Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Pathol Int ; 70(1): 12-20, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31872565

ABSTRACT

This study was to investigate the expression of coactivator-associated arginine methyltransferase 1 (CARM1) and miR-16-5p in cervical cancer (CC), and explore their roles in radioresistance. Western blot and immunohistochemistry were used to detect the expression of CARM1 in tissues and cells. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the expression of miR-16-5p. CC cells received different doses of X-ray exposure, and then cell counting kit-8 method and colony formation assay were used to detect cell proliferation. Apoptosis was detected by flow cytometry. Then we used Targetscan database to predict that CARM1 is a potential target of miR-16-5p, and further verified the targeting relationship between them by western blot, RT-PCR and dual luciferase reporter experiments. We demonstrated that CARM1 were highly expressed in CC tissues and radio-resistant CC cells, while miR-16-5p expression was low. Under irradiation, up-regulation of CARM1 can induce radiotherapy resistance of CC cells, while overexpression of miR-16-5p or CARM1 knockdown could inhibit the survival of CC cell and induced apoptosis. CARM1 was verified as a target for miR-16-5p. Besides, up-regulation of CARM1 reversed the increase in radiosensitivity induced by miR-16-5p. Collectively, we concluded that miR-16-5p promoted the radiosensitivity of CC cells by targeting CARM1.


Subject(s)
CARD Signaling Adaptor Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Guanylate Cyclase/biosynthesis , MicroRNAs/genetics , Radiation Tolerance/genetics , Uterine Cervical Neoplasms/pathology , CARD Signaling Adaptor Proteins/genetics , Female , Guanylate Cyclase/genetics , Humans , Uterine Cervical Neoplasms/genetics
2.
Development ; 141(1): 136-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24284209

ABSTRACT

The second messengers cAMP and cGMP modulate attraction and repulsion mediated by neuronal guidance cues. We find that the Drosophila receptor guanylyl cyclase Gyc76C genetically interacts with Semaphorin 1a (Sema-1a) and physically associates with the Sema-1a receptor plexin A (PlexA). PlexA regulates Gyc76C catalytic activity in vitro, and each distinct Gyc76C protein domain is crucial for regulating Gyc76C activity in vitro and motor axon guidance in vivo. The cytosolic protein dGIPC interacts with Gyc76C and facilitates Sema-1a-PlexA/Gyc76C-mediated motor axon guidance. These findings provide an in vivo link between semaphorin-mediated repulsive axon guidance and alteration of intracellular neuronal cGMP levels.


Subject(s)
Axons/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Guanylate Cyclase/metabolism , Motor Neurons/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Animals , Axons/metabolism , Carrier Proteins/metabolism , Catalysis , Cells, Cultured , Cyclic GMP/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Motor Neurons/metabolism , Protein Binding , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Sequence Deletion/genetics , Signal Transduction/genetics , Signal Transduction/physiology
3.
Am J Physiol Renal Physiol ; 311(1): F103-11, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27076649

ABSTRACT

Purinoceptors (adrengeric receptors and P2 receptors) are expressed on the cellular components of the glomerular filtration barrier, and their activation may affect glomerular permeability to albumin, which may ultimately lead to albuminuria, a well-established risk factor for the progression of chronic kidney disease and development of cardiovascular diseases. We investigated the mechanisms underlying the in vitro and in vivo purinergic actions on glomerular filter permeability to albumin by measuring convectional albumin permeability (Palb) in a single isolated rat glomerulus based on the video microscopy method. Primary cultured rat podocytes were used for the analysis of Palb, cGMP accumulation, PKG-Iα dimerization, and immunofluorescence. In vitro, natural nucleotides (ATP, ADP, UTP, and UDP) and nonmetabolized ATP analogs (2-meSATP and ATP-γ-S) increased Palb in a time- and concentration-dependent manner. The effects were dependent on P2 receptor activation, nitric oxide synthase, and cytoplasmic guanylate cyclase. ATP analogs significantly increased Palb, cGMP accumulation, and subcortical actin reorganization in a PKG-dependent but nondimer-mediated route in cultured podocytes. In vivo, 2-meSATP and ATP-γ-S increased Palb but did not significantly affect urinary albumin excretion. Both agonists enhanced the clathrin-mediated endocytosis of albumin in podocytes. A product of adenine nucleotides hydrolysis, adenosine, increased the permeability of the glomerular barrier via adrenergic receptors in a dependent and independent manner. Our results suggest that the extracellular nucleotides that stimulate an increase of glomerular Palb involve nitric oxide synthase and cytoplasmic guanylate cyclase with actin reorganization in podocytes.


Subject(s)
Albumins/metabolism , Albuminuria/metabolism , Kidney Glomerulus/metabolism , Purines/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Albuminuria/pathology , Animals , Cyclic GMP/metabolism , Endocytosis/drug effects , Female , Guanylate Cyclase/biosynthesis , In Vitro Techniques , Kidney Glomerulus/drug effects , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Permeability/drug effects , Podocytes/drug effects , Podocytes/metabolism , Primary Cell Culture , Purinergic P2 Receptor Agonists/pharmacology , Rats , Rats, Wistar
4.
J Pharmacol Exp Ther ; 352(3): 509-18, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25550199

ABSTRACT

The effect of proinflammatory cytokines on the expression and activity of soluble guanylyl cyclase (sGC) and cGMP-phosphodiesterases (PDEs) was determined in intestinal longitudinal smooth muscle. In control muscle cells, cGMP levels are regulated via activation of sGC and PDE5; the activity of the latter is regulated via feedback phosphorylation by cGMP-dependent protein kinase. In muscle cells isolated from muscle strips cultured with interleukin-1ß (IL-1ß) or tumor necrosis factor α (TNF-α) or obtained from the colon of TNBS (2,4,6-trinitrobenzene sulfonic acid)-treated mice, expression of inducible nitric oxide synthase (iNOS) was induced and sGC was S-nitrosylated, resulting in attenuation of nitric oxide (NO)-induced sGC activity and cGMP formation. The effect of cytokines on sGC S-nitrosylation and activity was blocked by the iNOS inhibitor 1400W [N-([3-(aminomethyl)phenyl]methyl)ethanimidamide dihydrochloride]. The effect of cytokines on cGMP levels measured in the absence of IBMX (3-isobutyl-1-methylxanthine), however, was partly reversed by 1400W or PDE1 inhibitor vinpocetine and completely reversed by a combination of 1400W and vinpocetine. Expression of PDE1A was induced and was accompanied by an increase in PDE1A activity in muscle cells isolated from muscle strips cultured with IL-1ß or TNF-α or obtained from the colon of TNBS-treated mice; the effect of cytokines on PDE1 expression and activity was blocked by MG132 (benzyl N-[(2S)-4-methyl-1-[[(2S)-4-methyl-1-[[(2S)-4-methyl-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]carbamate), an inhibitor of nuclear factor κB activity. NO-induced muscle relaxation was inhibited in longitudinal muscle cells isolated from muscle strips cultured with IL-1ß or TNF-α or obtained from the colon of TNBS-treated mice, and this inhibition was completely reversed by the combination of both 1400W and vinpocetine. Inhibition of smooth muscle relaxation during inflammation reflects the combined effects of decreased sGC activity via S-nitrosylation and increased cGMP hydrolysis via PDE1 expression.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/biosynthesis , Gene Expression Regulation, Enzymologic , Guanylate Cyclase/biosynthesis , Muscle Relaxation/physiology , Muscle, Smooth/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Animals , Cytokines/toxicity , Male , Mice , Mice, Inbred C57BL , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Soluble Guanylyl Cyclase
5.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L868-76, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25326578

ABSTRACT

Excess superoxide has been implicated in pulmonary hypertension (PH). We previously found lung overexpression of the antioxidant extracellular superoxide dismutase (EC-SOD) attenuates PH and pulmonary artery (PA) remodeling. Although comprising a small fraction of total SOD activity in most tissues, EC-SOD is abundant in arteries. We hypothesize that the selective loss of vascular EC-SOD promotes hypoxia-induced PH through redox-sensitive signaling pathways. EC-SOD(loxp/loxp) × Tg(cre/SMMHC) mice (SMC EC-SOD KO) received tamoxifen to conditionally deplete smooth muscle cell (SMC)-derived EC-SOD. Mice were exposed to hypobaric hypoxia for 35 days, and PH was assessed by right ventricular systolic pressure measurements and right ventricle hypertrophy. Vascular remodeling was evaluated by morphometric analysis and two-photon microscopy for collagen. We examined cGMP content and soluble guanylate cyclase expression and activity in lung, lung phosphodiesterase 5 (PDE5) expression and activity, and expression of endothelial nitric oxide synthase and GTP cyclohydrolase-1 (GTPCH-1), the rate-limiting enzyme in tetrahydrobiopterin synthesis. Knockout of SMC EC-SOD selectively decreased PA EC-SOD without altering total lung EC-SOD. PH and vascular remodeling induced by chronic hypoxia was augmented in SMC EC-SOD KO. Depletion of SMC EC-SOD did not impact content or activity of lung soluble guanylate cyclase or PDE5, yet it blunted the hypoxia-induced increase in cGMP. Although total eNOS was not altered, active eNOS and GTPCH-1 decreased with hypoxia only in SMC EC-SOD KO. We conclude that the localized loss of PA EC-SOD augments chronic hypoxic PH. In addition to oxidative inactivation of NO, deletion of EC-SOD seems to reduce eNOS activity, further compromising pulmonary vascular function.


Subject(s)
Hypertension, Pulmonary/therapy , Hypoxia/therapy , Superoxide Dismutase/genetics , Animals , Blood Pressure , Cyclic GMP/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Estrogen Antagonists/pharmacology , GTP Cyclohydrolase/biosynthesis , Guanylate Cyclase/biosynthesis , Hypertrophy, Right Ventricular/physiopathology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/biosynthesis , Pulmonary Artery/pathology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Signal Transduction , Soluble Guanylyl Cyclase , Tamoxifen/pharmacology
6.
Mol Hum Reprod ; 20(1): 77-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23894150

ABSTRACT

The molecular mechanism of stress-associated reproductive dysfunction is complex and largely unknown. This study was designed to systematically analyze molecular effects of systemic in vivo blockade of α1-adrenergic receptors (α1-ADRs) on stress-induced disturbance of cAMP/cGMP signaling in testosterone-producing Leydig cells using the following parameters (i) level of circulating stress hormones, LH and testosterone; (ii) level of main molecular markers of Leydig cell functionality (testosterone, Insl3, cAMP); (iii) expression of cAMP signaling (cAMP 'producers'/'effectors'/'removers') and (iv) expression of NO-cGMP signaling (NO-cGMP 'producers'/'effectors'/'removers'). The results showed that oral administration of α1-ADR blocker before stress increased cGMP and diminished stress-reduced cAMP production in Leydig cells. In the same cells, stress-induced effects on cAMP/cGMP signaling pathways elements were changed. Sustained in vivo α1-ADR blockade completely abolished stress-increased transcription of most abundantly expressed phosphodiesterase that remove cAMP (Pde4b) and potentiated stress-increased expression of PRKA, the main stimulator of Leydig cell steroidogenesis. In the same Leydig cells, stress-decreased NOS3 expression was abolished, while stress-increased GUCY1 (cGMP 'producer') and PRKG1 (cGMP 'effector') were potentiated. It is possible that all molecules mentioned could contribute, at least in part, in recovery of Leydig cell testosterone production. Presented data provide new role of α1-ADRs in stress-triggered disturbance of cAMP/cGMP signaling, and new molecular insights into the relationship between stress and mammalian reproduction. Regardless of whether the effects of α1-blocker + stress are direct or indirect, the results are important in terms of human reproductive health and the wide use of α1-ADR antagonists, alone or in combination, to treat post-traumatic stress disorders, hypertension, benign prostatic hyperplasia symptoms and potential drugs for prostate cancer prevention/treatment.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Leydig Cells/metabolism , Stress, Physiological/drug effects , AMP-Activated Protein Kinases/biosynthesis , Animals , Corticosterone/blood , Cyclic AMP/biosynthesis , Cyclic GMP/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type I/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Doxazosin/pharmacology , Epinephrine/blood , Guanylate Cyclase/biosynthesis , Insulin/biosynthesis , Luteinizing Hormone/blood , Male , Nitric Oxide Synthase Type III/biosynthesis , Proteins , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Cytoplasmic and Nuclear/biosynthesis , Signal Transduction , Soluble Guanylyl Cyclase , Testosterone/biosynthesis , Testosterone/blood
7.
Heart Fail Rev ; 18(2): 123-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22622468

ABSTRACT

The number of annual hospitalizations for heart failure (HF) and the mortality rates among patients hospitalized for HF remains unacceptably high. The search continues for safe and effective agents that improve outcomes when added to standard therapy. The nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway serves an important physiologic role in both vascular and non-vascular tissues, including regulation of myocardial and renal function, and is disrupted in the setting of HF, leading to decreased protection against myocardial injury, ventricular remodeling, and the cardio-renal syndrome. The impaired NO-sGC-cGMP pathway signaling in HF is secondary to reduced NO bioavailability and an alteration in the redox state of sGC, making it unresponsive to NO. Accordingly, increasing directly the activity of sGC is an attractive pharmacologic strategy. With the development of two novel classes of drugs, sGC stimulators and sGC activators, the hypothesis that restoration of NO-sGC-cGMP signaling is beneficial in HF patients can now be tested. Characterization of these agents in pre-clinical and clinical studies has begun with investigations suggesting both hemodynamic effects and organ-protective properties independent of hemodynamic changes. The latter could prove valuable in long-term low-dose therapy in HF patients. This review will explain the role of the NO-sGC-cGMP pathway in HF pathophysiology and outcomes, data obtained with sGC stimulators and sGC activators in pre-clinical and clinical studies, and a plan for the further clinical development to study these agents as HF therapy.


Subject(s)
Cyclic GMP/therapeutic use , Guanylate Cyclase/drug effects , Heart Failure/drug therapy , Hemodynamics/drug effects , Nitric Oxide/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Cyclic GMP/metabolism , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/metabolism , Heart Failure/physiopathology , Humans , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/biosynthesis , Soluble Guanylyl Cyclase , Treatment Outcome
8.
Clin Exp Pharmacol Physiol ; 40(3): 240-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23331131

ABSTRACT

The mechanisms by which the hexane extract of Curcuma comosa increases femoral blood flow (FBF) in ovariectomized rats are not known. Thus, the aim of the present study was to investigate the acute effects and modes of action of the diarylheptanoid (3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol (D3), a phyto-oestrogen isolated from C. comosa, on FBF in ovariectomized rats. On Day 7 after ovariectomy, rats were injected once intra-arterially with D3 (100, 200, 400 and 800 µg/kg), 17ß-oestradiol (E2; 1, 2, 4 and 8 µg/kg) or vehicle. In some experiments, rats were injected with N(G)-nitro-L-arginine methyl ester (L-NAME; 10 mg/kg) 120 min after 800 µg/kg D3 or 4 µg/kg E2. In other experiments, rats were injected with 10 mg/kg L-NAME, 900 µg/kg 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or 900 µg/kg ICI 182 780 30 min prior to the injection of 800 µg/kg D3 or 4 µg/kg E2. Mean arterial blood pressure (mABP) and FBF were recorded using a pressure transducer and a laser Doppler flow meter, respectively. Both D3 and E2 dose-dependently increased FBF without changing mABP or heart rate. The EC(50) at 120 min for D3 and E2 was 195.8 and 1.8 µg/kg, respectively. In addition, D3 and E2 dose-dependently decreased femoral vascular resistance (FVR). The EC(50) of D3 was about 100-fold greater than that of E2. The effects of D3 and E2 on FBF and FVR were diminished by intravenous injection of 10 mg/kg l-NAME. Furthermore, 30 min pretreatment with L-NAME (10 mg/kg), ODQ (900 µg/kg) or ICI 182 780 (900 µg/kg) blocked the effects of D3 and E2 on FBF and FVR. The results of the present study suggest that the phyto-oestrogen D3 increases FBF in ovariectomized rats via oestrogen receptor and nitric oxide-guanylyl cyclase signalling, which, in turn, relaxes femoral vascular resistance.


Subject(s)
Arterial Pressure/drug effects , Curcuma/chemistry , Femoral Artery/drug effects , Heptanol/analogs & derivatives , Nitric Oxide/biosynthesis , Phytoestrogens/pharmacology , Regional Blood Flow/drug effects , Animals , Arterial Pressure/physiology , Diarylheptanoids , Dose-Response Relationship, Drug , Female , Femoral Artery/metabolism , Femoral Artery/physiology , Guanylate Cyclase/biosynthesis , Heptanol/isolation & purification , Heptanol/pharmacology , Injections, Intra-Arterial , Molecular Structure , Ovariectomy , Phytoestrogens/isolation & purification , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Resistance/drug effects
9.
Biosci Biotechnol Biochem ; 77(8): 1689-93, 2013.
Article in English | MEDLINE | ID: mdl-23924731

ABSTRACT

Oxidative stress was induced in 12-week-old offspring of protein-restricted (9% protein) and control (20% protein) protein-restricted stroke-prone spontaneously hypertensive rats (SHRSP) by administering phorbol 12-myristate 13-acetate (PMA) for 4 weeks to determine the effects of oxidative stress on the vascular function of the SHRSP offspring. There was no significant difference in the blood pressure of offspring of the protein-restricted dams and control dams. The plasma diacron-reactive oxygen metabolite (dROM) level at 16 weeks of age was significantly higher in offspring of the protein-restricted dams, whereas the anti-oxidative enzyme activity was similar in both groups. Acetylcholine (Ach)-induced relaxation was significantly reduced in offspring of the protein-restricted dams. The expression of endothelial nitric oxide synthase (eNOS) was lower and the expression of soluble guanylic acid cyclase (sGC) was higher in offspring of the protein-restricted dams. These results indicate that SHRSP offspring of the protein-restricted dams were sensitive to oxidative stress, and displayed the vascular dysfunction.


Subject(s)
Diet, Protein-Restricted , Endothelium, Vascular/drug effects , Hypertension/metabolism , Oxidative Stress , Animals , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Guanylate Cyclase/biosynthesis , Hypertension/chemically induced , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Rats , Reactive Oxygen Species/metabolism , Tetradecanoylphorbol Acetate/administration & dosage , Vasodilation/drug effects , Vasodilation/physiology
10.
J Headache Pain ; 14: 74, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24004534

ABSTRACT

BACKGROUND: Nitric oxide (NO) is thought to play an important role in the pathophysiology of migraine. Infusion of the nitrovasodilator glyceroltrinitrate (nitroglycerin, GTN), which mobilizes NO in the organism, is an approved migraine model in humans. Calcitonin gene-related peptide (CGRP) is regarded as another key mediator in migraine. Increased plasma levels of CGRP have been found during spontaneous as well as nitrovasodilator-induced migraine attacks. The nociceptive processes and interactions underlying the NO and CGRP mediated headache are poorly known but can be examined in animal experiments. In the present study we examined changes in immunofluorescence of CGRP receptor components (CLR and RAMP1) and soluble guanylyl cyclase (sGC), the intracellular receptor for NO, in rat trigeminal ganglia after pretreatment with GTN. METHODS: Isoflurane anaesthetised rats were intravenously infused with GTN (1 mg/kg) or saline for four hours and two hours later the trigeminal ganglia were processed for immunohistochemistry. Different primary antibodies recognizing CLR, RAMP1, CGRP and sGC coupled to fluorescent secondary antibodies were used to examine immunoreactive cells in serial sections of trigeminal ganglia with epifluorescence and confocal laser scanning microscopy. Several staining protocols were examined to yield optimized immunolabeling. RESULTS: In vehicle-treated animals, 42% of the trigeminal ganglion neurons were immunopositive for RAMP1 and 41% for CLR. After GTN pretreatment CLR-immunopositivity was unchanged, while there was an increase in RAMP1-immunopositive neurons to 46%. RAMP1 and CLR immunoreactivity was also detected in satellite cells. Neurons immunoreactive for sGC were on average smaller than sGC-immunonegative neurons. The percentage of sGC-immunopositive neurons (51% after vehicle) was decreased after GTN infusion (48%). CONCLUSIONS: Prolonged infusion of GTN caused increased fractions of RAMP1- and decreased fractions of sGC-immunopositive neurons in the trigeminal ganglion. The observed alterations are likely immunophenotypic correlates of the pathophysiological processes underlying nitrovasodilator-induced migraine attacks and indicate that signalling via CGRP receptors but not sGC-mediated mechanisms may be enhanced through endogenous NO production.


Subject(s)
Guanylate Cyclase/biosynthesis , Migraine Disorders/metabolism , Receptors, Calcitonin Gene-Related Peptide/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Trigeminal Ganglion/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Immunohistochemistry , Male , Microscopy, Confocal , Migraine Disorders/chemically induced , Nitroglycerin/toxicity , Rats , Rats, Wistar , Soluble Guanylyl Cyclase , Vasodilator Agents/toxicity
11.
J Urol ; 188(1): 316-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22608751

ABSTRACT

PURPOSE: Gender difference and nitric oxide deficiency contribute to the progression of many chronic kidney diseases. In a model of unilateral ureteral obstruction relief we analyzed the impact of biological gender and nitric oxide/cyclic guanosine monophosphate signaling stimulation on renal disease severity and restoration. MATERIALS AND METHODS: Female and male rats underwent sham surgery or unilateral ureteral obstruction. After 5-day unilateral ureteral obstruction female and male rats were assigned to obstruction relief alone or obstruction relief plus 7-day treatment with the soluble guanylate cyclase stimulator BAY 41-8543. RESULTS: Compared to male rats with obstruction relief renal disease was less severe in female rats, which had significantly less tubulointerstitial matrix accumulation and tubular atrophy. In each gender group α1 and ß1-soluble guanylate cyclase was comparably and significantly increased but female rats produced significantly more cyclic guanosine monophosphate after treatment with the soluble guanylate cyclase stimulator. In each group BAY 41-8543 treatment was associated with significant amelioration of renal matrix protein expansion, macrophage infiltration, tubular apoptosis and atrophy. CONCLUSIONS: Female gender is protective for unilateral ureteral obstruction relief. This was linked to higher sensitivity of the soluble guanylate cyclase enzyme and cyclic guanosine monophosphate production in response to BAY 41-8543. In these female and male rats enhancing the signaling of nitric oxide/cyclic guanosine monophosphate with BAY 41-8543 significantly accelerated the restoration of renal architecture after obstruction relief and largely ameliorated the differences in disease severity due to the gender disparity.


Subject(s)
Gene Expression Regulation , Guanylate Cyclase/genetics , Kidney/physiology , Morpholines/therapeutic use , Pyrimidines/therapeutic use , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Recovery of Function , Ureteral Obstruction/drug therapy , Administration, Oral , Animals , Apoptosis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Guanylate Cyclase/biosynthesis , In Situ Nick-End Labeling , Male , Morpholines/administration & dosage , Pyrimidines/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/biosynthesis , Severity of Illness Index , Sex Factors , Soluble Guanylyl Cyclase , Treatment Outcome , Ureteral Obstruction/pathology , Ureteral Obstruction/physiopathology
12.
Mol Pharmacol ; 80(6): 1076-84, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21908708

ABSTRACT

The NO and cGMP signaling pathways are of broad physiological and pathological significance. We compared the NO/soluble guanylyl cyclase (sGC)/cGMP pathway in human glioma tissues and cell lines with that of healthy control samples and demonstrated that sGC expression is significantly lower in glioma preparations. Our analysis of GEO databases (National Cancer Institute) further revealed a statistically significant reduction of sGC transcript levels in human glioma specimens. On the other hand, the expression levels of particulate (membrane) guanylyl cyclases (pGC) and cGMP-specific phosphodiesterase (PDE) were intact in the glioma cells that we have tested. Pharmacologically manipulating endogenous cGMP generation in glioma cells through either stimulating pGC by ANP/BNP, or blocking PDE by 3-isobutyl-1-methylxanthine/zaprinast caused significant inhibition of proliferation and colony formation of glioma cells. Genetically restoring sGC expression also correlated inversely with glioma cells growth. Orthotopic implantation of glioma cells transfected with an active mutant form of sGC (sGCα1ß1(Cys105)) in athymic mice increased the survival time by 4-fold over the control. Histological analysis of xenografts overexpressing α1ß1(Cys105) sGC revealed changes in cellular architecture that resemble the morphology of normal cells. In addition, a decrease in angiogenesis contributed to glioma inhibition by sGC/cGMP therapy. Our study proposes the new concept that suppressed expression of sGC, a key enzyme in the NO/cGMP pathway, may be associated with an aggressive course of glioma. The sGC/cGMP signaling-targeted therapy may be a favorable alternative to chemotherapy and radiotherapy for glioma and perhaps other tumors.


Subject(s)
Antineoplastic Agents/metabolism , Gene Expression Regulation, Enzymologic , Glioma/enzymology , Glioma/prevention & control , Guanylate Cyclase/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Glioma/pathology , Guanylate Cyclase/physiology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Receptors, Cytoplasmic and Nuclear/physiology , Soluble Guanylyl Cyclase , Xenograft Model Antitumor Assays/methods
13.
Am J Physiol Endocrinol Metab ; 299(4): E544-50, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20663985

ABSTRACT

Sildenafil citrate (Viagra), a cGMP-selective phosphodiesterase (PDE) inhibitor, is widely used to treat erectile dysfunction and pulmonary arterial hypertension. In contrast to its well established action on erectile dysfunction, little is known on the action of sildenafil on cGMP/cAMP signaling and testicular steroidogenesis. This study was designed to assess the effects of prolonged sildenafil treatment on NO synthase-dependent signaling and steroidogenic function of rat Leydig cells. Male adult rats were treated with Viagra (1.25 mg/kg body wt) daily for 30 days. In our studies, serum testosterone and ex vivo testosterone production significantly increased in sildenafil-treated animals. Human chorionic gonadotropin-stimulated testosterone production and cAMP accumulation were also significantly higher in Leydig cells obtained from sildenafil-treated rats. The expression of soluble guanylyl cyclase (GUCY1) subunits (Gucy1a1, Gucy1b1) significantly increased; cAMP-specific Pde4a, cGMP-specific Pde6c, and dual Pde1c and Nos2 were inhibited and expression of Nos3, protein kinase G1 (Pkg1), and Pde5 remained unchanged. Treatment of purified Leydig cells with NO donor caused a dose-dependent increase in both testosterone and cGMP production. Testosterone and cGMP production was significantly higher in Leydig cells obtained from sildenafil-treated animals. The stimulatory effect of NO donor was significantly enhanced by saturating concentrations of hCG in both Leydig cells obtained from control and sildenafil-treated animals. Occurrence of mature steroidogenic acute regulatory protein also increased in sildenafil treated animals in accord with increased cAMP and cGMP production. In summary, inhibition of PDE activity during prolonged sildenafil treatment increased serum testosterone level and Leydig cells' steroidogenic capacity by coordinated stimulatory action on cAMP and cGMP signaling pathway.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Leydig Cells/drug effects , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Testis/drug effects , Testosterone/biosynthesis , Animals , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Leydig Cells/enzymology , Leydig Cells/metabolism , Male , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Phosphodiesterase 5 Inhibitors , Purines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sildenafil Citrate , Statistics, Nonparametric , Testis/cytology , Testis/enzymology , Testis/metabolism
14.
J Pharmacol Exp Ther ; 335(1): 85-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20605906

ABSTRACT

Heme oxygenase-1 knockout, H(mox)1(-/-), mice exhibit exacerbated vascular lesions after ischemia-reperfusion and mechanical injury. Surprisingly, we found no studies that reported contractile responses and sensitivity to vasorelaxants in H(mox)1(-/-) mice. The contractile responses [superior mesenteric arteries (SMA), from female H(mox)1(-/-) mice] exhibited increased sensitivity to phenylephrine (p < 0.001). Cumulative addition of acetylcholine relaxed SMA, with the residual contraction remaining 2 times higher in H(mox)1(-/-) mice (p < 0.001). Sodium nitroprusside (SNP, an NO donor) and 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). Branches from H(mox)1(-/-) mesenteric and renal arteries also showed reduced relaxation (p < 0.025). Relaxation of SMA was measured to 4-({(4-carboxybutyl) [2-(5-fluoro-2-{[4'-(trifluoromethyl) biphenyl-4-yl] methoxy}phenyl)ethyl]amino}benzoic acid (BAY 60-2770), which is a more effective activator of oxidized/heme-free sGC; and to 5-cyclopropyl-2-{1-(2-fluoro-benzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl}-pyrimidin-4-ylamine (BAY 41-2272), a more effective stimulator of reduced sGC. H(mox)1(-/-) arteries were 15 times more sensitive to BAY 60-2770 (p < 0.025) than were H(mox)1(+/+) arteries. Pretreatment with 1H-[1,2,4]oxadiazolo[3,4-a]quinoxalin-1-one (ODQ), an oxidizer of sGC, predictably shifted the BAY 60-2770 response of H(mox)1(+/+) to the left (p < 0.01) and BAY 41-2272 response to the right (p < 0.01). ODQ had little effect on the responses of H(mox)1(-/-) arteries, indicating that much of sGC was oxidized/heme-free. Western analyses of sGC in SMA indicated that both α1 and ß1 subunit levels were reduced to <50% of H(mox)1(+/+) level (p < 0.025). These findings support the hypothesis that the antioxidant function of H(mox)1 plays a significant role in the maintenance of sGC in a reduced state, which is resistant to degradation and is sensitive to NO. This function may be especially important in reducing vascular damage during ischemia-reperfusion injury.


Subject(s)
Guanylate Cyclase/metabolism , Heme Oxygenase-1/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Acetylcholine/metabolism , Acetylcholine/pharmacology , Animals , Arteries/drug effects , Arteries/metabolism , Blotting, Western , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Guanylate Cyclase/biosynthesis , Heme/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Mice , Mice, Knockout , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitrates/metabolism , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Oxidation-Reduction , Phenylephrine/pharmacology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Soluble Guanylyl Cyclase , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
15.
Amino Acids ; 39(2): 399-408, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20063108

ABSTRACT

Soluble guanylate cyclase (sGC), as a nitric oxide (NO) sensor, is a critical heme-containing enzyme in NO-signaling pathway of eukaryotes. Human sGC is a heterodimeric hemoprotein, composed of a alpha-subunit (690 AA) and a heme-binding beta-subunit (619 AA). Upon NO binding, sGC catalyzes the conversion of guanosine 5'-triphosphate (GTP) to 3',5'-cyclic guanosine monophosphate (cGMP). cGMP is a second messenger and initiates the nitric oxide signaling, triggering vasodilatation, smooth muscle relaxation, platelet aggregation, and neuronal transmission etc. The breakthrough of the bottle neck problem for sGC-mediated NO singling was made in this study. The recombinant human sGC beta1 subunit (HsGC beta 619) and its truncated N-terminal fragments (HsGC beta 195 and HsGC beta 384) were efficiently expressed in Escherichia coli and purified successfully in quantities. The three proteins in different forms (ferric, ferrous, NO-bound, CO-bound) were characterized by UV-vis and EPR spectroscopy. The homology structure model of the human sGC heme domain was constructed, and the mechanism for NO binding to sGC was proposed. The EPR spectra showed a characteristic of five-coordinated heme-nitrosyl species with triplet hyperfine splitting of NO. The interaction between NO and sGC was investigated and the schematic mechanism was proposed. This study provides new insights into the structure and NO-binding of human sGC. Furthermore, the efficient expression system of E. coli will be beneficial to the further studies on structure and activation mechanism of human sGC.


Subject(s)
Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Cloning, Molecular , Electron Spin Resonance Spectroscopy , Escherichia coli/enzymology , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/chemistry , Humans , Models, Molecular , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Soluble Guanylyl Cyclase , Spectrophotometry, Ultraviolet
16.
Eur Respir J ; 33(4): 785-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19129292

ABSTRACT

Pulmonary hypertension (PH) is associated with impaired production of the vasodilator nitric oxide (NO). Riociguat (BAY 63-2521; Bayer Healthcare AG, Wuppertal, Germany) acts directly on soluble guanylate cyclase, stimulating the enzyme and increasing sensitivity to low NO levels. The present study evaluates riociguat safety, tolerability and efficacy in patients with moderate-to-severe PH (pulmonary arterial hypertension, distal chronic thromboembolic PH or PH with mild to moderate interstitial lung disease). The optimal tolerated dose was identified by incremental dosing in four patients with PH; pharmacodynamic and pharmacokinetic parameters were assessed following single-dose administration (2.5 mg or 1 mg) in 10 and five patients with PH, respectively. All subjects (n = 19) were analysed for safety and tolerability. Riociguat had a favourable safety profile at single doses < or =2.5 mg. It significantly improved pulmonary haemodynamic parameters and cardiac index in patients with PH in a dose-dependent manner, to a greater extent than inhaled NO. Although riociguat also had significant systemic effects and showed no pulmonary selectivity, mean systolic blood pressure remained >110 mmHg. The present report is the first to describe the use of riociguat in patients with pulmonary hypertension. The drug was well-tolerated and superior to nitric oxide in efficacy and duration. Riociguat, therefore, has potential as a novel therapy for pulmonary hypertension and warrants further investigation.


Subject(s)
Guanylate Cyclase/biosynthesis , Guanylate Cyclase/physiology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Area Under Curve , Chromatography, High Pressure Liquid , Female , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Nitric Oxide Synthase Type II/pharmacology , Oxidation-Reduction , Pulmonary Circulation/physiology , Pyrimidines/pharmacokinetics , Soluble Guanylyl Cyclase , Treatment Outcome
17.
Protein Expr Purif ; 65(2): 122-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19174191

ABSTRACT

Compounds capable of stimulating soluble guanylate cyclase (sGC) activity might become important new tools to treat hypertension. While rational design of these drugs would be aided by elucidation of the sGC three-dimensional structure and molecular mechanism of activation, such efforts also require quantities of high quality enzyme that are challenging to produce. We implemented the titerless infected-cells preservation and scale-up (TIPS) methodology to express the heterodimeric sGC. In the TIPS method, small-scale insect cell cultures were first incubated with a recombinant baculovirus which replicated in the cells. The baculovirus-infected insect cells (BIIC) were harvested and frozen prior to cell lysis and the subsequent escape of the newly replicated virus into the culture supernatant. Thawed BIIC stocks were ultimately used for subsequent scale up. As little as 1 mL of BIIC was needed to infect a 100-L insect cell culture, in contrast to the usual 1L of high-titer, virus stock supernatants. The TIPS method eliminates the need and protracted time for titering virus supernatants, and provides stable, concentrated storage of recombinant baculovirus in the form of infected cells. The latter is particularly advantageous for virus stocks which are unstable, such as those for sGC, and provides a highly efficient alternative for baculovirus storage and expression. The TIPS process enabled efficient scale up to 100-L batches, each producing about 200mg of active sGC. Careful adjustment of expression culture conditions over the course of several 100-L runs provided uniform starting titers, specific activity, and composition of contaminating proteins that facilitated development of a process that reproducibly yielded highly active, purified sGC.


Subject(s)
Baculoviridae/genetics , Guanylate Cyclase/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Spodoptera/cytology , Spodoptera/metabolism , Animals , Baculoviridae/physiology , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Humans , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Soluble Guanylyl Cyclase , Spodoptera/virology , Time Factors
18.
J Pineal Res ; 46(2): 140-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19175857

ABSTRACT

Melatonin influences the second messenger cyclic guanosine 3',5'-monophosphate (cGMP) signaling pathway in pancreatic beta-cells via a receptor-mediated mechanism. In the present study, it was determined how the regulation of cGMP concentrations by melatonin proceeds. The results provide evidence that melatonin acts via the soluble guanylate cyclase (sGC), as molecular investigations demonstrated that long-term incubation with melatonin significantly reduced the expression levels of the sGC mRNA in rat insulinoma beta-cells (INS1) cells, whereas mRNA expression of membrane guanylate cyclases was unaffected. Incubation with melatonin abolished the S-nitrosoacetyl penicillamine-induced increase of cGMP concentrations in INS1 cells. In addition, the cGMP-inhibitory effect of melatonin was reversed by preincubation with the sGC inhibitors 1H-(1,2,4)oxadiazolo(4,3-alpha)quinoxalin-1-one and 4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one. Nitric oxide (NO) production was not influenced after 1 hr of melatonin application, but was influenced after a 4 hr incubation period. Preincubation of INS1 cells with the NO synthase inhibitor N(G)-monomethyl-l-arginine did not abolish the cGMP-inhibitory effect of melatonin. Transcripts of cyclic nucleotide-gated (CNG) channels were significantly reduced after melatonin treatment in a dose-dependent manner, indicating the involvement of these channels in mediating the melatonin effect in INS1 cells. The results of this study demonstrate that melatonin mediates its inhibitory effect on cGMP concentrations in pancreatic beta-cells by inhibiting the sGC, but does not influence NO concentration or NO synthase activity in short-term incubation experiments. In addition, it was demonstrated that melatonin is involved in modulation of CNG channel mRNA.


Subject(s)
Antioxidants/pharmacology , Cyclic GMP/metabolism , Insulin-Secreting Cells/metabolism , Melatonin/pharmacology , Receptors, Melatonin/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , RNA, Messenger/biosynthesis , Rats , Receptors, Melatonin/agonists , Signal Transduction/physiology
19.
Exp Hematol ; 73: 50-63.e2, 2019 05.
Article in English | MEDLINE | ID: mdl-30986496

ABSTRACT

The most frequently occurring genetic abnormality in pediatric B-lymphocyte-lineage acute lymphoblastic leukemia is the t(12;21) chromosomal translocation that results in a ETV6-RUNX1 (also known as TEL-AML1) fusion gene. Expression of ETV6-RUNX1 induces a preleukemic condition leading to acquisition of secondary driver mutations, but the mechanism is poorly understood. SPI-B (encoded by SPIB) is an important transcriptional activator of B-cell development and differentiation. We hypothesized that SPIB is directly transcriptionally repressed by ETV6-RUNX1. Using chromatin immunoprecipitation, we identified a regulatory region in the first intron of SPIB that interacts with ETV6-RUNX1. Mutation of the RUNX1 binding site in SPIB intron 1 prevented transcriptional repression in transient transfection assays. Next, we sought to determine to what extent gene expression in REH cells can be altered by ectopic SPI-B expression. SPI-B expression was forced using CRISPR-mediated gene activation and also using a retroviral vector. Forced expression of SPI-B resulted in altered gene expression and, at high levels, impaired cell proliferation and induced apoptosis. Finally, we identified CARD11 and CDKN1A (encoding p21) as transcriptional targets of SPI-B involved in regulation of proliferation and apoptosis. Taken together, this study identifies SPIB as an important target of ETV6-RUNX1 in regulation of B-cell gene expression in t(12;21) leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Leukemic , Introns , Oncogene Proteins, Fusion/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Response Elements , Transcription Factors/biosynthesis , Apoptosis/genetics , CARD Signaling Adaptor Proteins/biosynthesis , CARD Signaling Adaptor Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Humans , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/genetics , Translocation, Genetic
20.
J Neurosci ; 27(12): 3295-304, 2007 Mar 21.
Article in English | MEDLINE | ID: mdl-17376990

ABSTRACT

Children exposed to early parental loss from death or separation carry a greater risk for developing future psychiatric illnesses, such as major depression and anxiety. Monkeys experiencing maternal separation at 1 week of age show fewer social behaviors and an increase in self-comforting behaviors (e.g., thumb sucking) over development, whereas in contrast, monkeys experiencing maternal separation at 1 month of age show increased seeking of social comfort later in life. We sought to identify neural systems that may underlie these stress-induced behavioral changes by examining changes in mRNA content in amygdala tissue collected from 1 week separated, 1 month separated, and maternally reared infants at 3 months of age. mRNA from the right medial temporal lobe, primarily the amygdala, was analyzed using Affymetrix U133A 2.0 arrays. One gene, guanylate cyclase 1 alpha 3 (GUCY1A3), showed differential expression between the 1 week and maternally reared groups and the 1 week and 1 month groups; these changes were confirmed by in situ hybridization. The expression of this gene was positively correlated with acute social-comforting behavior (r = 0.923; p = 0.001) and longer-term close social behavior (r = 0.708; p = 0.015) and negatively correlated with self-comforting behaviors (r = -0.88; p < 0.001). Additional in situ hybridization studies of GUCY1A3 in normal monkeys showed that this gene is expressed at adult levels by 1 week of age and that its expression is greater in the amygdala than all other brain areas examined. We conclude that GUCY1A3 may contribute to the altered behavioral phenotypes that are differentially displayed depending on the age at which macaque infants experience an early-life stress.


Subject(s)
Amygdala/metabolism , Gene Expression Regulation, Developmental/physiology , Maternal Deprivation , Social Behavior , Age Factors , Animals , Animals, Newborn , Female , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Macaca mulatta , Maternal Behavior/physiology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
SELECTION OF CITATIONS
SEARCH DETAIL