Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Stem Cell Res Ther ; 13(1): 305, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35841070

ABSTRACT

BACKGROUND: High glucose-induced damage to the osteogenic differentiation of human periodontal ligament stem cells (PDLSCs) has long been a challenge to periodontal regeneration for diabetic individuals. Metformin is an anti-hyperglycemic drug that exhibits abundant biological activities associated with cell metabolism and downstream tissue regeneration. However, how metformin combats damage to PDLSC osteogenic differentiation under high glucose and the underlying mechanisms remain unknown. METHODS: Osteogenic differentiation of PDLSCs was assessed by alkaline phosphatase (ALP) staining, ALP activity, Alizarin Red staining and quantitative assay, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis. RNA-seq analysis was performed to screen target genes of metformin, and the effects of target genes were confirmed using lentivirus transfection. Western blot analysis was also used to detect the protein level of underlying signaling pathways. RESULTS: We found that osteogenic differentiation of PDLSCs under high glucose was decreased, and metformin addition enhanced this capacity of differentiation. Furthermore, the results of RNA-seq analysis showed that natriuretic peptide receptor 3 (NPR3) was upregulated in PDLSCs under high glucose and downregulated after metformin addition. When the underlying pathways involved were investigated, we found that upregulation of NPR3 can compromise the metformin-enhanced PDLSC osteogenic differentiation and activate the MAPK pathway (especially the p38 MAPK and Erk1/2 pathway), and that inhibition of the NPR3-mediated p38 MAPK or Erk1/2 pathway enhanced the osteogenic differentiation of PDLSCs under high glucose. CONCLUSIONS: The present study suggests that metformin may enhance the osteogenic differentiation of PDLSCs under high glucose via downregulation of NPR3 and inhibition of its downstream MAPK pathway. This is the first report identifying the involvement of NPR3-mediated MAPK pathway in the metformin-enhanced osteogenic differentiation, indicating that NPR3 antagonists, such as metformin, may be feasible therapeutics for periodontal tissue regeneration in diabetic individuals.


Subject(s)
MAP Kinase Signaling System , Metformin , Periodontal Ligament , Receptors, Atrial Natriuretic Factor , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Glucose/administration & dosage , Glucose/metabolism , Humans , MAP Kinase Signaling System/drug effects , Metformin/pharmacology , Osteogenesis/drug effects , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/metabolism , Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Invest Dermatol ; 140(9): 1856-1866.e7, 2020 09.
Article in English | MEDLINE | ID: mdl-32032577

ABSTRACT

Recurrent and intractable chronic itch is a worldwide problem, but mechanisms, especially the neural mechanisms, underlying chronic itch still remain unclear. In this study, we investigated the peripheral and spinal mechanisms responsible for prolonged itch in a mouse model of allergic contact dermatitis induced by squaric acid dibutylester. We found that repeated exposure of mice to squaric acid dibutylester evoked persistent spontaneous scratching and significantly aberrant cutaneous and systemic immune responses lasting for weeks. Squaric acid dibutylester-induced itch requires both nonhistaminergic and histaminergic pathways, which are likely relayed by GRPR and NPRA in the spinal cord, respectively. Employing genetic, pharmacologic, RNAscope assay, and cell-specific ablation methods, we dissected a neural circuit for prolonged itch formed as Grpr+ neurons act downstream of Npr1+ neurons in the spinal cord. Taken together, our data suggested that targeting GRPR and NPRA may provide effective treatments for allergic contact dermatitis-associated chronic pruritus.


Subject(s)
Dermatitis, Allergic Contact/immunology , Pruritus/immunology , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Bombesin/metabolism , Animals , Antipruritics/pharmacology , Antipruritics/therapeutic use , Chronic Disease , Cyclobutanes/immunology , Dermatitis, Allergic Contact/complications , Dermatitis, Allergic Contact/drug therapy , Disease Models, Animal , Humans , Male , Mice , Neural Pathways/drug effects , Neural Pathways/immunology , Neurons/metabolism , Pruritus/drug therapy , Pruritus/pathology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Bombesin/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/immunology , Skin/innervation , Spinal Cord/cytology , Spinal Cord/metabolism
3.
Sci Transl Med ; 11(500)2019 07 10.
Article in English | MEDLINE | ID: mdl-31292265

ABSTRACT

There is a major clinical need for new therapies for the treatment of chronic itch. Many of the molecular components involved in itch neurotransmission are known, including the neuropeptide NPPB, a transmitter required for normal itch responses to multiple pruritogens in mice. Here, we investigated the potential for a novel strategy for the treatment of itch that involves the inhibition of the NPPB receptor NPR1 (natriuretic peptide receptor 1). Because there are no available effective human NPR1 (hNPR1) antagonists, we performed a high-throughput cell-based screen and identified 15 small-molecule hNPR1 inhibitors. Using in vitro assays, we demonstrated that these compounds specifically inhibit hNPR1 and murine NPR1 (mNPR1). In vivo, NPR1 antagonism attenuated behavioral responses to both acute itch- and chronic itch-challenged mice. Together, our results suggest that inhibiting NPR1 might be an effective strategy for treating acute and chronic itch.


