Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
PLoS One ; 14(9): e0218229, 2019.
Article in English | MEDLINE | ID: mdl-31509532

ABSTRACT

ASB20123, a C-type natriuretic peptide/ghrelin chimeric peptide, was designed as a novel peptide and demonstrated full agonistic activity for natriuretic-peptide receptor B and a significantly longer half-life in plasma compared with the native peptide. We researched the toxicological profile of ASB20123, the correlation between the morphological change of the epiphyseal plate and bone and cartilage toxicity, and biomarkers to detect the toxicity. ASB20123 was systemically administered to male and female rats at daily dose levels of 0.5, 1.5, and 5.0 mg/kg/day for 4 weeks. In this study, toxicity was observed as changes related to bone and cartilage tissues, and no other toxicological changes were observed in all animals. Next, ASB20123 was administered to 12-month-old rats with a little epiphyseal plate. The toxic changes related to bone and cartilage tissues were not observed in any animal with a closed epiphyseal plate, indicating that the toxic changes were triggered by the growth-accelerating effect on the bone and cartilage. Furthermore, we searched for the biomarker related to the bone and cartilage toxicity using rats treated with ASB20123 at doses of 0.005, 0.05, 0.5, and 5.0 mg/kg/day for 4 weeks. A close correlation between necrosis/fibrosis in the epiphysis and metaphysis and thickness of the epiphyseal plate in the femur was confirmed in this study. A decrease in the bone mineral density (BMD) of the femur also was associated with the appearance of bone toxicity. These results indicated that the toxicity of ASB20123 was limited to bone- and cartilage-specific changes, and these changes were triggered by an excessive growth accelerating effect. Furthermore, our data suggested that the thickness of the epiphyseal plate and BMD could be reliable biomarkers to predict bone toxicity.


Subject(s)
Bone Development/drug effects , Bone and Bones/drug effects , Cartilage/drug effects , Natriuretic Peptide, C-Type/pharmacology , Animals , Biomarkers , Bone Density/drug effects , Bone and Bones/metabolism , Cartilage/metabolism , Epiphyses/drug effects , Female , Growth Plate/drug effects , Male , Natriuretic Peptide, C-Type/adverse effects , Natriuretic Peptide, C-Type/analogs & derivatives , Rats
2.
PLoS One ; 14(5): e0216340, 2019.
Article in English | MEDLINE | ID: mdl-31120905

ABSTRACT

C-type natriuretic peptide (CNP)-knockout (KO) rats exhibit impaired skeletal growth, with long bones shorter than those in wild-type (WT) rats. This study compared craniofacial morphology in the CNP-KO rat with that in the Spontaneous Dwarf Rat (SDR), a growth hormone (GH)-deficient model. The effects of subcutaneous administration of human CNP with 53 amino acids (CNP-53) from 5 weeks of age for 4 weeks on craniofacial morphology in CNP-KO rats were also investigated. Skulls of CNP-KO rats at 9 weeks of age were longitudinally shorter and the foramen magnum was smaller than WT rats. There were no differences in foramen magnum stenosis and midface hypoplasia between CNP-KO rats at 9 and 33 weeks of age. These morphological features were the same as those observed in CNP-KO mice and activated fibroblast growth factor receptor 3 achondroplasia-phenotype mice. In contrast, SDR did not exhibit foramen magnum stenosis and midface hypoplasia, despite shorter stature than in control rats. After administration of exogenous CNP-53, the longitudinal skull length and foramen magnum size in CNP-KO rats were significantly greater, and full or partial rescue was confirmed. The synchondrosis at the cranial base in CNP-KO rats is closed at 9 weeks, but not at 4 weeks of age. In contrast, synchondrosis closure in CNP-KO rats treated with CNP-53 was incomplete at 9 weeks of age. Administration of exogenous CNP-53 accelerated craniofacial skeletogenesis, leading to improvement in craniofacial morphology. As these findings in CNP-KO rats are similar to those in patients with achondroplasia, treatment with CNP-53 or a CNP analog may be able to restore craniofacial morphology and foramen magnum size as well as short stature.


