transforming-growth-factor-beta has been researched along with Hemorrhage* in 20 studies
1 review(s) available for transforming-growth-factor-beta and Hemorrhage
Article | Year |
---|---|
Hereditary haemorrhagic telangiectasia: a clinical and scientific review.
The autosomal-dominant trait hereditary haemorrhagic telangiectasia (HHT) affects 1 in 5-8000 people. Genes mutated in HHT (most commonly for endoglin or activin receptor-like kinase (ALK1)) encode proteins that modulate transforming growth factor (TGF)-beta superfamily signalling in vascular endothelial cells; mutations lead to the development of fragile telangiectatic vessels and arteriovenous malformations. In this article, we review the underlying molecular, cellular and circulatory pathobiology; explore HHT clinical and genetic diagnostic strategies; present detailed considerations regarding screening for asymptomatic visceral involvement; and provide overviews of management strategies. Topics: Activin Receptors, Type II; Antigens, CD; Endoglin; Hemorrhage; Humans; Mutation; Receptors, Cell Surface; Signal Transduction; Telangiectasia, Hereditary Hemorrhagic; Transforming Growth Factor beta | 2009 |
19 other study(ies) available for transforming-growth-factor-beta and Hemorrhage
Article | Year |
---|---|
Model-informed approach for risk management of bleeding toxicities for bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1.
Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domain of the transforming growth factor-β (TGF-β) receptor II fused to a human immunoglobulin G1 antibody blocking programmed death ligand 1 (PD-L1). The recommended phase 2 dose (RP2D) was selected based on phase 1 efficacy, safety, and pharmacokinetic (PK)-pharmacodynamic data, assuming continuous inhibition of PD-L1 and TGF-β is required. Here, we describe a model-informed dose modification approach for risk management of BA-associated bleeding adverse events (AEs).. The PK and AE data from studies NCT02517398, NCT02699515, NCT03840915, and NCT04246489 (n = 936) were used. Logistic regression analyses were conducted to evaluate potential relationships between bleeding AEs and BA time-averaged concentration (C. The probability of bleeding AEs increased with increasing C. A pragmatic model-informed approach for management of bleeding AEs was implemented in ongoing clinical trials of BA. This approach is expected to improve benefit-risk profile; however, its effectiveness will need to be evaluated based on safety data generated after implementation. Topics: B7-H1 Antigen; Clinical Studies as Topic; Hemorrhage; Humans; Immunologic Factors; Neoplasms; Risk Management; Transforming Growth Factor beta | 2022 |
Accumulation of versican facilitates wound healing: Implication of its initial ADAMTS-cleavage site.
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan in the extracellular matrix, and is expressed at high levels in tissues during development and remodeling in pathological conditions. Its core protein is cleaved at a region close to the N-terminal end of CSβ domain by several members of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, i.e., ADAMTS-1, 4, 5, 9, 15, and 20. Here, using a CRISPR/Cas9 system, we generated knock-in mice (V1R), which express an ADAMTS cleavage-resistant versican. Some V1R homozygote mice, termed R/R, exhibit syndactyly and organ hemorrhage. In wound healing experiments, R/R wound shows accumulation of versican and activated TGFβ-signaling in the early stage, leading to faster healing than wild type wound. Immunostaining for Ki67, CD31, smooth muscle α-actin, periostin demonstrates higher levels of overall cell proliferation and an increased number of endothelial cells and myofibroblasts. Immunostaining for CD11b and qRT-PCR for macrophage markers revealed increased levels of inflammatory cell infiltration, especially those of M1 macrophages. Cultured R/R dermal fibroblasts revealed increased deposition of versican, type I and III collagens, and hyaluronan, and upregulation of Smad2/3 signaling. Taken together, these results demonstrate that the cleavage site determines versican turnover and that versican plays a central role in the provisional matrix during the wound repair. Topics: ADAMTS Proteins; Animals; Cell Proliferation; Cells, Cultured; CRISPR-Cas Systems; Extracellular Matrix; Gene Knock-In Techniques; Hemorrhage; Male; Mice; Signal Transduction; Syndactyly; Transforming Growth Factor beta; Versicans; Wound Healing | 2020 |
A Potent Pan-TGFβ Neutralizing Monoclonal Antibody Elicits Cardiovascular Toxicity in Mice and Cynomolgus Monkeys.
Transforming growth factor β (TGFβ) signaling has been recently shown to reduce antitumor response to PD-L1 blockade, leading to a renewed enthusiasm in developing anti-TGFβ therapies for potential combination with cancer immunotherapy agents. Inhibition of TGFβ signaling in nonclinical toxicology species is associated with serious adverse toxicities including cardiac valvulopathies and anemia. Previously, cardiovascular toxicities have been thought to be limited to small molecule inhibitors of TGFβ receptor and not considered to be a liability associated with pan-TGFβ neutralizing monoclonal antibodies (mAbs). Here, we report the toxicity findings associated with a potent pan-TGFβ neutralizing mAb (pan-TGFβ mAb; neutralizes TGFβ1, 2, and 3) after 5 weekly intravenous doses of 10, 30, and 100 mg/kg, followed by a 4-week recovery period, in mice and cynomolgus monkeys. Mortality was observed due to acute bleeding and cardiovascular toxicity in mice at ≥ 30 mg/kg and prolonged menstruation in female monkeys at 100 mg/kg. Additional findings considered to be on-target exaggerated pharmacology included generalized bleeding and cardiovascular toxicity in mice and monkeys; histopathologic changes in the teeth, tongue, and skin in mice; and abnormal wound healing and microscopic pathology in the bone in monkeys. Importantly, our data indicate that the cardiovascular toxicities associated with the inhibition of TGFβ signaling are not limited to small molecule inhibitors but are also observed following administration of a potent pan-TGFβ inhibiting mAb. Topics: Animals; Antibodies, Monoclonal, Humanized; Antibodies, Neutralizing; Cardiotoxicity; Cardiovascular Diseases; Cell Line; Female; Heart; Hemorrhage; Humans; Macaca fascicularis; Male; Mice; Myocardium; Risk Assessment; Time Factors; Toxicity Tests; Toxicokinetics; Transforming Growth Factor beta | 2020 |
Shp2 in myocytes is essential for cardiovascular and neointima development.
Mutations in the PTPN11 gene, which encodes the protein tyrosine phosphatase Shp2, cause Noonan syndrome and LEOPARD syndrome, inherited multifaceted diseases including cardiac and vascular defects. However, the function of Shp2 in blood vessels, especially in vascular smooth muscle cells (VSMCs), remains largely unknown. We generated mice in which Shp2 was specifically deleted in VSMCs and embryonic cardiomyocytes using the SM22α-Cre transgenic mouse line. Conditional Shp2 knockout resulted in massive hemorrhage, cardiovascular defects and embryonic lethality at the late embryonic developmental stage (embryonic date 16.5). The thinning of artery walls in Shp2-knockout embryos was due to decreased VSMC number and reduced extracellular matrix deposition. Myocyte proliferation was decreased in Shp2-knockout arteries and hearts. Importantly, cardiomyocyte-specific Shp2-knockout did not cause similar vascular defects. Shp2 was required for TGFβ1-induced expression of ECM components, including collagens in VSMCs. In addition, collagens were sufficient to promote Shp2-inefficient VSMC proliferation. Finally, Shp2 was deleted in adult mouse VSMCs by using SMMHC-CreER Topics: Animals; Carotid Arteries; Cell Proliferation; Collagen; Cyclin D1; Embryo, Mammalian; Extracellular Matrix; Female; Heart; Hemorrhage; Integrases; Male; Mice, Knockout; Muscle, Smooth, Vascular; Myocardium; Myocytes, Smooth Muscle; Neointima; Protein Tyrosine Phosphatase, Non-Receptor Type 11; Rats; Signal Transduction; Smad2 Protein; Transforming Growth Factor beta | 2019 |
A Brain-Region-Specific Neural Pathway Regulating Germinal Matrix Angiogenesis.
Intimate communication between neural and vascular cells is critical for normal brain development and function. Germinal matrix (GM), a key primordium for the brain reward circuitry, is unique among brain regions for its distinct pace of angiogenesis and selective vulnerability to hemorrhage during development. A major neonatal condition, GM hemorrhage can lead to cerebral palsy, hydrocephalus, and mental retardation. Here we identify a brain-region-specific neural progenitor-based signaling pathway dedicated to regulating GM vessel development. This pathway consists of cell-surface sphingosine-1-phosphate receptors, an intracellular cascade including Gα co-factor Ric8a and p38 MAPK, and target gene integrin β8, which in turn regulates vascular TGF-β signaling. These findings provide insights into region-specific specialization of neurovascular communication, with special implications for deciphering potent early-life endocrine, as well as potential gut microbiota impacts on brain reward circuitry. They also identify tissue-specific molecular targets for GM hemorrhage intervention. Topics: Brain; Embryo, Mammalian; Enzyme Activation; Fingolimod Hydrochloride; Guanine Nucleotide Exchange Factors; Hemorrhage; Humans; Integrin beta Chains; Lysophospholipids; Mutation; Neostriatum; Neovascularization, Physiologic; Neural Pathways; Neural Stem Cells; Organ Specificity; p38 Mitogen-Activated Protein Kinases; Phenotype; Receptors, Lysosphingolipid; Reward; Signal Transduction; Sphingosine; Transforming Growth Factor beta | 2017 |
Expression Profile of Cytokines and Enzymes mRNA in Blood Leukocytes of Dogs with Leptospirosis and Its Associated Pulmonary Hemorrhage Syndrome.
Dogs with leptospirosis show similar organ manifestations and disease course as human patients, including acute kidney injury and pulmonary hemorrhage, making this naturally-occurring infection a good animal model for human leptospirosis. Expression patterns of cytokines and enzymes have been correlated with disease manifestations and clinical outcome in humans and animals. The aim of this study was to describe mRNA expression of pro- and anti-inflammatory mediators in canine leptospirosis and to compare it with other renal diseases to identify patterns characterizing the disease and especially its pulmonary form.. The mRNA abundance of cytokines (IL-1α, IL-1β, IL-8, IL-10, TNF-α, TGF-β) and enzymes (5-LO, iNOS) was measured prospectively in blood leukocytes from 34 dogs with severe leptospirosis and acute kidney injury, including 22 dogs with leptospirosis-associated pulmonary hemorrhages. Dogs with leptospirosis were compared to 14 dogs with acute kidney injury of other origin than leptospirosis, 8 dogs with chronic kidney disease, and 10 healthy control dogs. Canine leptospirosis was characterized by high 5-LO and low TNF-α expression compared to other causes of acute kidney injury, although the decreased TNF-α expression was also seen in chronic kidney disease. Leptospirosis-associated pulmonary hemorrhage was not characterized by a specific pattern, with only mild changes noted, including increased IL-10 and decreased 5-LO expression on some days in affected dogs. Fatal outcome from pulmonary hemorrhages was associated with low TNF-α, high IL-1β, and high iNOS expression, a pattern possibly expressed also in dogs with other forms of acute kidney injury.. The patterns of cytokine and enzyme expression observed in the present study indicate a complex pro- and anti-inflammatory response to the infection with leptospires. The recognition of these signatures may be of diagnostic and prognostic relevance for affected individuals and they may indicate options for newer therapies targeting the identified pathways. Topics: Acute Kidney Injury; Animals; Arachidonate 5-Lipoxygenase; Disease Models, Animal; Disease Progression; Dogs; Female; Gene Expression Regulation; Hemorrhage; Humans; Interleukin-1alpha; Interleukin-1beta; Interleukin-8; Leptospirosis; Leukocytes, Mononuclear; Lung Injury; Male; Nitric Oxide Synthase Type II; RNA, Messenger; Severity of Illness Index; Signal Transduction; Survival Analysis; Syndrome; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 2016 |
Essential role for thymosin β4 in regulating vascular smooth muscle cell development and vessel wall stability.
Compromised development of blood vessel walls leads to vascular instability that may predispose to aneurysm with risk of rupture and lethal hemorrhage. There is currently a lack of insight into developmental insults that may define the molecular and cellular characteristics of initiating and perpetrating factors in adult aneurismal disease.. To investigate a role for the actin-binding protein thymosin β4 (Tβ4), previously shown to be proangiogenic, in mural cell development and vascular wall stability.. Phenotypic analyses of both global and endothelial-specific loss-of-function Tβ4 mouse models revealed a proportion of Tβ4-null embryos with vascular hemorrhage coincident with a reduction in smooth muscle cell coverage of their developing vessels. Mechanistic studies revealed that extracellular Tβ4 can stimulate differentiation of mesodermal progenitor cells to a mature mural cell phenotype through activation of the transforming growth factor-beta (TGFβ) pathway and that reduced TGFβ signaling correlates with the severity of hemorrhagic phenotype in Tβ4-null vasculature.. Tβ4 is a novel endothelial secreted trophic factor that functions synergistically with TGFβ to regulate mural cell development and vascular wall stability. These findings have important implications for understanding congenital anomalies that may be causative for adult-onset vascular instability. Topics: Animals; Aorta; Cell Differentiation; Cells, Cultured; Coculture Techniques; Endothelial Cells; Genes, Reporter; Genotype; Gestational Age; Hemorrhage; Human Umbilical Vein Endothelial Cells; Humans; Mesenchymal Stem Cells; Mice; Mice, Inbred C57BL; Mice, Knockout; Muscle, Smooth, Vascular; Myocytes, Smooth Muscle; Paracrine Communication; Phenotype; Signal Transduction; Smad Proteins; Thymosin; Transfection; Transforming Growth Factor beta | 2012 |
Long-term effects of subcutaneously injected 2,3,7,8-tetrachlorodibenzo-p-dioxin on the liver of rhesus monkeys.
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) accumulates and remains stable in the fatty tissues and liver of rodents for a long time. Considering the pronounced difference between species, long-term, low dose hepatic effects of TCDD were investigated after subcutaneous administration of TCDD into rhesus monkeys during pregnancy. Macroscopic and histopathological examination of the liver carried out 4 y after TCDD administration demonstrated intrahepatic focal fatty changes, infarction, hemorrhage, microthrombi-formation, sinusoidal ectasia, small hepatocyte hyperplasia, and increased number of alpha-smooth muscle actin (alpha-SMA)-positive cells. An electron microscopic study disclosed sinusoidal endothelial cell degeneration and injury in the liver of TCDD-treated monkeys. Western blot analysis showed downregulation of aryl hydrocarbon receptor (AhR) protein expression and decreased level of vascular endothelial (VE) cadherin but increased expression levels of CYP1A1 and transforming growth factor beta (TGF-beta) protein in the liver tissues. These changes observed in TCDD-exposed monkeys indicated sinusoidal endothelial cell injury and impairment in intrasinusoidal microcirculation. Infarction, focal fatty change, and microthrombi-formation are considered to be closely associated with intrahepatic circulatory impairment. Increased number of alpha-SMA-positive cells and decreased level of VE cadherin expression in the liver tissues might also be associated with sinusoidal endothelial cell injury. In addition, downregulation of AhR expression and increased CYP1A1 protein levels in the liver were consistent with persistent effects of TCDD. Although it has been reported that TCDD induced endothelial cell injury, this is the first report to describe vascular disorders and protein expression in the liver after injection with TCDD in a primate model. Topics: Animals; Antigens, CD; Blotting, Northern; Cadherins; Chemical and Drug Induced Liver Injury; Cytochrome P-450 CYP1A1; Endothelial Cells; Fatty Liver; Female; Hemorrhage; Infarction; Injections, Subcutaneous; Liver Diseases; Macaca mulatta; Microscopy, Electron; Muscle, Smooth; Polychlorinated Dibenzodioxins; Pregnancy; Receptors, Aryl Hydrocarbon; Thrombosis; Time Factors; Transforming Growth Factor beta | 2007 |
Dosage-dependent requirement of BMP type II receptor for maintenance of vascular integrity.
Germ-line mutations in bone morphogenic protein type II receptor (Bmpr2) confer susceptibility to pulmonary arterial hypertension (PAH), which is characterized by obstructive vascular lesions in small arteries. The molecular and cellular mechanisms that account for the etiology of this disorder remain elusive, as does the role of Bmpr2 in postnatal tissue homeostasis. Here we show that in adult mice, stably silencing Bmpr2 expression by RNA interference does not increase pulmonary arterial resistance but results in severe mucosal hemorrhage, incomplete mural cell coverage on vessel walls, and gastrointestinal hyperplasia. We present evidence that BMP receptor signaling regulates vascular remodeling during angiogenesis by maintaining the expression of endothelial guidance molecules that promote vessel patterning and maturation and by counteracting growth factor-induced AKT activation. Attenuation of this function may cause vascular dysmorphogenesis and predisposition to angioproliferative diseases. Our findings provide a mechanistic link between PAH and other diseases associated with the BMP/TGF-beta pathways, such as hereditary hemorrhagic telangiectasia and juvenile polyposis syndrome. Topics: Adenomatous Polyposis Coli; Animals; Bone Morphogenetic Protein Receptors, Type II; Gene Dosage; Germ-Line Mutation; Hemorrhage; Homeostasis; Hypertension, Pulmonary; Mice; Mice, Knockout; Neovascularization, Pathologic; Proto-Oncogene Proteins c-akt; RNA Interference; Telangiectasia, Hereditary Hemorrhagic; Transforming Growth Factor beta | 2007 |
L-arginine improves wound healing after trauma-hemorrhage by increasing collagen synthesis.
Several studies indicate impaired wound healing after trauma and shock. Wound immune cell dysfunction seems to be responsible for altered wound healing after trauma-hemorrhage (T-H). In this respect, administration of the amino acid L-arginine normalized wound immune cell function under those conditions. It remains unknown, however, whether L-arginine improves impaired wound healing after T-H.. To study this, male C3H/HeN mice were subjected to a midline laparotomy (i.e., soft tissue trauma induced), and polyvinyl sponges were implanted subcutaneously at the wound site before hemorrhage (35 +/- 5 mm Hg for 90 minutes) or were subjected to sham operation. During resuscitation, mice received 300 mg/kg body weight L-arginine or saline (vehicle). Seven days thereafter, hydroxyproline (OHP), a metabolite of collagen synthesis, was measured in the wound fluid using high-performance liquid chromatography. Collagen types I and III were determined in the wound by Western blot analysis. In addition, wound breaking strength was measured 10 days after T-H or sham operation.. The results indicate that OHP was significantly decreased in T-H mice. L-arginine, however, restored depressed OHP in the wound fluid in the T-H animals. Similarly, L-arginine treatment prevented a significant depression of collagen I synthesis after T-H. Collagen III was not significantly affected by T-H or L-arginine. Most important, L-arginine increased maximal wound breaking strength after severe blood loss. Therefore, L-arginine improves wound healing after T-H by increasing collagen synthesis.. Because L-arginine improves wound healing, the results suggest that L-arginine might represent a novel and useful adjunct to fluid resuscitation for decreasing wound complications after trauma and severe blood loss. Topics: Analysis of Variance; Animals; Arginine; Blotting, Western; Chromatography, High Pressure Liquid; Collagen; Hemorrhage; Hydroxyproline; Male; Mice; Mice, Inbred C3H; Random Allocation; Transforming Growth Factor beta; Wound Healing; Wounds and Injuries | 2005 |
Inhibition of matrix metalloproteinase on hepatic transforming growth factor beta1 and caspase-3 activation in hemorrhage.
Hemorrhage initiates an inflammatory response that induces the systemic release of cytokines and sequestration of polymorphonuclear neutrophils. Sequestered polymorphonuclear neutrophils release proteases, including matrix metalloproteinases (MMPs) that degrade elements of the extracellular matrix, contributing to the morbidity and mortality seen from hemorrhage. Activation of MMPs may be associated with changes in transforming growth factor beta1 (TGF-beta1) and caspase-3 signaling pathways. In this study, the authors examined hemorrhage-induced changes in the expression of rat hepatic MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-l), TGF-beta1, and caspase-3 activities in the presence and absence of the MMP inhibitor hydroxamate.. Hemorrhagic shock was induced in fasted, anesthetized, and cannulated rats by rapid phlebotomy to a mean arterial pressure level of 40 mm Hg, maintained for 90 minutes by withdrawal and infusion of blood, followed by a resuscitation period of lactated Ringer's infusion. Rats received either hydroxamate (25 mg/kg) or vehicle by gavage before hemorrhage. Twenty-four hours after resuscitation, plasma and liver samples were collected. Liver MMP-9, TGF-beta1, and caspase-3 levels were quantified by Western immunoblotting. Plasma glutamic oxaloacetic transaminase (GOT) and plasma glutamic pyruvic transaminase (GPT) were determined enzymatically.. Plasma GOT, plasma GPT, and liver MMP-9, TGF-beta1, and caspase-3 levels were all significantly elevated at 24 hours postresuscitation when compared with the control values. Hepatic TIMP-1, an in vivo inhibitor of MMP-9, was unaltered at 24 hours. Hydroxamate treatment reduced GOT, GPT, MMP-9, TGF-beta1, and caspase-3 levels at 24 hours. The mortality of hemorrhaged untreated rats was 29% after 24 hours, and pretreatment with hydroxamate reduced mortality to 0%.. These results indicate the beneficial effects of MMP inhibitor in preventing an increase in GOT, GPT, MMP-9, TGF-beta1, and caspase-3 activity with the potential for improvement of hepatic injury due to hemorrhage. Topics: Alanine Transaminase; Animals; Aspartate Aminotransferases; Cardiopulmonary Resuscitation; Caspase 3; Caspases; Disease Models, Animal; Hemorrhage; Hydroxamic Acids; Liver Diseases; Male; Matrix Metalloproteinase 9; Matrix Metalloproteinase Inhibitors; Rats; Rats, Sprague-Dawley; Reference Values; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
Consequences of altered TGF-beta expression and responsiveness in breast cancer: evidence for autocrine and paracrine effects.
To characterize the impact of increased production of TGF-beta in a xenograft model of human breast cancer, TGF-beta-responsive MDA-231 cells were genetically modified by stable transfection so as to increase their production of active TGF-beta1. Compared with control cells, cells that produced increased amounts of TGF-beta proliferated in vitro more slowly. In vivo, however, tumors derived from these cells exhibited increased proliferation and grew at an accelerated pace. To evaluate the role of autocrine TGF-beta signaling, cells were also transfected with a dominant-negative truncated type II TGF-beta receptor (TbetaRII). Disruption of autocrine TGF-beta signaling in the TGF-beta-overexpressing cells reduced their in vivo growth rate. Co-inoculation of Matrigel with the TGF-beta-overexpressing cells expressing the truncated TbetaRII compensated for their diminished in vivo growth capacity, compared with the TGF-beta-overexpressing cells with an intact autocrine loop. Tissue invasion by the tumor was a distinctive feature of the TGF-beta-overexpressing cells, whether or not the autocrine loop was intact. Furthermore, tumors derived from TGF-beta-overexpressing cells, irrespective of the status of the autocrine TGF-beta-signaling pathway, had a higher incidence of lung metastasis. Consistent with the suggestion that TGF-beta's enhancement of invasion and metastasis is paracrine-based, we observed no significant differences among the cell clones in an in vitro invasion assay. Thus, in this experimental model system in vitro assays of cell proliferation and invasion do not accurately reflect in vivo observations, perhaps due to autocrine and paracrine effects of TGF-beta that influence the important in vivo-based phenomena of tumor growth, invasion, and metastasis. Topics: Animals; Autocrine Communication; Breast Neoplasms; Carcinoma, Ductal, Breast; Cell Division; Collagen; Culture Media, Conditioned; Drug Combinations; Female; Gene Expression Regulation, Neoplastic; Genes, Dominant; Hemorrhage; Humans; Laminin; Lung Neoplasms; Mice; Mice, Nude; Neoplasm Proteins; Neoplasm Transplantation; Paracrine Communication; Polymerase Chain Reaction; Protein Serine-Threonine Kinases; Proteoglycans; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; Recombinant Fusion Proteins; Sequence Deletion; Skin Ulcer; Transfection; Transforming Growth Factor beta; Transplantation, Heterologous; Tumor Cells, Cultured | 2002 |
Potential role of modifier genes influencing transforming growth factor-beta1 levels in the development of vascular defects in endoglin heterozygous mice with hereditary hemorrhagic telangiectasia.
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disorder because of mutations in the genes coding for endoglin (HHT1) or ALK-1 (HHT2). The disease is associated with haploinsufficiency and a murine model was obtained by engineering mice that express a single Endoglin allele. Of a total of 171 mice that were observed for 1 year, 50 developed clinical signs of HHT. Disease prevalence was high in 129/Ola strain (72%), intermediate in the intercrosses (36%), and low in C57BL/6 backcrosses (7%). Most mice first presented with an ear telangiectasia and/or recurrent external hemorrhage. One-third of mice with HHT showed severe vascular abnormalities such as dilated vessels, hemorrhages, liver and lung congestion, and/or brain and heart ischemia. Disease sequelae included stroke, hydrocephalus, fatal hemorrhage, and congestive heart failure. Thus the murine model reproduces the multiorgan manifestations of the human disease. Levels of circulating latent transforming growth factor (TGF)-beta1 were significantly lower in the 129/Ola than in the C57BL/6 strain. Intercrosses and 129/Ola mice expressing reduced endoglin also showed lower plasma TGF-beta1 levels than control. These data suggest that modifier genes involved in the regulation of TGF-beta1 expression act in combination with a single functional copy of endoglin in the development of HHT. Topics: Abnormalities, Multiple; Animals; Antigens, CD; Blood Vessels; Brain; Cerebral Hemorrhage; Disease Models, Animal; Endoglin; Gastrointestinal Hemorrhage; Genes; Heart Defects, Congenital; Heart Failure; Hemorrhage; Heterozygote; Liver; Lung; Lung Diseases; Mice; Mice, Inbred C57BL; Receptors, Cell Surface; Telangiectasia, Hereditary Hemorrhagic; Transforming Growth Factor beta; Transforming Growth Factor beta1; Vascular Cell Adhesion Molecule-1 | 2001 |
Sex differences in posttraumatic cytokine release of endotoxin-stimulated whole blood: relationship to the development of severe sepsis.
In experimental trauma-hemorrhage and sepsis, a sexual dimorphism of cell-mediated immune functions has been described, which has been related to higher susceptibility to and mortality from sepsis in males. Therefore, in the present study, sex differences with regard to cytokine release of endotoxin stimulated whole blood and its relation to the development of severe posttraumatic sepsis were investigated in blunt trauma patients with multiple injuries.. Eighty-four patients (25 female; 59 male) sustaining blunt injuries with an Injury Severity Score > 16 were enrolled in the study. Whole blood and serum were obtained during a 14-day period of hospitalization. The capacity of peripheral blood mononuclear cells to produce cytokines (tumor necrosis factor-alpha, interleukin [IL]-6, IL-8) was tested by using a whole blood assay. Serum samples were assayed for anti-inflammatory cytokines (IL-4, IL-10, and transforming growth factor beta1) and sex hormones (testosterone, estradiol, progesterone). Patients were monitored daily for sepsis criteria according to the ACCP/ SCCM consensus conference 1992.. Within the entire patient population, sex differences in posttraumatic cytokine release were not detectable. Male trauma patients developing severe sepsis (n = 16) presented with a significantly increased cytokine producing capacity in the early posttraumatic period (< or = 24 hours after admission to the emergency room) when compared with males with an uncomplicated recovery. In females, differences between the subgroups of patients with (n = 7) and without development of severe sepsis were not detectable. There were no differences in systemic levels of anti-inflammatory cytokines within the early posttraumatic period between the subgroups of male and female patients with and without development of severe sepsis. In females, differences in sex hormone levels were not detectable, whereas in males, development of severe sepsis later was found to coincide with significantly decreased testosterone and increased estradiol serum levels.. The present study demonstrates a sex-specific regulation of leukocyte function in patients with multiple injuries within the early posttraumatic period. In male patients with multiple injuries, increased cytokine-producing capacities may correspond to enhanced inflammatory responses, which increase susceptibility to sepsis, whereas in female patients, other regulatory mechanisms may be involved. Topics: Adult; Blood; Cytokines; Endotoxins; Estradiol; Female; Hemorrhage; Humans; Interleukin-10; Interleukin-4; Interleukin-6; Interleukin-8; Leukocytes, Mononuclear; Male; Middle Aged; Multiple Trauma; Progesterone; Prospective Studies; Sepsis; Severity of Illness Index; Sex Characteristics; Testosterone; Time Factors; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha; Wounds, Nonpenetrating | 2000 |
Hypertonic saline resuscitation restores hemorrhage-induced immunosuppression by decreasing prostaglandin E2 and interleukin-4 production.
It was previously shown that hypertonic saline (HTS) enhances in vivo and in vitro cellular immune function of normal mice and reverses in vitro prostaglandin E2 (PGE2)-induced immunosuppression of normal peripheral blood mononuclear cells. Hemorrhage induces immunosuppression despite adequate isotonic fluid resuscitation. The effects of HTS resuscitation on immunosuppression following hemorrhage were studied. A mouse model of hemorrhagic shock was used. Bleeding was performed through a catheter placed in the femoral artery. Phytohemagglutinin-induced splenocyte proliferation and interleukin (IL)-1, IL-2,IL-4, IL-6, IL-10, transforming growth factor beta, and PGE2 plasma levels were measured 2 and 24 hr following hemorrhage and resuscitation with lactated Ringer's and HTS. In vivo cellular immune function was measured using a contact hypersensitivity test. Suppression of splenocyte proliferation (40%) 24 hr following hemorrhage occurred after lactated Ringer's resuscitation. HTS prevented immunosuppression. In vivo cell-mediated immune function 24 hr after hemorrhage was improved by HTS. HTS-resuscitated animals showed significantly lower levels of IL-4 and PGE2, and slightly elevated levels of proinflammatory cytokines (IL-1, IL-2, and IL-6). HTS reverses hemorrhage-induced T-cell suppression by reducing the production and/or release of IL-4 and PGE2. Topics: Animals; Cell Division; Dinoprostone; Hemodynamics; Hemorrhage; Hypertonic Solutions; Immunosuppression Therapy; Interleukin-1; Interleukin-10; Interleukin-2; Interleukin-4; Interleukin-6; Isotonic Solutions; Mice; Mice, Inbred BALB C; Resuscitation; Ringer's Lactate; Sodium; Sodium Chloride; Spleen; Transforming Growth Factor beta | 1996 |
Cytokine gene expression in splenic macrophages and Kupffer cells following haemorrhage.
TNF-alpha, IL-1, IL-6 and TGF-beta are important macrophage-derived mediators which play the pleiotropic role in inflammatory, metabolic, hematopoietic and immunologic processes. Studies have shown that haemorrhagic shock without significant tissue trauma induces profound immunosuppression which is associated with elevated plasma levels of TNF-alpha IL-1, IL-6 as well as TGF-beta. Furthermore, Kupffer cells but not the splenic M phi isolated from post-haemorrhaged animals showed an increased capacity to release inflammatory cytokines in response to LPS stimulation in vitro. However, it remains unknown whether the innate (i.e. in the absence of LPS stimulation) cytokine genes expression in Kupffer cells and splenic M phi is affected by haemorrhage. To determine this, C3H/HeN male mice were bled to and maintained at a mean arterial blood pressure of 35 mmHg for 60 min, and then adequately resuscitated. Splenic macrophages and Kupffer cells were isolated at 1 h after haemorrhage. Total RNA was extracted and cytokine mRNA was detected by semi-quantitative reverse transcription and polymerase chain reaction (RT-PCR). The results demonstrate that haemorrhage significantly elevated the mRNA accumulation of TNF-alpha, IL-1 beta, TGF-beta while IL-6 gene expression in Kupffer cells and splenic M phi was only slightly increased. Since Kupffer cells but not the splenic M phi showed increased cytokine release, it could be concluded that the differential regulation of cytokine release by these two macrophage populations following haemorrhage may be due to the divergence of the cytokine at the translational level.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Cytokines; Gene Expression Regulation; Hemorrhage; Interleukin-1; Interleukin-6; Kupffer Cells; Macrophages; Male; Mice; Mice, Inbred C3H; Polymerase Chain Reaction; Protein Biosynthesis; RNA, Messenger; Spleen; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 1995 |
Anti-transforming growth factor-beta monoclonal antibodies prevent lung injury in hemorrhaged mice.
Acute lung injury, characterized as the adult respiratory distress syndrome (ARDS), is a common clinical occurrence following blood loss and injury. We previously found increased levels of transforming growth factor (TGF)-beta 1 mRNA in murine intraparenchymal mononuclear cells and in alveolar macrophages within 1 h after hemorrhage. Because TGF-beta has potent proinflammatory and immunoregulatory properties, we investigated the effect of blocking TGF-beta with mAb on hemorrhage-induced pathology, cytokine mRNA levels in lungs, as well as survival from pneumonia. Mice treated with anti-TGF-beta mAb showed normal pulmonary histology 3 days after hemorrhage and resuscitation in contrast to the mononuclear and neutrophil infiltrates, intraalveolar hemorrhage, and interstitial edema found in hemorrhaged mice either treated with control antibody or not treated with any antibody. Decreased mRNA levels for IL-1 beta, TNF-alpha, IL-6, IL-10, and IFN-gamma as compared with untreated, hemorrhaged controls were present in intraparenchymal pulmonary mononuclear cells following therapy with anti-TGF-beta. In contrast, therapy with anti-TGF-beta increased mRNA levels for IL-1 beta and TNF-alpha in alveolar macrophages and for TGF-beta in peripheral blood mononuclear cells collected 3 days after hemorrhage. Administration of anti-TGF-beta to hemorrhaged mice did not correct the enhanced susceptibility to Pseudomonas aeruginosa pneumonia that exists after hemorrhage. These results suggest that TGF-beta has an important role in hemorrhage-induced acute lung injury, but does not contribute to the post-hemorrhage depression in pulmonary antibacterial response. Topics: Animals; Antibodies, Monoclonal; Cytokines; Disease Models, Animal; Gene Expression; Hemorrhage; Leukocytes, Mononuclear; Lung; Macrophages, Alveolar; Male; Mice; Mice, Inbred BALB C; Monocytes; Pneumonia; Pseudomonas Infections; Respiratory Distress Syndrome; Resuscitation; RNA, Messenger; Transforming Growth Factor beta | 1994 |
Effects of hemorrhage on cytokine gene transcription.
Injury and blood loss are often followed by infection and the rapid development of organ system dysfunction, frequently involving mucosal sites, such as the lung and intestine. To examine possible mechanisms contributing to these conditions, we used semiquantitative polymerase chain reactions to determine cytokine mRNA expression among cellular populations isolated from mucosal and systemic anatomic sites of mice at predetermined time points following 30% blood volume hemorrhage with resuscitation 1 hr later. Within 1 hr after hemorrhage, significant increases were observed in mRNA levels for IL-1 alpha, IL-1 beta, IL-5, and TGF-beta in intraparenchymal pulmonary mononuclear cells. The levels of TGF-beta transcripts among alveolar macrophages were increased 1 hr following blood loss, and increase in IL-1 alpha transcripts was found starting 2 hr posthemorrhage. Cells from Peyer's patches showed significant increases in mRNA levels for IL-1 beta, IL-2, IL-5, IL-6, IFN-gamma, and TGF-beta during the 4 hr following hemorrhage. Significant increases in mRNA levels for IL-1 beta, TNF-alpha, and TGF-beta were present within 4 hr of blood loss among cells isolated from mesenteric lymph nodes. The expression of mRNA for most cytokines was not significantly altered in splenocytes or peripheral blood mononuclear cells at any time point following hemorrhage. These experiments demonstrate that blood loss, even if resuscitated, produces significant increases in proinflammatory and immunoregulatory cytokine gene transcription as early as 1 hr following hemorrhage. These posthemorrhage alterations in cytokine mRNA expression were particularly prominent at mucosal sites, suggesting a mechanism for the increased incidence of pulmonary and intestinal involvement in organ system failure following severe blood loss and injury. Topics: Animals; B-Lymphocytes; Cytokines; Disease Models, Animal; Gene Expression; Hemorrhage; Interferon-gamma; Interleukin-1; Interleukin-5; Interleukin-6; Macrophages, Alveolar; Male; Mice; Mice, Inbred BALB C; Monocytes; Peyer's Patches; Polymerase Chain Reaction; Receptors, Interleukin-2; Resuscitation; RNA, Messenger; T-Lymphocytes; Time Factors; Transcription, Genetic; Transforming Growth Factor beta | 1993 |
The release of transforming growth factor-beta following haemorrhage: its role as a mediator of host immunosuppression.
Haemorrhage in the absence of trauma is reported to induce a profound depression in cell-mediated immunity. Recent studies have drawn attention to the cytokine transforming growth factor-beta (TGF-beta) that, while important in wound healing, also has marked immunosuppressive effects. The aim of this study was to determine whether: (1) haemorrhage induces an increase in circulating TGF-beta and if this is associated with the loss of host immunoresponsiveness; and (2) administration of monoclonal antibody (mAb) to TGF-beta following haemorrhage ablates these changes. To determine this, C3H/HeN mice were bled to and maintained at a mean arterial pressure of 35 mmHg for 1 hr. This required removing approximately 50% of the circulating blood volume. Following this period of hypotension, the mice were adequately resuscitated. Blood samples obtained at 24 and 72 hr, but not at 2 hr, following haemorrhage showed a significant elevation in plasma TGF-beta levels when compared to shams. At 24 hr, the increase of TGF-beta in the plasma was associated with decreases in both concanavalin A (Con A)-induced splenocyte proliferation and splenic macrophage antigen presentation. Treating animals with neutralizing antibody (animals received 200 micrograms mAb against bovine TGF-beta 1,2,3/mouse intraarterially) not only reduced the levels of TGF-beta in the blood at 24 hr, but also restored splenocyte functions, such as Con A-induced proliferation, interleukin-2 (IL-2) release, and the capacity of splenic macrophages to present antigen. However, elevated levels of prostaglandin E2 (PGE2) seen in plasma during haemorrhage were only partially depressed by the antibody treatment. These results indicate that the release of TGF-beta contributes to the protracted (> or = 24 hr) suppression of cell-mediated immunity following haemorrhage. Topics: Animals; Dinoprostone; Hemorrhage; Immune Tolerance; Immunity, Cellular; Kinetics; Lymphocyte Activation; Macrophages; Male; Mice; Mice, Inbred C3H; Spleen; Transforming Growth Factor beta | 1993 |