Subject(s)
Ganglia, Spinal/metabolism , Pruritus/drug therapy , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Animals , Behavior, Animal , Cell-Free System , Dermatitis, Contact/drug therapy , Disease Models, Animal , Ganglia, Spinal/pathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pruritus/pathology , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
4.
Cell Physiol Biochem ; 48(6): 2337-2349, 2018.
Article in English | MEDLINE | ID: mdl-30114690

ABSTRACT

BACKGROUND/AIMS: Long noncoding RNAs (lncRNAs) constitute a large proportion of noncoding transcripts that have recently emerged as a new class of important regulators in cancers. LncRNA BCYRN1, also known as BC200, has a potential function in tumorigenesis. However, the clinical significance of BCYRN1 and its effect on colorectal cancer (CRC) progression remains unclear. METHODS: Quantitative reverse transcriptase PCR (qRT-PCR) was performed to investigate the expression of BCYRN1 in CRC tissues and cell lines. The biological function of BCYRN1 was also investigated through knockdown and overexpression of BCYRN1 in vitro. Microarray bioinformatics analysis was performed to analyze the putative targets of BCYRN1. RESULTS: The results showed that BCYRN1 expression was significantly upregulated in 96 CRC tumor tissues compared with para-carcinoma control tissues. Additionally, BCYRN1 overexpression was associated with larger tumor size and advanced pathological stages in CRC patients. In vitro BCYRN1 knockdown significantly inhibited the proliferation and apoptosis of CRC cells. Furthermore, NPR3 was identified to be a target of BCYRN1 and was downregulated by BCYRN1 knockdown. CONCLUSION: Together, we provide the first evidence that BCYRN1 plays an oncogenic role in CRC cells. BCYRN1 may be a promising prognostic biomarker and a potential therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms/pathology , RNA, Long Noncoding/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Female , G1 Phase Cell Cycle Checkpoints , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Transcriptome , Up-Regulation
5.
Biofactors ; 44(2): 95-108, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29336068

ABSTRACT

Isatin (indole-2,3-dione) is an oxidized indole. It is widely distributed in mammalian tissues and body fluids, where isatin concentrations vary significantly from <0.1 to > 10 µM. Isatin output is increased under conditions of stress. Exogenously administered isatin is characterized by low toxicity, mutagenicity, and genotoxicity in vivo. Cytotoxic effects of isatin on various cell cultures are usually observed at concentrations exceeding 100 µM. Binding of [3 H]isatin to rat brain sections is consistent with its physiological concentrations. Proteomic analysis of mouse and rat brain isatin-binding proteins revealed about 90 individual proteins, which demonstrated significant interspecies differences (rat versus mouse). Certain evidence exist that redox state(s) and possibly other types of posttranslational modifications regulate affinity of target proteins to isatin. Recent data suggest that interacting with numerous intracellular isatin binding proteins, isatin can act as a regulator of complex protein networks in norm and pathology. Physiological concentrations of isatin in vitro inhibit monoamine oxidase B and natriuretic peptide receptor guanylate cyclase, higher (neuroprotective) concentrations (50-400 µM) cause apoptosis of various (including malignant tumor) cell lines and influence expression of certain apoptosis-related genes. Being administered in vivo, isatin exhibits various behavioral effects; it attenuates manifestations of MPTP-induced parkinsonism and tumor growth in experimental animal models. © 2017 BioFactors, 44(2):95-108, 2018.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation/drug effects , Isatin/pharmacology , Neoplasms, Experimental/drug therapy , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line , Humans , Mice , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Rats , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Species Specificity
6.
Biochem Biophys Res Commun ; 491(2): 250-256, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28743500

