Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
3.
Facial Plast Surg ; 33(1): 17-19, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28226375

ABSTRACT

To determine if facial plastic and reconstructive surgeons still adhere to the classic nasal subunit principle as described by Burget and Menick. Observational survey. A Weill Cornell Medicine institutional review board approved electronic survey that was sent via e-mail to active members of the American Academy of Facial Plastic and Reconstructive Surgery (AAFPRS). The survey consisted of 32 multiple-choice questions pertaining to the operative management of small (22-30%), medium (50-58%), and large (75-81%) defects of each subunit of the nose, as well as demographic, provider, and practice characteristics. There were 111 responses to the survey (10.1% response rate). Ninety-eight percent of respondents reported familiarity with the subunit principle, and 59.6% considered the subunit principle in greater than 90% of cases. Almost three-quarters (70.4%) of respondents felt the subunit principle should be applied but could be modified based on the particular nasal defect, whereas 28.7% felt it was only sometimes helpful and was not mandatory for successful nasal reconstruction. Large defects of the tip and ala are generally treated by excision of the remaining subunit (79.4 and 80.6%, respectively). Fewer surgeons would excise the remaining subunit for large defects of the dorsum (39.8%), sidewall (38.8%), and soft tissue facet (18.4%). Simple repair without additional excision was the treatment of choice for small defects of the tip (58.2%), ala (59.2%), sidewall (65%), dorsum (68%), and soft tissue facet (71.8%). However, in many small- (up to 32%) and medium- (up to 51%) sized defects of the tip, ala, sidewall, and dorsum, respondents reported partial subunit excision. The majority of AAFPRS members abide to the classical subunit principle by completely excising the remaining subunit for large defects of the tip and ala. Many surgeons modify the subunit principle in small and medium defects.


Subject(s)
Nose Deformities, Acquired/surgery , Practice Patterns, Physicians' , Rhinoplasty/methods , Surgery, Plastic/methods , Adult , Clinical Competence , Humans , Middle Aged , Rhinoplasty/trends , Surveys and Questionnaires
4.
Immunol Cell Biol ; 93(6): 567-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25753271

ABSTRACT

Cystic fibrosis (CF) is the most common life-limiting genetically acquired respiratory disorder. Patients with CF have thick mucus obstructing the airways leading to recurrent infections, bronchiectasis and neutrophilic airway inflammation culminating in deteriorating lung function. Current management targets airway infection and mucus clearance, but despite recent advances in care, life expectancy is still only 40 years. We investigated whether activin A is elevated in CF lung disease and whether inhibiting activin A with its natural antagonist follistatin retards lung disease progression. We measured serum activin A levels, lung function and nutritional status in CF patients. We studied the effect of activin A on CF lung pathogenesis by treating newborn CF transgenic mice (ß-ENaC) intranasally with the natural activin A antagonist follistatin. Activin A levels were elevated in the serum of adult CF patients, and correlated inversely with lung function and body mass index. Follistatin treatment of newborn ß-ENaC mice, noted for respiratory pathology mimicking human CF, decreased the airway activin A levels and key features of CF lung disease including mucus hypersecretion, airway neutrophilia and levels of mediators that regulate inflammation and chemotaxis. Follistatin treatment also increased body weight and survival of ß-ENaC mice, with no evidence of local or systemic toxicity. Our findings demonstrate that activin A levels are elevated in CF and provide proof-of-concept for the use of the activin A antagonist, follistatin, as a therapeutic in the long-term management of lung disease in CF patients.


Subject(s)
Activins/antagonists & inhibitors , Cystic Fibrosis/complications , Follistatin/metabolism , Pneumonia/etiology , Pneumonia/metabolism , Activins/blood , Adult , Animals , Body Weight/drug effects , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Disease Models, Animal , Female , Follistatin/pharmacology , Humans , Inflammation Mediators/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Mucus/metabolism , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/pathology , Pneumonia/drug therapy , Pneumonia/pathology , Pneumonia/physiopathology , Respiratory Function Tests , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Young Adult
6.
Cytokine ; 69(2): 255-62, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052838

ABSTRACT

Activin A, a member of the transforming growth factor-ß superfamily, is stimulated early in inflammation via the Toll-like receptor (TLR) 4 signalling pathway, which is also activated in myocardial ischaemia-reperfusion. Neutralising activin A by treatment with the activin-binding protein, follistatin, reduces inflammation and mortality in several disease models. This study assesses the regulation of activin A and follistatin in a murine myocardial ischaemia-reperfusion model and determines whether exogenous follistatin treatment is protective against injury. Myocardial activin A and follistatin protein levels were elevated following 30 min of ischaemia and 2h of reperfusion in wild-type mice. Activin A, but not follistatin, gene expression was also up-regulated. Serum activin A did not change significantly, but serum follistatin decreased. These responses to ischaemia-reperfusion were absent in TLR4(-/-) mice. Pre-treatment with follistatin significantly reduced ischaemia-reperfusion induced myocardial infarction. In mouse neonatal cardiomyocyte cultures, activin A exacerbated, while follistatin reduced, cellular injury after 3h of hypoxia and 2h of re-oxygenation. Neither activin A nor follistatin affected hypoxia-reoxygenation induced reactive oxygen species production by these cells. However, activin A reduced cardiomyocyte mitochondrial membrane potential, and follistatin treatment ameliorated the effect of hypoxia-reoxygenation on cardiomyocyte mitochondrial membrane potential. Taken together, these data indicate that myocardial ischaemia-reperfusion, through activation of TLR4 signalling, stimulates local production of activin A, which damages cardiomyocytes independently of increased reactive oxygen species. Blocking activin action by exogenous follistatin reduces this damage.


Subject(s)
Activins/metabolism , Follistatin/metabolism , Myocardial Reperfusion Injury/metabolism , Activins/blood , Activins/genetics , Animals , Animals, Newborn , Cells, Cultured , Follistatin/genetics , Follistatin/pharmacology , Gene Expression Regulation/drug effects , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism
7.
Cytokine ; 61(1): 199-204, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23116663

ABSTRACT

Activin A, a transforming growth factor-ß family cytokine, plays a crucial role in regulating the onset and severity of many inflammatory conditions, such as acute lipopolysaccharide (LPS)-induced inflammation. Activin A is also implicated in type 2 diabetes (T2D), a disease characterised by insulin resistance, hyperglycaemia and chronic elevation of pro-inflammatory cytokines, including tumour necrosis factor (TNF-α). In the human, neutrophils contain activin A that can be released in response to TNF-α. Studies of inflammatory disease in vivo, however, generally use the mouse, so it is essential to know if murine neutrophils have similar properties. Regulation of activin A was investigated in bone marrow-derived neutrophil precursors (BMNPs) from 8 to 10 weeks old C57BL6/J male mice. The BMNPs contained 7-fold higher concentrations of activin A than bone marrow mononuclear cells. Release of activin A from isolated BMNPs was stimulated by TNF-α, but this was not due to increased activin A production. In contrast to TNF-α, LPS had no effect on isolated BMNPs, but stimulated activin A release and production in total bone marrow cell cultures. Moreover, activin A release in response to LPS, was not prevented in TNF-α null mice. Increased glucose and insulin had no effect on base-line activin A secretion by BMNPs in culture, but pre-treatment with insulin blocked the TNF-α induced release of activin A. These results indicate that murine neutrophils are a source of stored activin A, the release of which can be directly stimulated by TNF-α, although TNF-α is not the only stimulator of activin A release during inflammation. Furthermore, regulation of neutrophil activin A release by insulin may also play a role in the inflammation associated with T2D.


Subject(s)
Activins/metabolism , Bone Marrow Cells/metabolism , Insulin/pharmacology , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Glucose/pharmacology , Inflammation , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/genetics
8.
Biol Reprod ; 86(6): 171, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22423050

ABSTRACT

The current dogma is that the differential regulation of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesis and secretion is modulated by gonadotropin-releasing hormone (GnRH) pulse frequency and by changes in inhibins, activins, and follistatins both at the pituitary and at the peripheral level. To date no studies have looked at the overlapping function of these regulators in a combined setting. We tested the hypothesis that changes in GnRH pulse frequency alter the relative abundance of these regulators at the pituitary and peripheral levels in a manner consistent with changes in pituitary and circulating concentrations of FSH; that is, an increase in FSH will be accompanied by increased stimulatory input (activin) and/or reduced follistatin and inhibin. Ovariectomized ewes were subjected to a combination hypothalamic pituitary disconnection (HPD)-hypophyseal portal blood collection procedure. Hypophyseal portal and jugular blood samples were collected for a 6-h period from non-HPD ewes, HPD ewes, or HPD ewes administered GnRH hourly or every 3 h for 4 days. In the absence of endogenous hypothalamic and ovarian hormones that regulate gonadotropin secretion, 3-hourly pulses of GnRH increased pituitary content of FSH more than hourly GnRH, although these differences were not evident in the peripheral circulation. The results failed to support the hypothesis in that the preferential increase of pituitary content of FSH by the lower GnRH pulse frequency could be explained by changes in the pituitary content of inhibin A, follistatin, or activin B. Perhaps the effects of GnRH pulse frequency on FSH is due to changes in the balance of free versus bound amounts of these FSH regulatory proteins or to the involvement of other regulators not monitored in this study.


Subject(s)
Activins/blood , Follicle Stimulating Hormone/metabolism , Follistatin/blood , Gonadotropin-Releasing Hormone/metabolism , Inhibins/blood , Animals , Female , Follicle Stimulating Hormone/biosynthesis , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/blood , Hypothalamus/metabolism , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Pituitary Gland/metabolism , Sheep
9.
Am J Physiol Regul Integr Comp Physiol ; 303(6): R665-75, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22855279

ABSTRACT

Activin A, a member of the transforming growth factor-ß family, increases in the circulation within 1 h after administration of bacterial LPS. To clarify the origins of this rapid increase, the distribution of activin A and its binding protein, follistatin, and their production following LPS treatment, were assessed in adult male mice. In untreated mice, activin A was detectable in all 23 tissues examined, with highest mRNA expression (as measured by quantitative RT-PCR) was found in the liver, and the largest concentration of activin A protein (by ELISA) was found in the bone marrow. Likewise, follistatin mRNA and protein were present in all tissues, with highest expression in the vas deferens. Activin A and follistatin mRNA did not increase significantly in any tissue within the first hour after LPS, but activin A protein decreased by 35% in the bone marrow and increased 5-fold in the lung. No significant changes were observed in any other tissue. Activin A reached a peak in the circulation 1 h following LPS, and then declined. Cycloheximide, an inhibitor of protein translation, reduced this increase of activin A by more than 50%. Actinomycin D, an inhibitor of mRNA transcription, had no effect. Circulating follistatin did not increase until 4 h after LPS and was not affected by either inhibitor. These data indicate that the rapid increase in circulating activin A during LPS-induced inflammation is regulated at the posttranscriptional level, apparently from newly translated and stored protein, and implicate bone marrow-derived cells, and, in particular, neutrophils, as a significant source of this preformed activin A.


Subject(s)
Activins/metabolism , Follistatin/metabolism , Lipopolysaccharides/toxicity , Activins/blood , Activins/genetics , Animals , Dactinomycin/pharmacology , Follistatin/blood , Follistatin/genetics , Gene Expression Regulation/physiology , Male , Mice , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Immunol Cell Biol ; 89(8): 889-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21445090

ABSTRACT

Activin A, a member of the transforming growth factor-ß superfamily, is a critical early mediator of acute inflammation. Activin A release coincides with the release of tumour necrosis factor-α (TNF-α) in models of lipopolysaccharide (LPS)-induced inflammation. The source of circulating activin A during acute inflammation has not been identified and the potential contribution of leukocyte subsets was examined in the following study. Human leukocytes from healthy volunteers were fractionated using Ficoll gradients and cultured under serum-free conditions. Freshly isolated human neutrophils contained 20-fold more activin A than blood mononuclear cells as measured by enzyme-linked immunosorbent assay (ELISA), and both dimeric and monomeric forms of activin A were detected in these cells by western blotting. Activin A was predominantly immunolocalized in the neutrophil cytoplasm. Purified neutrophils secreted activin A in culture when stimulated by TNF-α, but were unable to respond to LPS directly. Although TNF-α stimulated activin A release from neutrophils within 1 h, activin subunit mRNA expression did not increase until 12 h of culture, and the amount of activin A released following TNF-α stimulation did not change between 1 and 12 h. Specific inhibition of the p38 MAP kinase signalling pathway blocked TNF-α-induced activin release, and the secretion of activin A was not due to TNF-α-induced neutrophil apoptosis. These data provide the first evidence that neutrophils are a significant source of mature, stored activin A. Stimulation of the release of neutrophil activin A by TNF-α may contribute to the early peak in circulating activin A levels during acute inflammation.


Subject(s)
Activins/metabolism , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Activins/blood , Apoptosis , Enzyme-Linked Immunosorbent Assay , Humans , Imidazoles/pharmacology , Inflammation/chemically induced , Inflammation Mediators , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/immunology , MAP Kinase Signaling System/drug effects , Neutrophils/cytology , Neutrophils/immunology , Pyridines/pharmacology , RNA, Messenger/biosynthesis , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
Biol Reprod ; 84(1): 87-96, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20739662

ABSTRACT

Prenatal testosterone excess in sheep leads to reproductive and metabolic disruptions that mimic those seen in women with polycystic ovary syndrome. Comparison of prenatal testosterone-treated sheep with prenatal dihydrotestosterone-treated sheep suggests facilitation of defects by androgenic as well as androgen-independent effects of testosterone. We hypothesized that the disruptive impact of prenatal testosterone on adult pathology may partially depend on its conversion to estrogen and consequent changes in maternal and fetal endocrine environments. Pregnant Suffolk sheep were administered either cottonseed oil (control) or testosterone propionate in cottonseed oil (100 mg, i.m. twice weekly), from Day 30 to Day 90 of gestation (term is ~147 d). Maternal (uterine) and fetal (umbilical) arterial samples were collected at Days 64-66, 87-90, and 139-140 (range; referred to as D65, D90, and D140, respectively) of gestation. Concentrations of gonadal and metabolic hormones, as well as differentiation factors, were measured using liquid chromatography/mass spectrometer, radioimmunoassay, or ELISA. Findings indicate that testosterone treatment produced maternal and fetal testosterone levels comparable to adult males and D65 control male fetuses, respectively. Testosterone treatment increased fetal estradiol and estrone levels during the treatment period in both sexes, supportive of placental aromatization of testosterone. These steroidal changes were followed by a reduction in maternal estradiol levels at term, a reduction in activin A availability, and induction of intrauterine growth restriction in D140 female fetuses. Overall, our findings provide the first direct evidence in support of the potential for both androgenic as well as estrogenic contribution in the development of adult reproductive and metabolic pathology in prenatal testosterone-treated sheep.


Subject(s)
Sheep/embryology , Testosterone Propionate/toxicity , Animals , Blood Glucose , Estrogens/blood , Estrogens/metabolism , Female , Gene Expression Regulation/physiology , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Leptin/blood , Male , Pregnancy , Prenatal Exposure Delayed Effects , Testosterone Propionate/blood , Testosterone Propionate/metabolism , Thyroxine/blood , Triiodothyronine/blood
12.
Cytokine ; 54(2): 154-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21320787

ABSTRACT

OBJECTIVE: Activin A, a member of transforming growth factor-ß superfamily, has been established as a critical cytokine released early in endotoxemia and other inflammatory syndromes. The release of activin A and its binding protein, follistatin during cardiopulmonary bypass (CPB) has not been previously reported. Our study aimed to define the pattern of activin A and follistatin release in a sheep CPB model. METHODS: Control group consisted of left thoractomy alone (n=6). CPB was performed using either unfractionated heparin (n=6) or lepirudin (n=6) as anticoagulant. Unlike heparin, lepirudin does not cause activin A and follistatin release on its own. Serum samples were assayed for activin A, follistatin, tumour necrosis factor-α and interleukin-6. RESULTS: Compared with the control group, CPB using lepirudin was associated with a biphasic release of activin A. The first peak occurred within the first hour of CPB and a second peak occurred within the early post-operative period, coincident with a large release of follistatin. Close correlation was found between follistatin and IL-6 in the control and lepirudin groups, indicative of a role for follistatin in the acute phase response. In contrast to the control and lepirudin groups, CPB using heparin resulted in a concurrent release of activin A and follistatin. CONCLUSIONS: CPB is a trigger for the release of biologically-active free activin A into the circulation, at levels considerably greater than that induced by surgery alone. Triggering release of this critical inflammatory cytokine suggests that activin A may contribute to the adverse outcomes associated with systemic inflammation in cardiac surgery.


Subject(s)
Activins/metabolism , Coronary Artery Bypass , Follistatin/metabolism , Models, Animal , Animals , Case-Control Studies , Interleukin-6/metabolism , Sheep , Tumor Necrosis Factor-alpha/metabolism
13.
Am J Respir Cell Mol Biol ; 42(6): 667-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19635933

ABSTRACT

Activin A is a member of the TGF-beta superfamily and plays a role in allergic inflammation and asthma pathogenesis. Recent evidence suggests that activin A regulates proinflammatory cytokine production and is regulated by inflammatory mediators. In a murine model of acute allergic airway inflammation, we observed previously that increased activin A concentrations in bronchoalveolar lavage (BAL) fluid coincide with Th2 cytokine production in lung-draining lymph nodes and pronounced mucus metaplasia in bronchial epithelium. We therefore hypothesized that IL-13, the key cytokine for mucus production, regulates activin A secretion into BAL fluid in experimental asthma. IL-13 increased BAL fluid activin A concentrations in naive mice and dose dependently induced activin A secretion from cultured human airway epithelium. A key role for IL-13 in the secretion of activin A into the BAL fluid during allergic airway inflammation was confirmed in IL-13-deficient mice. Eosinophils were not involved in this response because there was no difference in BAL fluid activin A concentrations between wild-type and eosinophil-deficient mice. Our data highlight an important role for IL-13 in the regulation of activin A intraepithelially and in BAL fluid in naive mice and during allergic airway inflammation. Given the immunomodulatory and fibrogenic effects of activin A, our findings suggest an important role for IL-13 regulation of activin A in asthma pathogenesis.


Subject(s)
Activins/metabolism , Asthma/metabolism , Epithelial Cells/metabolism , Interleukin-13/metabolism , Pneumonia/metabolism , Respiratory Mucosa/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors/metabolism , Animals , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Disease Models, Animal , Eosinophils/immunology , Eosinophils/metabolism , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Humans , Inhibin-beta Subunits/metabolism , Interleukin-13/administration & dosage , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-5/deficiency , Interleukin-5/genetics , Metaplasia , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin , Pneumonia/immunology , Pneumonia/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , Time Factors
14.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R760-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20071615

ABSTRACT

The impact of endotoxemia on cerebral endothelium and cerebral blood flow (CBF) regulation was studied in conscious newborn lambs. Bacterial endotoxin [LPS, 2 microg/kg iv] was infused on 3 consecutive days. Cerebrovascular function was assessed by monitoring CBF and cerebral vascular resistance (CVR) over 12 h each day and by the endothelium-dependent vasodilator bradykinin (BK) (n = 10). Inflammatory responses were assessed by plasma tumor necrosis factor-alpha (TNF-alpha, n = 5). Acutely, LPS disrupted the cerebral circulation within 1 h, with peak cerebral vasoconstriction at 3 h (CBF -28 and CVR +118%, P < 0.05) followed by recovery to baseline by 12 h. TNF-alpha and body temperature peaked approximately 1 h post-LPS. BK-induced vasodilatation (CVR -20%, P < 0.05) declined with each LPS infusion, was abolished after 3 days, and remained absent for at least the subsequent 5 days. Histological evidence of brain injury was found in four of five LPS-treated newborns. We conclude that endotoxin impairs cerebral perfusion in newborn lambs via two mechanisms: 1) acute vasoconstriction (over several hours); and 2) persistent endothelial dysfunction (over several days). Endotoxin-induced circulatory impairments may place the newborn brain at prolonged risk of CBF dysregulation and injury as a legacy of endotoxin exposure.


Subject(s)
Cerebrovascular Circulation/physiology , Cerebrovascular Disorders/chemically induced , Cerebrovascular Disorders/physiopathology , Endotoxemia/physiopathology , Lipopolysaccharides/toxicity , Acute Disease , Animals , Animals, Newborn , Body Temperature/drug effects , Body Temperature/physiology , Bradykinin/blood , Bradykinin/pharmacology , Brain/blood supply , Brain/pathology , Cerebrovascular Circulation/drug effects , Chronic Disease , Dose-Response Relationship, Drug , Drug Tolerance , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Endotoxemia/pathology , Macrophages/pathology , Nitrates/blood , Nitrites/blood , Sheep , Tumor Necrosis Factor-alpha/blood , Vascular Resistance/drug effects , Vascular Resistance/physiology , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/blood , Vasodilator Agents/pharmacology
15.
Clin Endocrinol (Oxf) ; 71(6): 867-73, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19486020

ABSTRACT

BACKGROUND AND OBJECTIVE: There are currently no sensitive and specific assays for activin B that could be utilized to study human biological fluids. The aim of this project was to develop and validate a 'total' activin B ELISA for use with human biological fluids and establish concentrations of activin B in the circulation and fluids from the reproductive organs. DESIGN: The new ELISA was validated and then used to measure activin B levels in the circulation of healthy participants, IVF patients, pregnant women and in ovarian follicular fluid and seminal plasma. PATIENTS AND MEASUREMENTS: Healthy adult subjects (n = 143), subjects from an IVF clinic (n = 27) and pregnancy groups (n = 29) were sampled. RESULTS: The sensitivity of the assay was 0.019 ng/ml. Validation of the activin B ELISA showed good recovery (90.7 +/- 9.8%) and linearity in biological fluid and cell culture media and low cross-reactivity with related analytes (inhibin B = 0.077% and activin A = 0.0034%). There was a negative correlation between activin B concentration (r = -0.281, P < 0.011) and females with increasing age. Patients attending IVF clinics had significantly lower levels of activin B compared with gender-matched control subjects. Ovarian follicular fluid and seminal plasma had 50-80 fold higher levels of activin B (mean = 5.35 and 3.66 ng/ml respectively) than sera (mean = 0.071 ng/ml). CONCLUSIONS: This fully validated ELISA for activin B offers a tremendous utility for measuring this protein in a variety of normal physiological processes and in various clinical pathologies.


Subject(s)
Activins/analysis , Adolescent , Adult , Aged , Enzyme-Linked Immunosorbent Assay , Female , Follicular Fluid/chemistry , Humans , Male , Middle Aged , Pregnancy , Semen/chemistry , Young Adult
16.
Physiol Rep ; 7(4): e13973, 2019 02.
Article in English | MEDLINE | ID: mdl-30785235

ABSTRACT

As the comparative pathophysiology of perinatal infection in the fetus and newborn is uncertain, this study contrasted the cerebral effects of endotoxemia in conscious fetal sheep and newborn lambs. Responses to intravenous bacterial endotoxin (lipopolysaccharide, LPS) or normal saline were studied on three consecutive days in fetal sheep (LPS 1 µg/kg, n = 5; normal saline n = 5) and newborn lambs (LPS 2 µg/kg, n = 10; normal saline n = 5). Cerebro-vascular function was assessed by monitoring cerebral blood flow (CBF) and cerebral vascular resistance (CVR) over 12 h each day, and inflammatory responses were assessed by plasma TNF alpha (TNF-α), nitrate and nitrite concentrations. Brain injury was quantified by counting both resting and active macrophages in the caudate nucleus and periventricular white matter (PVWM). An acute cerebral vasoconstriction (within 1 h of LPS injection) occurred in both the fetus (ΔCVR +53%) and newborn (ΔCVR +63%); subsequently prolonged cerebral vasodilatation occurred in the fetus (ΔCVR -33%) in association with double plasma nitrate/nitrite concentrations, but not in the newborn. Abundant infiltration of activated macrophages was observed in both CN and PVWM at each age, with the extent being 2-3 times greater in the fetus (P < 0.001). In conclusion, while the fetus and newborn experience a similar acute disruption of the cerebral circulation after LPS, the fetus suffers a more prolonged circulatory disruption, a greater infiltration of activated macrophages, and an exaggerated susceptibility to brain injury.


Subject(s)
Brain/embryology , Encephalitis/physiopathology , Fetal Diseases/physiopathology , Lipopolysaccharides/toxicity , Animals , Brain/growth & development , Brain/physiopathology , Cerebrovascular Circulation , Encephalitis/etiology , Female , Fetal Diseases/etiology , Macrophages/pathology , Male , Nitrates/blood , Nitrites/blood , Sheep , Tumor Necrosis Factor-alpha/blood , Vasoconstriction , Vasodilation
17.
Bioorg Med Chem Lett ; 18(14): 4146-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18539454

ABSTRACT

We describe a novel series of imidazopyridine substituted phenylalanines which are potent VLA-4 antagonists. A wide variety of substituents are tolerated as replacements for the pendant 3-pyridyl ring. A clear structure-activity relationship was identified around the substitution of the 3-amino-cyclobut-2-enone portion of the molecule.


Subject(s)
Chemistry, Pharmaceutical/methods , Integrin alpha4beta1/antagonists & inhibitors , Phenylalanine/chemistry , Pyridines/chemistry , Animals , Drug Design , Humans , Inhibitory Concentration 50 , Integrin alpha4beta1/blood , Mice , Models, Chemical , Molecular Conformation , Protein Binding , Rats , Structure-Activity Relationship
18.
Burns ; 34(1): 50-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17644256

ABSTRACT

Activin A is a member of the transforming growth factor-beta (TGF-beta) family of cytokines and growth factors and upregulation of this protein has been linked with a number of disease processes associated with chronic inflammation and fibrosis. Its potential involvement in burns has not yet been investigated. We therefore studied the localization of activin in tissue sections from excised mid- and deep dermal and full thickness cutaneous burn by immunohistochemistry. There was cell-specific temporal expression in tissues with prominent expression from day 4 onwards in lymphocytes and histiocytes and expression from day 8 onwards in reactive fibroblasts and endothelial cells. Immunopositivity over the first 18 days persisted in reactive fibroblasts and lymphocytes although the latter were in most circumstances decreasing in number. These data are consistent with activin A being central to the inflammatory and repair phases occurring in burnt skin and early scar formation. Modulation of activin expression and actions may, therefore, be a target for the management of burns.


Subject(s)
Activins/metabolism , Burns/metabolism , Fibroblasts/metabolism , Acute Disease , Burns/pathology , Dermatitis/metabolism , Dermatitis/pathology , Humans , Immunoenzyme Techniques , Lymphocytes/metabolism , Prospective Studies
19.
Cancer Res ; 66(11): 5686-95, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740706

ABSTRACT

The transforming growth factor-beta superfamily member activin and its antagonist, follistatin, act as a pleiotropic growth factor system that controls cell proliferation, differentiation, and apoptosis. Activin inhibits fibroblast growth factor 2-induced sprouting angiogenesis in vitro (spheroidal angiogenesis assay) and in vivo (Matrigel assay). To further study the role of the activin/follistatin system during angiogenesis and tumor progression, activin- and follistatin-expressing R30C mammary carcinoma cells were studied in mouse tumor experiments. Surprisingly, activin-expressing tumors grew much faster than follistatin-expressing tumors although they failed to induce increased angiogenesis (as evidenced by low microvessel density counts). Conversely, follistatin-expressing tumors were much smaller but had a dense network of small-diameter capillaries. Qualitative angioarchitectural analyses (mural cell recruitment, perfusion) revealed no major functional differences of the tumor neovasculature. Analysis of activin- and follistatin-expressing R30C cells identified a cell autonomous role of this system in controlling tumor cell growth. Whereas proliferation of R30C cells was not altered, follistatin-expressing R30C cells had an enhanced susceptibility to undergo apoptosis. These findings in experimental tumors are complemented by an intriguing case report of a human renal cell carcinoma that similarly shows a dissociation of angiogenesis and tumorigenesis during tumor progression. Collectively, the data shed further light into the dichotomous stimulating and inhibiting roles that the activin/follistatin system can exert during angiogenesis and tumor progression. Furthermore, the experiments provide a critical proof-of-principle example for the dissociation of angiogenesis and tumorigenesis, supporting the concept that tumor growth may not be dependent on increased angiogenesis as long as a minimal intratumoral microvessel density is maintained.


Subject(s)
Activins/biosynthesis , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/metabolism , Follistatin/biosynthesis , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Activins/genetics , Animals , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Renal Cell/pathology , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Follistatin/genetics , Humans , Kidney Neoplasms/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Transfection , Transplantation, Heterologous
20.
Mol Cell Endocrinol ; 253(1-2): 30-5, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16697104

ABSTRACT

Activin A and its binding protein, follistatin, are released into the circulation following acute systemic inflammation. In this study, we determined the activin and follistatin response of ovine aortic endothelial cells to lipopolysaccharide (LPS). Exposure to LPS for 1h, mimicking a transient inflammatory event, elicited significant increases in activin betaA subunit mRNA or activin A release, with larger, more prolonged increases evident with continuous exposure. On the other hand, follistatin increases were only evident with prolonged exposure to LPS and following increases in activin A release. While cell-associated activin A increased with LPS exposure, levels were lower than those secreted, whereas the opposite was apparent for follistatin. In summary, our findings suggest that vascular endothelial cells, while capable of releasing activin A and follistatin following inflammatory stimulation, are unlikely to be responsible for the rapid release of activin A in vivo following inflammatory challenge.


Subject(s)
Activins/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Follistatin/metabolism , Lipopolysaccharides/pharmacology , Animals , Cell Survival , Cells, Cultured , Sheep , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL