous tissues from rat and chick, animals in which synaptogenesis takes place at different times during their development. The levels of the protein increased significantly in rat cerebrum and cerebellum and in chick brain and retina during the periods corresponding to major synapse formation. In rat cerebrum, the enzyme appeared to be equally distributed between the cytosol and the particulate fraction; the level in both compartments increased during the major period of synapse formation. Thus, the development of calmodulin-dependent protein phosphatase closely parallels synaptogenesis, implicating a role in some synaptic function.
- Published
- 1983
6. [4] Assay of calmodulin by Ca2+-dependent phosphodiesterase
- Author
-
Tallant Ea, Wallace Rw, and Wai Yiu Cheung
- Subjects
chemistry.chemical_classification ,Calmodulin ,biology ,Proteolytic enzymes ,Phosphodiesterase ,Radioimmunoassay ,Adenosine ,Enzyme activator ,Enzyme ,chemistry ,Biochemistry ,Nucleotidase ,biology.protein ,medicine ,medicine.drug - Abstract
Publisher Summary Ca2+-dependent phosphodiesterase is a convenient calmodulin-dependent enzyme for use in determining tissue levels of calmodulin. The enzyme may be prepared in a calmodulin-deficient form by a single-column chromatography procedure. Moreover, the enzyme prepared in this manner is very stable and a single preparation of enzyme is sufficient for hundreds of calmodulin determinations. This chapter details the procedures to assay calmodulin based on its ability to stimulate a Ca2+-dependent phosphodiesterase from the bovine brain. It also discusses the pitfalls of the assay and the appropriate precautions to alleviate them. Calmodulin is assayed by its ability to stimulate phosphodiesterase activity as determined by a two-step assay procedure. During the first step of the assay, 2,8-3H-labeled cyclic adenosine 3'-5'-monophosphate (cAMP) is incubated with phosphodiesterase, which hydrolyzes the 3' bond producing adenosine 5'-monophosphate (5'-AMP). During the second step, 5'-AMP is quantitatively converted into adenosine and inorganic phosphate through the action of a 5'-nucleotidase. The adenosine is separated from unreacted cAMP by the batch use of an anion exchange resin. Snake venom is a convenient source of 5'-nucleotidase and exhibits no detectable phosphodiesterase activity. If the two steps of the assay are combined, phosphodiesterase is stimulated irreversibly by proteolytic enzymes present in the snake venom.
- Published
- 1983
7. [23] Purification and radioimmunoassay of calmodulin-dependent protein phosphatase from bovine brain
- Author
-
Wallace Rw, Wai Yiu Cheung, and Tallant Ea
- Subjects
Chromatography ,Ion exchange ,Calmodulin ,biology ,Chemistry ,Phosphatase ,Phosphodiesterase ,Radioimmunoassay ,chemistry.chemical_compound ,Affinity chromatography ,Bovine brain ,Biochemistry ,biology.protein ,Sodium dodecyl sulfate - Abstract
Publisher Summary This chapter first describes a simple procedure for purifying phosphatase from the bovine brain and discusses the development of a sensitive radioimmunoassay, including the establishment of its specificity and usefulness. Calmodulin-dependent protein phosphatase is purified from the bovine cerebral cortex by a procedure involving three chromatographic steps. Affi-Gel Blue chromatography first separates the phosphatase from calmodulin and Ca 2+ -dependent phosphodiesterase; next, a calmodulin-Sepharose affinity column resolves the bulk of the contaminants present; and finally, an ion exchange column removes the remaining contaminants. A double-antibody radioimmunoassay has been developed to measure quantitatively the amount of calmodulin-dependent phosphatase. The procedure was slightly modified to include some detergents, which allows the measurement of the protein in paniculate fractions. Two detergents are used in this procedure: sodium dodecyl sulfate (SDS), an anionic detergent, and Triton X-100, a nonionic detergent. The radioimmunoassay for calmodulin-dependent phosphatase is specific and sensitive and is accurate over a wide range of phosphatase levels. It has been useful in determining tissue levels, subcellular distribution, and ontogenic development of the phosphatase.
- Published
- 1983
8. Osteoarthritis as a Systemic Disease Promoted Prostate Cancer In Vivo and In Vitro.
- Author
-
Rosas S, Kwok A, Moore J, Shi L, Smith TL, Tallant EA, Kerr BA, and Willey JS
- Subjects
- Male, Animals, Mice, Cell Line, Tumor, Cell Movement drug effects, Humans, Disease Models, Animal, Interleukin-1alpha metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Cell Proliferation, Cartilage Oligomeric Matrix Protein metabolism, Cartilage Oligomeric Matrix Protein genetics, Osteoarthritis metabolism, Osteoarthritis pathology, Osteoarthritis etiology
- Abstract
Osteoarthritis (OA) is increasing worldwide, and previous work found that OA increases systemic cartilage oligomeric matrix protein (COMP), which has also been implicated in prostate cancer (PCa). As such, we sought to investigate whether OA augments PCa progression. Cellular proliferation and migration of RM1 murine PCa cells treated with interleukin (IL)-1α, COMP, IL-1α + COMP, or conditioned media from cartilage explants treated with IL-1α (representing OA media) and with inhibitors of COMP were assessed. A validated murine model was used for tumor growth and marker expression analysis. Both proliferation and migration were greater in PCa cells treated with OA media compared to controls ( p < 0.001), which was not seen with direct application of the stimulants. Migration and proliferation were not negatively affected when OA media was mixed with downstream and COMP inhibitors compared to controls ( p > 0.05 for all). Mice with OA developed tumors 100% of the time, whereas mice without OA only 83.4% ( p = 0.478). Tumor weight correlated with OA severity (Pearson correlation = 0.813, p = 0.002). Moreover, tumors from mice with OA demonstrated increased Ki-67 expression compared to controls (mean 24.56% vs. 6.91%, p = 0.004) but no difference in CD31, PSMA, or COMP expression ( p > 0.05). OA appears to promote prostate cancer in vitro and in vivo.
- Published
- 2024
- Full Text
- View/download PDF
9. Probiotic and Muscadine Grape Extract Interventions Shift the Gut Microbiome and Improve Metabolic Parameters in Female C57BL/6 Mice.
- Author
-
Newman TM, Wilson AS, Clear KYJ, Tallant EA, Gallagher PE, and Cook KL
- Subjects
- Female, Animals, Mice, Dysbiosis complications, RNA, Ribosomal, 16S, Diet, High-Fat, Mice, Inbred C57BL, Obesity metabolism, Inflammation metabolism, Gastrointestinal Microbiome, Vitis, Probiotics pharmacology
- Abstract
Obesity and Western-like diet consumption leads to gut microbiome dysbiosis, which is associated with the development of cardio-metabolic diseases and poor health outcomes. The objective of this study was to reduce Western diet-mediated gut microbial dysbiosis, metabolic dysfunction, and systemic inflammation through the administration of a novel combined intervention strategy (oral probiotic bacteria supplements and muscadine grape extract (MGE)). To do so, adult female C57BL/6 mice were fed a low-fat control or Western-style diet and sub-grouped into diet alone, probiotic intervention, antibiotic treatments, MGE supplementation, a combination of MGE and probiotics, or MGE and antibiotics for 13 weeks. Mouse body weight, visceral adipose tissue (VAT), liver, and mammary glands (MG) were weighed at the end of the study. Fecal 16S rRNA sequencing was performed to determine gut bacterial microbiome populations. Collagen, macrophage, and monocyte chemoattractant protein-1 (MCP-1) in the VAT and MG tissue were examined by immunohistochemistry. Adipocyte diameter was measured in VAT. Immunohistochemistry of intestinal segments was used to examine villi length, muscularis thickness, and goblet cell numbers. We show that dietary interventions in Western diet-fed mice modulated % body weight gain, visceral adiposity, MG weight, gut microbial populations, and inflammation. Intervention strategies in both diets effectively reduced VAT and MG fibrosis, VAT and MG macrophages, adipocyte diameter, and VAT and MG MCP-1. Interventions also improved intestinal health parameters. In conclusion, dietary intervention with MGE and probiotics modulates several microbial, inflammatory, and metabolic factors reducing poor health outcomes associated with Western diet intake.
- Published
- 2023
- Full Text
- View/download PDF
10. Study design and methods for the pilot study of muscadine grape extract supplement to improve fatigue among older adult cancer survivors (FOCUS) trial.
- Author
-
Klepin HD, Tooze JA, Bitting RL, Davis B, Pleasant K, Melo AC, Cook K, Soto-Pantoja DR, Tallant EA, and Gallagher PE
- Subjects
- Humans, Aged, Quality of Life, Pilot Projects, Fatigue drug therapy, Fatigue etiology, Double-Blind Method, Dietary Supplements, Cancer Survivors, Vitis, Neoplasms complications, Neoplasms drug therapy
- Abstract
Introduction: Fatigue is a prevalent symptom among both cancer survivors and older adults. Negative consequences of fatigue include increased sedentary behavior, decreased physical activity and function, and lower quality of life. Few pharmacologic interventions improve fatigue. Our preclinical and clinical data show promising effects of a muscadine grape extract supplement (MGES) on oxidative stress, mitochondrial bioenergetics, the microbiome, and the symptom of fatigue. This pilot study seeks to translate these observations to cancer survivorship by testing the preliminary effect of MGE supplementation on older adult cancer survivors with self-reported fatigue., Materials and Methods: We designed a double-blinded placebo-controlled pilot study to evaluate preliminary efficacy of MGE supplementation versus placebo on fatigue among older adult cancer survivors (aged ≥65 years) who report baseline fatigue. Sixty-four participants will be enrolled and randomized 1:1 to twice daily MGES (four tablets twice daily) versus placebo for 12 weeks. The primary outcome is change in Patient-Reported Outcomes Measurement Information System (PROMIS) Fatigue score from baseline to 12 weeks. Secondary outcomes are change in self-reported physical function, physical fitness (6-min walk test), self-reported physical activity, global quality of life (QOL), and the Fried frailty index. Correlative biomarker assays will assess changes in 8-hydroxy-2 deoxyguanosine, peripheral blood mitochondrial function, inflammatory markers, and the gut microbiome., Discussion: This pilot study builds on preclinical and clinical observations to estimate effects of MGE supplementation on fatigue, physical function, QOL, and biologic correlates in older adult cancer survivors. Trial registration #: CT.govNCT04495751; IND 152908., Competing Interests: Declaration of Competing Interest The authors have no conflicts of interest to report., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Targeted UHPLC-MS Analysis Reveals Disparate Polyphenol Composition and Concentration in Muscadine Grape Supplements with Proportional Antioxidant Activity.
- Author
-
Chappell MC, Duncan AV, Melo AC, Schaich CL, Pirro NT, Diz DI, Tallant EA, and Gallagher PE
- Abstract
Muscadine grape supplements (MGS) with high polyphenol content are a potential therapeutic option to combat oxidative stress; however, the precise identity and concentration of individual phenolics in commercially processed MGSs is not well defined. We probed for 17 phenolic compounds by ultra-high pressure liquid chromatography and mass spectroscopy from distinct lots of four commercially processed MGSs composed of MG seed and/or skin waste products. The total phenolic content (TPC) and antioxidant capacity were highest in a dried water-extract MGS as compared to three ground seed and/or skin products. The TPC was not different between MGS lots from individual companies and remained stable for 3 years without microbial contamination. The extract MGS had the highest concentration of epicatechin, ellagic acid, gallic acid, procyanidin B2, catechin and catechin gallate compared to the other supplements. Only ellagic acid and gallic acid were detected in all four MGSs, while catechin and catechin gallate were below detection in two supplements. Based on gram weight, only the extract MGS prevented the angiotensin II-induced increase in malondialdehyde and 4-hydroxynonenol in rat H9c2 cardiomyocytes as well as upregulated superoxide dismutase and catalase. This study demonstrates that commercial MGSs differ in phenolic composition and concentration, resulting in disparate antioxidant activity.
- Published
- 2022
- Full Text
- View/download PDF
12. A Polyphenol-Rich Extract from Muscadine Grapes Prevents Hypertension-Induced Diastolic Dysfunction and Oxidative Stress.
- Author
-
Patil PD, Melo AC, Westwood BM, Tallant EA, and Gallagher PE
- Abstract
Muscadine grapes are abundant in dietary polyphenols, but their effect on hypertension-induced cardiac damage is limited. This study assessed whether a muscadine grape skin/seed extract supplement (MGES) prevents hypertension-induced cardiac damage and oxidative stress. Male Sprague Dawley rats were treated for four weeks with drinking water, angiotensin II (Ang II) to induce hypertension, MGES, or both Ang II and MGES. Cardiac function assessed by echocardiography showed that Ang II increased systolic blood pressure while MGES alone or in combination with Ang II had no effect. Ang II increased E/e', an indicator of left ventricular filling pressure and diastolic dysfunction, by 41% compared to Control and co-treatment with MGES prevented the Ang II-mediated increase, suggesting that the extract attenuated hypertension-induced diastolic function. Ang II infusion increased urinary 8-hydroxy-2'-deoxyguanosine and cardiac 4-hydroxynonenal and malondialdehyde, which were prevented by the extract. The antioxidant enzymes catalase and superoxide dismutase 1 activity and mRNA were increased significantly in animals treated with MGES alone or in combination with Ang II, suggesting that the extract upregulates oxidative stress defense mechanisms in cardiac tissue. Thus, MGES may serve as a medical food to protect the heart from hypertension-induced diastolic dysfunction caused in part by excessive reactive oxygen species production.
- Published
- 2022
- Full Text
- View/download PDF
13. Angiotensin-(1-7) reduces doxorubicin-induced aortic arch dysfunction in male and female juvenile Sprague Dawley rats through pleiotropic mechanisms.