ABSTRACT

Npr1 gene (coding for NPR-A) and Npr2 gene (coding for NPR-B) are identified as intrinsic anti-hypertrophic genes that opposes abnormal cardiac remodeling. However, the functional role of Npr1 and Npr2 genes during cardiac hypertrophic growth is not well understood. Hence, the present investigation was aimed to study the effect of Npr1 and Npr2 gene silencing, respectively on ß-AR activation induced cardiac hypertrophic growth in H9c2 cells in vitro. The control, Npr1, and Npr2 gene suppressed H9c2 cells, respectively were treated with ISO (10-5 M) for 48 h. The mRNA and protein expression profile of NPR-A, NPR-B, PKG-I and cGMP were analyzed by qPCR, Western blotting, ELISA, and immunofluorescence methods, respectively. A marked increase in cell size (30.10 ± 0.51 µm vs 61.83 ± 0.43 µm, 2-fold) accompanied by elevated hypertrophic marker genes (α-sk and ß-MHC 3-fold, respectively) expression was observed in Npr1 gene suppressed H9c2 cells as compared with control cells. In contrast, the Npr2 gene suppression in H9c2 cells neither altered the cell size nor the level of hypertrophic marker genes expression. Upon exposure to Isoproterenol, the Npr1 suppressed H9c2 cells exhibited further increase in cell size (1.5 fold), whereas, no significant increase in cell size or marker genes expression was noticed in Npr2 suppressed cells. Moreover, the intracellular cGMP level was down-regulated by 2-fold in Npr1 suppressed cells, while, no significant change was observed in Npr2 suppressed cells. Together, these results suggest that Npr1, not Npr2 gene function is positively associated with the initiation of cardiac fetal gene program and development of cardiac hypertrophic growth.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Atrial Natriuretic Factor/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Size/drug effects , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Gene Expression Regulation , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptors, Adrenergic, beta-1/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction
7.
Bioorg Med Chem Lett ; 27(15): 3542-3545, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28596054

ABSTRACT

The natriuretic peptide (NP) system is a critical endocrine, autocrine, and paracrine system and has been investigated for potential use against cardiovascular and metabolic diseases. The clearance of NPs is regulated by the proteolysis of neutral endopeptidase (NEP) and by endocytosis via natriuretic peptide receptor-3 (NPR3). A linear NPR3-selective peptide, [Cha8]-ANP(7-16)-NH2 (1), showed potent binding affinity for NPR3 but poor predicted chemical stability due to its free thiol group. A 12-mer peptide (9) without a thiol group was designed by the hybridization of two NPR3-binding peptides: a linear ANP fragment peptide analog and musclin, a murine member of the bHLH family of transcription factors, possessed high binding affinity and strict selectivity for NPR3. To increase the proteolytic resistance of 9, amino acid substitutions at the cleavage sites led to hydroxyacetyl-[d-Phe5,d-Hyp7,Cha8,d-Ser9,Hyp11,Arg(Me)14]-ANP(5-15)-NHCH3 (23), showing high and selective binding affinity for NPR3 over NPR1 and excellent stability in mouse serum. Compound 23 increased intracellular cGMP concentrations in primary cultured adipocytes, and continuous administration induced substantial plasma cGMP elevation in mice, suggesting its potential to clarify the physiological role of NPR3 and its therapeutic application.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Muscle Proteins/pharmacology , Peptide Fragments/pharmacology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Transcription Factors/pharmacology , Amino Acid Sequence , Animals , Atrial Natriuretic Factor/administration & dosage , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/chemistry , Mice , Mice, Inbred C57BL , Muscle Proteins/administration & dosage , Muscle Proteins/blood , Muscle Proteins/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/chemistry , Receptors, Atrial Natriuretic Factor/metabolism , Transcription Factors/administration & dosage , Transcription Factors/blood , Transcription Factors/chemistry
8.
J Biol Chem ; 292(24): 10220-10229, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28450398

