Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Kaohsiung J Med Sci ; 37(12): 1038-1047, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34558803

ABSTRACT

Coronary atherosclerotic heart disease (CAD) is a chronic disease caused by multiple risk factors. Aberrant expression of long non-coding RNAs (lncRNAs) has been regarded as an independent risk factor of CAD. This study evaluated lncRNA myocardial infarction-associated transcription (MIAT) expression in CAD patients and its clinical significance. Totally, 155 CAD patients and 76 non-CAD controls were enrolled. MIAT expression was detected using reverse transcription quantitative polymerase chain reaction. The clinical diagnostic significance of MIAT was evaluated by plotting the receiver operating characteristic (ROC) curve. The levels of inflammatory cytokines were detected using enzyme-linked immunosorbent assay. microRNA (miR)-29b-3p expression and pregnancy-associated plasma protein A (PAPPA) level were detected. MIAT expression in CAD patients (4.23 [1.22-6.50]) was higher than that in non-CAD controls (1.64 [0.05-2.93]) (p < 0.01) and had an independent correlation with CAD. The area under ROC curve of predicting CAD was calculated as 0.790, the specificity as 71.40%, and the sensitivity as 70.00%. MIAT expression was positively correlated with the C-reactive protein level (r = 0.769, p < 0.0001) and pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), and IL-8 levels, while negatively correlated with the anti-inflammatory cytokine IL-10. MIAT was positively correlated with Gensini score and had an independent correlation with it. LncRNA MIAT sponged miR-29b-3p and miR-29b-3p targeted PAPPA. In conclusion, lncRNA MIAT was upregulated in the peripheral blood of CAD patients and elicited clinical diagnostic significance. MIAT participated in the development of CAD via miR-29b-3p/PAPPA axis. This study provides insights into a potential target for the diagnosis and treatment of CAD.


Subject(s)
Coronary Artery Disease/etiology , Coronary Stenosis/etiology , Inflammation/etiology , RNA, Long Noncoding/physiology , Aged , C-Reactive Protein/analysis , Coronary Artery Disease/genetics , Coronary Stenosis/genetics , Female , Humans , Male , Middle Aged , Pregnancy-Associated Plasma Protein-A/analysis , Pregnancy-Associated Plasma Protein-A/physiology , RNA, Long Noncoding/blood , Up-Regulation
2.
Sci Transl Med ; 12(571)2020 11 25.
Article in English | MEDLINE | ID: mdl-33239385

ABSTRACT

Pregnancy is a physiological state of continuous adaptation to changing maternal and fetal nutritional needs, including a reduction of maternal insulin sensitivity allowing for appropriately enhanced glucose availability to the fetus. However, excessive insulin resistance in conjunction with insufficient insulin secretion results in gestational diabetes mellitus (GDM), greatly increasing the risk for pregnancy complications and predisposing both mothers and offspring to future metabolic disease. Here, we report a signaling pathway connecting pregnancy-associated plasma protein A (PAPPA) with adipose tissue expansion in pregnancy. Adipose tissue plays a central role in the regulation of insulin sensitivity, and we show that, in both mice and humans, pregnancy caused remodeling of adipose tissue evidenced by altered adipocyte size, vascularization, and in vitro expansion capacity. PAPPA is known to be a metalloprotease secreted by human placenta that modulates insulin-like growth factor (IGF) bioavailability through prolteolysis of IGF binding proteins (IGFBPs) 2, 4, and 5. We demonstrate that recombinant PAPPA can stimulate ex vivo human adipose tissue expansion in an IGFBP-5- and IGF-1-dependent manner. Moreover, mice lacking PAPPA displayed impaired adipose tissue remodeling, pregnancy-induced insulin resistance, and hepatic steatosis, recapitulating multiple aspects of human GDM. In a cohort of 6361 pregnant women, concentrations of circulating PAPPA are inversely correlated with glycemia and odds of developing GDM. These data identify PAPPA and the IGF signaling pathway as necessary for the regulation of maternal adipose tissue physiology and systemic glucose homeostasis, with consequences for long-term metabolic risk and potential for therapeutic use.


Subject(s)
Diabetes, Gestational , Insulin Resistance , Pregnancy-Associated Plasma Protein-A/physiology , Adipose Tissue , Animals , Blood Glucose , Female , Humans , Mice , Pregnancy , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/pharmacology
3.
Mol Cell Endocrinol ; 518: 110967, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32739295

ABSTRACT

Growth hormone (GH) and its mediator, insulin-like growth factor-1 (IGF-1), have long been recognized as central to human growth physiology. IGF-1 is known to complex with IGF binding proteins as well as with the acid labile subunit (ALS) in order to prolong its half-life in circulation. Factors regulating the bioavailability of IGF-1 (i.e. the balance between free and bound IGF-1) were less well understood. Recently, pregnancy-associated plasma protein-A2 (PAPP-A2) was discovered as a protease which specifically cleaves IGF-binding protein (IGFBP)-3 and -5. PAPP-A2 deficient patients present with characteristic findings including growth failure, elevated total IGF-1 and -2, IGFBPs, and ALS, but decreased percentage of free to total IGF-1. Additionally, patients with PAPP-A2 deficiency have impairments in glucose metabolism and bone mineral density (BMD). Treatment with recombinant human IGF-1 (rhIGF-1) improved height SD scores, growth velocity, body composition, and dysglycemia. Mouse models recapitulate many of the human findings of PAPP-A2 deficiency. This review summarizes the function of PAPP-A2 and its contribution to the GH-IGF axis through an examination of PAPP-A2 deficient patients and mouse models, thereby emphasizing the importance of the regulation of IGF-1 bioavailability in human growth.


Subject(s)
Genetic Diseases, Inborn/genetics , Growth and Development/genetics , Pregnancy-Associated Plasma Protein-A/genetics , Animals , Female , Genetic Diseases, Inborn/metabolism , Human Growth Hormone/metabolism , Human Growth Hormone/physiology , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mutation , Pregnancy-Associated Plasma Protein-A/physiology , Signal Transduction/genetics
4.
Cell Signal ; 73: 109698, 2020 09.
Article in English | MEDLINE | ID: mdl-32569826

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common genetic diseases implicated in the development of end stage renal disease (ESRD). Although FDA has recently approved a drug against ADPKD, there is still a great need for development of alternative management strategies for ADPKD. Understanding the different mechanisms that lead to cystogenesis and cyst expansion in ADPKD is imperative to develop new therapies against ADPKD. Recently, we demonstrated that caloric restriction can prevent the development of cystic disease in animal models of ADPKD and through these studies identified a new role for pregnancy associated plasma protein-A (PAPP-A), a component of the insulin-like growth factors (IGF) pathway, in the pathogenesis of this disease. The PAPP-A-IGF pathway plays an important role in regulation of cell growth, differentiation, and transformation and dysregulation of this pathway has been implicated in many diseases. Several indirect studies support the involvement of IGF-1 in the pathogenesis of ADPKD. However, it was only recently that we described a direct role for a component of this pathway in pathogenesis of ADPKD, opening a new avenue for the therapeutic approaches for this cystic disease. The present literature review will critically discuss the evidence that supports the role of components of IGF pathway in the pathogenesis of ADPKD and discuss the pharmacological implications of PAPP-A-IGF axis in this disease.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Polycystic Kidney, Autosomal Dominant , Pregnancy-Associated Plasma Protein-A/physiology , Animals , Cell Differentiation , Cell Proliferation , Humans , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology
5.
Circ J ; 83(3): 515-523, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30662023

ABSTRACT

BACKGROUND: Recent studies have suggested that pregnancy-associated plasma protein-A (PAPP-A) is involved in the pathogenesis of atherosclerosis. This study aim is to investigate the role and mechanisms of PAPP-A in reverse cholesterol transport (RCT) and inflammation during the development of atherosclerosis. Methods and Results: PAPP-A was silenced in apolipoprotein E (apoE-/-) mice with administration of PAPP-A shRNA. Oil Red O staining of the whole aorta root revealed that PAPP-A knockdown reduced lipid accumulation in aortas. Oil Red O, hematoxylin and eosin (HE) and Masson staining of aortic sinus further showed that PAPP-A knockdown alleviated the formation of atherosclerotic lesions. It was found that PAPP-A knockdown reduced the insulin-like growth factor 1 (IGF-1) levels and repressed the PI3K/Akt pathway in both aorta and peritoneal macrophages. The expression levels of LXRα, ABCA1, ABCG1, and SR-B1 were increased in the aorta and peritoneal macrophages from apoE-/-mice administered with PAPP-A shRNA. Furthermore, PAPP-A knockdown promoted RCT from macrophages to plasma, the liver, and feces in apoE-/-mice. In addition, PAPP-A knockdown elevated the expression and secretion of monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6), tumor necrosis factor-α, and interleukin-1ß through the nuclear factor kappa-B (NF-κB) pathway. CONCLUSIONS: The present study results suggest that PAPP-A promotes the development of atherosclerosis in apoE-/-mice through reducing RCT capacity and activating an inflammatory response.