- Author
-
Rahimi O, Melo AC, Westwood B, Grier RDM, Tallant EA, and Gallagher PE
- Subjects
- Angiotensin I, Animals, Doxorubicin, Female, Humans, Male, Peptide Fragments, Pulse Wave Analysis, Rats, Rats, Sprague-Dawley, Angiotensin II, Aorta, Thoracic
- Abstract
Doxorubicin (Dox), an effective chemotherapeutic, can cause cumulative dose-dependent cardiovascular toxicity, which may manifest as vascular dysfunction leading to long-term end-organ damage. Currently, there are no effective treatments to mitigate Dox-induced vascular damage in cancer patients, particularly pediatric patients. We showed that angiotensin-(1-7) [Ang-(1-7)], an endogenous peptide hormone, mitigated cardiac damage in Dox-treated juvenile rats. In this study assessing aortic stiffness, juvenile male and female rats were administered a clinically equivalent dose of Dox (21-24 mg/kg) over 6 weeks, in the presence and absence of Ang-(1-7) [24 µg/kg/h]. Aortic function was measured using echocardiography. Ang-(1-7) reduced the Dox-mediated increase in pulse wave velocity, a measure of arterial stiffness (males: p < 0.05; females: p < 0.001) as compared in control animals. Dox decreased aortic lumen diameter (p < 0.0001) and increased wall thickness (p < 0.01) in males, which was attenuated by Ang-(1-7). In male but not female aortic arches, Dox increased media hypertrophy (p < 0.05) and reduced elastin content (p < 0.001), which were prevented by Ang-(1-7). Conversely, Dox increased fibrosis (p < 0.0001) in juvenile female rats, which was reduced by Ang-(1-7). Adjunct Ang-(1-7) prevented the Dox-induced increase in total cell and nuclear pERK1/2 in the aortic intima and media of male rats and nuclear pSMAD2 in the intimal and medial regions of the aortic arches of both sexes. These results demonstrate that Ang-(1-7) attenuated Dox-induced aortic dysfunction in both sexes of juvenile rats, albeit through different mechanisms, suggesting that Ang-(1-7) may serve as an effective adjuvant to ameliorate cardiovascular and long-term end-organ damage in pediatric patients produced by anthracyclines., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
14. Phase I Study of Muscadine Grape Extract for Patients With Advanced Cancer.
- Author
-
Bitting RL, Tooze JA, Isom S, Petty WJ, Grant SC, Desnoyers RJ, Thomas A, Thomas CY, Alistar AT, Golden SL, Pleasant K, Chappell MC, Tallant EA, Gallagher PE, and Klepin HD
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Follow-Up Studies, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Metastasis, Neoplasms pathology, Prognosis, Tissue Distribution, Neoplasms drug therapy, Plant Extracts pharmacokinetics, Plant Extracts therapeutic use, Vitis chemistry
- Abstract
Objective: Preclinical studies with muscadine grape extract (MGE) show antitumor activity and decreased systemic inflammation. This phase I study (NCT02583269) assessed safety and tolerability of a proprietary MGE preparation in patients with advanced solid tumors., Methods: Patients with metastatic or unresectable cancers who were progressing on standard therapies were assigned to MGE in a standard 3+3 design. Five dose levels were tested (320 to 1600 mg total phenolics/d). Safety and maximum-tolerated dose were assessed after 4 weeks. Patients were evaluated for response at 8 weeks and continued on MGE if clinically stable. Secondary outcomes were response, survival, adherence, fatigue, and quality of life (QOL)., Results: In total, 23 patients (lung, n=7; gastrointestinal, n=7; genitourinary, n=6; other, n=3) received MGE capsules by mouth twice daily. The cohort [median age 72 years, 48% Eastern Cooperative Oncology Group (ECOG) 2] was heavily pretreated. After 4 weeks on MGE, possibly attributable adverse events grade 2 or higher were fatigue (n=1), decreased lymphocyte count (n=1), and constipation (n=2), including 1 dose-limiting toxicity for grade 3 constipation. Maximum-tolerated dose was not reached. No partial responses were observed. Median time on therapy was 8 weeks, with 29% of patients treated beyond 16 weeks and a median overall survival of 7.2 months. QOL and fatigue levels were stable from baseline to 8 weeks. Higher MGE dose was correlated with improvement in self-reported physical well-being QOL at 8 weeks (r=0.6; P=0.04)., Conclusions: MGE is safe and well-tolerated in heavily pretreated and older cancer patients. The potential anticancer properties and the effects of MGE on physical well-being and QOL metrics will be evaluated in future studies., Competing Interests: The authors declare no conflicts of interest., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
15. Angiotensin-(1-7) reduces doxorubicin-induced cardiac dysfunction in male and female Sprague-Dawley rats through antioxidant mechanisms.
- Author
-
Rahimi O, Kirby J, Varagic J, Westwood B, Tallant EA, and Gallagher PE
- Subjects
- Animals, Cardiotoxicity, Catalase metabolism, Female, Heart Diseases etiology, Heart Rate, Male, Malondialdehyde metabolism, Mitral Valve physiopathology, Myocardium metabolism, NADPH Oxidases genetics, NADPH Oxidases metabolism, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Superoxide Dismutase metabolism, Angiotensin I therapeutic use, Antineoplastic Agents toxicity, Antioxidants therapeutic use, Doxorubicin toxicity, Heart Diseases drug therapy, Peptide Fragments therapeutic use
- Abstract
Doxorubicin (Dox) is an effective chemotherapeutic for a variety of pediatric malignancies. Unfortunately, Dox administration often results in a cumulative dose-dependent cardiotoxicity that manifests with marked oxidative stress, leading to heart failure. Adjunct therapies are needed to mitigate Dox cardiotoxicity and enhance quality of life in pediatric patients with cancer. Angiotensin-(1-7) [Ang-(1-7)] is an endogenous hormone with cardioprotective properties. This study investigated whether adjunct Ang-(1-7) attenuates cardiotoxicity resulting from exposure to Dox in male and female juvenile rats. Dox significantly reduced body mass, and the addition of Ang-(1-7) had no effect. However, adjunct Ang-(1-7) prevented Dox-mediated diastolic dysfunction, including markers of decreased passive filling as measured by reduced early diastole mitral valve flow velocity peak ( E ) ( P < 0.05) and early diastole mitral valve annulus peak velocity ( e' ; P < 0.001) and increased E/e' ( P < 0.001), an echocardiographic measure of diastolic dysfunction. Since Dox treatment increases reactive oxygen species (ROS), the effect of Ang-(1-7) on oxidative by-products and enzymes that generate or reduce ROS was investigated. In hearts of male and female juvenile rats, Dox increased NADPH oxidase 4 ( P < 0.05), a major cardiovascular NADPH oxidase isozyme that generates ROS, as well as 4-hydroxynonenal ( P < 0.001) and malondialdehyde ( P < 0.001), markers of lipid peroxidation; Ang-(1-7) prevented these effects of Dox. Cotreatment with Dox and Ang-(1-7) increased the antioxidant enzymes SOD1 (male: P < 0.05; female: P < 0.01) and catalase ( P < 0.05), which likely contributed to reduced ROS. These results demonstrate that Ang-(1-7) prevents diastolic dysfunction in association with a reduction in ROS, suggesting that the heptapeptide hormone may serve as an effective adjuvant to improve Dox-induced cardiotoxicity. NEW & NOTEWORTHY Ang-(1-7) is a clinically safe peptide hormone with cardioprotective and antineoplastic properties that could be used as an adjuvant therapy to improve cancer treatment and mitigate the long-term cardiotoxicity associated with doxorubicin in pediatric patients with cancer.
- Published
- 2020
- Full Text
- View/download PDF
16. A Polyphenol-Rich Extract From Muscadine Grapes Inhibits Triple-Negative Breast Tumor Growth.
- Author
-
Collard M, Gallagher PE, and Tallant EA
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Proliferation, Humans, Mice, Mice, Nude, Polyphenols, Plant Extracts pharmacology, Triple Negative Breast Neoplasms, Vitis
- Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that tends to affect young women and has a high propensity to metastasize. No targeted treatments are available for this type of breast cancer due to a lack of estrogen or progesterone receptors or overexpression of human epidermal growth factor receptor type 2 overexpression. Currently, patients have no therapeutic options once standard of care is complete, indicating a need for safe and effective therapies to slow or prevent the progression of TNBC to metastatic disease. Studies showed that isolated polyphenols or polyphenol-rich muscadine grape extracts polyphenols inhibit the proliferation of various cancer cells including breast cancer. A proprietary muscadine grape extract (MGE) was administered to nude mice with human MDA-MB-231 TNBC atumors for 4 weeks to determine the effect of the extract on tumor growth. MGE decreased tumor volume in association with a reduction in the proliferative markers Ki67 and cyclin D1. To determine the molecular mechanisms for the MGE-induced reduction in tumor growth, mouse 4T1, MDA-MB-231, or human BT-549 TNBC cells were treated with MGE, and various signaling pathways were investigated. MGE reduced c-Met, differentially abrogated ERK/MAPK and AKT signaling, and decreased a downstream targets of ERK/MAPK and AKT pathways, cyclin D1. Cyclin D1 reduction was associated with retinoblastoma activation and cell cycle arrest in MDA-MB-231 TNBC cells. MGE-regulated molecular signaling pathways were functionally associated with a dose-dependent reduction in cell proliferation. The pluripotency of MGE and high index of safety and tolerability suggest that the extract may serve as a therapeutic to reduce TNBC progression to metastatic disease.
- Published
- 2020
- Full Text
- View/download PDF
17. Stabilization of Angiotensin-(1-7) by key substitution with a cyclic non-natural amino acid.
- Author
-
Wester A, Devocelle M, Tallant EA, Chappell MC, Gallagher PE, and Paradisi F
- Subjects
- Humans, Protein Stability, Amino Acid Substitution, Angiotensin I chemical synthesis, Angiotensin I chemistry, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases chemistry, Peptide Fragments chemical synthesis, Peptide Fragments chemistry, Peptidyl-Dipeptidase A chemistry, Proteolysis
- Abstract
Angiotensin-(1-7) [Ang-(1-7)], a heptapeptide hormone of the renin-angiotensin-aldosterone system, is a promising candidate as a treatment for cancer that reflects its anti-proliferative and anti-angiogenic properties. However, the peptide's therapeutic potential is limited by the short half-life and low bioavailability resulting from rapid enzymatic metabolism by peptidases including angiotensin-converting enzyme (ACE) and dipeptidyl peptidase 3 (DPP 3). We report the facile assembly of three novel Ang-(1-7) analogues by solid-phase peptide synthesis which incorporates the cyclic non-natural δ-amino acid ACCA. The analogues containing the ACCA substitution at the site of ACE cleavage exhibit complete resistance to human ACE, while substitution at the DDP 3 cleavage site provided stability against DPP 3 hydrolysis. Furthermore, the analogues retain the anti-proliferative properties of Ang-(1-7) against the 4T1 and HT-1080 cancer cell lines. These results suggest that ACCA-substituted Ang-(1-7) analogues which show resistance against proteolytic degradation by peptidases known to hydrolyze the native heptapeptide may be novel therapeutics in the treatment of cancer.
- Published
- 2017
- Full Text
- View/download PDF
18. Programs to Recruit and Retain a More Diverse Workforce in Biomedical Sciences Research.
- Author
-
Gwathmey TM, Tallant EA, Howlett AC, and Diz DI
- Abstract
To improve overall healthcare and to reduce health disparities, efforts must focus on increasing the diversity of personnel trained in the biomedical sciences. Here, we describe the development, implementation, and relative outcomes of three pipeline training programs in biomedical sciences research designed to increase workforce diversity institutionally, regionally, and nationally. We report on their effectiveness in improving the recruitment and retention of underrepresented minorities with the long-term goal of remedying health inequities and disparities.
- Published
- 2016
19. Angiotensin-(1-7) Attenuates Skeletal Muscle Fibrosis and Stiffening in a Mouse Model of Extremity Sarcoma Radiation Therapy.
- Author
-
Willey JS, Bracey DN, Gallagher PE, Tallant EA, Wiggins WF, Callahan MF, Smith TL, and Emory CL
- Subjects
- Analysis of Variance, Animals, Biopsy, Needle, Disease Models, Animal, Fibrosis etiology, Fibrosis prevention & control, Hindlimb, Immunohistochemistry, Male, Mice, Mice, Inbred Strains, Muscle Neoplasms pathology, Muscle Neoplasms therapy, Muscle, Skeletal pathology, Random Allocation, Reference Values, Sarcoma, Experimental pathology, Sensitivity and Specificity, Spasm pathology, Angiotensin I administration & dosage, Muscle, Skeletal drug effects, Muscle, Skeletal radiation effects, Peptide Fragments administration & dosage, Sarcoma, Experimental therapy, Spasm prevention & control
- Abstract
Background: Radiation-induced fibrosis (RIF) of musculoskeletal tissue is a common complication of radiation therapy for extremity soft-tissue sarcoma, with no standardized strategy for prevention and treatment. Angiotensin-(1-7) (Ang-[1-7]), a well-tolerated endogenous heptapeptide hormone with antitumor and antifibrotic properties, was tested as a radioprotectant for RIF and stiffening of irradiated muscles., Methods: Male CD-1 mice were randomized to one of three treatment groups: control, simulated sarcoma radiation therapy to the gastrocnemius and soleus muscles, or radiation therapy along with continuous Ang-(1-7) delivery initiated three days before radiation therapy. The biologically equivalent dose of radiation (∼100.3 Gy) absorbed by normal musculature during the course of radiation therapy for extremity sarcoma was delivered by means of four dose fractions of 7.3 Gy over two weeks. Fibrosis (n = 5 per group) and mechanical properties (n = 4 to 6 per group) of the muscles were measured at six weeks and four months after radiation therapy, and the intramuscular concentration of the profibrotic cytokines transforming growth factor-beta (TGF-β) and connective tissue growth factor (CTGF) (n = 8 to 10 per group) were measured at six weeks., Results: Interstitial (p < 0.01) and perivascular (p < 0.05) fibrosis increased significantly in the muscles treated with radiation therapy alone versus the nonirradiated controls at both six weeks (interstitial, +89%; perivascular, +112%) and four months (interstitial, +154%; perivascular, +88%). The muscles treated with radiation alone also exhibited increased tension (p < 0.01) versus nonirradiated controls at both six weeks (+779%) and four months (+1761%) when placed under 5% strain, and at four months (+1390%; p < 0.001) under 10% strain. At four months, muscle stiffness had increased in the mice treated with radiation therapy alone (+90%; p = 0.002) compared with nonirradiated controls. TGF-β production was also greater in this group at six weeks (+37%; p = 0.06) versus control. Ang-(1-7) administration prevented RIF and stiffening, with no differences observed for any other outcome between those receiving radiation therapy with Ang-(1-7) and the nonirradiated controls. Likewise, Ang-(1-7) mitigated the increase in TGF-β and CTGF concentration from radiation therapy., Conclusions: Ang-(1-7) attenuated RIF, stiffening, and production of profibrotic cytokines that were elevated in mouse skeletal muscles after simulated radiation therapy for extremity sarcoma., Clinical Relevance: Ang-(1-7) may serve as a potential therapy for the prevention of RIF in patients who require radiation therapy as adjuvant treatment for soft-tissue sarcoma., (Copyright © 2016 by The Journal of Bone and Joint Surgery, Incorporated.)
- Published
- 2016
- Full Text
- View/download PDF
20. Angiotensin-(1-7) prevents angiotensin II-induced fibrosis in cremaster microvessels.
- Author
-
Carver KA, Smith TL, Gallagher PE, and Tallant EA
- Subjects
- Angiotensin II pharmacology, Animals, Arterioles metabolism, Arterioles pathology, Arterioles physiopathology, Chronic Disease, Connective Tissue Growth Factor metabolism, Dual Specificity Phosphatase 1 metabolism, Fibrosis metabolism, Fibrosis pathology, Fibrosis physiopathology, Hypertension drug therapy, Hypertension pathology, Hypertension physiopathology, MAP Kinase Signaling System drug effects, Male, Mitogen-Activated Protein Kinase 3 metabolism, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Phosphorylation drug effects, Rats, Rats, Inbred Lew, Smad2 Protein metabolism, Smad3 Protein metabolism, Transforming Growth Factor beta metabolism, Vasoconstrictor Agents pharmacology, Angiotensin I pharmacology, Antihypertensive Agents pharmacology, Blood Pressure drug effects, Hypertension metabolism, Muscle, Skeletal blood supply, Muscle, Skeletal metabolism, Peptide Fragments pharmacology
- Abstract
Objective: The effect of the heptapeptide hormone Ang-(1-7) on microvascular fibrosis in rats with Ang II-induced hypertension was investigated, since vascular fibrosis/remodeling plays a prominent role in hypertension-induced end-organ damage and Ang-(1-7) inhibits vascular growth and fibrosis., Methods: Fibrosis of cremaster microvessels was studied in male Lewis rats infused with Ang II and/or Ang-(1-7)., Results: Ang II elevated systolic blood pressure by approximately 40 mmHg, while blood pressure was not changed by Ang-(1-7). Ang II increased perivascular fibrosis surrounding 20-50 μm arterioles as well as interstitial fibrosis; coadministration of Ang-(1-7) prevented the increases in fibrosis. The fibrotic factor CTGF and phospho-Smad 2/3, which upregulates CTGF, were increased by Ang II; this effect was prevented by coadministration of Ang-(1-7). Although TGF-β phosphorylates Smad 2/3, TGF-β was no different among treatment groups. In contrast, Ang II increased the MAP kinase phospho-ERK1/2, which also phosphorylates Smad; p-ERK was reduced by Ang-(1-7). Ang-(1-7), in the presence or absence of Ang II, upregulated the MAP kinase phosphatase DUSP1., Conclusions: These results suggest that Ang-(1-7) increases DUSP1 to reduce MAP kinase/Smad/CTGF signaling and decrease fibrosis in resistance arterioles, to attenuate end-organ damage associated with chronic hypertension., (© 2014 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
21. Angiotensin-(1-7): a peptide hormone with anti-cancer activity.
- Author
-
Gallagher PE, Arter AL, Deng G, and Tallant EA
- Subjects
- Amino Acid Sequence, Angiotensin I chemical synthesis, Angiotensin I chemistry, Angiotensin I therapeutic use, Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Cell Proliferation drug effects, Clinical Trials as Topic, Humans, Neoplasms blood supply, Neoplasms drug therapy, Neoplasms pathology, Peptide Fragments chemical synthesis, Peptide Fragments chemistry, Peptide Fragments therapeutic use, Peptide Hormones chemical synthesis, Peptide Hormones chemistry, Peptide Hormones therapeutic use, Angiotensin I pharmacology, Antineoplastic Agents pharmacology, Peptide Fragments pharmacology, Peptide Hormones pharmacology
- Abstract
The development of peptides as therapeutic agents has progressed such that these small molecules of less than fifty amino acids are currently in use for the treatment of a variety of pathologies. This review focuses on the pre-clinical studies and clinical trials assessing the anti-cancer properties of angiotensin-(1-7) [Ang-(1-7)], an endogenous heptapeptide hormone of the renin-angiotensin system. Ang-(1-7) mediates biological responses by activating mas, a unique G protein- coupled receptor, thereby providing specific targeted actions when used as a therapeutic agent. Studies in in vitro as well as in vivo mouse models demonstrated that the heptapeptide hormone reduced proliferation of human cancer cells and xenograft tumors. This attenuation was concomitant with decreased angiogenesis, cancer associated fibrosis, osteoclastogenesis, tumor-induced inflammation and metastasis as well as altered regulation of growth promoting cellular signaling pathways. In three clinical trials, Ang-(1-7) was well tolerated with limited toxic or quality-of-life side effects and showed clinical benefit in cancer patients with solid tumors. Taken together, these studies suggest that Ang-(1-7) may serve as a first-in-class peptide chemotherapeutic agent, reducing cancer growth and metastases by pleiotrophic mechanisms as well as targeting the tumor microenvironment.
- Published
- 2014
- Full Text
- View/download PDF
22. Angiotensin-(1-7) attenuates metastatic prostate cancer and reduces osteoclastogenesis.
- Author
-
Krishnan B, Smith TL, Dubey P, Zapadka ME, Torti FM, Willingham MC, Tallant EA, and Gallagher PE
- Subjects
- Adenocarcinoma secondary, Aged, Animals, Bone Marrow Cells drug effects, Bone Marrow Cells pathology, Bone Neoplasms drug therapy, Bone Neoplasms secondary, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Male, Mice, Mice, Nude, Mice, SCID, Middle Aged, Neoplasm Metastasis drug therapy, Osteoclasts pathology, Vascular Endothelial Growth Factor A metabolism, Xenograft Model Antitumor Assays, Adenocarcinoma drug therapy, Angiotensin I pharmacology, Antineoplastic Agents pharmacology, Osteoclasts drug effects, Peptide Fragments pharmacology, Prostatic Neoplasms drug therapy
- Abstract
Background: Angiotensin-(1-7) [Ang-(1-7)] is an endogenous, heptapeptide hormone with anti-proliferative and anti-angiogenic properties. The primary objective of this study was to determine whether Ang-(1-7) effectively reduces prostate cancer metastasis in mice., Methods: Human PC3 prostate cancer cells were injected into the aortic arch via the carotid artery of SCID mice pre-treated with Ang-(1-7) or injected into the tibia of athymic mice, administered Ang-(1-7) for 5 weeks beginning 2 weeks post-injection. Tumor growth and volume were determined by bioluminescent and magnetic resonance imaging. The presence of tumors was confirmed by hematoxylin and eosin staining; TRAP histochemistry was used to identify osteolytic lesions. The effect of Ang-(1-7) on osteoclastogenesis was assessed in differentiated bone marrow cells., Results: Pre-treatment with Ang-(1-7) prevented metastatic tumor formation following intra-aortic injection of PC3 cells, while 83% of untreated mice developed tumors in metastatic sites. Circulating VEGF was significantly higher in control mice compared to mice administered Ang-(1-7). A 5-week regimen of the heptapeptide hormone attenuated intra-tibial tumor growth; Ang-(1-7) was significantly higher in the tibia of treated mice than in control animals. Osteoclastogenesis was reduced by 50% in bone marrow cells differentiated in the presence of Ang-(1-7), suggesting that the heptapeptide hormone prevents the formation of osteolytic lesions to reduce tumor survival in the bone microenvironment., Conclusions: These findings suggest that Ang-(1-7) may serve as an anti-angiogenic and anti-metastatic agent for advanced prostate cancer. By extension, the heptapeptide hormone may provide effective therapy for bone metastasis produced from primary tumors of the lung and breast., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
23. Angiotensin-(1-7) reduces proliferation and angiogenesis of human prostate cancer xenografts with a decrease in angiogenic factors and an increase in sFlt-1.
- Author
-
Krishnan B, Torti FM, Gallagher PE, and Tallant EA
- Subjects
- Adenocarcinoma blood supply, Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Cell Line, Tumor, Humans, Male, Mice, Mice, Nude, Placenta Growth Factor, Pregnancy Proteins metabolism, Prostatic Neoplasms blood supply, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Vascular Endothelial Growth Factor A metabolism, Xenograft Model Antitumor Assays, Adenocarcinoma drug therapy, Angiotensin I pharmacology, Antineoplastic Agents pharmacology, Cell Proliferation drug effects, Neovascularization, Pathologic drug therapy, Peptide Fragments pharmacology, Prostatic Neoplasms drug therapy, Vascular Endothelial Growth Factor Receptor-1 metabolism
- Abstract
Background: Prostate cancer is the most frequently diagnosed malignancy and the second-leading cause of cancer death in men. The purpose of this study was to determine the anti-proliferative and anti-angiogenic efficacy of angiotensin-(1-7) [Ang-(1-7)], an endogenous peptide hormone, in human prostate cancer xenografts., Methods: Human LNCaP prostate cancer cells were injected into the flank of athymic mice and tumors were treated with Ang-(1-7) for 54 days. Tumor growth and angiogenesis were determined by immunohistochemistry and western blot hybridization., Results: Ang-(1-7) markedly reduced the volume and wet weight of LNCaP xenograft tumors. Histological analysis of tumor sections from saline-treated mice showed increased Ki67 immunoreactivity and enhanced phosphorylation of the MAP kinases ERK1/2 compared to tumors from Ang-(1-7)-treated mice, suggesting that the heptapeptide reduces cell proliferation. Intratumoral vessel density was decreased in Ang-(1-7)-treated mice with an associated reduction in vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), suggesting that the heptapeptide attenuates vascularization by reducing angiogenic factors. Ang-(1-7) administration markedly increased the soluble fraction of VEGF receptor 1 (sFlt-1), with a concomitant reduction in VEGF receptors 1 and 2. sFlt-1 serves as a decoy receptor that traps VEGF and PlGF, making the ligands unavailable to membrane-bound VEGF receptors and preventing activation of pro-angiogenic signaling., Conclusions: The decrease in PlGF and VEGF coupled with the increase in sFlt-1 suggests that Ang-(1-7) may serve as a novel anti-angiogenic therapy for prostate cancer. Further, the pleiotropic mechanisms of action by Ang-(1-7) may limit angiogenic resistance that occurs with VEGF inhibitors or receptor blockers., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
24. Differences in oxidative stress status and expression of MKP-1 in dorsal medulla of transgenic rats with altered brain renin-angiotensin system.
- Author
-
Nautiyal M, Katakam PV, Busija DW, Gallagher PE, Tallant EA, Chappell MC, and Diz DI
- Subjects
- Angiotensin I metabolism, Angiotensin II metabolism, Angiotensinogen genetics, Angiotensinogen metabolism, Animals, Blood Pressure, Disease Models, Animal, Dual Specificity Phosphatase 1 genetics, Gene Expression Regulation, Hypertension genetics, Hypertension physiopathology, Ion Channels genetics, Ion Channels metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Male, Mitochondria enzymology, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, NADPH Oxidases metabolism, Oligonucleotides, Antisense metabolism, Peptide Fragments metabolism, Phosphorylation, Rats, Rats, Sprague-Dawley, Rats, Transgenic, Reactive Oxygen Species metabolism, Renin genetics, Renin metabolism, Signal Transduction, Uncoupling Protein 2, Dual Specificity Phosphatase 1 metabolism, Hypertension enzymology, Medulla Oblongata enzymology, Oxidative Stress, Renin-Angiotensin System genetics
- Abstract
ANG II-stimulated production of reactive oxygen species (ROS) through NADPH oxidase is suggested to activate MAPK pathways, which are implicated in neurally mediated pressor effects of ANG II. Emerging evidence suggests that ANG-(1-7) up regulates MAPK phosphatases to reduce MAPK signaling and attenuate actions of ANG II. Whether angiotensin peptides participate in long-term regulation of these systems in the brain is not known. Therefore, we determined tissue and mitochondrial ROS, as well as expression and activity of MAPK phosphatase-1 (MKP-1) in brain dorsal medullary tissue of hypertensive transgenic (mRen2)27 rats exhibiting higher ANG II/ANG-(1-7) tone or hypotensive transgenic rats with targeted decreased glial expression of angiotensinogen, ASrAOGEN (AS) exhibiting lower ANG II/ANG-(1-7) tone compared with normotensive Sprague-Dawley (SD) rats that serve as the control strain. Transgenic (mRen2)27 rats showed higher medullary tissue NADPH oxidase activity and dihydroethidium fluorescence in isolated mitochondria vs. SD or AS rats. Mitochondrial uncoupling protein 2 was lower in AS and unchanged in (mRen2)27 compared with SD rats. MKP-1 mRNA and protein expression were higher in AS and unchanged in (mRen2)27 compared with SD rats. AS rats also had lower phosphorylated ERK1/2 and JNK consistent with higher MKP-1 activity. Thus, an altered brain renin-angiotensin system influences oxidative stress status and regulates MKP-1 expression. However, there is a dissociation between these effects and the hemodynamic profiles. Higher ROS was associated with hypertension in (mRen2)27 and normal MKP-1, whereas the higher MKP-1 was associated with hypotension in AS, where ROS was normal relative to SD rats.