ABSTRACT

Activating mutations in the receptor for C-type natriuretic peptide (CNP), guanylyl cyclase B (GC-B, also known as Npr2 or NPR-B), increase cellular cGMP and cause skeletal overgrowth, but how these mutations affect GTP catalysis is poorly understood. The A488P and R655C mutations were compared with the known mutation V883M. Neither mutation affected GC-B concentrations. The A488P mutation decreased the EC50 5-fold, increased Vmax 2.6-fold, and decreased the Km 13-fold, whereas the R655C mutation decreased the EC50 5-fold, increased the Vmax 2.1-fold, and decreased the Km 4.7-fold. Neither mutation affected maximum activity at saturating CNP concentrations. Activation by R655C did not require disulfide bond formation. Surprisingly, the A488P mutant only activated the receptor when it was phosphorylated. In contrast, the R655C mutation converted GC-B-7A from CNP-unresponsive to CNP-responsive. Interestingly, neither mutant was activated by ATP, and the Km and Hill coefficient of each mutant assayed in the absence of ATP were similar to those of wild-type GC-B assayed in the presence of ATP. Finally, 1 mm 2,4,6,-trinitrophenyl ATP inhibited all three mutants by as much as 80% but failed to inhibit WT-GC-B. We conclude that 1) the A488P and R655C missense mutations result in a GC-B conformation that mimics the allosterically activated conformation, 2) GC-B phosphorylation is required for CNP-dependent activation by the A488P mutation, 3) the R655C mutation abrogates the need for phosphorylation in receptor activation, and 4) an ATP analog selectively inhibits the GC-B mutants, indicating that a pharmacologic approach could reduce GC-B dependent human skeletal overgrowth.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Bone Diseases, Developmental/genetics , Enzyme Inhibitors/pharmacology , Models, Molecular , Mutation , Natriuretic Peptide, C-Type/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Allosteric Regulation , Amino Acid Substitution , Bone Diseases, Developmental/metabolism , Cyclic GMP/metabolism , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Kinetics , Mutagenesis, Site-Directed , Mutation, Missense , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Receptors, Atrial Natriuretic Factor/chemistry , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
9.
Free Radic Biol Med ; 99: 593-607, 2016 10.
Article in English | MEDLINE | ID: mdl-27634171

ABSTRACT

Natriuretic peptide receptor A (NPRA), the major receptor for atrial natriuretic peptide (ANP), has been implicated in tumorigenesis; however, the role of ANP-NPRA signaling in the development of gastric cancer remains unclear. Immunohistochemical analyses indicated that NPRA expression was positively associated with gastric tumor size and cancer stage. NPRA inhibition by shRNA induced G2/M cell cycle arrest, cell death, and autophagy in gastric cancer cells, due to accumulation of reactive oxygen species (ROS). Either genetic or pharmacologic inhibition of autophagy led to caspase-dependent cell death. Therefore, autophagy induced by NPRA silencing may represent a cytoprotective mechanism. ROS accumulation activated c-Jun N-terminal kinase (JNK) and AMP-activated protein kinase (AMPK). ROS-mediated activation of JNK inhibited cell proliferation by disturbing cell cycle and decreased cell viability. In addition, AMPK activation promoted autophagy in NPRA-downregulated cancer cells. Overall, our results indicate that the inhibition of NPRA suppresses gastric cancer development and targeting NPRA may represent a promising strategy for the treatment of gastric cancer.


Subject(s)
Autophagy/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Reactive Oxygen Species/agonists , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Anthracenes/pharmacology , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Nude , Neoplasm Staging , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Regul Integr Comp Physiol ; 311(1): R104-14, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27101299

ABSTRACT

Cardiac natriuretic peptides (NP) are involved in cardiorenal regulation and in lipolysis. The NP activity is largely dependent on the ratio between the signaling receptor NPRA and the clearance receptor NPRC. Lipolysis increases when NPRC is reduced by starving or very-low-calorie diet. On the contrary, insulin is an antilipolytic hormone that increases sodium retention, suggesting a possible functional link with NP. We examined the insulin-mediated regulation of NP receptors in differentiated human adipocytes and tested the association of NP receptor expression in visceral adipose tissue (VAT) with metabolic profiles of patients undergoing renal surgery. Differentiated human adipocytes from VAT and Simpson-Golabi-Behmel Syndrome (SGBS) adipocyte cell line were treated with insulin in the presence of high-glucose or low-glucose media to study NP receptors and insulin/glucose-regulated pathways. Fasting blood samples and VAT samples were taken from patients on the day of renal surgery. We observed a potent insulin-mediated and glucose-dependent upregulation of NPRC, through the phosphatidylinositol 3-kinase pathway, associated with lower lipolysis in differentiated adipocytes. No effect was observed on NPRA. Low-glucose medium, used to simulate in vivo starving conditions, hampered the insulin effect on NPRC through modulation of insulin/glucose-regulated pathways, allowing atrial natriuretic peptide to induce lipolysis and thermogenic genes. An expression ratio in favor of NPRC in adipose tissue was associated with higher fasting insulinemia, HOMA-IR, and atherogenic lipid levels. Insulin/glucose-dependent NPRC induction in adipocytes might be a key factor linking hyperinsulinemia, metabolic syndrome, and higher blood pressure by reducing NP effects on adipocytes.


Subject(s)
Adipocytes/metabolism , Glucose/pharmacology , Heart/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Natriuresis/drug effects , Natriuretic Peptides/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Adipocytes/drug effects , Aged , Cells, Cultured , Female , Humans , Insulin/blood , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Lipids/blood , Male , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
11.
J Pharmacol Exp Ther ; 356(3): 596-603, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26669425

ABSTRACT

B-type natriuretic peptide (BNP)-natriuretic peptide receptor A (NPRA) and gastrin-releasing peptide (GRP)-GRP receptor (GRPR) systems contribute to spinal processing of itch. However, pharmacological and anatomic evidence of these two spinal ligand-receptor systems are still not clear. The aim of this study was to determine the spinal functions of BNP-NPRA and GRP-GRPR systems for regulating scratching activities in mice by using pharmacological and immunohistochemical approaches. Our results showed that intrathecal administration of BNP (0.3-3 nmol) dose dependently elicited scratching responses, which could be blocked by the NPRA antagonist (Arg6,ß-cyclohexyl-Ala8,D-Tic16,Arg17,Cys18)-atrial natriuretic factor(6-18) amide (A71915). However, A71915 had no effect on intrathecal GRP-induced scratching. In contrast, pretreatment with a GRPR antagonist (D-Tpi6,Leu13ψ(CH2-NH)-Leu14)bombesin(6-14) (RC-3095) inhibited BNP-induced scratching. Immunostaining revealed that NPRA proteins colocalize with GRP, but not GRPR, in the superficial area of dorsal horn, whereas BNP proteins do not colocalize with either GRP or GRPR in the dorsal horn. Intradermal administration of ligands including endothelin-1, U-46619, bovine adrenal medulla 8-22, and Ser-Leu-Ile-Gly-Arg-Leu-NH2 (SLIGRL) increased scratching bouts at different levels of magnitude. Pretreatment with intrathecal A71915 did not affect scratching responses elicited by all four pruritogens, whereas pretreatment with RC-3095 only inhibited SLIGRL-induced scratching. Interestingly, immunostaining showed that RC-3095, but not A71915, inhibited SLIGRL-elicited c-Fos activation in the spinal dorsal horn, which was in line with behavioral outcomes. These findings demonstrate that: 1) BNP-NPRA system may function upstream of the GRP-GRPR system to regulate itch in the mouse spinal cord, and 2) both NPRA and GRPR antagonists may have antipruritic efficacy against centrally, but not peripherally, elicited itch.


Subject(s)
Gastrin-Releasing Peptide/physiology , Natriuretic Peptide, Brain/physiology , Pruritus/metabolism , Receptors, Atrial Natriuretic Factor/physiology , Receptors, Bombesin/physiology , Spinal Cord/metabolism , Animals , Atrial Natriuretic Factor/pharmacology , Atrial Natriuretic Factor/therapeutic use , Bombesin/analogs & derivatives , Bombesin/pharmacology , Bombesin/therapeutic use , Gastrin-Releasing Peptide/antagonists & inhibitors , Male , Mice , Natriuretic Peptide, Brain/antagonists & inhibitors , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Pruritus/drug therapy , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Bombesin/antagonists & inhibitors , Spinal Cord/drug effects , Tetrahydroisoquinolines/pharmacology , Tetrahydroisoquinolines/therapeutic use
12.
Am J Physiol Cell Physiol ; 308(7): C557-69, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25631869

ABSTRACT

The biological effects of atrial natriuretic peptide (ANP) are mediated by natriuretic peptide receptors (NPRs), which can either activate guanylyl cyclase (NPRA and NPRB) or inhibit adenylyl cyclase (NPRC) to modulate intracellular cGMP or cAMP, respectively. During cardiac development, ANP serves as an early maker of differentiating atrial and ventricular chamber myocardium. As development proceeds, expression of ANP persists in the atria but declines in the ventricles. Currently, it is not known whether ANP is secreted or the ANP-NPR signaling system plays any active role in the developing ventricles. Thus the primary aims of this study were to 1) examine biological activity of ANP signaling systems in embryonic ventricular myocardium, and 2) determine whether ANP signaling modulates proliferation/differentiation of undifferentiated cardiac progenitor cells (CPCs) and/or cardiomyocytes. Here, we provide evidence that ANP synthesized in embryonic day (E)11.5 ventricular myocytes is actively secreted and processed to its biologically active form. Notably, NPRA and NPRC were detected in E11.5 ventricles and exogenous ANP stimulated production of cGMP in ventricular cell cultures. Furthermore, we showed that exogenous ANP significantly decreased cell number and DNA synthesis of CPCs but not cardiomyocytes and this effect could be reversed by pretreatment with the NPRA receptor-specific inhibitor A71915. ANP treatment also led to a robust increase in nuclear p27 levels in CPCs compared with cardiomyocytes. Collectively, these data provide evidence that in the developing mammalian ventricles ANP plays a local paracrine role in regulating the balance between CPC proliferation and differentiation via NPRA/cGMP-mediated signaling pathways.


Subject(s)
Atrial Natriuretic Factor/biosynthesis , Cell Cycle/physiology , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/biosynthesis , Signal Transduction/physiology , Animals , Atrial Natriuretic Factor/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/drug effects , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Signal Transduction/drug effects
13.
World J Surg Oncol ; 12: 174, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24894887

ABSTRACT

The receptor for the cardiac hormone atrial natriuretic peptide (ANP), natriuretic peptide receptor A (NPR-A), has been reported to be expressed in lung cancer, prostate cancer and ovarian cancer. NPR-A expression and signaling is important for tumor growth; its deficiency protects C57BL/6 mice from lung, skin and ovarian cancers. This suggests that NPR-A is a new marker and a new target for cancer therapy. Recently, NPR-A has been demonstrated to be expressed in pre-implantation embryos and in embryonic stem cells, which has a novel role in the maintenance of self-renewal and pluripotency of embryonic stem cells. A nanoparticle-formulated interfering RNA for NPR-A attenuated B16 melanoma tumors in mice. Ectopic expression of a plasmid encoding NP73-102, the NH2-terminal peptide of the ANP prohormone which downregulates NPR-A expression, also suppressed lung metastasis of A549 cells in nude mice and tumorigenesis of Line 1 cells in immunocompetent BALB/c mice. These results suggest that NPR-A is involved in tumorigenesis and a new target for cancer therapy. This review focuses on structure, abnormal functions and carcinogenic mechanisms of NPR-A to investigate its role in tumorigenesis.


Subject(s)
Molecular Targeted Therapy , Neoplasms/prevention & control , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Animals , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , RNA, Small Interfering/genetics , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism
14.
Stem Cell Rev Rep ; 10(4): 561-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24798243

ABSTRACT

Embryonic stem cells (ESCs) have the ability to self-renew indefinitely and they can give unlimited source of cells and tissues for cellular therapies. Recently, the natriuretic peptide receptor A (NPR-A) has been recognized as an important regulator for the self-renewal of ESCs. To gain insights into possible novel mechanisms involved in NPR-A pathway that presumably regulates self-renewal and survival of ESCs, we utilized a comprehensive label-free proteomics technology in our study. Targeting of NPR-A gene with small interfering RNA (siRNA) resulted in the inhibition of ESCs self-renewal. Coherently, quantitative label-free shotgun proteomic analysis identified differentially expressed proteins involved in several biological processes, including cell cycle regulation, cell proliferation, cell fate specification, and apoptosis. Interestingly, in addition to Oct4 Nanog, and Sox2, other proteins involved in ESCs self-renewal were down-regulated after NPR-A knockdown, such as heterogeneous nuclear ribonucleoprotein A2/B1 (ROA2), non-POU domain-containing octamer-binding protein (Nono), nucleoplasmin (Npm1), histone H2A type 1-B/E (histone H2A.2), SW1/SNF complex (Brg1), polycomb protein Suz12 (Suz12), and cyclin-dependent kinase 4 (Cdk4). Furthermore, several protein candidates involved in early differentiation and cell death were up-regulated or down-regulated as a result of NPR-A knockdown, including importin subunit alpha-4 (Impα4), importin-5 (Ipo5), H3 histones, core histone macro-H2A.1 (H2A.y), apurine/apyrimidine endonuclease 1 (Apex1), 78-kDa glucose-regulated protein (Grp78), and programmed cell death 5 (Pdcd5). Overall, these findings depict a comprehensive view to our understanding of the pathways involved in the role of NPR-A in maintaining ESC functions.


Subject(s)
Biomarkers/analysis , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Proteomics/methods , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Gene Ontology , Gene Regulatory Networks , Immunoenzyme Techniques , Mice , Nucleophosmin , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Development ; 141(2): 335-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353062

ABSTRACT

Organ development is a highly regulated process involving the coordinated proliferation and differentiation of diverse cellular populations. The pathways regulating cell proliferation and their effects on organ growth are complex and for many organs incompletely understood. In all vertebrate species, the cardiac natriuretic peptides (ANP and BNP) are produced by cardiomyocytes in the developing heart. However, their role during cardiogenesis is not defined. Using the embryonic zebrafish and neonatal mammalian cardiomyocytes we explored the natriuretic peptide signaling network during myocardial development. We observed that the cardiac natriuretic peptides ANP and BNP and the guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2 are functionally redundant during early cardiovascular development. In addition, we demonstrate that low levels of the natriuretic peptides preferentially activate Npr3, a receptor with Gi activator sequences, and increase cardiomyocyte proliferation through inhibition of adenylate cyclase. Conversely, high concentrations of natriuretic peptides reduce cardiomyocyte proliferation through activation of the particulate guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2, and activation of protein kinase G. These data link the cardiac natriuretic peptides in a complex hierarchy modulating cardiomyocyte numbers during development through opposing effects on cardiomyocyte proliferation mediated through distinct cyclic nucleotide signaling pathways.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cell Proliferation , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Heart/embryology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
17.
Naunyn Schmiedebergs Arch Pharmacol ; 387(1): 5-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24297249

ABSTRACT

We found previously that stimulation of natriuretic peptide receptor (NPR)-B by C-type natriuretic peptide (CNP) in failing rat ventricle potentiates ß1-adrenoceptor (ß1-AR)-mediated inotropic response to noradrenaline through cGMP-mediated inhibition of phosphodiesterase (PDE) 3, thereby enhancing cAMP-mediated signalling. Increased cAMP-mediated signalling is deleterious in chronic heart failure (HF; basis for the use of ß-blockers in HF) and we propose to consider NPR-B antagonists as new HF treatment in addition to conventional therapy. Since there is no NPR-B-selective antagonist available for clinical studies, we aimed at identifying a novel small molecule (non-peptide) NPR-B antagonist. An assay was developed and high throughput screening performed on a chemical library of about 20,000 small molecule compounds (<500 Da) to identify NPR-B antagonists based on inhibition of CNP-stimulated cGMP production in NPR-B-expressing HEK293 cells. The screen revealed several potential NPR-B antagonists, of which six were selected for further studies. Three showed selective NPR-B vs NPR-A inhibition and three were partially selective. The compounds mediated reversible, noncompetitive inhibition and most likely act as allosteric modulators binding outside the agonist binding site of NPR-B. In rat ventricular muscle strips, the potentiating effect of CNP upon ß1-AR-evoked inotropic effects could be attenuated by at least one of these compounds. We identified several small molecule NPR-B antagonists by high throughput screening and show in a functional heart preparation that blocking NPR-B stimulation with a small molecule compound can reduce the potentiating effect of CNP on the ß1-AR-mediated inotropic response to noradrenaline.


Subject(s)
Heart Failure/diagnosis , Heart Failure/drug therapy , High-Throughput Screening Assays/statistics & numerical data , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Amino Acid Sequence , Animals , Drug Evaluation, Preclinical/methods , HEK293 Cells , Heart Failure/genetics , Heart Ventricles/drug effects , High-Throughput Screening Assays/methods , Humans , Molecular Sequence Data , Organ Culture Techniques , Rats , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism
18.
Endocrinology ; 154(9): 3401-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23787120

ABSTRACT

In preovulatory ovarian follicles, the oocyte is maintained in meiotic prophase arrest by natriuretic peptide precursor C (NPPC) and its receptor natriuretic peptide receptor 2 (NPR2). LH treatment results in the decrease of NPR2 guanylyl cyclase activity that promotes resumption of meiosis. We investigated the regulatory mechanism of LH-activated epidermal growth factor (EGF) receptor signaling on NPR2 function. Cumulus cell-oocyte complex is cultured in the medium with 30 nM NPPC to prevent oocyte spontaneous maturation. In this system, EGF could stimulate oocyte meiotic resumption after 4 hours of incubation. Further study showed that EGF elevated intracellular calcium concentrations of cumulus cells and decreased cGMP levels in cumulus cells and oocytes, and calcium-elevating reagents ionomycin and sphingosine-1-phosphate mimicked the effects of EGF on oocyte maturation and cGMP levels. EGF-mediated cGMP levels and meiotic resumption could be reversed by EGF receptor inhibitor AG1478 and the calcium chelator bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetra(acetoxymethyl)-ester. EGF also decreased the expression of Npr2 mRNA in cumulus cells, which may not be involved in meiotic resumption, because the block of NPR2 protein de novo synthesis by cycloheximide had no effect on NPPC and EGF-mediated oocyte maturation. However, EGF had no effect on oocyte maturation when meiotic arrest was maintained in the present of cGMP analog 8-bromoadenosine-cGMP. These results suggest that EGF receptor signaling induces meiotic resumption by elevating calcium concentrations of cumulus cells to decrease NPR2 guanylyl cyclase activity.


