Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
2.
J Pharmacol Exp Ther ; 388(2): 495-505, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37827703

ABSTRACT

The chemical warfare agent sulfur mustard and its structural analog nitrogen mustard (NM) cause severe vesicating skin injuries. The pathologic mechanisms for the skin injury following mustard exposure are poorly understood; therefore, no effective countermeasure is available. Previous reports demonstrated the protective activity of carvedilol, a US Food and Drug Administration (FDA)-approved ß-blocker, against UV radiation-induced skin damage. Thus, the current study evaluated the effects of carvedilol on NM-induced skin injuries in vitro and in vivo. In the murine epidermal cell line JB6 Cl 41-5a, ß-blockers with different receptor subtype selectivity were examined. Carvedilol and both of its enantiomers, R- and S-carvedilol, were the only tested ligands statistically reducing NM-induced cytotoxicity. Carvedilol also reduced NM-induced apoptosis and p53 expression. In SKH-1 mice, NM increased epidermal thickness, damaged skin architecture, and induced nuclear factor κB (NF-κB)-related proinflammatory genes as assessed by RT2 Profiler PCR (polymerase chain reaction) Arrays. To model chemical warfare scenario, 30 minutes after exposure to NM, 10 µM carvedilol was applied topically. Twenty-four hours after NM exposure, carvedilol attenuated NM-induced epidermal thickening, Ki-67 expression, a marker of cellular proliferation, and multiple proinflammatory genes. Supporting the in vitro data, the non-ß-blocking R-enantiomer of carvedilol had similar effects as racemic carvedilol, and there was no difference between carvedilol and R-carvedilol in the PCR array data, suggesting that the skin protective effects are independent of the ß-adrenergic receptors. These data suggest that the ß-blocker carvedilol and its enantiomers can be repurposed as countermeasures against mustard-induced skin injuries. SIGNIFICANCE STATEMENT: The chemical warfare agent sulfur mustard and its structural analog nitrogen mustard cause severe vesicating skin injuries for which no effective countermeasure is available. This study evaluated the effects of US Food and Drug Administration (FDA)-approved ß-blocker carvedilol on nitrogen mustard-induced skin injuries to repurpose this cardiovascular drug as a medical countermeasure.


Subject(s)
Chemical Warfare Agents , Mustard Gas , Animals , Mice , Mechlorethamine/toxicity , Mechlorethamine/metabolism , Carvedilol/pharmacology , Carvedilol/therapeutic use , Carvedilol/metabolism , Chemical Warfare Agents/toxicity , Mustard Gas/pharmacology , Mustard Gas/toxicity , Skin , Adrenergic beta-Antagonists/pharmacology
4.
Nanomaterials (Basel) ; 13(5)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36903807

ABSTRACT

The R-carvedilol enantiomer, present in the racemic mixture of the chiral drug carvedilol, does not bind to the ß-adrenergic receptors, but exhibits skin cancer preventive activity. For skin delivery, R-carvedilol-loaded transfersomes were prepared using various ratios of drug, lipids, and surfactants, and characterized for particle size, zeta potential, encapsulation efficiency, stability, and morphology. Transfersomes were compared for in vitro drug release and ex vivo skin penetration and retention. Skin irritation was evaluated by viability assay on murine epidermal cells and reconstructed human skin culture. Single-dose and repeated-dose dermal toxicity was determined in SKH-1 hairless mice. Efficacy was evaluated in SKH-1 mice exposed to single or multiple ultraviolet (UV) radiations. Transfersomes released the drug at a slower rate, but significantly increased skin drug permeation and retention compared with the free drug. The transfersome with a drug-lipid-surfactant ratio of 1:3:0.5 (T-RCAR-3) demonstrated the highest skin drug retention and was selected for further studies. T-RCAR-3 at 100 µM did not induce skin irritation in vitro and in vivo. Topical treatment with T-RCAR-3 at 10 µM effectively attenuated acute UV-induced skin inflammation and chronic UV-induced skin carcinogenesis. This study demonstrates feasibility of using R-carvedilol transfersome for preventing UV-induced skin inflammation and cancer.

5.
Cancers (Basel) ; 15(3)2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36765542

ABSTRACT

The current study evaluated the effects of the ß-blocker carvedilol on benzo(a)pyrene (B(a)P) and its active metabolite benzo(a)pyrene diol epoxide (BPDE)-induced lung toxicity, inflammation and carcinogenesis and explored the potential mechanisms. Carvedilol blocked the BPDE-induced malignant transformation of human bronchial epithelial cells BEAS-2B. In BEAS-2B cells, B(a)P strongly activated ELK-1, a transcription factor regulating serum response element (SRE) signaling, which was attenuated by carvedilol. Carvedilol also inhibited the B(a)P-induced AhR/xenobiotic responsive element (XRE) and mRNA expression of CYP1A1 and attenuated B(a)P-induced NF-κB activation. In a B(a)P-induced acute lung toxicity model in CD-1/IGS mice, pretreatment with carvedilol for 7 days before B(a)P exposure effectively inhibited the B(a)P-induced plasma levels of lactate dehydrogenase and malondialdehyde, inflammatory cell infiltration and histopathologic abnormalities in the lung, and upregulated the expression of GADD45α, caspase-3 and COX-2 in the lung. In a B(a)P-induced lung carcinogenesis model in A/J mice, carvedilol treatment for 20 weeks did not affect body weight but significantly attenuated tumor multiplicity and volume. These data reveal a previously unexplored role of carvedilol in preventing B(a)P-induced lung inflammation and carcinogenesis by inhibiting the cross-talk of the oncogenic transcription factors ELK-1, AhR and NF-κB.

6.
Adv Pharmacol ; 94: 141-182, 2022.
Article in English | MEDLINE | ID: mdl-35659371

ABSTRACT

The idea of personalized medicine came to fruition with sequencing the human genome; however, aside from a few cases, the genetic revolution has yet to materialize. Cardiovascular diseases are the leading cause of death globally, and hypertension is a common prelude to nearly all cardiovascular diseases. Thus, hypertension is an ideal candidate disease to apply tenants of personalized medicine to lessen cardiovascular disease. Herein is a survey that visually depicts the polymorphisms in the top eight antihypertensive targets. Although there are numerous genome-wide association studies regarding cardiovascular disease, few studies look at the effects of receptor polymorphisms on drug treatment. With 17,000+ polymorphisms in the combined target proteins examined, it is expected that some of the clinical variability in the treatment of hypertension is due to polymorphisms in the drug targets. Recent advances in techniques and technology, such as high throughput examination of single mutations, structure prediction, computational power for modeling, and CRISPR models of point mutations, allow for a relatively rapid and comprehensive examination of the effects of known and future polymorphisms on drug affinity and effects. As hypertension is easy to measure and has a plethora of clinically viable ligands, hypertension makes an excellent disease to study pharmacogenomics in the lab and the clinic. If the promises of personalized medicine are to materialize, a concerted effort to examine the effects polymorphisms have on drugs is required. A clinician with the knowledge of a patient's genotype can then prescribe drugs that are optimal for treating that specific patient.


Subject(s)
Cardiovascular Diseases , Hypertension , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Genome-Wide Association Study , Humans , Hypertension/drug therapy , Hypertension/genetics , Pharmacogenetics
7.
Int J Pharm ; 611: 121302, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34793935

ABSTRACT

The ß-blocker carvedilol prevents ultraviolet (UV)-induced skin cancer, but systemic drug administration may cause unwanted cadiovascular effects. To overcome this limitation, a topical delivery system based on transfersome (T-CAR) was characterized ex vivo and in vivo. T-CAR was visualized by Transmission Electron Microscopy as nanoparticles of spherical and unilamellar structure. T-CAR incorporated into carbopol gel and in suspension showed similar drug permeation and deposition profiles in Franz diffusion cells loaded with porcine ear skin. In mice exposed to a single dose UV, topical T-CAR gel (10 µM) significantly reduced UV-induced skin edema and cyclobutane pyrimidine dimer formation. In mice exposed to chronic UV radiation for 25 weeks, topical T-CAR gel (10 µM) significantly delayed the incidence of tumors, reduced tumor number and burden, and attenuated Ki-67 and COX-2 expression. The T-CAR gel was subsequently examined for skin deposition, systemic absorption and cardiovascular effects in mice. In mice treated with repeated doses of T-CAR gel (100 µM), the drug was undetectable in plasma, the heart rate was unaffected, but skin deposition was significantly higher than mice treated with oral carvedilol (32 mg/kg/day). These data indicate that the carbopol-based T-CAR gel holds great promise for skin cancer prevention with negligible systemic effects.