Subject(s)
Constriction, Pathologic , Face/abnormalities , Foramen Magnum/pathology , Natriuretic Peptide, C-Type/deficiency , Natriuretic Peptide, C-Type/therapeutic use , Achondroplasia/drug therapy , Animals , Bone Development , Humans , Rats , Time Factors
3.
PLoS One ; 14(2): e0212680, 2019.
Article in English | MEDLINE | ID: mdl-30794654

ABSTRACT

C-type natriuretic peptide (CNP) and its receptor natriuretic peptide receptor B (NPR-B) are physiological potent positive regulators of endochondral bone growth; therefore, the CNP/NPR-B signaling pathway is one of the most promising therapeutic targets for treating growth failure and dwarfism. In this article, we summarized the pharmacological properties of a novel CNP analog peptide ASB20123 as a therapeutic agent for short stature. ASB20123, one of the CNP/ghrelin chimeric peptides, is composed of CNP(1-22) and human ghrelin(12-28, E17D). Compared to CNP(1-22), ASB20123 showed similar agonist activity for NPR-B and improved biokinetics with a longer plasma half-life in rats. In addition, the distribution of ASB20123 to the cartilage was higher than that of CNP(1-22) after single subcutaneous (sc) injection to mice. These results suggested that the C-terminal part of ghrelin, which has clusters of basic amino acid residues and a BX7B motif, might contribute to the retention of ASB20123 in the extracellular matrix of the growth plate. Multiple sc doses of ASB20123 potently stimulated skeletal growth in rats in a dose-dependent manner, and sc infusion was more effective than bolus injection at the same dose. Our data indicated that high plasma levels of ASB20123 would not necessarily be required for bone growth acceleration. Thus, pharmaceutical formulation approaches for sustained-release dosage forms to allow chronic exposure to ASB20123 might be suitable to ensure drug effectiveness and safety.


Subject(s)
Bone Development/drug effects , Cartilage , Dwarfism , Growth Plate , Natriuretic Peptide, C-Type/pharmacology , Signal Transduction/drug effects , Animals , Cartilage/growth & development , Cartilage/pathology , Dwarfism/drug therapy , Dwarfism/metabolism , Dwarfism/pathology , Growth Plate/metabolism , Growth Plate/pathology , Mice , Mice, Inbred ICR , Natriuretic Peptide, C-Type/chemistry , Rats , Rats, Sprague-Dawley
4.
Hum Mol Genet ; 28(1): 74-83, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239744

ABSTRACT

Cardio-facio-cutaneous (CFC) syndrome, a genetic disorder caused by germline mutations in BRAF, KRAS, MAP2K1 and MAP2K2, is characterized by growth retardation, heart defects, dysmorphic facial appearance and dermatologic abnormalities. We have previously reported that knock-in mice expressing the CFC syndrome-associated mutation, Braf Q241R, showed growth retardation because of gastrointestinal dysfunction. However, other factors associated with growth retardation, including chondrogenesis and endocrinological profile, have not been examined. Here, we show that 3- and 4-week-old BrafQ241R/+ mice have decreased body weight and length, as well as reduced growth plate width in the proximal tibiae. Furthermore, proliferative and hypertrophic chondrocyte zones of the growth plate were reduced in BrafQ241R/+ mice compared with Braf+/+ mice. Immunohistological analysis revealed that extracellular signal-regulated kinase (ERK) activation was enhanced in hypertrophic chondrocytes in BrafQ241R/+ mice. In accordance with growth retardation and reduced growth plate width, decreased serum levels of insulin-like growth factor 1 (IGF-1) and IGF binding protein 3 (IGFBP-3) were observed in BrafQ241R/+ mice at 3 and 4 weeks of age. Treatment with C-type natriuretic peptide (CNP), a stimulator of endochondral bone growth and a potent inhibitor of the FGFR3-RAF1-MEK/ERK signaling, increased body and tail lengths in Braf+/+ and BrafQ241R/+ mice. In conclusion, ERK activation in chondrocytes and low serum IGF-1/IGFBP-3 levels could be associated with the growth retardation observed in BrafQ241R/+ mice. Our data also suggest that CNP is a potential therapeutic target in CFC syndrome.


Subject(s)
Ectodermal Dysplasia/metabolism , Failure to Thrive/metabolism , Heart Defects, Congenital/metabolism , Natriuretic Peptide, C-Type/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Animals , Chondrocytes/physiology , Disease Models, Animal , Ectodermal Dysplasia/physiopathology , Facies , Failure to Thrive/physiopathology , Germ-Line Mutation , Growth Disorders/metabolism , Heart Defects, Congenital/physiopathology , Humans , Insulin-Like Growth Factor I/analysis , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred ICR , Mutation , Natriuretic Peptide, C-Type/metabolism , Proto-Oncogene Proteins B-raf/physiology
5.
PLoS One ; 13(9): e0204172, 2018.
Article in English | MEDLINE | ID: mdl-30235256

ABSTRACT

Signaling by C-type natriuretic peptide (CNP) and its receptor, natriuretic peptide receptor-B, is a pivotal stimulator of endochondral bone growth. We recently developed CNP knockout (KO) rats that exhibit impaired skeletal growth with early growth plate closure. In the current study, we further characterized the phenotype and growth plate morphology in CNP-KO rats, and the effects of exogenous CNP in rats. We used CNP-53, an endogenous form of CNP consisting of 53 amino acids, and administered it for four weeks by continuous subcutaneous infusion at 0.15 or 0.5 mg/kg/day to four-week old CNP-KO and littermate wild type (WT) rats. We demonstrated that CNP-KO rats were useful as a reproducible animal model for skeletal dysplasia, due to their impairment in endochondral bone growth. There was no significant difference in plasma bone-turnover markers between the CNP-KO and WT rats. At eight weeks of age, growth plate closure was observed in the distal end of the tibia and the calcaneus of CNP-KO rats. Continuous subcutaneous infusion of CNP-53 significantly, and in a dose-dependent manner, stimulated skeletal growth in CNP-KO and WT rats, with CNP-KO rats being more sensitive to the treatment. CNP-53 also normalized the length of long bones and the growth plate thickness, and prevented growth plate closure in the CNP-KO rats. Using organ culture experiment of fetal rat tibia, gene set enrichment analysis indicated that CNP might have a negative influence on mitogen activated protein kinase signaling cascades in chondrocyte. Our results indicated that CNP-KO rats might be a valuable animal model for investigating growth plate physiology and the mechanism of growth plate closure, and that CNP-53, or its analog, may have the potential to promote growth and to prevent early growth plate closure in the short stature.


Subject(s)
Growth Plate/growth & development , Natriuretic Peptide, C-Type/deficiency , Natriuretic Peptide, C-Type/pharmacology , Animals , Biomarkers/blood , Body Weight/drug effects , Bone Remodeling , Female , Gene Knockout Techniques , Growth Plate/drug effects , Growth Plate/pathology , Humans , Hypertrophy , Ligands , MAP Kinase Signaling System/drug effects , Male , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Tibia/drug effects , Tibia/pathology
6.
PLoS One ; 13(3): e0194812, 2018.
Article in English | MEDLINE | ID: mdl-29566041

ABSTRACT

We have previously investigated the physiological role of C-type natriuretic peptide (CNP) on endochondral bone growth, mainly with mutant mouse models deficient in CNP, and reported that CNP is indispensable for physiological endochondral bone growth in mice. However, the survival rate of CNP knockout (KO) mice fell to as low as about 70% until 10 weeks after birth, and we could not sufficiently analyze the phenotype at the adult stage. Herein, we generated CNP KO rats by using zinc-finger nuclease-mediated genome editing technology. We established two lines of mutant rats completely deficient in CNP (CNP KO rats) that exhibited a phenotype identical to that observed in mice deficient in CNP, namely, a short stature with severely impaired endochondral bone growth. Histological analysis revealed that the width of the growth plate, especially that of the hypertrophic chondrocyte layer, was markedly lower and the proliferation of growth plate chondrocytes tended to be reduced in CNP KO rats. Notably, CNP KO rats did not have malocclusions and survived for over one year after birth. At 33 weeks of age, CNP KO rats persisted significantly shorter than wild-type rats, with closed growth plates of the femur in all samples, which were not observed in wild-type rats. Histologically, CNP deficiency affected only bones among all body tissues studied. Thus, CNP KO rats survive over one year, and exhibit a deficit in endochondral bone growth and growth retardation throughout life.