- Published
- 2012
- Full Text
- View/download PDF
25. Angiotensin-(1-7) abrogates mitogen-stimulated proliferation of cardiac fibroblasts.
- Author
-
McCollum LT, Gallagher PE, and Tallant EA
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, Collagen antagonists & inhibitors, Collagen biosynthesis, Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, DNA antagonists & inhibitors, DNA biosynthesis, Dual Specificity Phosphatase 1 genetics, Dual Specificity Phosphatase 1 metabolism, Endothelin-1 pharmacology, Fibroblasts cytology, Fibrosis complications, Fibrosis metabolism, Fibrosis physiopathology, Gene Expression Regulation drug effects, Heart Diseases complications, Heart Diseases metabolism, Heart Diseases physiopathology, Mitogen-Activated Protein Kinase 1 genetics, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 genetics, Mitogen-Activated Protein Kinase 3 metabolism, Myocardium cytology, Phosphorylation drug effects, Prostaglandin-Endoperoxide Synthases genetics, Prostaglandin-Endoperoxide Synthases metabolism, Rats, Signal Transduction drug effects, Angiotensin I pharmacology, Angiotensin II pharmacology, Cell Proliferation drug effects, Fibroblasts drug effects, Peptide Fragments pharmacology
- Abstract
Previous studies showed that angiotensin-(1-7) [Ang-(1-7)] attenuates cardiac remodeling by reducing both interstitial and perivascular fibrosis. Although a high affinity binding site for Ang-(1-7) was identified on cardiac fibroblasts, the molecular mechanisms activated by the heptapeptide hormone were not identified. We isolated cardiac fibroblasts from neonatal rat hearts to investigate signaling pathways activated by Ang-(1-7) that participate in fibroblast proliferation. Ang-(1-7) reduced (3)H-thymidine, -leucine and -proline incorporation into cardiac fibroblasts stimulated with serum or the mitogen endothelin-1 (ET-1), demonstrating that the heptapeptide hormone decreases DNA, protein and collagen synthesis. The reduction in DNA synthesis by Ang-(1-7) was blocked by the AT((1-7)) receptor antagonist [d-Ala(7)]-Ang-(1-7), showing specificity of the response. Treatment of cardiac fibroblasts with Ang-(1-7) reduced the Ang II- or ET-1-stimulated increase in phospho-ERK1 and -ERK2. In contrast, Ang-(1-7) increased dual-specificity phosphatase DUSP1 immunoreactivity and mRNA, suggesting that the heptapeptide hormone increases DUSP1 to reduce MAP kinase phosphorylation and activity. Incubation of cardiac fibroblasts with ET-1 increased cyclooxygenase 2 (COX-2) and prostaglandin synthase (PGES) mRNAs, while Ang-(1-7) blocked the increase in both enzymes, suggesting that the heptapeptide hormone alters the concentration and the balance between the proliferative and anti-proliferative prostaglandins. Collectively, these results indicate that Ang-(1-7) participates in maintaining cardiac homeostasis by reducing proliferation and collagen production by cardiac fibroblasts in association with up-regulation of DUSP1 to reduce MAP kinase activities and attenuation of the synthesis of mitogenic prostaglandins. Increased Ang-(1-7) or agents that enhance production of the heptapeptide hormone may prevent abnormal fibrosis that occurs during cardiac pathologies., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
26. Angiotensin peptides and lung cancer.
- Author
-
Gallagher PE, Cook K, Soto-Pantoja D, Menon J, and Tallant EA
- Subjects
- Animals, Humans, Peptide Fragments metabolism, Peptide Fragments pharmacology, Angiotensins metabolism, Angiotensins pharmacology, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Renin-Angiotensin System drug effects, Renin-Angiotensin System physiology
- Abstract
Lung cancer is a leading cause of death in both men and women, with over 1,000,000 new cases diagnosed worldwide annually and a 5-year survival rate of only 14%, a figure that has improved little in the past thirty years. This poor prognosis suggests a need for novel approaches for the treatment and prevention of lung cancer. The renin-angiotensin system is an established, primary regulator of blood pressure, homeostasis, and natriuresis; however, compelling evidence indicates that the angiotensin peptides also play a role in cell proliferation and inflammation. Angiotensin II is a vasoconstrictor, a mitogen, and an angiogenic factor, while angiotensin-(1-7) has vasodilator, anti-proliferative, and anti-angiogenic properties. This review focuses on studies examining the renin-angiotensin system in pulmonary cancers and whether clinical intervention of this pathway may serve as an effective chemotherapeutic and/or chemopreventive modality for lung cancer.
- Published
- 2011
- Full Text
- View/download PDF
27. Angiotensin peptides and central autonomic regulation.
- Author
-
Diz DI, Arnold AC, Nautiyal M, Isa K, Shaltout HA, and Tallant EA
- Subjects
- Aging physiology, Angiotensin I deficiency, Animals, Baroreflex physiology, Blood Pressure, Humans, Hypertension etiology, Hypertension genetics, MAP Kinase Signaling System, Medulla Oblongata physiology, Peptide Fragments deficiency, Phosphatidylinositol 3-Kinases physiology, Angiotensin I physiology, Angiotensin II physiology, Autonomic Nervous System physiology, Brain physiology, Peptide Fragments physiology
- Abstract
Aging, hypertension, and fetal-programmed cardiovascular disease are associated with a functional deficiency of angiotensin (Ang)-(1-7) in the brain dorsomedial medulla. The resulting unrestrained activity of Ang II in brainstem regions negatively impacts resting mean arterial pressure, sympathovagal balance, and baroreflex sensitivity for control of heart rate. The differential effects of Ang II and Ang-(1-7) may be related to the cellular sources of these peptides as well as different precursor pathways. Long-term alterations of the brain renin-angiotensin system may influence signaling pathways including phosphoinositol-3-kinase and mitogen-activated protein kinase and their downstream mediators, and as a consequence may influence metabolic function. Differential regulation of signaling pathways in aging and hypertension by Ang II versus Ang-(1-7) may contribute to the autonomic dysfunction accompanying these states., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
28. Angiotensin-(1-7) reduces fibrosis in orthotopic breast tumors.
- Author
-
Cook KL, Metheny-Barlow LJ, Tallant EA, and Gallagher PE
- Subjects
- Animals, Blotting, Western, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinoma, Ductal, Breast metabolism, Carcinoma, Ductal, Breast pathology, Dual Specificity Phosphatase 1 metabolism, Female, Fibronectins metabolism, Fibrosis metabolism, Fibrosis pathology, Fibrosis prevention & control, Fluorescent Antibody Technique, Humans, Immunoenzyme Techniques, Lung Diseases, Interstitial metabolism, Lung Diseases, Interstitial pathology, Mice, Mice, Nude, Mitogen-Activated Protein Kinase 3 metabolism, Phosphorylation drug effects, Transforming Growth Factor beta metabolism, Tumor Cells, Cultured, Angiotensin I pharmacology, Antihypertensive Agents pharmacology, Breast Neoplasms prevention & control, Carcinoma, Ductal, Breast prevention & control, Lung Diseases, Interstitial prevention & control, Peptide Fragments pharmacology
- Abstract
Angiotensin-(1-7) [Ang-(1-7)] is an endogenous 7-amino acid peptide hormone of the renin-angiotensin system that has antiproliferative properties. In this study, Ang-(1-7) inhibited the growth of cancer-associated fibroblasts (CAF) and reduced fibrosis in the tumor microenvironment. A marked decrease in tumor volume and weight was observed in orthotopic human breast tumors positive for the estrogen receptor (BT-474 or ZR-75-1) and HER2 (BT-474) following Ang-(1-7) administration to athymic mice. Ang-(1-7) concomitantly reduced interstitial fibrosis in association with a significant decrease in collagen I deposition, along with a similar reduction in perivascular fibrosis. In CAFs isolated from orthotopic breast tumors, the heptapeptide markedly attenuated in vitro growth as well as reduced fibronectin, transforming growth factor-β (TGF-β), and extracellular signal-regulated kinase 1/2 kinase activity. An associated increase in the mitogen-activated protein kinase (MAPK) phosphatase DUSP1 following treatment with Ang-(1-7) suggested a potential mechanism by which the heptapeptide reduced MAPK signaling. Consistent with these in vitro observations, immunohistochemical analysis of Ang-(1-7)-treated orthotopic breast tumors revealed reduced TGF-β and increased DUSP1. Together, our findings indicate that Ang-(1-7) targets the tumor microenvironment to inhibit CAF growth and tumor fibrosis., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
29. Decreased cardiac Ang-(1-7) is associated with salt-induced cardiac remodeling and dysfunction.
- Author
-
Varagic J, Ahmad S, Brosnihan KB, Groban L, Chappell MC, Tallant EA, Gallagher PE, and Ferrario CM
- Subjects
- Angiotensin II blood, Angiotensin-Converting Enzyme 2, Animals, Blood Pressure, Male, Peptidyl-Dipeptidase A genetics, Peptidyl-Dipeptidase A metabolism, Proto-Oncogene Mas, Proto-Oncogene Proteins metabolism, RNA, Messenger metabolism, Rats, Rats, Inbred SHR, Receptors, G-Protein-Coupled metabolism, Renin blood, Angiotensin I metabolism, Peptide Fragments metabolism, Sodium Chloride, Dietary toxicity, Ventricular Dysfunction, Left etiology, Ventricular Remodeling
- Abstract
Objective: Angiotensin II has a critical role in the regulation of blood pressure and cell growth and excess activity of the peptide is implicated in the pathogenesis of salt-induced cardiovascular injury. On the other hand, the role of counteracting angiotensin-(1-7) in cardiac structural and functional responses to high salt diet has not been elucidated. Therefore, the present study examined the changes in cardiac angiotensin-(1-7), its forming enzyme angiotensin converting enzyme 2 (ACE2) and receptor mas in response to a high salt diet in spontaneously hypertensive rats (SHR)., Methods: Eight-week-old male spontaneously hypertensive rats (SHR) were given an 8% salt diet for 5 weeks (n = 8). Age- and gender-matched controls received standard chow (n = 6)., Results: Salt excess increased arterial pressure (p < 0.05) and plasma renin and angiotensin II concentrations (p < 0.05). Salt-induced left ventricular remodeling and diastolic dysfunction were associated with diminished levels of angiotensin-(1-7) in the heart (p < 0.05) and no changes in cardiac angiotensin II levels. Exposure to high salt intake decreased cardiac ACE2 mRNA and protein level (p < 0.05). There was no difference in the protein levels of angiotensin II type 1 and mas receptors between the two experimental groups., Conclusion: The adverse cardiac effects of excessive salt intake may result not only from the undesirable action of angiotensin II but may also be a consequence of diminished protective effects of the angiotensin-(1-7).
- Published
- 2010
- Full Text
- View/download PDF
30. Phase I and pharmacokinetic study of angiotensin-(1-7), an endogenous antiangiogenic hormone.
- Author
-
Petty WJ, Miller AA, McCoy TP, Gallagher PE, Tallant EA, and Torti FM
- Subjects
- Adult, Aged, Aged, 80 and over, Angiogenesis Inhibitors therapeutic use, Angiotensin I therapeutic use, Biomarkers, Tumor metabolism, Cohort Studies, Dose-Response Relationship, Drug, Female, Humans, Male, Middle Aged, Models, Biological, Peptide Fragments therapeutic use, Peptides chemistry, Angiogenesis Inhibitors pharmacokinetics, Angiotensin I pharmacokinetics, Neoplasms blood, Neoplasms drug therapy, Peptide Fragments pharmacokinetics
- Abstract
Purpose: Angiotensin-(1-7) [Ang-(1-7)] is an endogenous peptide hormone of the renin-angiotensin system with antiproliferative and antiangiogenic properties. The primary objective of this study was to establish the recommended phase II dose of Ang-(1-7) for treating patients with advanced cancer. Secondary objectives were to assess toxicities, pharmacokinetics, clinical activity, and plasma biomarkers., Experimental Design: Patients with advanced solid tumors refractory to standard therapy were treated with escalating doses of Ang-(1-7) in cohorts of three patients. Ang-(1-7) was administered by s.c. injection once daily for 5 days on a 3-week cycle. Tumor measurements were done every two cycles and treatment was continued until disease progression or unacceptable toxicity., Results: Eighteen patients were enrolled. Dose-limiting toxicities encountered at the 700 microg/kg dose included stroke (grade 4) and reversible cranial neuropathy (grade 3). Other toxicities were generally mild. One patient developed a 19% reduction in tumor measurements. Three additional patients showed clinical benefit with stabilization of disease lasting more than 3 months. On day 1, Ang-(1-7) administration led to a decrease in plasma placental growth factor (PlGF) levels in patients with clinical benefit (P = 0.04) but not in patients without clinical benefit (P = 0.25). On day 5, PlGF levels remained lower in patients with clinical benefit compared with patients without clinical benefit (P = 0.04)., Conclusions: Ang-(1-7) is a first-in-class antiangiogenic drug with activity for treating cancer that is linked to reduction of plasma PlGF levels. The recommended phase II dose is 400 microg/kg for this administration schedule.
- Published
- 2009
- Full Text
- View/download PDF
31. Angiotensin-(1-7) inhibits tumor angiogenesis in human lung cancer xenografts with a reduction in vascular endothelial growth factor.
- Author
-
Soto-Pantoja DR, Menon J, Gallagher PE, and Tallant EA
- Subjects
- Angiotensin I administration & dosage, Animals, Blotting, Western, Cell Line, Tumor, Cells, Cultured, Chick Embryo, Chorioallantoic Membrane blood supply, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells physiology, Humans, Injections, Subcutaneous, Lung Neoplasms blood supply, Lung Neoplasms pathology, Mice, Mice, Nude, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Neovascularization, Physiologic drug effects, Peptide Fragments administration & dosage, Proto-Oncogene Mas, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Reverse Transcriptase Polymerase Chain Reaction, Vascular Endothelial Growth Factor A genetics, Angiotensin I pharmacology, Lung Neoplasms drug therapy, Neovascularization, Pathologic prevention & control, Peptide Fragments pharmacology, Vascular Endothelial Growth Factor A metabolism, Xenograft Model Antitumor Assays
- Abstract
Angiotensin-(1-7) [Ang-(1-7)] is an endogenous seven-amino acid peptide hormone with antiproliferative properties. Our previous studies showed that Ang-(1-7) inhibits the growth of human lung cancer cells in vitro and reduces the size of human lung tumor xenografts in vivo. In the current study, s.c. injection of Ang-(1-7) not only caused a significant reduction in human A549 lung tumor growth but also markedly decreased vessel density, suggesting that the heptapeptide inhibits angiogenesis to reduce tumor size. A decrease in human endothelial cell tubule formation in Matrigel was observed following a 16 h incubation with Ang-(1-7), with a maximal reduction at a 10 nmol/L concentration. Ang-(1-7) had similar antiangiogenic effects in the chick chorioallantoic membrane, causing a >50% decrease in neovascularization. The Ang-(1-7)-induced reduction in both endothelial cell tubule formation and vessel formation in the chick was completely blocked by the specific Ang-(1-7) receptor antagonist [d-proline(7)]-Ang-(1-7), suggesting that these biological actions are mediated by an AT((1-7)) receptor. Ang-(1-7) significantly reduced vascular endothelial growth factor-A protein and mRNA in tumors from mice treated with the heptapeptide compared with saline controls as well as in the parent A549 human lung cancer cells in culture. These results suggest that Ang-(1-7) may attenuate tumor angiogenesis by reducing vascular endothelial growth factor-A, a primary proangiogenic protein. Taken together, this study shows that Ang-(1-7) exhibits significant antiangiogenic activity and may be a novel therapeutic agent for lung cancer treatment targeting a specific AT((1-7)) receptor.
- Published
- 2009
- Full Text
- View/download PDF
32. Regulation of ACE2 in cardiac myocytes and fibroblasts.
- Author
-
Gallagher PE, Ferrario CM, and Tallant EA
- Subjects
- Angiotensin I, Angiotensin II analogs & derivatives, Angiotensin II metabolism, Angiotensin II pharmacology, Angiotensin II Type 1 Receptor Blockers pharmacology, Angiotensin-Converting Enzyme 2, Animals, Animals, Newborn, Atrial Natriuretic Factor metabolism, Butadienes pharmacology, Cells, Cultured, Endothelin-1 metabolism, Fibroblasts drug effects, Flavonoids pharmacology, Gene Expression Regulation, Enzymologic, Heart Ventricles cytology, Heart Ventricles drug effects, Losartan pharmacology, Mitogen-Activated Protein Kinase 1 metabolism, Myocytes, Cardiac drug effects, Nitriles pharmacology, Peptide Fragments metabolism, Peptide Fragments pharmacology, Peptidyl-Dipeptidase A genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Mas, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Fibroblasts enzymology, Heart Ventricles enzymology, Myocytes, Cardiac enzymology, Peptidyl-Dipeptidase A metabolism
- Abstract
Angiotensin-converting enzyme 2 (ACE2) preferentially forms angiotensin-(1-7) [ANG-(1-7)] from ANG II. We showed that cardiac ACE2 is elevated following treatment of coronary artery-ligated rats with AT1 receptor blockers (ARBs). Cardiac myocytes and fibroblasts were isolated from neonatal rats to determine the molecular mechanisms for the ACE2 upregulation by ARB treatment. ANG II significantly reduced ACE2 activity and downregulated ACE2 mRNA in cardiac myocytes, effects blocked by the ARB losartan, indicating that ANG II regulates ACE2. ANG II also reduced ACE2 mRNA in cardiac fibroblasts; however, no enzyme activity was detected, reflecting the limited expression of ACE2 in these cells. Endothelin-1 (ET-1) also significantly reduced myocyte ACE2 mRNA. The reduction in ACE2 mRNA by ANG II or ET-1 was blocked by inhibitors of mitogen-activated protein kinase kinase 1, suggesting that ANG II or ET-1 activates extracellular signal-regulated kinase (ERK) 1/ERK2 to reduce ACE2. Although ACE2 mRNA was not affected by ANG-(1-7), both the ANG II- and ET-1-mediated reductions in ACE2 mRNA were blocked by the heptapeptide. The ANG-(1-7) modulatory effect was prevented by the ANG-(1-7) receptor antagonist [D-Ala7]-ANG-(1-7), indicating that the ANG-(1-7) response was mediated by a specific AT(1-7) receptor. Myocyte treatment with atrial natriuretic peptide (ANP) also reversed the ACE2 mRNA downregulation by ANG II or ET-1, whereas treatment with ANP alone was ineffective. These results indicate that multiple hypertrophic and anti-hypertropic peptides regulate ACE2 production in myocytes, suggesting that ACE2 expression in the heart is dependent upon the compliment and concentration of regulatory molecules.
- Published
- 2008
- Full Text
- View/download PDF
33. MAP kinase/phosphatase pathway mediates the regulation of ACE2 by angiotensin peptides.
- Author
-
Gallagher PE, Ferrario CM, and Tallant EA
- Subjects
- Angiotensin I, Angiotensin II analogs & derivatives, Angiotensin II pharmacology, Angiotensin-Converting Enzyme 2, Animals, Cells, Cultured, Flavonoids pharmacology, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Mitogen-Activated Protein Kinase Phosphatases antagonists & inhibitors, Mitogen-Activated Protein Kinases antagonists & inhibitors, Muscle, Smooth, Vascular drug effects, Myocytes, Smooth Muscle drug effects, Okadaic Acid pharmacology, Peptide Fragments pharmacology, Peptidyl-Dipeptidase A genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Mas, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled metabolism, Vanadates pharmacology, Angiotensin II metabolism, MAP Kinase Signaling System drug effects, Mitogen-Activated Protein Kinase Phosphatases metabolism, Mitogen-Activated Protein Kinases metabolism, Muscle, Smooth, Vascular enzymology, Myocytes, Smooth Muscle enzymology, Peptide Fragments metabolism, Peptidyl-Dipeptidase A metabolism
- Abstract
Angiotensin-converting enzyme 2 (ACE2) catalyzes the conversion of the vasoconstrictor angiotensin II (ANG II) to the vasodilatory peptide angiotensin-(1-7) [ANG-(1-7)]. We showed that treatment of hypertensive rats with the AT(1) receptor antagonist olmesartan increased ACE2 mRNA and protein in the thoracic aorta, suggesting that endogenous ANG II tonically reduces the enzyme. We now report that ANG II downregulates ACE2 activity and mRNA in rat aortic vascular smooth muscle cells (VSMCs) to reduce the conversion of ANG II to ANG-(1-7). Although ANG-(1-7) alone had no effect on the regulation of ACE2 mRNA, the heptapeptide prevented the ANG II-mediated reduction in ACE2 mRNA, an effect blocked by the selective ANG-(1-7) receptor antagonist [d-Ala(7)]-ANG-(1-7). The reduction in ACE2 mRNA by ANG II was also prevented by the mitogen-activated protein (MAP) kinase kinase inhibitor PD98059. Treatment of VSMCs with ANG II increased ERK1/ERK2 activity, which was significantly reduced by pretreatment with ANG-(1-7). Blockade of the ANG II-mediated reduction in ACE2 mRNA and increase in MAP kinase activity by ANG-(1-7) was prevented by pretreatment with sodium vanadate, a tyrosine phosphatase inhibitor, or okadaic acid, a serine-threonine phosphatase inhibitor, suggesting that the heptapeptide activates a MAP kinase phosphatase. This study is the first to show that the MAP kinase-phosphatase pathway is a primary molecular mechanism for regulating ACE2 to maintain the balance between ANG II and ANG-(1-7). The modulatory role of ANG-(1-7) in the regulation of ACE2 by ANG II suggests a complex interplay between the two peptides that is mediated by specific receptors to activate distinct signaling pathways.
- Published
- 2008
- Full Text
- View/download PDF
34. Angiotensin-(1-7) inhibits growth of human lung adenocarcinoma xenografts in nude mice through a reduction in cyclooxygenase-2.
- Author
-
Menon J, Soto-Pantoja DR, Callahan MF, Cline JM, Ferrario CM, Tallant EA, and Gallagher PE
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma genetics, Adenocarcinoma pathology, Animals, Cell Growth Processes drug effects, Cell Line, Tumor, Cyclooxygenase 2 biosynthesis, Cyclooxygenase 2 genetics, Humans, Lung Neoplasms enzymology, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Mice, Mice, Nude, RNA, Messenger biosynthesis, RNA, Messenger genetics, Xenograft Model Antitumor Assays, Adenocarcinoma drug therapy, Angiotensin I pharmacology, Cyclooxygenase 2 metabolism, Lung Neoplasms drug therapy, Peptide Fragments pharmacology
- Abstract
Angiotensin-(1-7) [Ang-(1-7)] is an endogenous peptide of the renin-angiotensin system with vasodilator and antiproliferative properties. Our previous studies showed that Ang-(1-7) reduced serum-stimulated growth of human lung cancer cells in vitro through activation of a unique AT((1-7)) receptor. The current study investigates the effect of Ang-(1-7) on lung tumor growth in vivo, using a human lung tumor xenograft model. Athymic mice with tumors resulting from injection of A549 human lung cancer cells were treated for 28 days with either i.v. saline or Ang-(1-7), delivered by implanted osmotic mini-pumps. Treatment with Ang-(1-7) reduced tumor volume by 30% compared with the size before treatment; in contrast, tumor size in the saline-treated animals increased 2.5-fold. These results correlate with a reduction in the proliferation marker Ki67 in the Ang-(1-7)-infused tumors when compared with the saline-infused tumor tissues. Treatment with Ang-(1-7) significantly reduced cyclooxygenase-2 (COX-2) mRNA and protein in tumors of Ang-(1-7)-infused mice when compared with mice treated with saline as well as in the parent A549 human lung cancer cells in tissue culture. These results suggest that Ang-(1-7) may decrease COX-2 activity and proinflammatory prostaglandins to inhibit lung tumor growth. In contrast, the heptapeptide had no effect on COX-1 mRNA in xenograft tumors or A549 cells. Because Ang-(1-7), a peptide with antithrombotic properties, reduces growth through activation of a selective AT((1-7)) receptor, our results suggest that the heptapeptide represents a novel treatment for lung cancer by reducing COX-2.
- Published
- 2007
- Full Text
- View/download PDF
35. Oxidative stress and glomerular filtration barrier injury: role of the renin-angiotensin system in the Ren2 transgenic rat.
- Author
-
Whaley-Connell AT, Chowdhury NA, Hayden MR, Stump CS, Habibi J, Wiedmeyer CE, Gallagher PE, Tallant EA, Cooper SA, Link CD, Ferrario C, and Sowers JR
- Subjects
- Albuminuria metabolism, Albuminuria pathology, Albuminuria physiopathology, Angiotensin II Type 1 Receptor Blockers pharmacology, Angiotensin-Converting Enzyme 2, Animals, Animals, Genetically Modified, Blood Pressure, Disease Models, Animal, Hypertension, Renal pathology, Kidney Glomerulus metabolism, Kidney Glomerulus pathology, Kidney Glomerulus physiopathology, Male, Malondialdehyde metabolism, Mice, Microscopy, Electron, Transmission, NADPH Oxidases metabolism, Neprilysin genetics, Neprilysin metabolism, Peptidyl-Dipeptidase A genetics, Peptidyl-Dipeptidase A metabolism, Podocytes pathology, Podocytes ultrastructure, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Renin metabolism, Tetrazoles pharmacology, Valine analogs & derivatives, Valine pharmacology, Valsartan, Hypertension, Renal metabolism, Hypertension, Renal physiopathology, Oxidative Stress physiology, Renin genetics, Renin-Angiotensin System physiology
- Abstract
TG(mRen2)27 (Ren2) transgenic rats overexpress the mouse renin gene, manifest hypertension, and exhibit increased tissue ANG II levels and oxidative stress. Evidence indicates that elevated tissue ANG II contributes to oxidative stress, increases in glomerular macromolecular permeability, and consequent albuminuria. Furthermore, angiotensin type 1 receptor (AT1R) blockers reduce albuminuria and slow progression of renal disease. However, it is not known whether improvements in glomerular filtration barrier integrity and albuminuria during treatment are related to reductions in oxidative stress and/or kidney renin-angiotensin system (RAS) activity. To investigate the renal protective effects of AT1R blockade, we treated young (6-7 wk old) male Ren2 rats with valsartan (Ren2-V; 30 mg/kg) for 3 wk and measured urine albumin, kidney malondialdehyde (MDA), RAS component mRNAs, and NADPH oxidase subunits (gp91(phox) and Rac1) compared with age-matched untreated Ren2 and Sprague-Dawley (S-D) rats. Basement membrane thickness, slit pore diameter and number, and foot process base width were measured by transmission electron microscopy (TEM). Results indicate that AT1R blockade lowered systolic blood pressure (30%), albuminuria (91%), and kidney MDA (80%) in Ren2-V compared with untreated Ren2 rats. Increased slit pore number and diameter and reductions in basement membrane thickness and podocyte foot process base width were strongly associated with albuminuria and significantly improved following AT1R blockade. AT1R blockade was also associated with increased angiotensin-converting enzyme-2 and neprilysin expression, demonstrating a beneficial shift in balance of renal RAS. Thus reductions in blood pressure, albuminuria, and tissue oxidative stress with AT1R blockade were associated with improved indexes of glomerular filtration barrier integrity and renal RAS in Ren2 rats.
- Published
- 2006
- Full Text
- View/download PDF
36. Effect of angiotensin II blockade on a new congenic model of hypertension derived from transgenic Ren-2 rats.
- Author
-
Jessup JA, Gallagher PE, Averill DB, Brosnihan KB, Tallant EA, Chappell MC, and Ferrario CM
- Subjects
- Angiotensin I blood, Angiotensin I urine, Angiotensin II antagonists & inhibitors, Angiotensin II blood, Angiotensin II metabolism, Angiotensin-Converting Enzyme 2, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Animals, Congenic, Animals, Genetically Modified, Crosses, Genetic, Hypertension, Renal metabolism, Kinetics, Lisinopril pharmacology, Losartan pharmacology, Male, Peptide Fragments blood, Peptide Fragments urine, Peptidyl-Dipeptidase A genetics, Peptidyl-Dipeptidase A physiology, RNA, Messenger metabolism, Rats, Rats, Inbred Lew, Renin-Angiotensin System drug effects, Renin-Angiotensin System physiology, Time Factors, Angiotensin II Type 1 Receptor Blockers pharmacology, Angiotensin Receptor Antagonists, Hypertension, Renal genetics, Models, Genetic, Peptidyl-Dipeptidase A drug effects, Renin genetics
- Abstract
The generation of the Lew.Tg(mRen2) congenic hypertensive rat strain, developed through a backcross of the hypertensive (mRen2)27 transgenic rat with normotensive Lewis rats, provides a new model by which primary hypertension can be studied without the genetic variability found in the original strain. The purpose of this study was to characterize the Lew.Tg(mRen2) rats by dually investigating the effects of type 1 angiotensin II (ANG II) receptor (AT(1)) blockade and angiotensin-converting enzyme (ACE) activity inhibition on the ANG-(1-7)/ACE2 axis of the renin-angiotensin system in this new hypertensive model. The control of blood pressure elicited by 12-day administration of either lisinopril (mean difference change = 92 +/- 2, P < 0.05) or losartan (mean difference change = 69 +/- 2, P < 0.05) was associated with 54% and 33% increases in cardiac ACE2 mRNA and 54% and 43% increases in cardiac ACE mRNA, respectively. Lisinopril induced a 3.1-fold (P < 0.05) increase in renal cortical expression of ACE2, whereas losartan increased ACE2 mRNA 3.5-fold (P < 0.05). Both treatment regimens increased renal ACE mRNA 2.6-fold (P < 0.05). The two therapies augmented ACE2 protein activity, as well as increased cardiac and renal AT(1) receptor mRNAs. ACE inhibition reduced plasma ANG II levels (81%, P < 0.05) and increased plasma ANG-(1-7) (265%, P < 0.05), whereas losartan had no effect on the peptides. In contrast with what had been shown in normotensive rats, ACE inhibition decreased renal ANG II excretion and transiently decreased ANG-(1-7) excretion, whereas losartan treatment was associated with a consistent decrease in ANG-(1-7) urinary excretion rates. In response to the treatments, the expression of both renal cortical renin and angiotensinogen mRNAs was significantly augmented. The paradoxical effects of blockade of ANG II synthesis and activity on urinary excretion rates of the peptides and plasma angiotensins levels suggest that, in Lew.Tg(mRen2) congenic rats, a failure of compensatory ACE2 and ANG-(1-7)-dependent vasodepressor mechanisms may contribute both to the development and progression of hypertension driven by increased formation of endogenous ANG II.
- Published
- 2006
- Full Text
- View/download PDF
37. Cardioprotective role for angiotensin-(1-7) and angiotensin converting enzyme 2 in the heart.
- Author
-
Tallant EA, Ferrario CM, and Gallagher PE
- Abstract
Angiotensin-(1-7), a biologically active peptide of the renin-angiotensin system, is cardioprotective following ischemia/reperfusion and reduces cardiac hypertrophy. A recently discovered homolog of angiotensin converting enzyme (ACE), ACE2, is present in the heart and synthesizes angiotensin-(1-7) from angiotensin II. Cardiac ACE2 is elevated following inhibition of Ang II subtype 1 (AT(1)) receptors or blockade of angiotensin II production, suggesting that angiotensin-(1-7) plays a role in the beneficial effects of AT(1) receptor antagonists and ACE inhibitors in the heart. An increase in ACE2 activity and the production of angiotensin-(1-7) may thus represent a novel therapy for heart failure following myocardial infarction.
- Published
- 2006
- Full Text
- View/download PDF
38. Distinct roles for ANG II and ANG-(1-7) in the regulation of angiotensin-converting enzyme 2 in rat astrocytes.
- Author
-
Gallagher PE, Chappell MC, Ferrario CM, and Tallant EA
- Subjects
- Angiotensin II Type 1 Receptor Blockers metabolism, Angiotensin-Converting Enzyme 2, Animals, Astrocytes cytology, Carboxypeptidases genetics, Cells, Cultured, Cerebellum cytology, Female, Imidazoles metabolism, Losartan metabolism, Medulla Oblongata metabolism, Peptidyl-Dipeptidase A, Pregnancy, Pyridines metabolism, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Tetrazoles metabolism, Valine analogs & derivatives, Valine metabolism, Valsartan, Vasoconstrictor Agents metabolism, Angiotensin I metabolism, Angiotensin II metabolism, Antihypertensive Agents metabolism, Astrocytes metabolism, Carboxypeptidases metabolism, Gene Expression Regulation, Peptide Fragments metabolism
- Abstract
Angiotensin-converting enzyme 2 (ACE2) is a homolog of ACE that preferentially forms angiotensin-(1-7) [ANG-(1-7)] from angiotensin II (ANG II). Incubation of neonatal rat cerebellar or medullary astrocytes with ANG II reduced ACE2 mRNA by approximately 60%, suggesting transcriptional regulation of the enzyme. In contrast, ANG II had no effect on ACE mRNA in astrocytes isolated from either brain region, demonstrating a differential regulation of the two enzymes by ANG II. The ANG II-mediated reduction in ACE2 mRNA was blocked by the angiotensin type 1 (AT(1)) receptor antagonists losartan or valsartan; the angiotensin type 2 (AT(2)) antagonist PD123319 was ineffective. The reduction in ACE2 mRNA by ANG II also was associated with a 50% decrease in cerebellar and medullary ACE2 protein, which was blocked by losartan. Treatment of medullary astrocytes with ANG-(1-7), the product of ACE2 hydrolysis of ANG II, did not affect ACE2 mRNA; however, ANG-(1-7) prevented the ANG II-mediated reduction in ACE2 mRNA. The addition of [d-Ala(7)]-ANG-(1-7), a selective AT((1-7)) receptor antagonist, blocked the inhibitory actions of ANG-(1-7). These data are the first to demonstrate transcriptional regulation of ACE2 by ANG II and ANG-(1-7). Because ACE2 preferentially converts ANG II to ANG-(1-7), downregulation of the enzyme by ANG II constitutes a novel positive feed-forward system within the brain that may favor ANG II-mediated neural responses. Furthermore, the modulatory role of ANG-(1-7) in the transcriptional regulation of ACE2 by ANG II suggests a complex interplay between these peptides that is mediated by distinct receptor systems.
- Published
- 2006
- Full Text
- View/download PDF
39. Angiotensin-(1-7) inhibits growth of cardiac myocytes through activation of the mas receptor.
- Author
-
Tallant EA, Ferrario CM, and Gallagher PE
- Subjects
- Animals, Animals, Newborn, Cell Division drug effects, Cells, Cultured, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Oligonucleotides, Antisense pharmacology, Proto-Oncogene Mas, Proto-Oncogene Proteins genetics, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Receptors, G-Protein-Coupled, Ventricular Remodeling physiology, Angiotensin I pharmacology, Antihypertensive Agents pharmacology, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Peptide Fragments pharmacology, Proto-Oncogene Proteins metabolism
- Abstract
Peptide hormones such as ANG II and endothelin contribute to cardiac remodeling after myocardial infarction by stimulating myocyte hypertrophy and myofibroblast proliferation. In contrast, angiotensin-(1-7) [ANG-(1-7)] infusion after myocardial infarction reduced myocyte size and attenuated ventricular dysfunction and remodeling. We measured the effect of ANG-(1-7) on protein and DNA synthesis in cultured neonatal rat myocytes to assess the role of the heptapeptide in cell growth. ANG-(1-7) significantly attenuated either fetal bovine serum- or endothelin-1-stimulated [(3)H]leucine incorporation into myocytes with no effect on [(3)H]thymidine incorporation. [d-Ala(7)]-ANG-(1-7), the selective ANG type 1-7 (AT(1-7)) receptor antagonist, blocked the ANG-(1-7)-mediated reduction in protein synthesis in cardiac myocytes, whereas the AT(1) and AT(2) angiotensin peptide receptors were ineffective. Serum-stimulated ERK1/ERK2 mitogen-activated protein kinase activity was significantly decreased by ANG-(1-7) in myocytes, a response that was also blocked by [d-Ala(7)]-ANG-(1-7). Both rat heart and cardiac myocytes express the mRNA for the mas receptor, and a 59-kDa immunoreactive protein was identified in both extracts of rat heart and cultured myocytes by Western blot hybridization with the use of an antibody to mas, an ANG-(1-7) receptor. Transfection of cultured myocytes with an antisense oligonucleotide to the mas receptor blocked the ANG-(1-7)-mediated inhibition of serum-stimulated MAPK activation, whereas a sense oligonucleotide was ineffective. These results suggest that ANG-(1-7) reduces the growth of cardiomyocytes through activation of the mas receptor. Because ANG-(1-7) is elevated after treatment with angiotensin-converting enzyme inhibitors or AT(1) receptor blockers, ANG-(1-7) may contribute to their beneficial effects on cardiac dysfunction and ventricular remodeling after myocardial infarction.
- Published
- 2005
- Full Text
- View/download PDF
40. Angiotensin II stimulates arachidonic acid release from bone marrow stromal cells.
- Author
-
Richmond RS, Tallant EA, Gallagher PE, Ferrario CM, and Strawn WB
- Subjects
- Angiotensin II Type 1 Receptor Blockers pharmacology, Angiotensin II Type 2 Receptor Blockers, Animals, Arachidonic Acid antagonists & inhibitors, Cells, Cultured, Humans, Imidazoles pharmacology, Losartan pharmacology, Macaca fascicularis, Macrophage Colony-Stimulating Factor metabolism, Pyridines pharmacology, Receptor, Angiotensin, Type 1 metabolism, Receptor, Angiotensin, Type 2 metabolism, Angiotensin II pharmacology, Arachidonic Acid metabolism, Bone Marrow Cells metabolism, Stromal Cells metabolism
- Abstract
Introduction: Angiotensin II (Ang II) is recognised as a regulator of haematopoiesis, but its actions within the bone marrow are not fully understood. Support of haematopoiesis by bone marrow stromal cells (MSC) is dependent on factors that include arachidonic acid and macrophage colony stimulating factor (MCSF), both of which are increased by Ang II stimulation in other tissues. To further elucidate the mechanisms of Ang II-regulated haematopoiesis, we determined whether Ang II-stimulation alters arachidonic acid release and MCSF secretion from MSC., Methods: Cynomolgus monkey MSC isolated from bone marrow aspirates and the human HS-5 stromal cell line were studied for Ang II-mediated arachidonic acid (AA) release, while secretion of MCSF in response to Ang II was studied in HS-5 cells. Cells were labelled overnight with 3H-AA and the release of 3H-AA was measured in culture medium following 20 minutes stimulation with Ang II, alone or in combination with the AT1- or AT2-receptor antagonists, losartan and PD 123319, respectively. MCSF secretion into culture medium was measured using an enzyme immunoassay following 24 hours of treatment with Ang II alone or in combination with losartan or PD 123319. Phorbol-myristate-acetate, known to stimulate release of AA and MCSF, was used as a positive control in both experiments., Results: In response to Ang II, release of 3H-AA from monkey and human MSC was increased (p<0.05) to 147+/-4% and 124+/-3% of control, respectively. The AT1- and AT2-receptor antagonists, losartan and PD 123319, individually reduced Ang II-stimulated 3H-AA release. In contrast, Ang II had no effect on secretion of MCSF from HS-5 cells., Conclusions: These results provide mechanistic evidence for Ang II-mediated haematopoiesis through AA release that may, in part, explain Ang II-facilitated recovery of haematopoiesis in experimental myelosuppression and the anaemias associated with Ang II receptor blockade.
- Published
- 2004
- Full Text
- View/download PDF
41. Inhibition of human lung cancer cell growth by angiotensin-(1-7).
- Author
-
Gallagher PE and Tallant EA
- Subjects
- Cell Line, Tumor, Culture Media, DNA Replication drug effects, Humans, Kinetics, Lung Neoplasms pathology, Mitogen-Activated Protein Kinase 3 metabolism, Angiotensin I pharmacology, Antineoplastic Agents pharmacology, Cell Division drug effects, Peptide Fragments pharmacology
- Abstract
Angiotensin-(1-7) [Ang-(1-7)] is an endogenous peptide hormone of the renin-angiotensin system with vasodilator and anti-proliferative properties. Human adenocarcinoma SK-LU-1 and A549 cells as well as non-small lung cancer SK-MES-1 cells were treated with serum in the presence and absence of Ang-(1-7), to determine whether Ang-(1-7) inhibits the growth of lung cancer cells. Ang-(1-7) caused a significant reduction in serum-stimulated growth in all three lung cancer cell lines. Treatment with Ang-(1-7) resulted in both a dose- and time-dependent reduction in serum-stimulated DNA synthesis in all three cell lines, with IC(50)'s in the sub-nanomolar range. The Ang-(1-7) receptor antagonist [D-Ala(7)]-Ang-(1-7) blocked the attenuation of the serum-stimulated DNA synthesis of SK-LU-1 cells by Ang-(1-7), while neither AT(1) nor AT(2) angiotensin receptor subtype antagonists prevented the response to the heptapeptide. MAS mRNA and protein, a receptor for Ang-(1-7), was detected in the three lung cancer cell lines, suggesting that the anti-proliferative effect of Ang-(1-7) in the cancer cells may be mediated by the non-AT(1), non-AT(2), AT((1-7)) receptor MAS. Other angiotensin peptides [Ang I, Ang II, Ang-(2-8), Ang-(3-8) and Ang-(3-7)] did not attenuate mitogen-stimulated DNA synthesis of SK-LU-1 cells, demonstrating that Ang-(1-7) selectively inhibits SK-LU-1 cancer cell growth. Pre-treatment of SK-LU-1 cells with 10 nM Ang-(1-7) reduced serum-stimulated phosphorylation of extracellular signal-regulated kinase (ERK)1 and ERK2, indicating that the anti-proliferative effects may occur, at least in part, through inhibition of the ERK signal transduction pathway. The results of this study suggest that Ang-(1-7) inhibits lung cancer cell growth through the activation of an angiotensin peptide receptor and may represent a novel chemotherapeutic and chemopreventive treatment for lung cancer.
- Published
- 2004
- Full Text
- View/download PDF
42. Upregulation of angiotensin-converting enzyme 2 after myocardial infarction by blockade of angiotensin II receptors.
- Author
-
Ishiyama Y, Gallagher PE, Averill DB, Tallant EA, Brosnihan KB, and Ferrario CM
- Subjects
- Angiotensin I biosynthesis, Angiotensin I blood, Angiotensin I genetics, Angiotensin II blood, Angiotensin-Converting Enzyme 2, Animals, Carboxypeptidases genetics, Carboxypeptidases physiology, Cardiomyopathy, Hypertrophic etiology, Coronary Vessels, Disease Models, Animal, Enzyme Induction drug effects, Imidazoles therapeutic use, Ligation, Losartan therapeutic use, Male, Myocardial Infarction complications, Myocardial Infarction physiopathology, Olmesartan Medoxomil, Peptide Fragments biosynthesis, Peptide Fragments genetics, Peptidyl-Dipeptidase A biosynthesis, Peptidyl-Dipeptidase A genetics, Pyridines pharmacology, RNA, Messenger biosynthesis, Rats, Rats, Inbred Lew, Receptor, Angiotensin, Type 1 physiology, Renin-Angiotensin System drug effects, Renin-Angiotensin System physiology, Tetrazoles therapeutic use, Ventricular Remodeling drug effects, Angiotensin II physiology, Angiotensin II Type 1 Receptor Blockers, Carboxypeptidases biosynthesis, Cardiomyopathy, Hypertrophic prevention & control, Imidazoles pharmacology, Losartan pharmacology, Myocardial Infarction drug therapy, Myocardium enzymology, Tetrazoles pharmacology
- Abstract
We investigated in Lewis normotensive rats the effect of coronary artery ligation on the expression of cardiac angiotensin-converting enzymes (ACE and ACE 2) and angiotensin II type-1 receptors (AT1a-R) 28 days after myocardial infarction. Losartan, olmesartan, or the vehicle (isotonic saline) was administered via osmotic minipumps for 28 days after coronary artery ligation or sham operation. Coronary artery ligation caused left ventricular dysfunction and cardiac hypertrophy. These changes were associated with increased plasma concentrations of angiotensin I, angiotensin II, angiotensin-(1-7), and serum aldosterone, and reduced AT1a-R mRNA. Cardiac ACE and ACE 2 mRNAs did not change. Both angiotensin II antagonists attenuated cardiac hypertrophy; olmesartan improved ventricular contractility. Blockade of the AT1a-R was accompanied by a further increase in plasma concentrations of the angiotensins and reduced serum aldosterone levels. Both losartan and olmesartan completely reversed the reduction in cardiac AT1a-R mRNA observed after coronary artery ligation while augmenting ACE 2 mRNA by approximately 3-fold. Coadministration of PD123319 did not abate the increase in ACE 2 mRNA induced by losartan. ACE 2 mRNA correlated significantly with angiotensin II, angiotensin-(1-7), and angiotensin I levels. These results provide evidence for an effect of angiotensin II blockade on cardiac ACE 2 mRNA that may be due to direct blockade of AT1a receptors or a modulatory effect of increased angiotensin-(1-7).
- Published
- 2004
- Full Text
- View/download PDF
43. Molecular mechanisms of inhibition of vascular growth by angiotensin-(1-7).
- Author
-
Tallant EA and Clark MA
- Subjects
- Angiotensin I, Angiotensin II antagonists & inhibitors, Animals, Cell Division drug effects, Cells, Cultured, Cyclic AMP physiology, Mitogen-Activated Protein Kinases metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Prostaglandins physiology, Rats, Rats, Sprague-Dawley, Angiotensin II pharmacology, Muscle, Smooth, Vascular drug effects, Peptide Fragments pharmacology
- Abstract
Angiotensin (Ang) peptides play a critical role in regulating vascular reactivity and structure. We showed that Ang-(1-7) reduced smooth muscle growth after vascular injury and attenuated the proliferation of vascular smooth muscle cells (VSMCs). This study investigated the molecular mechanisms of the antiproliferative effects of Ang-(1-7) in cultured rat aortic VSMCs. Ang-(1-7) caused a dose-dependent release of prostacyclin from VSMCs, with a maximal release of 277.9+/-25.2% of basal values (P<0.05) by 100 nmol/L Ang-(1-7). The cyclooxygenase inhibitor indomethacin significantly attenuated growth inhibition by Ang-(1-7). In contrast, neither a lipoxygenase inhibitor nor a cytochrome p450 epoxygenase inhibitor prevented the antiproliferative effects of Ang-(1-7). These results suggest that Ang-(1-7) inhibits vascular growth by releasing prostacyclin. Ang-(1-7) caused a dose-dependent release of cAMP, which might result from prostacyclin-mediated activation of adenylate cyclase. The cAMP-dependent protein kinase inhibitor Rp-adenosine-3',5'-cyclic monophosphorothioate attenuated the Ang-(1-7)-mediated inhibition of serum-stimulated thymidine incorporation. Finally, Ang-(1-7) inhibited Ang II stimulation of mitogen-activated protein kinase activities (ERK1/2). Incubation of VSMCs with concentrations of Ang-(1-7) up to 1 micromol/L had no effect on ERK1/2 activation. However, preincubation with increasing concentrations of Ang-(1-7) caused a dose-dependent reduction in Ang II-stimulated ERK1/2 activities. Ang-(1-7) (1 micromol/L) reduced 100 nmol/L Ang II-stimulated ERK1 and ERK2 activation by 42.3+/-6.2% and 41.2+/-4.2%, respectively (P<0.01). These results suggest that Ang-(1-7) inhibits vascular growth through the release of prostacyclin, through the prostacyclin-mediated production of cAMP and activation of cAMP-dependent protein kinase, and by attenuation of mitogen-activated protein kinase activation.
- Published
- 2003
- Full Text
- View/download PDF
44. Angiotensin-(1-7) reduces renal angiotensin II receptors through a cyclooxygenase-dependent mechanism.
- Author
-
Clark MA, Tallant EA, Tommasi E, Bosch S, and Diz DI
- Subjects
- Angiotensin Receptor Antagonists, Animals, Dose-Response Relationship, Drug, Kidney chemistry, Male, Protein Binding drug effects, Protein Binding physiology, Rats, Rats, Sprague-Dawley, Receptors, Angiotensin analysis, Angiotensin I pharmacology, Kidney drug effects, Kidney metabolism, Peptide Fragments pharmacology, Prostaglandin-Endoperoxide Synthases metabolism, Receptors, Angiotensin metabolism
- Abstract
In the kidney, angiotensin-(1-7) [Ang-(1-7)] exhibits diuretic and natriuretic properties associated with an increase in prostaglandin production. The prohypertensive effects of Ang II are attenuated in rats infused with Ang-(1-7), consistent with recent work showing that Ang-(1-7) downregulates AT1 receptors in Chinese hamster ovary-AT1A or vascular smooth muscle cells. To determine whether exposure to Ang-(1-7) reduces AT1 receptors in the kidney through an increase in prostaglandin production, kidney slices from Sprague-Dawley rats were incubated with 10 n -1 microM Ang-(1-7) in the presence or absence of 5 microM meclofenamate, a cyclooxygenase inhibitor. Following these treatments, the kidney slices were retrieved, frozen, and sectioned for determination of [125I]-Ang II binding using in vitro receptor autoradiography. Greater than 90% of the specific binding was competed for by losartan, indicating that the majority of binding was to the AT1 receptor. Incubation of kidney slices with 1 microM Ang-(1-7) caused a 20% reduction in [125I]-Ang II binding (n = 8) in the cortical tubulointerstitium, which was prevented when Ang-(1-7)-treated slices were incubated in the presence of 5 microM meclofenamate (1 +/- 2% increase, n = 8; p < 0.05). Incubation with 5 microM meclofenamate alone had no effect on [125I]-Ang II binding (-3 +/- 3%). The decrease in [125I]-Ang II binding with Ang-(1-7) was also blocked by the Ang-(1-7) antagonist [d-Ala7]-Ang-(1-7). Treatment with 1 microM [d-Ala7]-Ang-(1-7) alone had no effect on [125I]-Ang II binding (-3 +/- 6% of control). Pretreatment with 1 microM Ang II caused a similar reduction in [125I]-Ang II binding in the cortical tubulointerstitium. Neither Ang-(1-7) nor Ang II had any effect on [125I]-Ang II binding in the glomeruli and the area of the vasa recta of the kidney. These original findings suggest that prior exposure to Ang-(1-7) or Ang II causes a modest decrease in the number of AT1 receptors in the cortical tubulointerstitial area of the kidney. The reduction in Ang II binding by Ang-(1-7) was blocked by meclofenamate and [d-Ala7]-Ang-(1-7), suggesting that cyclooxygenase products released through activation of a novel receptor participate in this effect.
- Published
- 2003
- Full Text
- View/download PDF
45. Downregulation of the AT1A receptor by pharmacologic concentrations of Angiotensin-(1-7).
- Author
-
Clark MA, Tallant EA, and Diz DI
- Subjects
- Angiotensin II metabolism, Animals, CHO Cells, Cricetinae, Dose-Response Relationship, Drug, Inositol Phosphates metabolism, Radioligand Assay, Receptor, Angiotensin, Type 1, Receptors, Angiotensin genetics, Angiotensin I pharmacology, Antihypertensive Agents pharmacology, Down-Regulation physiology, Peptide Fragments pharmacology, Receptors, Angiotensin metabolism
- Abstract
Angiotensin (Ang)-(1-7), the amino terminal heptapeptide fragment of Ang II, is an endogenous Ang peptide with vasodilatory and antiproliferative actions. Because Ang II causes vasoconstriction and promotes growth through activation of Ang type 1 (AT1) receptors, we investigated whether the actions of Ang-(1-7) are due to its regulation of these receptors. Studies were performed in CHO cells stably transfected with the AT1A receptor. Ang-(1-7) competed poorly with [125I]-Ang II for the AT1A binding site and was ineffective at shifting the IC50 for Ang II competition with [125I]-Ang II for binding to the AT1A receptor. However, if CHO-AT1A cells were pretreated with Ang-(1-7) and then treated with acidic glycine to remove surface-bound ligand, the heptapeptide caused a concentration-dependent reduction in Ang II binding, with a maximal inhibition to 67.8 +/- 4.6% of total (p < 0.05) at 1 microM Ang-(1-7) compared with a reduction to 24% of total by 10 nM Ang II. Ang-(1-7) pretreatment caused a small but significant decrease in the affinity of [125I]-Ang II for the AT1A receptor and a significant reduction in the total number of binding sites. The Ang-(1-7)-induced reduction in binding was rapid (occurring as early as 5 min after exposure to the peptide), was maintained for 30 min during continued exposure of the cells to Ang-(1-7), and rapidly recovered after removal of the heptapeptide. The AT1 receptor antagonist L-158,809 reduced the Ang-(1-7)-induced downregulation of the AT1A receptor, suggesting that interactions with AT1A receptors mediate the regulatory events. Pretreatment with 1 microM or 10 microM Ang-(1-7) significantly reduced inositol phosphate production in response to 10 nM Ang II. The decrease in binding and responsiveness of the AT1A receptor after exposure to micromolar concentrations of Ang-(1-7) suggests that the heptapeptide downregulates the AT1A receptor to reduce responses to Ang II. Because downregulation of the receptor only occurred at micromolar concentrations of the heptapeptide, our findings suggest that Ang-(1-7) is not a potent antagonist at the AT1A receptor. However, when the balance between Ang II and Ang-(1-7) is shifted in favor of Ang-(1-7), such as during inhibition of Ang-converting enzyme, some contribution of this mechanism may come into play.
- Published
- 2001
- Full Text
- View/download PDF
46. Angiotensin-(1-7) downregulates the angiotensin II type 1 receptor in vascular smooth muscle cells.
- Author
-
Clark MA, Diz DI, and Tallant EA
- Subjects
- Analysis of Variance, Angiotensin I administration & dosage, Angiotensin II antagonists & inhibitors, Angiotensin II metabolism, Angiotensin Receptor Antagonists, Animals, Binding, Competitive, Cells, Cultured, Down-Regulation, Imidazoles pharmacology, Male, Peptide Fragments administration & dosage, Rats, Rats, Sprague-Dawley, Receptors, Angiotensin drug effects, Regression Analysis, Tetrazoles pharmacology, Type C Phospholipases metabolism, Angiotensin I metabolism, Muscle, Smooth, Vascular metabolism, Peptide Fragments metabolism, Receptors, Angiotensin metabolism
- Abstract
Angiotensin (Ang)-(1-7) is a biologically active peptide of the renin-angiotensin system that has both vasodilatory and antiproliferative activities that are opposite the constrictive and proliferative effects of angiotensin II (Ang II). We studied the actions of Ang-(1-7) on the Ang II type 1 (AT(1)) receptor in cultured rat aortic vascular smooth muscle cells to determine whether the effects of Ang-(1-7) are due to its regulation of the AT(1) receptor. Ang-(1-7) competed poorly for [(125)I]Ang II binding to the AT(1) receptor on vascular smooth muscle cells, with an IC(50) of 2.0 micromol/L compared with 1.9 nmol/L for Ang II. The pretreatment of vascular smooth muscle cells with Ang-(1-7) followed by treatment with acidic glycine to remove surface-bound peptide resulted in a significant decrease in [(125)I]Ang II binding; however, reduced Ang II binding was observed only at micromolar concentrations of Ang-(1-7). Scatchard analysis of vascular smooth muscle cells pretreated with 1 micromol/L Ang-(1-7) showed that the reduction in Ang II binding resulted from a loss of the total number of binding sites [B(max) 437.7+/-261.5 fmol/mg protein in Ang-(1-7)-pretreated cells compared with 607.5+/-301.2 fmol/mg protein in untreated cells, n=5, P<0.05] with no significant effect on the affinity of Ang II for the AT(1) receptor. Pretreatment with the AT(1) receptor antagonist L-158,809 blocked the reduction in [(125)I]Ang II binding by Ang-(1-7) or Ang II. Pretreatment of vascular smooth muscle cells with increasing concentrations of Ang-(1-7) reduced Ang II-stimulated phospholipase C activity; however, the decrease was significant (81.2+/-6.4%, P<0.01, n=5) only at 1 micromol/L Ang-(1-7). These results demonstrate that pharmacological concentrations of Ang-(1-7) in the micromolar range cause a modest downregulation of the AT(1) receptor on vascular cells and a reduction in Ang II-stimulated phospholipase C activity. Because the antiproliferative and vasodilatory effects of Ang-(1-7) are observed at nanomolar concentrations of the heptapeptide, these responses to Ang-(1-7) cannot be explained by competition of Ang-(1-7) at the AT(1) receptor or Ang-(1-7)-mediated downregulation of the vascular AT(1) receptor.
- Published
- 2001
- Full Text
- View/download PDF
47. State-of-the-Art lecture. Antiproliferative actions of angiotensin-(1-7) in vascular smooth muscle.
- Author
-
Tallant EA, Diz DI, and Ferrario CM
- Subjects
- Animals, Cardiovascular Diseases metabolism, Cardiovascular Diseases pathology, Cardiovascular Diseases physiopathology, Cell Division physiology, Humans, Angiotensin I physiology, Angiotensin II physiology, Muscle, Smooth, Vascular pathology, Muscle, Smooth, Vascular physiology, Peptide Fragments physiology
- Abstract
Hemodynamic factors, circulating hormones, paracrine factors, and intracrine factors influence vascular smooth muscle growth and plasticity. The well-characterized role of angiotensin II in the modulation of vascular tone and cell function may be critically involved in the mechanisms by which vascular smooth muscle responds to signals associated with vascular endothelial dysfunction and increases in oxidative stress. Studies from this laboratory suggest that the trophic actions of angiotensin II may be intrinsically regulated by angiotensin-(1-7), a separate product of the angiotensin system derived from the common substrate, angiotensin I. Exposure of cultured vascular smooth muscle cells to angiotensin-(1-7) inhibited the trophic actions of angiotensin II and reduced the expression of the mitogenic effects of both normal serum and platelet-derived growth factor. The growth-inhibitory actions of angiotensin-(1-7) were blocked by the selective D-alanine(7)-angiotensin-(1-7) antagonist and the nonselective angiotensin receptor blocker sarcosine(1)-threonine(8)-angiotensin II. In contrast, subtype-selective antagonists for the AT(1) and AT(2) receptors had no effect on the inhibitory actions of angiotensin-(1-7), a finding that is consistent with the pharmacological characterization of a high-affinity (125)I-labeled angiotensin-(1-7) binding site in the vasculature by use of selective and nonselective angiotensin II receptor antagonists. The relevance of these findings to the proliferative response of vascular smooth muscle cells after endothelial injury was confirmed by assessment of the effect of a 12-day infusion of angiotensin-(1-7) on neointimal formation. In these experiments, the proliferative response produced by injuring the carotid artery was inhibited by angiotensin-(1-7) through a mechanism that could not be explained by changes in arterial pressure. Because plasma angiotensin-(1-7) increased to levels comparable to those found in animals and human subjects given therapeutic doses of angiotensin-converting enzyme inhibitors, angiotensin-(1-7) may be one factor participating in the reversal of vascular proliferation during inhibition of angiotensin II formation or activity.
- Published
- 1999
- Full Text
- View/download PDF
48. Angiotensin II AT1-receptor blockade inhibits monocyte activation and adherence in transgenic (mRen2)27 rats.
- Author
-
Strawn WB, Gallagher PE, Tallant EA, Ganten D, and Ferrario CM
- Subjects
- Angiotensin II blood, Angiotensin II drug effects, Animals, Animals, Genetically Modified, Antihypertensive Agents pharmacology, Blood Cell Count drug effects, Blood Pressure drug effects, Cell Adhesion, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular pathology, Female, Heart Rate drug effects, Hematocrit, Hydralazine, Hypertension genetics, Losartan pharmacology, Male, Monocytes cytology, Monocytes drug effects, Nitroblue Tetrazolium metabolism, Peptide Fragments blood, Peptide Fragments drug effects, Rats, Rats, Sprague-Dawley, Receptor, Angiotensin, Type 1, Receptor, Angiotensin, Type 2, Renin-Angiotensin System drug effects, Angiotensin Receptor Antagonists, Hypertension metabolism, Monocytes metabolism
- Abstract
This study investigated whether angiotensin II AT1-receptor blockade with losartan inhibits endothelium-monocyte interactions originating from long-term activation of the renin-angiotensin system in hypertensive transgenic rats [TGR(mRen2)27]. The number of circulating activated monocytes, monocytes adhered to thoracic aorta endothelium, and the extent of endothelial cell injury were compared in adult male transgenic (mRen2)27 and age-matched Hannover Sprague-Dawley (SD) rats after 12 days of continuous subcutaneous administration of saline (120 microl/24 h), losartan (10 mg/kg/24 h), or the vasodilator hydralazine (3 mg/kg/24 h). At the doses administered in this experiment, both losartan and hydralazine normalized mRen2 rat blood pressures equal to values in similarly treated SD rats. Compared with saline infusion, administration of either antihypertensive in mRen2 rats reduced (p<0.05) endothelial cell injury, but only losartan significantly (p<0.05) decreased the number of activated circulating and endothelium-adherent monocytes. Infusion of antihypertensives in SD rats had no effect on blood pressures, monocyte activity, or endothelial injury compared with saline administration. These findings suggest that the recruitment and infiltration of leukocytes into the subendothelium associated with renin-angiotensin system-induced hypertension is partly mediated by pressure-independent AT1-receptor pathways.
- Published
- 1999
- Full Text
- View/download PDF
49. Angiotensin-(1-7) reduces smooth muscle growth after vascular injury.
- Author
-
Strawn WB, Ferrario CM, and Tallant EA
- Subjects
- Angiotensin I, Angiotensin II administration & dosage, Animals, Carotid Artery, Common cytology, Catheterization adverse effects, Cell Division drug effects, Infusions, Intravenous, Male, Peptide Fragments administration & dosage, Rats, Rats, Sprague-Dawley, Tunica Intima cytology, Angiotensin II pharmacology, Carotid Artery Injuries, Carotid Artery, Common drug effects, Peptide Fragments pharmacology, Tunica Intima drug effects, Tunica Intima injuries
- Abstract
Regulation of vascular smooth muscle cell growth is critical to the maintenance of normal blood flow and vessel patency. Angiotensin-(1-7) [Ang-(1-7)] inhibits proliferation of vascular smooth muscle cells in vitro and opposes the mitogenic effects of angiotensin II. The present study investigated whether Ang-(1-7) inhibits vascular smooth muscle cell growth in vivo by determining its effect on neointimal formation and medial remodeling in balloon-injured carotid arteries. The carotid arteries of adult male Sprague-Dawley rats were injured with a balloon embolectomy catheter. Ang-(1-7) in saline (24 microg/kg per hour) or saline alone was infused intravenously for 12 days after injury. Pumps containing bromodeoxyuridine were implanted at the same time to determine DNA synthesis. Intravenous infusion increased plasma Ang-(1-7) to 166. 0+/-41.2 fmol/mL (n=6) compared with 46.9+/-4.2 fmol/mL (n=8) in saline-infused rats. Plasma concentrations of Ang II were not changed by Ang-(1-7) infusion. Elevation in circulating Ang-(1-7) had no effect on either blood pressure or heart rate compared with saline controls. Histomorphometric analysis of carotid arteries indicated that Ang-(1-7) infusion significantly reduced neointimal area compared with rats infused with saline (0.063+/-0.011 versus 0. 100+/-0.009 mm2; P<0.05). In contrast, Ang-(1-7) infusion had no effect on medial area of the injured or the contralateral uninjured artery compared with saline controls. Ang-(1-7) infusion also reduced the rate of DNA synthesis in both the neointima and the media of the injured vessels. Therefore, exogenous Ang-(1-7) inhibited vascular smooth muscle cell proliferation associated with balloon-catheter injury. Similar increases in endogenous plasma Ang-(1-7) and inhibition of neointimal growth were observed in rats after angiotensin-converting enzyme inhibitor or angiotensin type 1 receptor antagonist administration, suggesting that Ang-(1-7) may contribute to the in vivo antiproliferative effects of these agents on vascular smooth muscle.
- Published
- 1999
- Full Text
- View/download PDF
50. Angiotensin-(1-7): a novel vasodilator of the coronary circulation.
- Author
-
Brosnihan KB, Li P, Tallant EA, and Ferrario CM
- Subjects
- Angiotensin I, Animals, Bradykinin pharmacology, Dogs, Angiotensin II pharmacology, Coronary Circulation drug effects, Peptide Fragments pharmacology, Vasodilator Agents pharmacology
- Abstract
Angiotensin-(1-7) [Ang-(1-7)] possesses novel biological functions that are distinct from angiotensin II (Ang II). In coronary arteries, the octapeptide Ang II and the heptapeptide Ang-(1-7) exert opposing actions. Ang II elicits vasoconstriction and Ang-(1-7) is a vasodilator. Ang-(1-7) elicits vasodilation by an endothelium-dependent release of nitric oxide. Further, the vasorelaxant activity is markedly attenuated by the bradykinin (BK) B2 receptor antagonist icatibant and does not appear to be associated with the synthesis and release of prostaglandins. Ang-(1-7) vasodilation is mediated by a non-AT1/AT2 receptor, since [Sar1Thr8]-Ang II, but neither candesartan, an AT1 receptor antagonist, nor PD123319, an AT2 receptor antagonist, blocked the response. Specific and high affinity binding of 125I-Ang-(1-7) to the endothelial layer of canine coronary arteries was demonstrated using in vitro emulsion autoradiography. Binding was effectively competed for by either unlabeled Ang-(1-7) or the specific Ang-(1-7) antagonist [D-Ala7]-Ang-(1-7). Additionally, Ang-(1-7) potentiated synergistically BK-induced vasodilation. The EC50 of BK vasodilation (2.45 +/- 0.51 nmol/L vs 0.37 +/- 0.08 nmol/L) was shifted 6.6-fold left-ward in the presence of 2 mumol/L concentration of Ang-(1-7). The potentiated response was specific for BK, since Ang-(1-7) did not augment the vasodilation produced by either acetylcholine or sodium nitroprusside; further, it was specific for Ang-(1-7), since neither Ang I nor Ang II augmented the BK response. In contrast to the vasodilator actions of Ang-(1-7), the potentiated response was not blocked by candesartan, PD123319 or [Sar1Thr8]-Ang II. Novel studies from our group demonstrate that Ang-(1-7) is both a substrate and inhibitor for angiotensin converting enzyme (ACE). Ang-(1-7) was shown to retard the degradation of 125I-[Tyr0]-BK in coronary rings. These studies describe novel actions of Ang-(1-7) as a vasodilator and a local synergistic modulator of kinin-induced vasodilation in coronary arteries.
- Published
- 1998
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.