Subject(s)
Atherosclerosis/etiology , Cholesterol/metabolism , Inflammation/etiology , Pregnancy-Associated Plasma Protein-A/physiology , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/pathology , Biological Transport , Female , Humans , Lipid Metabolism/drug effects , Macrophages/metabolism , Mice , Mice, Knockout, ApoE , NF-kappa B/metabolism , Pregnancy , Pregnancy-Associated Plasma Protein-A/pharmacology
6.
Growth Horm IGF Res ; 42-43: 1-7, 2018.
Article in English | MEDLINE | ID: mdl-30025286

ABSTRACT

Although implicated in cardiovascular disease, little is known about the fat surrounding the heart. In humans, epicardial fat is the visceral fat depot of the heart, which directly contacts the myocardium. This strategically placed fat depot is thought to produce bioactive molecules that could affect cardiac function. A major limitation in understanding the biology of epicardial fat is its restricted access in humans and its seeming absence in commonly-used experimental animal models. Although laboratory mice do not have epicardial fat per se, they do have a fat depot around the heart. In this study, we found that mouse pericardial fat has the molecular signature, small adipocyte size, and resistance to differentiation consistent with visceral fat. In addition, we show that mouse pericardial fat is regulated by pregnancy-associated plasma protein-A (PAPP-A), a key modulator of local insulin-like growth factor bioavailability. PAPP-A is highly expressed in mouse pericardial fat at levels equivalent to those in mesenteric visceral fat and 10-fold higher than in subcutaneous inguinal fat (P = .0003). Cultured pre-adipocytes isolated from pericardial fat show 2-fold increased PAPP-A secretion compared to pre-adipocytes isolated from inguinal fat. Furthermore, PAPP-A knock-out mice fed a high fat diet for 20 weeks have significantly reduced pericardial fat (by 60%; P < .0001) compared to wild-type littermates. There was no significant difference in inguinal fat between wild-type and PAPP-A knock-out mice. These data characterize a new mouse model of visceral-like pericardial fat and lay a foundation for understanding its role in human heart disease.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiology , Pericardium/physiology , Pregnancy-Associated Plasma Protein-A/physiology , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Cells, Cultured , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pericardium/cytology
7.
Endokrynol Pol ; 68(6): 682-691, 2017.
Article in English | MEDLINE | ID: mdl-29238946

ABSTRACT

Short stature is the main problem that paediatric endocrinologists have to grapple with. Endocrine disorders account for only 5% of patients with short stature, but this is still one of the most common causes of reports to the endocrine clinic and hospitalisation in the endocrine department. A properly functioning growth hormone/insulin-like growth factor (GH/IGF) axis is one of the most important factors in proper growth. A lot of genetic defects in this axis lead to syndromes marked by impaired growth, like Laron syndrome, muta-tions in the STAT5B, insulin-like growth factor 1 (IGF1), and insulin-like growth factor 1 receptor (IGF1R) and mutations in the acid labile subunit (ALS). Two proteases important for the proper functioning of the GH/IGF axis: pregnancy-associated plasma protein-A (PAPP-A) and pregnancy-associated plasma protein-A2 (PAPP-A2), have been described. The first description of the new syndrome of growth failure associated with mutation in the PAPP-A2 gene was given by Andrew Dauber et al. This review evaluates the current data concerning PAPP-A2 function, and particularly the effect of PAPP-A2 mutation on growth.


Subject(s)
Growth/genetics , Mutation , Pregnancy-Associated Plasma Protein-A/physiology , Animals , Female , Growth Hormone , Humans , Insulin-Like Growth Factor I , Male , Mice , Pregnancy-Associated Plasma Protein-A/metabolism
8.
J Endocrinol ; 231(1): 71-80, 2016 10.
Article in English | MEDLINE | ID: mdl-27519211

ABSTRACT

Insulin-like growth factors (IGFs) are implicated in the development of diabetic nephropathy (DN) and are shown to increase proliferation and extracellular matrix production in mesangial cells. The IGF system is complex and is composed of ligands, receptors, six binding proteins (IGF BPs) and a novel zinc metalloproteinase - pregnancy-associated plasma protein (PAPP)-A. PAPP-A increases the local bioavailability of IGF through the cleavage of IGF BP-4. Mesangial expansion is a major component of DN, and PAPP-A is shown to be increased in the glomeruli of patients with DN. Therefore, we determined the expression of PAPP-A and components of the IGF system in normal human mesangial cells (HMCs) and their regulation by factors known to be involved in DN. Under basal conditions, HMCs expressed PAPP-A, IGF1 receptor and all six IGF BPs. Interleukin (IL)-1ß was the most potent stimulus for PAPP-A expression (5-fold) followed by tumor necrosis factor (TNF)-α (2.5-fold). This PAPP-A was secreted, cell associated and proteolytically active. IL1ß also increased IGF BP-1expression (3-fold) with either reduction or no effect on other IGF BPs. Generally, TNF-α treatment decreased IGF BP expression. No treatment effect on PAPP-A or IGF BPs was seen with IL6, IGFs, advanced glycation end products or prolonged hyperglycemia. In addition, stimulation of HMCs with IGF1 alone or IGF1 complexed to wild-type, but not protease-resistant, IGF BP-4 led to increased [(3)H]-thymidine incorporation. In conclusion, these novel findings of PAPP-A and its regulation by proinflammatory cytokines, as well as the comprehensive analysis of the IGF system regulation in HMCs, suggest a mechanism by which inflammatory states such as DN can impact IGF activity in the kidney.


Subject(s)
Diabetic Nephropathies/metabolism , Glomerular Mesangium/metabolism , Pregnancy-Associated Plasma Protein-A/physiology , Animals , Cytokines/metabolism , Glycation End Products, Advanced/metabolism , Humans , Inflammation , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice , Receptor, IGF Type 1/metabolism
9.
Arch. cardiol. Méx ; 86(2): 148-156, abr.-jun. 2016. tab, graf
Article in Spanish | LILACS | ID: biblio-838364

ABSTRACT

Resumen La aterosclerosis es una enfermedad que involucra múltiples mecanismos fisiopatológicos cuyo conocimiento no se ha dilucidado por completo. Con frecuencia, los avances científicos sobre la fisiopatología aterogénica generan que a diversas moléculas no consideradas previamente en el panorama de dicha enfermedad se les atribuyan acciones sobre el inicio o progresión de la misma. Un ejemplo representativo es el estudio de un nuevo mecanismo involucrado en el proceso aterogénico, consistente en la asociación entre el sistema de factores de crecimiento similares a la insulina (IGF) y la proteína plasmática A asociada al embarazo (PAPP-A). El sistema IGF es una familia de péptidos compuesto por 3 hormonas peptídicas, 4 receptores transmembranales y 6 proteínas transportadoras. El factor de crecimiento similar a la insulina tipo 1 (IGF-1) es el principal ligando del sistema IGF involucrado en la aterosclerosis coronaria y ejerce sus efectos mediante la activación del receptor IGF-1R en células de músculo liso vascular de las arterias coronarias o en macrófagos de placas ateroscleróticas. En células de músculo liso vascular promueve la migración y previene la apoptosis aumentando la estabilidad de la placa, y en macrófagos disminuye el transporte reverso de colesterol propiciando la formación de células espumosas. La regulación de la biodisponibilidad de IGF-1 en el endotelio se lleva a cabo por las proteasas de proteínas IGFBP, principalmente por la PAPP-A. En la presente revisión se abordan los mecanismos involucrados entre el sistema IGF y la PAPP-A en aterosclerosis coronaria con énfasis en los efectos moleculares producidos en células de músculo liso vascular y en macrófagos.


Abstract Atherosclerosis is a condition that involves multiple pathophysiological mechanisms and whose knowledge has not been fully elucidated. Often, scientific advances on the atherogenic pathophysiology generate that molecules not previously considered in the scene of this disease, were attributed actions on the onset or progression of it. A representative example is the study of a new mechanism involved in the atherogenic process, consisting of the association between the insulin-like growth factor (IGF) system and pregnancy-associated plasma protein-A (PAPP-A). Insulin-like growth factor system is a family of peptides that include 3 peptide hormones, 4 transmembrane receptors and 6 binding proteins. Insulin-like growth factor-1 (IGF-1) is the main ligand of the IGF system involved in coronary atherosclerosis. IGF-1 exerts its effects via activation of the IGF-1R receptor on vascular smooth muscle cells or macrophages. In vascular smooth muscle cells promotes migration and prevents apoptosis which increases plaque stability while in macrophages reduces reverse cholesterol transport leading to the formation of foam cells. Regulation of IGF-1 endothelial bioavailability is carried out by IGFBP proteases, mainly by PAPP-A. In this review, we address the mechanisms between IGF system and PAPP-A in atherosclerosis with emphasis on molecular effects on vascular smooth muscle cells and macrophages.