Subject(s)
Bone Diseases, Developmental/genetics , Natriuretic Peptide, C-Type/genetics , Animals , Bone Development/genetics , Bone Diseases, Developmental/mortality , Bone Diseases, Developmental/pathology , Dwarfism/genetics , Dwarfism/pathology , Female , Gene Deletion , Gene Knockout Techniques , Growth Plate/pathology , Osteogenesis/genetics , Rats , Rats, Inbred F344 , Rats, Transgenic
7.
Bioorg Med Chem ; 25(24): 6680-6694, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153628

ABSTRACT

Natriuretic peptide receptor A (NPR-A) agonists were evaluated in vivo by optimizing the structure of quinazoline derivatives to improve agonistic activity for rat NPR-A. A 1,4-Cis-aminocyclohexylurea moiety at 4-position and hydroxy group of d-alaninol at 2-position on the quinazoline ring were found to be important factors in improving rat NPR-A activity. We identified potent quinazoline and pyrido[2,3-d]pyrimidine derivatives against rat NPR-A, with double-digit nanomolar EC50 values. The in vivo results showed that compound 56b administered at 1.0 mg/kg/min significantly increased plasma cGMP concentration and urine volume in rats. We discovered novel potent NPR-A agonists that showed agonistic effects similar to those of atrial natriuretic peptide.


Subject(s)
Drug Discovery , Quinazolines/pharmacology , Receptors, Atrial Natriuretic Factor/agonists , Animals , Dose-Response Relationship, Drug , Humans , Male , Molecular Structure , Quinazolines/chemical synthesis , Quinazolines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
8.
Peptides ; 97: 16-21, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28899838

ABSTRACT

C-type natriuretic peptide (CNP) and its receptor, natriuretic peptide receptor B (NPR-B), are potent positive regulators of endochondral bone growth, making the CNP pathway one of the most promising therapeutic targets for the treatment of growth failure. However, the administration of exogenous CNP is not fully effective, due to its rapid clearance in vivo. Modification of CNP to potentially druggable derivatives may result in increased resistance to proteolytic degradation, longer plasma half-life (T1/2), and better distribution to target tissues. In the present study, we designed and evaluated CNP/ghrelin chimeric peptides as novel CNP derivatives. We have previously reported that the ghrelin C-terminus increases peptide metabolic stability. Therefore, we combined the 17-membered, internal disulfide ring portion of CNP with the C-terminal portion of ghrelin. The resultant peptide displayed improved biokinetics compared to CNP, with increased metabolic stability and longer plasma T1/2. Repeated subcutaneous administration of the chimeric peptide to mice resulted in a significant acceleration in longitudinal growth, whereas CNP(1-22) did not. These results suggest that the ghrelin C-terminus improves the stability of CNP, and the chimeric peptide may be useful as a novel therapeutic agent for growth failure and short stature.


Subject(s)
Ghrelin , Natriuretic Agents , Natriuretic Peptide, C-Type , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Ghrelin/administration & dosage , Ghrelin/chemistry , Ghrelin/pharmacokinetics , Ghrelin/pharmacology , Injections, Subcutaneous , Male , Mice , Mice, Inbred ICR , Natriuretic Agents/administration & dosage , Natriuretic Agents/chemistry , Natriuretic Agents/pharmacokinetics , Natriuretic Agents/pharmacology , Natriuretic Peptide, C-Type/administration & dosage , Natriuretic Peptide, C-Type/analogs & derivatives , Natriuretic Peptide, C-Type/pharmacokinetics , Natriuretic Peptide, C-Type/pharmacology , Osteogenesis/drug effects , Protein Stability , Proteolysis , Rats , Rats, Sprague-Dawley
9.
Peptides ; 33(2): 279-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22286034

ABSTRACT

Ghrelin consists of 28 amino acid residues with an octanoyl modification at the third serine residue. Recently we have found that the C-terminal part of ghrelin protects the ester bond of 3-octanoyled serine from plasma esterases and plays the essential role to prolong the plasma half-life and to show its biological activity in vivo. In the present study, we researched whether the C-terminal part of ghrelin has a potential to prolong the plasma half-life of motilin, by comparing the pharmacokinetics of various chimeric peptides of ghrelin and motilin. Motilin is another gastro-intestinal peptide hormone related with ghrelin structurally, binding to the same family of G protein-coupled receptors. Chimeric peptides were designed to be composed of motilin(1-12) fragment, the active core binding to the motilin receptor, GPR38, and C-terminal part of ghrelin. The modification of motilin(1-12) fragment by C-terminal part of ghrelin hardly influenced its agonist activity to GPR38 and almost all these chimeric peptides showed more than two times longer plasma half-lives than motilin in rats. From the relationship between structures of chimeric peptides and their corresponding plasma half-lives, the mid-region of ghrelin rich in basic amino acids ((15)RKESKK(20)) was considered to be the most important in prolonging the plasma half-life of motilin. The deletion of these fragments or replacement of 17th glutamic acid with a neutral amino acid resulted in short plasma half-lives. In conclusion, our data suggested that the C-terminal part of ghrelin has a potential to improve the biokinetics of motilin probably by a metabolic stabilizing effect.


Subject(s)
Ghrelin/pharmacokinetics , Motilin/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Biotransformation , CHO Cells , Cricetinae , Ghrelin/chemistry , Ghrelin/pharmacology , Half-Life , Humans , Male , Molecular Sequence Data , Motilin/chemistry , Motilin/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Fragments/pharmacology , Protein Stability , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/agonists , Receptors, Neuropeptide/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
10.
Peptides ; 32(5): 1001-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21291937

ABSTRACT

Ghrelin is an endogenous ligand for growth hormone secretagogue receptor 1a (GHS-R1a), and consists of 28 amino acid residues with octanoyl modification at Ser(3). The previous studies have revealed that N-terminal part of ghrelin including modified Ser(3) is the active core for the activation of GHS-R1a. On the other hand, the role of C-terminal (8-28) region in ghrelin has not been clarified yet. In the present study, we prepared human ghrelin, C-terminal truncated ghrelin derivatives and anamorelin, a small molecular GHS compound which supposedly mimics the N-terminal active core, and examined GHS-R1a agonist activity in vitro, pharmacokinetic (PK) profile and growth hormone (GH) releasing activity in rats. All compounds demonstrated potent GHS-R1a agonist activities in vitro. Although the lack of C-terminal two amino acids did not modify PK profile and GH releasing activity, the deletion of C-terminal 8 and 20 amino acids affected them, and ghrelin(1-7)-Lys-NH(2) exhibited very short plasma half-life and low GH releasing activity in vivo. In rat plasma, ghrelin(1-7)-Lys-NH(2) was degraded more rapidly than ghrelin, suggesting that C-terminal part of ghrelin protected octanoylation of Ser(3) from plasma esterases. Subdiaphragmatic vagotomy significantly attenuated GH response to ghrelin but not to anamorelin. These results suggest that the C-terminal part of ghrelin has an important role in the biological activity in vivo. We also found that ghrelin stimulated GH release mainly via a vagal nerve pathway but anamorelin augmented GH release possibly by directly acting on brain in rats.


Subject(s)
Ghrelin/pharmacology , Ghrelin/pharmacokinetics , Animals , Calcium/metabolism , Drug Stability , Enzyme-Linked Immunosorbent Assay , Female , Growth Hormone/pharmacology , Half-Life , Humans , Liver/drug effects , Liver/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/agonists , Receptors, Ghrelin/metabolism , Vagotomy
SELECTION OF CITATIONS
SEARCH DETAIL