Subject(s)
Pharmaceutical Preparations , Skin Neoplasms , Absorption, Physiological , Animals , Carvedilol , Mice , Skin Neoplasms/prevention & control , Swine , Ultraviolet Rays
8.
Cancer Prev Res (Phila) ; 14(5): 527-540, 2021 05.
Article in English | MEDLINE | ID: mdl-33648941

ABSTRACT

Skin cancer is the most common malignancy worldwide and is rapidly rising in incidence, representing a significant public health challenge. The ß-blocker, carvedilol, has shown promising effects in preventing skin cancer. However, as a potent ß-blocker, repurposing carvedilol to an anticancer agent is limited by cardiovascular effects. Carvedilol is a racemic mixture consisting of equimolar S- and R-carvedilol, whereas the R-carvedilol enantiomer does not possess ß-blocking activity. Because previous studies suggest that carvedilol's cancer preventive activity is independent of ß-blockade, we examined the skin cancer preventive activity of R-carvedilol compared with S-carvedilol and the racemic carvedilol. R- and S-carvedilol were equally effective in preventing EGF-induced neoplastic transformation of the mouse epidermal JB6 Cl 41-5a (JB6 P+) cells and displayed similar attenuation of EGF-induced ELK-1 activity. R-carvedilol appeared slightly better than S-carvedilol against UV-induced intracellular oxidative stress and release of prostaglandin E2 from the JB6 P+ cells. In an acute UV-induced skin damage and inflammation mouse model using a single irradiation of 300 mJ/cm2 UV, topical treatment with R-carvedilol dose dependently attenuated skin edema and reduced epidermal thickening, Ki-67 staining, COX-2 protein, and IL6 and IL1ß mRNA levels similar to carvedilol. In a chronic UV (50-150 mJ/cm2) induced skin carcinogenesis model in mice with pretreatment of test agents, topical treatment with R-carvedilol, but not racemic carvedilol, significantly delayed and reduced skin squamous cell carcinoma development. Therefore, as an enantiomer present in an FDA-approved agent, R-carvedilol may be a better option for developing a safer and more effective preventive agent for skin carcinogenesis. PREVENTION RELEVANCE: In this study, we demonstrated the skin cancer preventive activity of R-carvedilol, the non-ß-blocking enantiomer present in the racemic ß-blocker, carvedilol. As R-carvedilol does not have ß-blocking activity, such a preventive treatment would not lead to common cardiovascular side effects of ß-blockers.


Subject(s)
Carcinogenesis/drug effects , Carvedilol/administration & dosage , Epidermis/drug effects , Neoplasms, Experimental/prevention & control , Skin Neoplasms/prevention & control , Animals , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Carvedilol/chemistry , Epidermal Cells , Epidermal Growth Factor/toxicity , Epidermis/pathology , Epidermis/radiation effects , Female , HEK293 Cells , Humans , Mice , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Stereoisomerism , Ultraviolet Rays/adverse effects
9.
Pharmaceutics ; 12(12)2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33260886

ABSTRACT

The ß-blocker carvedilol has been shown to prevent skin carcinogenesis in vitro and in vivo. Since systemic absorption of the ß-blocker may cause cardiovascular disturbance, we developed a carvedilol loaded transfersome for skin-targeted delivery. Transfersomes were prepared using phospholipids and surfactants at various ratios and characterized. One formulation (F18) selected for further analysis was composed of carvedilol, soy phosphatidylcholine, and Tween-80 at a ratio of 1:3:0.5, which had a particle size of 115.6 ± 8.7 nm, a zeta potential of 11.34 ± 0.67 mV, and an encapsulation efficiency of 93.7 ± 5.1%. F18 inhibited EGF-induced neoplastic transformation of mouse epidermal JB6 P+ cells at non-toxic concentrations, while only high concentrations induced cytotoxicity in JB6 P+ and human keratinocytes HaCaT. Compared to the free drug, F18 released through the dialysis membrane and permeated through the porcine ear skin at a slower rate, but similarly depositing the drug in the epidermis and dermis of the skin. Consistently, surface application of F18 on reconstructed full-thickness human skin showed slower drug permeation, while it suppressed ultraviolet-induced DNA damage, inflammatory gene expression, and apoptosis. These data indicate that transfersome is a promising topical delivery system of carvedilol for preventing ultraviolet-induced skin damage and carcinogenesis.

10.
PLoS Comput Biol ; 16(11): e1007719, 2020 11.
Article in English | MEDLINE | ID: mdl-33237899

ABSTRACT

Angiotensin II type 1 receptor (AT1R) blockers (ARBs) are among the most prescribed drugs. However, ARB effectiveness varies widely, which may be due to non-synonymous single nucleotide polymorphisms (nsSNPs) within the AT1R gene. The AT1R coding sequence contains over 100 nsSNPs; therefore, this study embarked on determining which nsSNPs may abrogate the binding of selective ARBs. The crystal structure of olmesartan-bound human AT1R (PDB:4ZUD) served as a template to create an inactive apo-AT1R via molecular dynamics simulation (n = 3). All simulations resulted in a water accessible ligand-binding pocket that lacked sodium ions. The model remained inactive displaying little movement in the receptor core; however, helix 8 showed considerable flexibility. A single frame representing the average stable AT1R was used as a template to dock Olmesartan via AutoDock 4.2, MOE, and AutoDock Vina to obtain predicted binding poses and mean Boltzmann weighted average affinity. The docking results did not match the known pose and affinity of Olmesartan. Thus, an optimization protocol was initiated using AutoDock 4.2 that provided more accurate poses and affinity for Olmesartan (n = 6). Atomic models of 103 of the known human AT1R polymorphisms were constructed using the molecular dynamics equilibrated apo-AT1R. Each of the eight ARBs was then docked, using ARB-optimized parameters, to each polymorphic AT1R (n = 6). Although each nsSNP has a negligible effect on the global AT1R structure, most nsSNPs drastically alter a sub-set of ARBs affinity to the AT1R. Alterations within N298 -L314 strongly effected predicted ARB affinity, which aligns with early mutagenesis studies. The current study demonstrates the potential of utilizing in silico approaches towards personalized ARB therapy. The results presented here will guide further biochemical studies and refinement of the model to increase the accuracy of the prediction of ARB resistance in order to increase overall ARB effectiveness.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Imidazoles/therapeutic use , Precision Medicine , Tetrazoles/therapeutic use , Angiotensin II Type 1 Receptor Blockers/chemistry , Humans , Imidazoles/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Polymorphism, Single Nucleotide , Receptor, Angiotensin, Type 1/genetics , Reproducibility of Results , Tetrazoles/chemistry
11.
Int J Mol Sci ; 21(3)2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31991834

ABSTRACT

The ß-blocker carvedilol prevents ultraviolet (UV)-induced skin cancer, but the mechanism is unknown. Since carvedilol possesses antioxidant activity, this study investigated whether carvedilol prevents oxidative photodamage of skin, a precursor event in skin carcinogenesis. The effects of carvedilol, metoprolol (a ß-blocker without antioxidant property), and 4-hydroxycarbazole (4-OHC, a carvedilol synthesis intermediate and a free radical scavenger) were compared on UV- or H2O2-induced cell death and reactive oxygen species (ROS) production in murine epidermal JB6 P+ cells. Although carvedilol attenuated cell death, metoprolol and 4-OHC failed to show protective effects. As expected, increased cellular ROS induced by H2O2 or UV was abolished by carvedilol and 4-OHC, but not by metoprolol. Consistently, carvedilol attenuated the formation of UV-induced cyclobutane pyrimidine dimers (CPDs) and release of prostaglandin E2 in JB6 P+ cells. Carvedilol's activity was further confirmed in full thickness 3D human reconstituted skin, where carvedilol attenuated UV-mediated epidermal thickening, the number of Ki-67 and p53 positive cells as well as CPD formation. Based on pathway-specific Polymerase Chain Reaction (PCR) Array analysis, carvedilol treatment in many cases normalized UV-induced expression changes in DNA repair genes. Thus, carvedilol's photoprotective activity is not attributed to ß-blockade or direct ROS-scavenging capacity, but likely via DNA repair regulation.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carvedilol/pharmacology , Epidermal Cells/drug effects , Epidermal Cells/radiation effects , Ultraviolet Rays/adverse effects , Animals , Cell Culture Techniques , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Cytokines/metabolism , DNA Damage/drug effects , Dinoprostone/metabolism , Epidermal Cells/metabolism , Humans , Hydrogen Peroxide , Inflammation Mediators , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
PLoS One ; 14(5): e0217038, 2019.
Article in English | MEDLINE | ID: mdl-31107911

ABSTRACT

Carvedilol is reported to prevent cancers in humans and animal models. However, a molecular mechanism has yet to be established, and the extent to which other ß-blockers are chemopreventive remains relatively unknown. A comparative pharmacological approach was utilized with the expectation that a mechanism of action could be devised. JB6 Cl 41-5a (JB6 P+) murine epidermal cells were used to elucidate the chemopreventative properties of ß-blockers, as JB6 P+ cells recapitulate in vivo tumor promotion and chemoprevention. The initial hypothesis was that ß-blockers that are GRK/ß-arrestin biased agonists, like carvedilol, are chemopreventive. Sixteen ß-blockers of different classes, isoproterenol, and HEAT HCl were individually co-administered with epidermal growth factor (EGF) to JB6 P+ cells to examine the chemopreventative properties of each ligand. Cytotoxicity was examined to ensure that the anti-transformation effects of each ligand were not due to cellular growth inhibition. Many of the examined ß-blockers suppressed EGF-induced JB6 P+ cell transformation in a non-cytotoxic and concentration-dependent manner. However, the IC50 values are high for the most potent inhibitors (243, 326, and 431 nM for carvedilol, labetalol, and alprenolol, respectively) and there is no correlation between pharmacological properties and inhibition of transformation. Therefore, the role of α1- and ß2-adrenergic receptors (AR) was examined by standard competition assays and shRNA targeting ß2-ARs, the only ß-AR expressed in JB6 P+ cells. The results reveal that pharmacological inhibition of α1- and ß2-ARs and genetic knockdown of ß2-ARs did not abrogate carvedilol-mediated inhibition of EGF-induced JB6 P+ cell transformation. Furthermore, topical administration of carvedilol protected mice from UV-induced skin damage, while genetic ablation of ß2-ARs increased carvedilol-mediated effects. Therefore, the prevailing hypothesis that the chemopreventive property of carvedilol is mediated through ß-ARs is not supported by this data.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carvedilol/pharmacology , Epidermal Growth Factor/metabolism , Receptors, Adrenergic/metabolism , Alprenolol/pharmacology , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Inhibitory Concentration 50 , Labetalol/pharmacology , Ligands , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/drug therapy , Ultraviolet Rays
13.
Mol Carcinog ; 57(8): 997-1007, 2018 08.
Article in English | MEDLINE | ID: mdl-29626349

ABSTRACT

Recent studies suggest that the ß-blocker drug carvedilol prevents skin carcinogenesis but the mechanism is unknown. Carvedilol is one of a few ß-blockers identified as biased agonist based on an ability to promote ß-arrestin-mediated processes such as ERK phosphorylation. To understand the role of phosphoproteomic signaling in carvedilol's anticancer activity, the mouse epidermal JB6 P+ cells treated with EGF, carvedilol, or their combination were analyzed using the Phospho Explorer Antibody Array containing 1318 site-specific and phospho-specific antibodies of over 30 signaling pathways. The array data indicated that both EGF and carvedilol increased phosphorylation of ERK's cytosolic target P70S6 K while its nuclear target ELK-1 were activated only by EGF; Furthermore, EGF-induced phosphorylation of ELK-1 and c-Jun was attenuated by carvedilol. Subcellular fractionation analysis indicated that ERK nuclear translocation induced by EGF was blocked by co-treatment with carvedilol. Western blot and luciferase reporter assays confirmed that the biased ß-blockers carvedilol and alprenolol blocked EGF-induced phosphorylation and activation of c-Jun/AP-1 and ELK-1. Consistently, both carvedilol and alprenolol strongly prevented EGF-induced neoplastic transformation of JB6 P+ cells. Remarkably, oral carvedilol treatment significantly inhibited the growth of A375 melanoma xenograft in SCID mice. As nuclear translocation of ERK is a key step in carcinogenesis, inhibition of this event is proposed as a novel anticancer mechanism for biased ß-blockers such as carvedilol.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Anticarcinogenic Agents/therapeutic use , Carcinogenesis/drug effects , Carvedilol/therapeutic use , Melanoma/prevention & control , Adrenergic beta-Antagonists/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carvedilol/pharmacology , Epidermal Growth Factor/metabolism , HEK293 Cells , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Proteome/metabolism , Proto-Oncogene Proteins c-jun/metabolism
14.
Cancer Prev Res (Phila) ; 10(10): 598-606, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28912118

ABSTRACT

In previous studies, the ß-blocker carvedilol inhibited EGF-induced epidermal cell transformation and chemical carcinogen-induced mouse skin hyperplasia. As exposure to ultraviolet (UV) radiation leads to skin cancer, the present study examined whether carvedilol can prevent UV-induced carcinogenesis. Carvedilol absorbs UV like a sunscreen; thus, to separate pharmacological from sunscreen effects, 4-hydroxycarbazole (4-OHC), which absorbs UV to the same degree as carvedilol, served as control. JB6 P+ cells, an established epidermal model for studying tumor promotion, were used for evaluating the effect of carvedilol on UV-induced neoplastic transformation. Both carvedilol and 4-OHC (1 µmol/L) blocked transformation induced by chronic UV (15 mJ/cm2) exposure for 8 weeks. However, EGF-mediated transformation was inhibited by only carvedilol but not by 4-OHC. Carvedilol (1 and 5 µmol/L), but not 4-OHC, attenuated UV-induced AP-1 and NF-κB luciferase reporter activity, suggesting a potential anti-inflammatory activity. In a single-dose UV (200 mJ/cm2)-induced skin inflammation mouse model, carvedilol (10 µmol/L), applied topically after UV exposure, reduced skin hyperplasia and the levels of cyclobutane pyrimidine dimers, IL1ß, IL6, and COX-2 in skin. In SKH-1 mice exposed to gradually increasing levels of UV (50-150 mJ/cm2) three times a week for 25 weeks, topical administration of carvedilol (10 µmol/L) after UV exposure increased tumor latency compared with control (week 18 vs. 15), decreased incidence and multiplicity of squamous cell carcinomas, while 4-OHC had no effect. These data suggest that carvedilol has a novel chemopreventive activity and topical carvedilol following UV exposure may be repurposed for preventing skin inflammation and cancer. Cancer Prev Res; 10(10); 598-606. ©2017 AACR.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carbazoles/pharmacology , Carcinogenesis/drug effects , Neoplasms, Radiation-Induced/prevention & control , Propanolamines/pharmacology , Skin Neoplasms/prevention & control , Ultraviolet Rays/adverse effects , Administration, Cutaneous , Animals , Anticarcinogenic Agents/therapeutic use , Carbazoles/therapeutic use , Carcinogenesis/radiation effects , Carvedilol , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Disease Models, Animal , Epidermal Cells , Epidermal Growth Factor/metabolism , Epidermis/drug effects , Epidermis/pathology , Epidermis/radiation effects , Female , HEK293 Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Mice , Mice, Hairless , NF-kappa B/metabolism , Propanolamines/therapeutic use , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Sunscreening Agents/pharmacology , Transcription Factor AP-1/metabolism
15.
Physiol Rep ; 3(12)2015 Dec.
Article in English | MEDLINE | ID: mdl-26702076

ABSTRACT

The effects of heat acclimation on water intake and urine output responses to thermal dehydration and other thirst stimuli were studied in male Sprague-Dawley rats. Rats were heat acclimated by continuous exposure to a 34°C environment for at least 6 weeks. Thermal dehydration-induced thirst was brought about by exposing the heat-acclimated rats and control rats housed at 24°C to a 37.5°C environment for 4 h without access to food or water. Heat acclimation reduced evaporative and urinary water losses and the increases in plasma sodium and osmolality during thermal dehydration, which led to a reduction in thermal dehydration-induced thirst. Heat acclimation reduced the rate of rehydration following thermal dehydration but did not alter the final rehydration level, indicating that heat acclimation does not alter the primary control of thermal dehydration-induced thirst. Heat acclimation did not alter water intake or urine output following administration of hypertonic saline, which selectively stimulates intracellular thirst, but led to greater water intake following administration of angiotensin II, which plays an important role in extracellular/volemic thirst, and following water deprivation, which activates both thirst pathways. Cardiovascular responses to angiotensin II were not altered by heat acclimation. Heat acclimation thus reduces water loss during heat exposure in rats, but does not have major effects on thermal dehydration-induced or extracellular thirst but does appear to alter volemic thirst.

17.
Cancer Prev Res (Phila) ; 8(1): 27-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25367979

ABSTRACT

The stress-related catecholamine hormones and the α- and ß-adrenergic receptors (α- and ß-AR) may affect carcinogenesis. The ß-AR GRK/ß-arrestin biased agonist carvedilol can induce ß-AR-mediated transactivation of the EGFR. The initial purpose of this study was to determine whether carvedilol, through activation of EGFR, can promote cancer. Carvedilol failed to promote anchorage-independent growth of JB6 P(+) cells, a skin cell model used to study tumor promotion. However, at nontoxic concentrations, carvedilol dose dependently inhibited EGF-induced malignant transformation of JB6 P(+) cells, suggesting that carvedilol has chemopreventive activity against skin cancer. Such effect was not observed for the ß-AR agonist isoproterenol and the ß-AR antagonist atenolol. Gene expression, receptor binding, and functional studies indicate that JB6 P(+) cells only express ß2-ARs. Carvedilol, but not atenolol, inhibited EGF-mediated activator protein-1 (AP-1) activation. A topical 7,12-dimethylbenz(α)anthracene (DMBA)-induced skin hyperplasia model in SENCAR mice was utilized to determine the in vivo cancer preventative activity of carvedilol. Both topical and oral carvedilol treatment inhibited DMBA-induced epidermal hyperplasia (P < 0.05) and reduced H-ras mutations; topical treatment being the most potent. However, in models of established cancer, carvedilol had modest to no inhibitory effect on tumor growth of human lung cancer A549 cells in vitro and in vivo. In conclusion, these results suggest that the cardiovascular drug carvedilol may be repurposed for skin cancer chemoprevention, but may not be an effective treatment of established tumors. More broadly, this study suggests that ß-ARs may serve as a novel target for cancer prevention.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Anticarcinogenic Agents/therapeutic use , Carbazoles/therapeutic use , Propanolamines/therapeutic use , Skin Neoplasms/prevention & control , 9,10-Dimethyl-1,2-benzanthracene/chemistry , Animals , Atenolol/therapeutic use , Carvedilol , Cell Adhesion , Cell Line , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Isoproterenol/therapeutic use , Mice , Mutation , Neoplasm Transplantation , Transcription Factor AP-1/metabolism
18.
PLoS One ; 8(8): e71980, 2013.
Article in English | MEDLINE | ID: mdl-23977191

ABSTRACT

Nebivolol, a third generation ß-adrenoceptor (ß-AR) antagonist (ß-blocker), causes vasodilation by inducing nitric oxide (NO) production. The mechanism via which nebivolol induces NO production remains unknown, resulting in the genesis of much of the controversy regarding the pharmacological action of nebivolol. Carvedilol is another ß-blocker that induces NO production. A prominent pharmacological mechanism of carvedilol is biased agonism that is independent of Gαs and involves G protein-coupled receptor kinase (GRK)/ß-arrestin signaling with downstream activation of the epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK). Due to the pharmacological similarities between nebivolol and carvedilol, we hypothesized that nebivolol is also a GRK/ß-arrestin biased agonist. We tested this hypothesis utilizing mouse embryonic fibroblasts (MEFs) that solely express ß2-ARs, and HL-1 cardiac myocytes that express ß1- and ß2-ARs and no detectable ß3-ARs. We confirmed previous reports that nebivolol does not significantly alter cAMP levels and thus is not a classical agonist. Moreover, in both cell types, nebivolol induced rapid internalization of ß-ARs indicating that nebivolol is also not a classical ß-blocker. Furthermore, nebivolol treatment resulted in a time-dependent phosphorylation of ERK that was indistinguishable from carvedilol and similar in duration, but not amplitude, to isoproterenol. Nebivolol-mediated phosphorylation of ERK was sensitive to propranolol (non-selective ß-AR-blocker), AG1478 (EGFR inhibitor), indicating that the signaling emanates from ß-ARs and involves the EGFR. Furthermore, in MEFs, nebivolol-mediated phosphorylation of ERK was sensitive to pharmacological inhibition of GRK2 as well as siRNA knockdown of ß-arrestin 1/2. Additionally, nebivolol induced redistribution of ß-arrestin 2 from a diffuse staining pattern into more intense punctate spots. We conclude that nebivolol is a ß2-AR, and likely ß1-AR, GRK/ß-arrestin biased agonist, which suggests that some of the unique clinically beneficial effects of nebivolol may be due to biased agonism at ß1- and/or ß2-ARs.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Arrestins/metabolism , Benzopyrans/pharmacology , Ethanolamines/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism , Animals , Arrestins/genetics , Carbazoles/pharmacology , Carvedilol , Cell Line , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , MAP Kinase Signaling System , Mice , Nebivolol , Nitric Oxide/metabolism , Phosphorylation , Propanolamines/pharmacology , Protein Processing, Post-Translational , Protein Transport , Receptors, Adrenergic, beta-2/metabolism , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
19.
Am J Physiol Renal Physiol ; 304(8): F1105-13, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23389452

ABSTRACT

Blockade of the angiotensin (ANG) II receptor type 1 (AT(1)R) with angiotensin receptor blockers (ARBs) is widely used in the treatment of hypertension. However, ARBs are variably effective in reducing blood pressure, likely due, in part, to polymorphisms in the ARB binding pocket of the AT(1)R. Therefore, we need a better understanding of variations/polymorphisms that alter binding of ARBs in heterogeneous patient populations. The opossum proximal tubule cell (OKP) line is commonly used in research to evaluate renal sodium handling and therefore blood pressure. Investigating this issue, we found natural sequence variations in the opossum AT(1)R paralleling those observed in the human AT(1)R. Therefore, we posited that these sequence variations may explain ARB resistance. We demonstrate that OKP cells express AT(1)R mRNA, bind (125)I-labeled ANG II, and exhibit ANG II-induced phosphorylation of Jak2. However, Jak2 phosphorylation is not inhibited by five different ARBs commonly used to treat hypertension. Additionally, nonradioactive ANG II competes (125)I-ANG II efficiently, whereas a 10-fold molar excess of olmesartan and the ANG II receptor type 2 blocker PD-123319 is unable to block (125)I-ANG II binding. In contrast, ANG II binding to OKP cells stably expressing rat AT(1A)Rs, which have a conserved AT(1)R-binding pocket with human AT(1)R, is efficiently inhibited by olmesartan. A novel observation was that resistance to ARB binding to opossum AT(1)Rs correlates with variations from the human receptor at positions 108, 163, 192, and 198 within the ARB-binding pocket. These observations highlight the potential utility of evaluating AT(1)R polymorphisms within the ARB-binding pocket in various hypertensive populations.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Imidazoles/pharmacology , Kidney Tubules, Proximal/drug effects , Opossums/genetics , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/genetics , Tetrazoles/pharmacology , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Binding Sites , Cell Line , Drug Resistance/genetics , Humans , Iodine Radioisotopes , Janus Kinase 2/metabolism , Kidney Tubules, Proximal/cytology , Phylogeny , Polymorphism, Genetic/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Messenger/genetics , Rats , Receptor, Angiotensin, Type 1/metabolism , Species Specificity , Vasoconstrictor Agents/metabolism , Vasoconstrictor Agents/pharmacology
20.
Cell Physiol Biochem ; 30(5): 1148-58, 2012.
Article in English | MEDLINE | ID: mdl-23171819

ABSTRACT

BACKGROUND: In the renal collecting duct, vasopressin regulates water permeability by a process that involves stimulation of adenylyl cyclase activity, cAMP production and subsequent translocation of water channel aquaporin-2 (AQP2) into the apical plasma membrane. We have previously shown that in cos 1 cells in vitro, both adenylyl cyclase activity and cAMP production can be regulated by VACM-1, a cul 5 gene that forms complexes involved in protein ubiquitination and subsequent degradation. METHODS: To extend these observations further, the effects of changes in hydration state on the expression of VACM-1 at the mRNA and the protein level were examined in rats deprived of water (WD) for 24 hrs. RESULTS: In the kidney of WD rats Western blot analyses of kidney tissue showed that the decrease in VACM-1 protein concentration was correlated with the increase in the AQP2 protein level. The immunostaining data suggested that VACM-1/cul5 may be decreased in renal collecting duct but increases in the vasculature of the inner medullary region in response to WD. To determine the possible consequences of the WD dependent decrease in VACM-1/cul5, we next examined the effects of VACM-1 expression on AQP2 protein in vitro. Immunocytochemistry and Western blot analyses data indicate that VACM-1/cul5 expression in MDCK line stably expressing AQP2 gene and in cos 1 cells co-transfected with the AQP2 and VACM-1/cul5 cDNAs decreased AQP2 protein concentration when compared to the vector transfected control groups. CONCLUSION: In summary, our data demonstrate that VACM-1 is involved in the regulation of AQP2 protein concentration and may play a role in regulating water balance.


Subject(s)
Aquaporin 2/analysis , Cullin Proteins/metabolism , Receptors, Vasopressin/metabolism , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cullin Proteins/genetics , Dogs , Kidney/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...