Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Physiol Rep ; 9(20): e15058, 2021 10.
Article in English | MEDLINE | ID: mdl-34676684

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) disrupts renal parenchyma through progressive expansion of fluid-filled cysts. The only approved pharmacotherapy for ADKPD involves the blockade of the vasopressin type 2 receptor (V2R). V2R is a GPCR expressed by a subset of renal tubular cells and whose activation stimulates cyclic AMP (cAMP) accumulation, which is a major driver of cyst growth. The ß3-adrenergic receptor (ß3-AR) is a GPCR expressed in most segments of the murine nephron, where it modulates cAMP production. Since sympathetic nerve activity, which leads to activation of the ß3-AR, is elevated in patients affected by ADPKD, we hypothesize that ß3-AR might constitute a novel therapeutic target. We find that administration of the selective ß3-AR antagonist SR59230A to an ADPKD mouse model (Pkd1fl/fl ;Pax8rtTA ;TetO-Cre) decreases cAMP levels, producing a significant reduction in kidney/body weight ratio and a partial improvement in kidney function. Furthermore, cystic mice show significantly higher ß3-AR levels than healthy controls, suggesting a correlation between receptor expression and disease development. Finally, ß3-AR is expressed in human renal tissue and localizes to cyst-lining epithelial cells in patients. Thus, ß3-AR is a potentially interesting target for the development of new treatments for ADPKD.


Subject(s)
Cyclic AMP/metabolism , Epithelial Cells/drug effects , Kidney/drug effects , Polycystic Kidney, Autosomal Dominant/drug therapy , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/chemistry , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Case-Control Studies , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/etiology , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology
2.
Eur J Pharmacol ; 905: 174180, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34015319

ABSTRACT

Systemic arterial hypertension (SAH) is a chronic disease of multifactorial origin and one of the main risk factors for major adverse cardiovascular events (MACE), which are the leading causes of morbidity and mortality worldwide. The pharmacological treatment of SAH involves five main classes of drugs, and Nebivolol (NEB) is one of those drugs, belonging to the class of third generation ß1-adrenoceptors selective blockers. NEB is composed of a racemic mixture of two enantiomers: d-nebivolol, which exerts antagonist effects on ß1-adrenoceptors, and l-nebivolol, a vascular ß3 receptor agonist. There are several studies that report different actions of NEB, not only for the treatment of SAH, but also as an antioxidant agent or even as a protector of renal damage. The aim of this systematic review was to investigate the available evidence regarding the effects of NEB on kidney diseases, evaluating its possible renoprotective action.


Subject(s)
Nebivolol/pharmacology , Nebivolol/therapeutic use , Protective Agents/pharmacology , Protective Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Adrenergic beta-3 Receptor Antagonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/therapeutic use , Animals , Humans , Hypertension/drug therapy , Menopause/drug effects , Receptors, Adrenergic, beta-3
3.
J Neurosci Res ; 99(2): 621-637, 2021 02.
Article in English | MEDLINE | ID: mdl-32954502

ABSTRACT

Oxidative stress has been implicated in a variety of neurodegenerative disorders, such as Alzheimer's and Parkinson's disease. Astrocytes play a significant role in maintaining survival of neurons by supplying antioxidants such as glutathione (GSH) to neurons. Recently, we found that noradrenaline increased the intracellular GSH concentration in astrocytes via ß3 -adrenoceptor stimulation. These observations suggest that noradrenaline protects neurons from oxidative stress-induced death by increasing the supply of GSH from astrocytes to neurons via the stimulation of ß3 -adrenoceptor in astrocytes. In the present study, we examined the protective effect of noradrenaline against H2 O2 -induced neurotoxicity using two different mixed cultures: the mixed culture of human astrocytoma U-251 MG cells and human neuroblastoma SH-SY5Y cells, and the mouse primary cerebrum mixed culture of neurons and astrocytes. H2 O2 -induced neuronal cell death was significantly attenuated by pretreatment with noradrenaline in both mixed cultures but not in single culture of SH-SY5Y cells or in mouse cerebrum neuron-rich culture. The neuroprotective effect of noradrenaline was inhibited by SR59230A, a selective ß3 -adrenoceptor antagonist, and CL316243, a selective ß3 -adrenoceptor agonist, mimicked the neuroprotective effect of noradrenaline. DL-buthionine-[S,R]-sulfoximine, a GSH synthesis inhibitor, negated the neuroprotective effect of noradrenaline in both mixed cultures. MK571, which inhibits the export of GSH from astrocytes mediated by multidrug resistance-associated protein 1, also prevented the neuroprotective effect of noradrenaline. These results suggest that noradrenaline protects neurons against H2 O2 -induced death by increasing the supply of GSH from astrocytes via ß3 -adrenoceptor stimulation.


Subject(s)
Astrocytes/drug effects , Glutathione/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Astrocytes/metabolism , Astrocytoma , Brain/cytology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Coculture Techniques , Dioxoles/pharmacology , Humans , Hydrogen Peroxide/toxicity , Mice , Mice, Inbred C57BL , Neuroblastoma , Oxidative Stress , Propanolamines/pharmacology , Propionates/pharmacology , Quinolines/pharmacology
4.
Cells ; 9(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33297453

ABSTRACT

ß3-adrenoreceptor (ß3-AR), a G-protein coupled receptor, has peculiar regulatory properties in response to oxygen and widespread localization. ß3-AR is expressed in the most frequent neoplasms, also occurring in pregnant women, and its blockade reduces tumor growth, indicating ß3-AR-blockers as a promising alternative to antineoplastic drugs during pregnancy. However, ß3-AR involvement in prenatal morphogenesis and the consequences of its blockade for the fetus remain unknown. In this study, after the demonstrated expression of ß3-AR in endothelial and smooth muscle cells of ductus arteriosus (DA), C57BL/6 pregnant mice were acutely treated at 18.5 of gestational day (GD) with indomethacin or with the selective ß3-AR antagonist SR59230A, or chronically exposed to SR59230A from 15.5 to 18.5 GD. Six hours after the last treatment, fetuses were collected. Furthermore, newborn mice were treated straight after birth with BRL37344, a ß3-AR agonist, and sacrificed after 7 h. SR59230A, at the doses demonstrated effective in reducing cancer progression (10 and 20 mg/kg) in acute and chronic mode, did not induce fetal DA constriction and did not impair the DA ability to close after birth, whereas at the highest dose (40 mg/kg), it was shown to cause DA constriction and preterm-delivery. BRL37344 administered immediately after birth did not alter the physiological DA closure.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Ductus Arteriosus, Patent/metabolism , Ductus Arteriosus/metabolism , Receptors, Adrenergic, beta-3/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Disease Progression , Ductus Arteriosus/drug effects , Ductus Arteriosus, Patent/drug therapy , Ethanolamines/pharmacology , Female , Indomethacin/pharmacology , Male , Maternal Exposure , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy, Animal , Propanolamines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Time Factors
5.
Sci Rep ; 10(1): 19488, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33173084

ABSTRACT

The naked mole-rat (NMR) is a heterothermic mammal that forms eusocial colonies consisting of one reproductive female (queen), several reproductive males, and subordinates. Despite their heterothermy, NMRs possess brown adipose tissue (BAT), which generally induces thermogenesis in cold and some non-cold environments. Previous studies suggest that NMR-BAT induces thermogenesis by cold exposure. However, detailed NMR-BAT characteristics and whether NMR-BAT thermogenesis occurs in non-cold environments are unknown. Here, we show beta-3 adrenergic receptor (ADRB3)-dependent thermogenic potential of NMR-BAT, which contributes to thermogenesis in the isolated queen in non-cold environments (30 °C). NMR-BAT expressed several brown adipocyte marker genes and showed noradrenaline-dependent thermogenic activity in vitro and in vivo. Although our ADRB3 inhibition experiments revealed that NMR-BAT thermogenesis slightly delays the decrease in body temperature in a cold environment (20 °C), it was insufficient to prevent the decrease in the body temperatures. Even at 30 °C, NMRs are known to prevent the decrease of and maintain their body temperature by heat-sharing behaviors within the colony. However, isolated NMRs maintained their body temperature at the same level as when they are in the colony. Interestingly, we found that queens, but not subordinates, induce BAT thermogenesis in this condition. Our research provides novel insights into NMR thermoregulation.


Subject(s)
Adipose Tissue, Brown/metabolism , Body Temperature Regulation/physiology , Body Temperature/physiology , Thermogenesis/physiology , Adipose Tissue, Brown/diagnostic imaging , Adipose Tissue, Brown/drug effects , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Body Temperature/drug effects , Body Temperature Regulation/genetics , Cold Temperature , Female , Gene Expression Regulation/drug effects , Male , Mole Rats , Norepinephrine/pharmacology , Oxygen Consumption/drug effects , Positron Emission Tomography Computed Tomography , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Thermogenesis/genetics
6.
Front Immunol ; 11: 2098, 2020.
Article in English | MEDLINE | ID: mdl-32983164

ABSTRACT

Understanding the mechanisms of immune tolerance is currently one of the most important challenges of scientific research. Pregnancy affects the immune system balance, leading the host to tolerate embryo alloantigens. Previous reports demonstrated that ß-adrenergic receptor (ß-AR) signaling promotes immune tolerance by modulation of NK and Treg, mainly through the activation of ß2-ARs, but recently we have demonstrated that also ß3-ARs induce an immune-tolerant phenotype in mice bearing melanoma. In this report, we demonstrate that ß3-ARs support host immune tolerance in the maternal microenvironment by modulating the same immune cells populations as recently demonstrated in cancer. Considering that ß3-ARs are modulated by oxygen levels, we hypothesize that hypoxia, through the upregulation of ß3-AR, promotes the biological shift toward a tolerant immunophenotype and that this is the same trick that embryo and cancer use to create an aura of immune-tolerance in a competent immune environment. This study confirms the analogies between fetal development and tumor progression and suggests that the expression of ß3-ARs represents one of the strategies to induce fetal and tumor immune tolerance.


Subject(s)
Cell Hypoxia/physiology , Immune Tolerance/physiology , Models, Immunological , Placenta/metabolism , Pregnancy, Animal/immunology , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Decidua/immunology , Female , Immunocompetence , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Placenta/immunology , Pregnancy , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/biosynthesis , Receptors, Adrenergic, beta-3/genetics , T-Lymphocytes, Regulatory/immunology , Up-Regulation
7.
Int J Mol Sci ; 21(12)2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32545695

ABSTRACT

ß-adrenergic signaling is known to be involved in cancer progression; in particular, beta3-adrenoreceptor (ß3-AR) is associated with different tumor conditions. Currently, there are few data concerning ß3-AR in myeloid malignancies. Here, we evaluated ß3-AR in myeloid leukemia cell lines and the effect of ß3-AR antagonist SR59230A. In addition, we investigated the potential role of ß3-AR blockade in doxorubicin resistance. Using flow cytometry, we assessed cell death in different in vitro myeloid leukemia cell lines (K562, KCL22, HEL, HL60) treated with SR59230A in hypoxia and normoxia; furthermore, we analyzed ß3-AR expression. We used healthy bone marrow cells (BMCs), peripheral blood mononuclear cells (PBMCs) and cord blood as control samples. Finally, we evaluated the effect of SR59230A plus doxorubicin on K562 and K562/DOX cell lines; K562/DOX cells are resistant to doxorubicin and show P-glycoprotein (P-gp) overexpression. We found that SR59230A increased cancer cell lines apoptosis especially in hypoxia, resulting in selective activity for cancer cells; moreover, ß3-AR expression was higher in malignancies, particularly under hypoxic condition. Finally, we observed that SR59230A plus doxorubicin increased doxorubicin resistance reversion mainly in hypoxia, probably acting on P-gp. Together, these data point to ß3-AR as a new target and ß3-AR blockade as a potential approach in myeloid leukemias.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid/metabolism , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , K562 Cells , Leukemia, Myeloid/drug therapy , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism
8.
J Pharmacol Exp Ther ; 374(1): 84-92, 2020 07.
Article in English | MEDLINE | ID: mdl-32332112

ABSTRACT

Urinary incontinence is defined as an involuntary leakage of urine and is categorized into three types: stress urinary incontinence (SUI), urge urinary incontinence (UUI), and mixed urinary incontinence, which includes symptoms of SUI and UUI. As the underlying mechanisms of SUI and UUI are different, no drug is approved to treat all three types of urinary incontinence. TAS-303 is a selective norepinephrine reuptake inhibitor and has therapeutic potential for patients with SUI. In this report, we describe newly discovered pharmacological properties of TAS-303 and its effects on bladder function. Radioligand binding studies showed that TAS-303 inhibits M3 muscarinic receptor binding, with a Ki value of 547 nM. TAS-303 at 1, 3, and 10 mg/kg dose-dependently prolonged the intercontraction interval of carbachol-induced detrusor overactivity in rats, exhibiting a maximal effect that was comparable to tolterodine. These effects may result from coordinated regulation of bladder afferent activity via M3 muscarinic inhibition and ß3 adrenoreceptor activation by norepinephrine elevation due to norepinephrine transporter inhibition. Moreover, TAS-303 at the effective dose for bladder function did not induce dry mouth or constipation in rats, showing that this compound may have a lower risk of antimuscarinic side effects. Thus, TAS-303 is expected to be a new profile agent with therapeutic potential for all types of urinary incontinence. SIGNIFICANCE STATEMENT: Urinary incontinence is categorized into stress, urge, and mixed urinary incontinence, but because the underlying mechanisms of each differ, no drugs are available that treat all three. TAS-303 has therapeutic potential for stress urinary incontinence. This study describes newly discovered pharmacological properties of TAS-303, which ameliorated bladder afferent activity partly via M3 muscarinic inhibition, indicating improvement in urge urinary incontinence, and highlights the potential of TAS-303 as a new therapeutic agent for all types of urinary incontinence.


Subject(s)
Carbachol/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Urinary Bladder, Overactive/drug therapy , Urinary Bladder, Overactive/physiopathology , Urinary Bladder/drug effects , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Atropine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Muscle Contraction/drug effects , Rats , Receptors, Muscarinic/metabolism , Urinary Bladder/physiopathology , Urinary Bladder, Overactive/chemically induced , Urinary Bladder, Overactive/metabolism
9.
Eur J Pharmacol ; 878: 173096, 2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32259514

ABSTRACT

Urgency is regarded as a core symptom of overactive bladder (OAB) and may correspond to detrusor overactivity (DO). One of the causes of OAB in men is bladder outlet obstruction (BOO) associated with benign prostatic hyperplasia (BPH). Vibegron is a novel selective ß3-adrenoceptor agonist recently approved for the treatment of OAB. However, in OAB patients with BPH (BPH/OAB), the effects of vibegron on storage functions, especially DO and voiding functions have not been fully investigated. In this study, we evaluated the effects of a single administration of vibegron on storage function (particularly focusing on non-voiding contractions [NVC] considered a surrogate marker for DO) and voiding functions, using a rat model of partial BOO as a model for BPH/OAB. Furthermore, the utility of vibegron in combination with imidafenacin (an antimuscarinic) or silodosin (an α1A-adrenoceptor blocker) was evaluated. Six weeks after establishment of BOO, the frequency and amplitude of NVC, evaluated by cystometrography, increased. Vibegron inhibited the frequency of NVC without affecting voiding function (micturition pressure, residual volume, and voiding efficiency). Imidafenacin and silodosin also inhibited the frequency of NVC; however, the inhibitory effects of vibegron were stronger than those of imidafenacin or silodosin. The combination of vibegron with imidafenacin or silodosin additively inhibited the frequency of NVC without worsening the voiding function. These results suggest the possibility that vibegron is effective as a single agent for the amelioration of storage symptoms in BPH/OAB patients and is useful in combination with either antimuscarinics or α1-adrenoceptor blockers.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Muscarinic Antagonists/pharmacology , Prostatic Hyperplasia/drug therapy , Urinary Bladder Neck Obstruction/drug therapy , Urinary Bladder, Overactive/drug therapy , Animals , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Male , Pyrimidinones , Pyrrolidines , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/metabolism , Urinary Bladder/drug effects , Urinary Bladder Neck Obstruction/complications , Urination/physiology
10.
Int J Mol Sci ; 21(4)2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32093135

ABSTRACT

Although there is an increasing evidence that cancer stem cell (CSC) niches in the tumor microenvironment (TME) plays a crucial role in sustaining solid tumors progression, several molecular players involved in this regulation still remain unknown. The role of ß-adrenergic signaling in enhancing tumor growth through ß2-adrenoreceptors (ß2-ARs) has been confirmed in different cancer models, but the role played by the ß3-adrenergic receptor (ß3-AR) has recently emerged. Previous studies showed that ß3-AR promotes cancer growth through the activation of different stromal cells in the TME, and leads to melanoma malignancy progression through inflammation, angiogenesis, and immunotolerance. Here we show that in B16 melanoma-bearing mice, the pharmacological ß3-AR blockade is able to reduce the expression of CSC markers, and to induce a differentiated phenotype of hematopoietic subpopulations in TME. In particular, cytofluorimetric analysis (FACS) of the tumor mass shows that ß3-AR antagonist SR59230A promotes hematopoietic differentiation as indicated by increased ratios of lymphoid/hematopoietic stem cells (HSCs) and of myeloid progenitor cells/HSCs, and increases the number of Ter119 and natural killer (NK) precursor cells, and of granulocyte precursors, indicating active hematopoiesis within the tumor tissue. Moreover, pharmacological antagonism of ß3-AR induces mesenchymal stem cell (MSC) differentiation into adipocytes subtracting a potential renewal of the stem compartment by these cells. Here we demonstrate that ß3-AR blockade in the TME by inducing the differentiation of different stromal cells at the expense of stemness traits could possibly have a favorable effect on the control of melanoma progression.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Melanoma, Experimental/metabolism , Neoplasm Proteins , Neoplastic Stem Cells/metabolism , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Male , Melanoma, Experimental/pathology , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism
11.
Oncogene ; 39(2): 368-384, 2020 01.
Article in English | MEDLINE | ID: mdl-31477835

ABSTRACT

Neuroblastoma (NB) is the most frequently observed among extracranial pediatric solid tumors. It displays an extreme clinical heterogeneity, in particular for the presentation at diagnosis and response to treatment, often depending on cancer cell differentiation/stemness. The frequent presence of elevated hematic and urinary levels of catecholamines in patients affected by NB suggests that the dissection of adrenergic system is crucial for a better understanding of this cancer. ß3-adrenoreceptor (ß3-AR) is the last identified member of adrenergic receptors, involved in different tumor conditions, such as melanoma. Multiple studies have shown that the dysregulation of the bioactive lipid sphingosine 1-phosphate (S1P) metabolism and signaling is involved in many pathological diseases including cancer. However, whether S1P is crucial for NB progression and aggressiveness is still under investigation. Here we provide experimental evidence that ß3-AR is expressed in NB, both human specimens and cell lines, where it is critically involved in the activation of proliferation and the regulation between stemness/differentiation, via its functional cross-talk with sphingosine kinase 2 (SK2)/S1P receptor 2 (S1P2) axis. The specific antagonism of ß3-AR by SR59230A inhibits NB growth and tumor progression, by switching from stemness to cell differentiation both in vivo and in vitro through the specific blockade of SK2/S1P2 signaling.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Neuroblastoma/drug therapy , Receptors, Adrenergic, beta-3/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , Sphingosine-1-Phosphate Receptors/genetics , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lysophospholipids/metabolism , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Neurons/drug effects , Propanolamines/pharmacology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tumor Hypoxia/drug effects
12.
Am J Physiol Endocrinol Metab ; 317(6): E1055-E1062, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31526290

ABSTRACT

Zinc-α2-glycoprotein (AZGP1) is a newly identified adipokine that is associated with lipid metabolism and vascular fibrosis. Although adipokines contribute to lipid dysfunction and its related diseases, including stroke and coronary heart disease (CHD), the role of AZGP1 remains unclear. In this study, the role of AZGP1 in atherosclerosis and CHD was investigated. Serum AZGP1 levels from control (n = 84) and CHD (n = 91) patients were examined by ELISA and its relationship with various clinical parameters was analyzed. Immunohistochemistry and immunofluorescence were used to detect the expression of AZGP1 and its receptor in coronary atherosclerotic arteries. THP-1 and human embryonic kidney 293 cells were used to verify its anti-inflammatory role in atherosclerosis. Serum AZGP1 levels in CHD patients were lower than controls (P < 0.01) and independently associated with CHD prevalence (P = 0.021). AZGP1 levels also inversely correlated with the Gensini score. Immunohistochemistry and immunofluorescence showed that AZGP1 and its receptor ß3-adrenoceptor (ß3-AR) colocalized in lipid-rich areas of atherosclerotic plaques, particularly around macrophages. In vitro, AZGP1 had no effect on foam cell formation but showed anti-inflammatory effects through its regulation of JNK/AP-1 signaling. In summary, AZGP1 is an anti-inflammatory agent that can be targeted for CHD treatment.


Subject(s)
Carrier Proteins/metabolism , Coronary Artery Disease/metabolism , Coronary Disease/metabolism , Glycoproteins/metabolism , Inflammation/metabolism , Plaque, Atherosclerotic/metabolism , Adipokines , Adrenergic beta-3 Receptor Antagonists/pharmacology , Case-Control Studies , Coronary Artery Disease/pathology , Cross-Sectional Studies , Cytokines/drug effects , Cytokines/metabolism , Female , Foam Cells/metabolism , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophages/drug effects , Macrophages/metabolism , Male , Middle Aged , Plaque, Atherosclerotic/pathology , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , THP-1 Cells , Transcription Factor AP-1/metabolism
13.
J Vasc Res ; 56(6): 320-332, 2019.
Article in English | MEDLINE | ID: mdl-31550717

ABSTRACT

BACKGROUND: Perivascular adipose tissue (PVAT) reduces vascular tone in isolated arteries in vitro, however there are no studies of PVAT effects on vascular tone in vivo. In vitro adipocyte ß3-adrenoceptors play a role in PVAT function via secretion of the vasodilator adiponectin. OBJECTIVE: We have investigated the effects of PVAT on vessel diameter in vivo, and the contributions of ß3-adrenoceptors and adiponectin. METHOD: In anaesthetised rats, sections of the intact mesenteric bed were visualised and the diameter of arteries was recorded. Arteries were stimulated with electrical field stimulation (EFS), noradrenaline (NA), arginine-vasopressin (AVP), and acetylcholine (Ach). RESULTS: We report that in vivo, stimulation of PVAT with EFS, NA, and AVP evokes a local anti-constrictive effect on the artery, whilst PVAT exerts a pro-contractile effect on arteries subjected to Ach. The anti-constrictive effect of PVAT stimulated with EFS and NA was significantly reduced using ß3-adrenoceptor inhibition, and activation of ß3-adrenoceptors potentiated the anti-constrictive effect of vessels stimulated with EFS, NA, and AVP. The ß3-adrenoceptor agonist had no effect on mesenteric arteries with PVAT removed. A blocking peptide for adiponectin receptor 1 polyclonal antibody reduced the PVAT anti-constrictive effect in arteries stimulated with EFS and NA, indicating that adiponectin may be the anti-constrictive factor released upon ß3-adrenoceptor activation. CONCLUSIONS: These results clearly demonstrate that PVAT plays a paracrine role in regulating local vascular tone in vivo, and therefore may contribute to the modulation of blood pressure. This effect is mediated via adipocyte ß3-adrenoceptors, which may trigger release of the vasodilator adiponectin.


Subject(s)
Adiponectin/metabolism , Adipose Tissue/metabolism , Mesenteric Arteries/metabolism , Paracrine Communication , Receptors, Adrenergic, beta-3/metabolism , Vasoconstriction , Vasodilation , Adipose Tissue/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Electric Stimulation , Male , Mesenteric Arteries/drug effects , Paracrine Communication/drug effects , Rats, Wistar , Receptors, Adrenergic, beta-3/drug effects , Signal Transduction , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
Eur J Pharmacol ; 858: 172468, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31226249

ABSTRACT

As heart failure (HF) is a growing public health problem worldwide, rapid therapeutic development is required to improve HF management. Decreased myocardial contractility in HF is associated with the persistent sympathetic activation of ß1/ß2-adrenoceptors (ß1/ß2-ARs). Although it is initially activated to compensate for a decline in myocardial contractility, it plays a pivotal role in organ damage and functional deterioration over time, resulting in the desensitization of receptors involved. The third ß-AR subtype, ß3-AR, is resistant to desensitization, and as a result, the expression of this subtype is enhanced in human failing myocardium. In addition, this upregulation and the stimulation of this subtype have been demonstrated to mediate cardioprotective effects such as antihypertrophic, antioxidant and antifibrotic effects via various signaling pathways in different cell types. However, the role of this attractive therapeutic intervention in heart diseases must be clarified through clinical trials.


Subject(s)
Heart Diseases/drug therapy , Molecular Targeted Therapy/methods , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Antagonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/therapeutic use , Animals , Gene Expression Regulation/drug effects , Heart Diseases/metabolism , Heart Failure/drug therapy , Heart Failure/metabolism , Humans
15.
Biochem Biophys Res Commun ; 514(1): 295-300, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31030945

ABSTRACT

Neuroblastoma (NB), the most common extracranial solid tumor in childhood, always leads to an unfavorable prognosis. ß3-adrenergic receptor (ß3-AR) signaling plays an important role in lipid metabolism. Although previous studies have focused mainly on the role of ß2-AR in tumor cells; there are few studies about the cancer-related function of ß3-AR. Herein, we showed that ß3-AR expression was significantly increased in clinical NB tissue compared with that in the less malignant ganglioneuroma (GN) and ganglioneuroblastoma (GNB) tissues. Further cellular assays demonstrated that treatment of NB cells with SR59230A (a specific ß3-AR antagonist) suppressed NB cells growth and colony formation, and siRNA knockdown of ß3-AR expression also inhibited NB cell proliferation. The mechanistic study revealed that ß3-AR knockdown and SR59230A inhibited the phosphorylation and thereby the activation of the mTOR/p70S6K pathway. Activation of the mTOR pathway with the activator MHY1485 reversed the inhibitory effect of SR59230A on NB cell growth. Above all, our study clarifies a novel regulatory role of ß3-AR in NB cell growth and provides a potent therapeutic strategy for this disease by specific targeting of the ß3-AR pathway.


Subject(s)
Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adrenergic beta-3 Receptor Antagonists/pharmacology , Cell Proliferation/drug effects , Epinephrine/pharmacology , Gene Knockdown Techniques , Humans , Molecular Targeted Therapy , Morpholines/pharmacology , Neuroblastoma/pathology , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/genetics , Signal Transduction/drug effects , Triazines/pharmacology , Tumor Cells, Cultured
16.
Cells ; 8(4)2019 04 16.
Article in English | MEDLINE | ID: mdl-30995798

ABSTRACT

The beta-3 adrenergic receptor (ß3-AR) is by far the least studied isotype of the beta-adrenergic sub-family. Despite its study being long hampered by the lack of suitable animal and cellular models and inter-species differences, a substantial body of literature on the subject has built up in the last three decades and the physiology of ß3-AR is unraveling quickly. As will become evident in this work, ß3-AR is emerging as an appealing target for novel pharmacological approaches in several clinical areas involving metabolic, cardiovascular, urinary, and ocular disease. In this review, we will discuss the most recent advances regarding ß3-AR signaling and function and summarize how these findings translate, or may do so, into current clinical practice highlighting ß3-AR's great potential as a novel therapeutic target in a wide range of human conditions.


Subject(s)
Receptors, Adrenergic, beta-3/metabolism , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Epinephrine , Humans , Norepinephrine , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects
17.
Cardiovasc Drugs Ther ; 33(2): 163-177, 2019 04.
Article in English | MEDLINE | ID: mdl-30729348

ABSTRACT

The ß3-AR (beta3-adrenergic receptor) is resistant to short-term agonist-promoted desensitization and delivers a constant intracellular signal, making this receptor a potential target in acute myocardial infarction (AMI). AIM: To investigate whether selective modulation of ß3-AR prior to or during ischemia and/or reperfusion may be cardioprotective. METHODS: Isolated perfused rat hearts were exposed to 35-min regional ischemia (RI) and 60-min reperfusion. The ß3-AR agonist (BRL37344, 1 µM) or antagonist (SR59230A, 0.1 µM) was applied: (i) before RI (PreT) or (ii) last 10 min of RI (PerT) or (iii) onset of reperfusion (PostT) or (iv) during both PerT+PostT. Nitric oxide (NO) involvement was assessed, using the NOS inhibitor, L-NAME (50 µM). Endpoints were functional recovery, infarct size (IS), cGMP levels, and Western blot analysis of eNOS, ERKp44/p42, PKB/Akt, and glycogen synthase kinase-3ß (GSK-3ß). RESULTS: Selective treatment with BRL significantly reduced IS. L-NAME abolished BRL-mediated cardioprotection. BRL (PreT) and BRL (PerT) significantly increased cGMP levels (which were reduced by L-NAME) and PKB/Akt phosphorylation. BRL (PostT) produced significantly increased cGMP levels, PKB/Akt, and ERKp44/p42 phosphorylation. BRL (PerT+PostT) caused significant eNOS, PKB/Akt, ERKp44/p42, and GSK-3ß phosphorylation. CONCLUSION: ß3-AR activation by BRL37344 induced significant cardioprotection regardless of the experimental protocol. However, the pattern of intracellular signaling with each BRL treatment differed to some degree and suggests the involvement of cGMP, eNOS, ERK, GSK-3ß, and particularly PKB/Akt activation. The data also suggest that clinical application of ß3-AR stimulation should preferably be incorporated during late ischemia or/and early reperfusion.


Subject(s)
Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Receptors, Adrenergic, beta-3/drug effects , Animals , Cyclic GMP/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hemodynamics/drug effects , Isolated Heart Preparation , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Rats, Wistar , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Time Factors
18.
Oxid Med Cell Longev ; 2019: 6346529, 2019.
Article in English | MEDLINE | ID: mdl-31934266

ABSTRACT

The role of ROS and RNS is a long-standing debate in cancer. Increasing the concentration of ROS reaching the toxic threshold can be an effective strategy for the reduction of tumor cell viability. On the other hand, cancer cells, by maintaining intracellular ROS concentration at an intermediate level called "mild oxidative stress," promote the activation of signaling that favors tumor progression by increasing cell viability and dangerous tumor phenotype. Many chemotherapeutic treatments induce cell death by rising intracellular ROS concentration. The persistent drug stimulation leads tumor cells to simulate a process called hormesis by which cancer cells exhibit a biphasic response to exposure to drugs used. After a first strong response to a low dose of chemotherapeutic agent, cancer cells start to decrease the response even if high doses of drugs were used. In this framework, ß3-adrenoreceptors (ß3-ARs) fit with an emerging antioxidant role in cancer. ß3-ARs are involved in tumor proliferation, angiogenesis, metastasis, and immune tolerance. Its inhibition, by the selective ß3-ARs antagonist (SR59230A), leads cancer cells to increase ROS concentration thus inducing cell death and to decrease NO levels thus inhibiting angiogenesis. In this review, we report an overview on reactive oxygen biology in cancer cells focusing on ß3-ARs as new players in the antioxidant pathway.


Subject(s)
Antioxidants/metabolism , Neoplasms/metabolism , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic , Oxidative Stress , Propanolamines/pharmacology , Reactive Oxygen Species/metabolism
19.
Biol Psychiatry ; 85(3): 226-236, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30336931

ABSTRACT

BACKGROUND: Homeostatic plasticity in mesolimbic dopamine (DA) neurons plays an essential role in mediating resilience to social stress. Recent evidence implicates an association between stress resilience and projections from the locus coeruleus (LC) to the ventral tegmental area (VTA) (LC→VTA) DA system. However, the precise circuitry and molecular mechanisms of the homeostatic plasticity in mesolimbic DA neurons mediated by the LC→VTA circuitry, and its role in conferring resilience to social defeat stress, have not been described. METHODS: In a well-established chronic social defeat stress model of depression, using projection-specific electrophysiological recordings and optogenetic, pharmacological, and molecular profiling techniques, we investigated the functional role and molecular basis of an LC→VTA circuit in conferring resilience to social defeat stress. RESULTS: We found that LC neurons projecting to the VTA exhibit enhanced firing activity in resilient, but not susceptible, mice. Optogenetically mimicking this firing adaptation in susceptible mice reverses their depression-related behaviors, and induces reversal of cellular hyperactivity and homeostatic plasticity in VTA DA neurons projecting to the nucleus accumbens. Circuit-specific molecular profiling studies reveal that α1- and ß3-adrenergic receptors are highly expressed in VTA→nucleus accumbens DA neurons. Pharmacologically activating these receptors induces similar proresilient effects at the ion channel and cellular and behavioral levels, whereas antagonizing these receptors blocks the proresilient effect of optogenetic activation of LC→VTA circuit neurons in susceptible mice. CONCLUSIONS: These findings reveal a key role of the LC→VTA circuit in mediating homeostatic plasticity in stress resilience and reveal α1- and ß3-adrenergic receptors as new molecular targets for therapeutically promoting resilience.


Subject(s)
Locus Coeruleus/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta-3/physiology , Resilience, Psychological , Ventral Tegmental Area/physiology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Behavior, Animal/physiology , Dopaminergic Neurons/physiology , Homeostasis/physiology , Locus Coeruleus/drug effects , Male , Mice , Neural Pathways/physiology , Neuronal Plasticity/physiology , Resilience, Psychological/drug effects , Stress, Psychological/physiopathology , Ventral Tegmental Area/drug effects
20.
Oxid Med Cell Longev ; 2018: 6816508, 2018.
Article in English | MEDLINE | ID: mdl-30538804

ABSTRACT

The early phases of embryonic development and cancer share similar strategies to improve their survival in an inhospitable environment: both proliferate in a hypoxic and catecholamine-rich context, increasing aerobic glycolysis. Recent studies show that ß3-adrenergic receptor (ß3-AR) is involved in tumor progression, playing an important role in metastasis. Among ß-adrenergic receptors, ß3-AR is the last identified member of this family, and it is involved in cancer cell survival and induction of stromal reactivity in the tumor microenvironment. ß3-AR is well known as a strong activator of uncoupling protein 1 (UCP1) in brown fat tissue. Interestingly, ß3-AR is strongly expressed in early embryo development and in many cancer tissues. Induction of uncoupling protein 2 (UCP2) has been related to cancer metabolic switch, leading to accelerated glycolysis and reduced mitochondrial activity. In this study, for the first time, we demonstrate that ß3-AR is able to promote this metabolic shift in both cancer and embryonic stem cells, inducing specific glycolytic cytoplasmic enzymes and a sort of mitochondrial dormancy through the induction of UCP2. The ß3-AR/UCP2 axis induces a strong reduction of mitochondrial activity by reducing ATP synthesis and mitochondrial reactive oxygen species (mtROS) content. These effects are reverted by SR59230A, the specific ß3-AR antagonist, causing an increase in mtROS. The increased level of mtROS is neutralized by a strong antioxidant activity in embryonic stem cells, but not in cancer stem cells, where it causes a dramatic reduction in tumor cell viability. These results lead to the possibility of a selective antitumor therapeutic use of SR59230A. Notably, we demonstrate the presence of ß3-AR within the mitochondrial membrane in both cell lines, leading to the control of mitochondrial dormancy.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Embryonic Stem Cells/metabolism , Melanoma/metabolism , Mitochondria/metabolism , Propanolamines/pharmacology , Animals , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/pathology , Humans , Melanoma/pathology , Mice , Mitochondria/drug effects , Receptors, Adrenergic, beta-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...