Subject(s)
Humans , Animals , Pregnancy-Associated Plasma Protein-A/physiology , Coronary Artery Disease/etiology , Insulin-Like Growth Factor I/physiology
10.
Ann Endocrinol (Paris) ; 77(2): 90-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27155776

ABSTRACT

Firstly discovered as a placental protein present abundantly in the circulation of pregnant women, pregnancy-associated plasma protein-A (PAPP-A) is widely expressed in multiple tissues. PAPP-A is a metalloproteinase that is able to specifically cleave three insulin-like growth factor binding proteins (IGFBPs): IGFBP-2, -4 and -5. PAPP-A binds tightly to glycosaminoglycans present on the surface of cells, thus functioning within tissues as a growth-promoting enzyme, releasing bioactive IGF in close proximity to the IGF receptor. Pro-MBP and stanniocalcin-2 (STC2) appear to be the main inhibitors of PAPP-A activity, by forming a covalent complex with the protease. According to in vivo experiments, IGFBP-4 is believed to be the main PAPP-A substrate to regulate IGF bioavailability. The regulation of PAPP-A includes transcriptional control of its gene, competing reactions with other IGFBPs potentially sequestering IGF from IGFBP-4 and hence antagonizing PAPP-A-mediated IGF activation, and proteolytic inhibition of PAPP-A. Finally, PAPP-A may serve as a therapeutic target to indirectly inhibit IGF signalling in tissues where this is driven by increased PAPP-A activity. By taking advantage of the intricate interaction between PAPP-A and IGFBP-4, highly specific and selective inhibition of PAPP-A is possible.


Subject(s)
Pregnancy-Associated Plasma Protein-A/physiology , Somatomedins/physiology , Animals , Female , Humans , Pregnancy , Signal Transduction/physiology
11.
J Thromb Thrombolysis ; 42(2): 225-32, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27007282

ABSTRACT

Pregnancy-associated plasma protein-A (PAPP-A) is a metalloproteinase with a controversial role in pathophysiology of cardiovascular disease. It seems involved in progression of atherosclerosis and is widely represented in atherosclerotic plaque. PAPP-A plasma levels are elevated in patients with acute coronary syndromes (ACS), thus it has been suggested that it might be a prognostic marker for developing major cardiovascular events. However, the pathophysiological link(s) between PAPP-A and ACS are still unknown. Several studies have indicated that tissue factor (TF) plays a pivotal role in the pathophysiology of ACS by triggering the formation of intracoronary thrombi following endothelial injury. This study investigates whether PAPP-A, at concentrations measurable in ACS patients, might induce TF expression in human endothelial cells in culture (HUVEC). In HUVEC, PAPP-A induced TF-mRNA transcription as demonstrated by real time PCR and expression of functionally active TF as demonstrated by FACS analysis and pro-coagulant activity assay. PAPP-A induced TF expression through the activation of Akt/NF-κB axis, as demonstrated by luciferase assay and by suppression of TF-mRNA transcription as well as of TF expression/activity by Akt and NF-κB inhibitors. These data indicate that PAPP-A promotes TF expression in human endothelial cells and support the hypothesis that this proteinase, besides being involved in progression of atherosclerosis, does not represent an independent risk factor for adverse cardiovascular events, but it rather might play an "active" role in the pathophysiology of ACS as an effector molecule able to induce a pro-thrombotic phenotype in endothelial cells.


Subject(s)
Blood Coagulation , Pregnancy-Associated Plasma Protein-A/physiology , Thromboplastin/physiology , Acute Coronary Syndrome , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , NF-kappa B/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism
12.
Arch Cardiol Mex ; 86(2): 148-56, 2016.
Article in Spanish | MEDLINE | ID: mdl-26906607

ABSTRACT

Atherosclerosis is a condition that involves multiple pathophysiological mechanisms and whose knowledge has not been fully elucidated. Often, scientific advances on the atherogenic pathophysiology generate that molecules not previously considered in the scene of this disease, were attributed actions on the onset or progression of it. A representative example is the study of a new mechanism involved in the atherogenic process, consisting of the association between the insulin-like growth factor (IGF) system and pregnancy-associated plasma protein-A (PAPP-A). Insulin-like growth factor system is a family of peptides that include 3 peptide hormones, 4 transmembrane receptors and 6 binding proteins. Insulin-like growth factor-1 (IGF-1) is the main ligand of the IGF system involved in coronary atherosclerosis. IGF-1 exerts its effects via activation of the IGF-1R receptor on vascular smooth muscle cells or macrophages. In vascular smooth muscle cells promotes migration and prevents apoptosis which increases plaque stability while in macrophages reduces reverse cholesterol transport leading to the formation of foam cells. Regulation of IGF-1 endothelial bioavailability is carried out by IGFBP proteases, mainly by PAPP-A. In this review, we address the mechanisms between IGF system and PAPP-A in atherosclerosis with emphasis on molecular effects on vascular smooth muscle cells and macrophages.


Subject(s)
Coronary Artery Disease/etiology , Insulin-Like Growth Factor I/physiology , Pregnancy-Associated Plasma Protein-A/physiology , Animals , Humans
13.
Exp Gerontol ; 70: 131-4, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26325589

ABSTRACT

Obesity is on the rise in westernized countries, and visceral obesity in particular is associated with enhanced risk of developing metabolic disease and accelerated aging. Various dietary restriction regimens have been shown to extend healthy lifespan in a variety of species. However, identification of alternative approaches that could be more acceptable to humans is actively being pursued. We have shown previously that mice deficient in pregnancy-associated plasma protein-A (PAPP-A) have an extended healthy lifespan on a regular chow diet. In this study, we determined the lifespan of PAPP-A knock-out (KO) and wild-type (WT) littermates fed a high fat diet (HFD) starting at 12 months of age. PAPP-A KO and WT mice had equivalent weight gain as measured over 25 weeks on HFD. However, PAPP-A KO mice on HFD had a significant increase in lifespan (P=0.018). Body composition and tissue pathology were assessed in a separate cohort of mice after 30 weeks on HFD. Percent body fat was equivalent in the two groups. However, there was a decrease in visceral fat depot weights and an increase in serum adiponectin levels in PAPP-A KO compared to WT mice. Major pathological differences were seen in kidney, heart and testes, with PAPP-A KO mice having little, if any, evidence of inflammation, mineralization, or degeneration in these tissues compared to WT mice. Thus, PAPP-A is a novel drug target with the potential to promote healthy longevity without a need for dietary restriction.


Subject(s)
Diet, High-Fat/adverse effects , Pregnancy-Associated Plasma Protein-A/deficiency , Adipose Tissue/pathology , Aging/pathology , Animals , Body Composition/physiology , Disease Models, Animal , Heart Diseases/etiology , Heart Diseases/pathology , Intra-Abdominal Fat/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Longevity/physiology , Male , Mice, Knockout , Organ Size/physiology , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/physiology , Survival Analysis , Testicular Diseases/etiology , Testicular Diseases/pathology , Weight Gain/physiology
14.
Growth Horm IGF Res ; 25(6): 274-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26385171

ABSTRACT

OBJECTIVE: Pregnancy associated plasma protein-A2 (PAPP-A2) is a protease that cleaves insulin-like growth factor binding protein-5 (IGFBP-5), the most abundant IGFBP in bone. Deletion of Pappa2 reduces postnatal growth and bone length in mice. The aim of this study was to determine whether locally produced PAPP-A2 is required for normal bone growth. DESIGN: We deleted Pappa2 primarily in osteoblasts by crossing conditional Pappa2 deletion mice with mice expressing Cre recombinase under the control of the Sp7 (Osterix) promoter. Effects of disrupting Pappa2 in Sp7-expressing cells were examined by measuring body mass and tail length at 3, 6, 10 and 12 weeks of age and bone dimensions at 12 weeks. RESULTS: Body mass, tail length, and linear bone dimensions were significantly reduced at all ages by osteoblast-specific Pappa2 deletion. Mice homozygous for the conditional Pappa2 deletion allele and carrying the Cre transgene were smaller than controls carrying the Cre transgene, whereas mice homozygous for the conditional Pappa2 deletion allele were not smaller than controls when comparing mice not carrying the transgene. This result unambiguously demonstrates that PAPP-A2 produced by Sp7 expressing cells is required for normal growth. However, constitutive Pappa2 deletion had greater effects than osteoblast-specific Pappa2 deletion for many traits, indicating that post-natal growth is also affected by other sources of PAPP-A2. Immunohistochemistry revealed that PAPP-A2 localized in the epiphysis and metaphysis as well as osteoblasts, consistent with a role in bone growth. CONCLUSION: Locally-produced PAPP-A2 is required for normal bone growth.


Subject(s)
Bone Development/genetics , Bone and Bones/metabolism , Carrier Proteins/metabolism , Osteoblasts/metabolism , Pregnancy-Associated Plasma Protein-A/genetics , Animals , Epiphyses , Female , Gene Knockout Techniques , Genotype , Immunohistochemistry , Male , Mice , Mice, Transgenic , Phenotype , Pregnancy-Associated Plasma Protein-A/physiology
15.
J Struct Biol ; 192(1): 59-66, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26306763

ABSTRACT

Pregnancy-associated plasma protein-A (PAPP-A) serves to increase local insulin-like growth factor (IGF) stimulation of proliferation and differentiation in many tissues through proteolysis of inhibitory IGF-binding proteins. The purpose of this study was to investigate the effects of PAPP-A on tendon structure and mechanical properties. A total of 30 tails from 6-month-old mice were tested with 10 tails in each of following groups: PAPP-A knockout (KO), skeletal-specific PAPP-A overexpressing transgenic (Tg) and wild type (WT). Morphologically, the total tail cross-sectional area (CSA), individual tissue CSAs of bone, muscle and tendon, and fascicle diameter were measured. A fascicle pullout test was performed to assess stiffness and strength of interfascicular structures. Fascicles were mechanically characterized through low and high displacement rate uniaxial tension tests providing modulus at each rate, hysteresis area and stress relaxation ratio. The KO mice had a smaller total tail CSA (p<0.05), fascicle diameter (p<0.05), absolute tendon CSA (p<0.05), fast and slow stiffness (p<0.05 for both) and larger hysteresis area (p<0.05) compared to WT and Tg mice. On the other hand, the Tg mice had a larger fascicle diameter (p<0.05), absolute tendon CSA (p<0.05), higher interfascicular strength and stiffness (p<0.05) and lower fascicular modulus at low displacement rates (p<0.05) compared to WT and KO mice. Tg mice also had larger total tail CSA area (p<0.05) and smaller hysteresis area (p<0.05) than KO mice, and larger normalized tendon CSA (p<0.05) than WT mice. Based on these data, we conclude that PAPP-A affects fascicle structure, thereby affecting tendon phenotype.


Subject(s)
Pregnancy-Associated Plasma Protein-A/physiology , Tendons/physiology , Animals , Biomechanical Phenomena , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout
16.
Reprod Domest Anim ; 50(4): 659-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031184

ABSTRACT

Cystic ovarian disease (COD) is one of the main causes of infertility in dairy cattle. It has been shown that intra-ovarian factors, such as members of the insulin-like growth factor (IGF) system, may contribute to follicular persistence. The bioavailability of IGF to initiate its response by binding to specific receptors (IGFRs) depends on interactions with related compounds, such as pregnancy-associated plasma protein A (PAPP-A). The aim of this study was to determine IGFR1 and PAPP-A expression both in follicles at different stages of development and in cysts, to evaluate the roles in the etiopathogenesis of COD in cattle. The mRNA expression of PAPP-A was higher in granulosa cells of large tertiary follicles than in cysts, whereas the protein PAPP-A present in the follicular fluid from these follicles showed no differences. Although no PAPP-A mRNA expression was detected in smaller tertiary follicles, in their follicular fluid, this protease was detected in lesser concentration than in cysts. The mRNA expression of IGFR1 was lower in granulosa cells from cystic follicles than in those from tertiary ones. However, the protein expression of this receptor presented the highest levels in cystic structures, probably to increase the possibility of IGF response. The data obtained would indicate that animals with COD have an altered regulation of the IGF system in the ovary, which could be involved in the pathogenesis of this disease in cattle.


Subject(s)
Cattle Diseases/physiopathology , Ovarian Cysts/veterinary , Pregnancy-Associated Plasma Protein-A/physiology , Receptor, IGF Type 1/physiology , Animals , Cattle , Cattle Diseases/etiology , Female , Follicular Fluid/chemistry , Gene Expression , Granulosa Cells/chemistry , Immunohistochemistry , Ovarian Cysts/chemistry , Ovarian Cysts/physiopathology , Ovarian Follicle/chemistry , Pregnancy , Pregnancy-Associated Plasma Protein-A/analysis , Pregnancy-Associated Plasma Protein-A/genetics , RNA, Messenger/analysis , Receptor, IGF Type 1/analysis , Receptor, IGF Type 1/genetics
17.
PLoS Comput Biol ; 11(5): e1004293, 2015 May.
Article in English | MEDLINE | ID: mdl-26020769

ABSTRACT

Inter-cellular communication with stromal cells is vital for cancer cells. Molecules involved in the communication are potential drug targets. To identify them systematically, we applied a systems level analysis that combined reverse network engineering with causal effect estimation. Using only observational transcriptome profiles we searched for paracrine factors sending messages from activated hepatic stellate cells (HSC) to hepatocellular carcinoma (HCC) cells. We condensed these messages to predict ten proteins that, acting in concert, cause the majority of the gene expression changes observed in HCC cells. Among the 10 paracrine factors were both known and unknown cancer promoting stromal factors, the former including Placental Growth Factor (PGF) and Periostin (POSTN), while Pregnancy-Associated Plasma Protein A (PAPPA) was among the latter. Further support for the predicted effect of PAPPA on HCC cells came from both in vitro studies that showed PAPPA to contribute to the activation of NFκB signaling, and clinical data, which linked higher expression levels of PAPPA to advanced stage HCC. In summary, this study demonstrates the potential of causal modeling in combination with a condensation step borrowed from gene set analysis [Model-based Gene Set Analysis (MGSA)] in the identification of stromal signaling molecules influencing the cancer phenotype.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Pregnancy-Associated Plasma Protein-A/physiology , Cell Adhesion Molecules/metabolism , Cell Communication , Cell Line, Tumor , Computational Biology , Drug Design , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatic Stellate Cells/cytology , Humans , Liver/metabolism , Oligonucleotide Array Sequence Analysis , Placenta Growth Factor , Pregnancy Proteins/metabolism , Proteomics , Signal Transduction , Transcriptome
18.
J Gerontol A Biol Sci Med Sci ; 70(3): 263-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24691093

ABSTRACT

Work in yeast has shown that longevity extension induced by nutrient deprivation, altered ribosomal function, or diminished target of rapamycin action requires the activity of GCN4. We hypothesized that increased activity of ATF4, the mammalian equivalent of yeast GCN4, might be characteristic of mutations that extend mouse life span. Fibroblasts from the skin of two such mutants (Snell dwarf and PAPP-A knockout) were found to have higher levels of ATF4 protein and expression of several ATF4 target genes in responses to amino acid withdrawal, cadmium, hydrogen peroxide, and tunicamycin. ATF4 pathways were also elevated in liver of both kinds of long-lived mutant mice. Thus, a connection between ATF4 pathways and longevity may have deep evolutionary roots, and further studies of ATF4 mechanisms may provide insights into the links between cellular stress resistance, protein translation control, and aging.


Subject(s)
Activating Transcription Factor 4/metabolism , Aging/physiology , Fibroblasts/physiology , Liver/metabolism , Skin/metabolism , Activating Transcription Factor 4/genetics , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Culture Techniques , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Liver/pathology , Mice , Mice, Knockout , Pregnancy-Associated Plasma Protein-A/physiology , RNA, Messenger/metabolism , Skin/pathology , Stress, Physiological/physiology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
19.
Growth Horm IGF Res ; 24(6): 251-5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25179152

ABSTRACT

UNLABELLED: Mice deficient in pregnancy-associated plasma protein-A (PAPP-A), an IGF binding protein protease, have been shown to be resistant to experimentally induced atherosclerosis and diabetic nephropathy, and, in the laboratory environment, live 30-40% longer than wild-type littermates in association with delayed incidence and occurrence of age-related neoplasms and degenerative diseases. OBJECTIVE: PAPP-A is highly expressed in the cerebellum and hippocampus of the mouse brain. Therefore, the studies presented here were aimed at determining motor behavior, learning and retention in PAPP-A knock-out (KO) mice compared to wild-type (WT) littermates with age. DESIGN: Balance and coordination were assessed using an accelerating rotarod; learning and memory were assessed in a Stone T-maze. RESULTS: Time on the rotarod decreased with age but there was no significant difference between PAPP-A KO and WT mice at any of the testing ages. Latency to reach the goal box and number of errors committed in the Stone T-maze did not change with age and there were no significant differences between PAPP-A KO and WT mice. CONCLUSION: Lack of PAPP-A in mice did not impact central regulation of coordination, learning or memory.


Subject(s)
Longevity/physiology , Maze Learning , Memory/physiology , Motor Neurons/physiology , Pregnancy-Associated Plasma Protein-A/physiology , Rotarod Performance Test , Animals , Female , Male , Mice , Mice, Knockout , Pregnancy
20.
J Pathol ; 233(4): 344-56, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24931331

ABSTRACT

Aberrant mitosis is a common feature of cancer, yet little is known about the altered genes causing mitotic defects. We screened human tumours for cells with morphological signatures of highly specific mitotic defects previously assigned to candidate genes in a genome-wide RNA interference screen carried out in HeLa cells (www.mitocheck.org). We discovered a striking enrichment of early mitotic configurations indicative of prophase/prometaphase delay in breast cancer. Promoter methylation analysis of MitoCheck candidate genes assigned to the corresponding 'mitotic delay' class linked this defect to epigenetic silencing of the gene encoding pregnancy-associated plasma protein-A (PAPPA), a secreted protease. PAPPA silencing was highly prevalent in precursor lesions and invasive breast cancer. Experimental manipulation of PAPPA protein levels in human mammary epithelial cells and in breast cancer cell lines demonstrates that progression through early mitosis is dependent on PAPPA function, and that breast cancer cells become more invasive after down-regulation of this protease. PAPPA regulates mitotic progression through modulating the IGF-1 signalling pathway resulting in activation of the forkhead transcription factor FoxM1, which drives a transcriptional cluster of essential mitotic genes. Our results show that PAPPA has a critical function in normal cell division and is targeted early in breast cancer development.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Epigenomics , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Mitosis/physiology , Pregnancy-Associated Plasma Protein-A/physiology , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Middle Aged , Phenotype , Pregnancy-Associated Plasma Protein-A/genetics , RNA Interference/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...