Subject(s)
Calcium Signaling , Cumulus Cells/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Oocytes/cytology , Oogenesis , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Animals, Outbred Strains , Calcium Ionophores/pharmacology , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Luteinizing Hormone/metabolism , Lysophospholipids/metabolism , Mice , Natriuretic Peptide, C-Type/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Precursors/metabolism , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tissue Culture Techniques
19.
Biosci Rep ; 33(3)2013 May 24.
Article in English | MEDLINE | ID: mdl-23586811

ABSTRACT

In this study, Tyr808 in GC-B (guanylate cyclase-B), a receptor of the CNP (C-type natriuretic peptide), has been shown to be a critical regulator of GC-B activity. In searching for phosphorylation sites that could account for suppression of GC-B activity by S1P (sphingosine-1-phosphate), mutations were introduced into several candidate serine/threonine and tyrosine residues. Although no novel phosphorylation sites that influenced the suppression of GC-B were identified, experiments revealed that mutations in Tyr808 markedly enhanced GC-B activity. CNP-stimulated activities of the Y808F and Y808A mutants were greater than 30-fold and 70-fold higher, respectively, than that of WT (wild-type) GC-B. The Y808E and Y808S mutants were constitutively active, expressing 270-fold higher activity without CNP stimulation than WT GC-B. Those mutations also influenced the sensitivity of GC-B to a variety of inhibitors, including S1P, Na3VO4 and PMA. Y808A, Y808E and Y808S mutations markedly weakened S1P- and Na3VO4-dependent suppression of GC-B activity, whereas Y808E and Y808S mutations rather elevated cGMP production. Tyr808 is conserved in all membrane-bound GCs and located in the niche domain showing sequence similarity to a partial fragment of the HNOBA (haem nitric oxide binding associated) domain, which is found in soluble GC and in bacterial haem-binding kinases. This finding provides new insight into the activation mechanism of GCs.


Subject(s)
Point Mutation , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Tyrosine/genetics , Enzyme Activation , HeLa Cells , Humans , Phosphorylation , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Tyrosine/metabolism
20.
Regul Pept ; 181: 9-16, 2013 Feb 10.
Article in English | MEDLINE | ID: mdl-23327998

ABSTRACT

In recent years, accumulating evidence suggests that atrial natriuretic peptide (ANP), a hormone widely known as a result of its significant effects on the cardiovascular system mediated by natriuretic peptide receptor A (NPRA), may play a nonnegligible role in the regulation of immune responses. In this study, we firstly investigated whether ANP signaling could regulate the differentiation and capacity of Th17 cells and discovered ANP-dose (10(-8)-10(-6)M) dependently indeed suppressed the differentiation of Th17 cells along with the reduced IL-17 production by polarizing naïve CD4(+) T cells isolated from splenocytes to Th17 phenotype in vitro. Moreover, ANP primarily signals through NPRA and cGMP-dependent protein kinase (PKG) which could be antagonized when pretreated with either ANP/NPRA signaling antagonist or PKG inhibitor. In addition, we also found that ANP signaling could upregulate the levels of phosphorylation of Akt which was hypothesized to be implicated in ANP-induced inhibition of Th17 development in our studies, and the effect of ANP on the development of murine Th17 cells seemed to be partially reversed when an inhibitor of phosphatidylinositol 3'-kinase (PI3K)/Akt had been performed in advance. Briefly, we showed for the first time that ANP signaling could suppress murine Th17 cell development from naïve CD4(+) T cells in vitro through NPRA/PKG pathway and the PI3K-Akt signal was implicated in the ANP-mediated suppression of Th17 development.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Th17 Cells/drug effects , Animals , Atrial Natriuretic Factor/immunology , Atrial Natriuretic Factor/pharmacology , Cell Differentiation , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/immunology , Gene Expression Regulation/drug effects , Interleukin-17/biosynthesis , Interleukin-17/immunology , Mice , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/immunology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/immunology , Signal Transduction/drug effects , Th17 Cells/cytology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL