Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Endocrinol Invest ; 44(7): 1475-1482, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33155181

ABSTRACT

PURPOSE: Xeroderma pigmentosum (XP) is an autosomal recessive disease with defective DNA repair, a markedly increased risk of skin cancer, and premature aging. Reports from North Africa have described thyroid nodules in XP patients, but thyroid nodule prevalence has never been determined in XP patients enrolled in our natural history study at the National Institutes of Health (NIH). METHODS: We performed thyroid ultrasound examinations on all 29 XP patients examined from 2011 to 2019 and assessed nodule malignancy using the Thyroid Imaging Reporting and Data System. Thyroid nodule prevalence was also obtained from comparison cohorts. DNA sequencing was performed on thyroid tissue from XP patients who had surgery for thyroid cancer. RESULTS: Thyroid nodules were identified in 18/29 XP patients (62%). The median age of patients with thyroid nodules in our XP cohort (20Ā years) was younger than that of three comparison groups: 36Ā years (California study-208 subjects), 48Ā years (Korean study-24,757 subjects), and 52Ā years (NIH-682 research subjects). Multiple (2-4) thyroid nodules were found in 12/18 (67%) of the patients with nodules. Autopsy examination revealed follicular adenomas in 4/8 (50%) additional XP patients. DNA sequencing revealed rare mutations in two other XP patients with papillary thyroid cancer. CONCLUSIONS: XP patients have an increased incidence of thyroid nodules at an early age in comparison to the general population. These finding confirm another premature aging feature of XP.


Subject(s)
Aging, Premature/physiopathology , Thyroid Nodule/epidemiology , Xeroderma Pigmentosum/complications , Adolescent , Adult , Child , Female , Follow-Up Studies , Humans , Male , Maryland/epidemiology , Middle Aged , Prognosis , Thyroid Nodule/etiology , Thyroid Nodule/pathology , Young Adult
4.
Br J Cancer ; 105(8): 1210-7, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21897392

ABSTRACT

BACKGROUND: Activin A is a multi-functional cytokine belonging to the transforming growth factor-Ɵ (TGF-Ɵ) superfamily; however, the effect of activin A on angiogenesis remains largely unclear. We found that inhibin Ɵ A subunit (INHBA) mRNA is overexpressed in gastric cancer (GC) specimens and investigated the effect of activin A, a homodimer of INHBA, on angiogenesis in GC. METHODS: Anti-angiogenic effects of activin A via p21 induction were evaluated using human umbilical vein endothelial cells (HUVECs) in vitro and a stable INHBA-introduced GC cell line in vivo. RESULTS: Compared with TGF-Ɵ, activin A potently inhibited the cellular proliferation and tube formation of HUVECs with induction of p21. A promoter assay and a chromatin immunoprecipitation assay revealed that activin A directly regulates p21 transcriptional activity through Smads. Stable p21-knockdown significantly enhanced the cellular proliferation of HUVECs. Notably, stable p21-knockdown exhibited a resistance to activin-mediated growth inhibition in HUVECs, indicating that p21 induction has a key role on activin A-mediated growth inhibition in vascular endothelial cells. Finally, a stable INHBA-introduced GC cell line exhibited a decrease in tumour growth and angiogenesis in vivo. CONCLUSION: Our findings highlight the suppressive role of activin A, unlike TGF-Ɵ, on tumour growth and angiogenesis in GC.


Subject(s)
Activins/physiology , Neovascularization, Pathologic/prevention & control , Stomach Neoplasms/blood supply , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Smad2 Protein/metabolism , Stomach Neoplasms/pathology
5.
Clin Genet ; 77(4): 365-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20002457

ABSTRACT

The effects of DNA repair and transcription gene abnormalities in human pre-natal life have never been studied. Trichothiodystrophy (TTD) is a rare (affected frequency of 10(-6)) recessive disorder caused by mutations in genes involved in nucleotide excision repair (NER) pathway and in transcription. Based on our novel clinical observations, we conducted a genetic epidemiologic study to investigate gestational outcomes associated with TTD. We compared pregnancies resulting in TTD-affected offspring (n = 24) with respect to abnormalities during their antenatal and neonatal periods to pregnancies resulting in their unaffected siblings (n = 18), accounting for correlation, and to population reference values. Significantly higher incidence of several severe gestational complications was noted in TTD-affected pregnancies. Small for gestational age (SGA) <10th percentile [Relative risk (RR ) = 9.3, 95% CI = 1.4-60.5, p = 0.02], SGA <3rd percentile (RR = 7.2, 95% CI = 1.1-48.1, p = 0.04), and neonatal intensive care unit (NICU) hospitalization (RR = 6.4, 95% CI = 1.4-29.5, p = 0.02) occurred more frequently among TTD-affected neonates compared with their unaffected siblings. Compared with reference values from general obstetrical population, pregnancies that resulted in TTD-affected infants were significantly more likely to be complicated by hemolysis, elevated liver enzymes and low platelets (HELLP) syndrome (RR = 35.7, 95% CI = 7.6-92.5, p = 0.0002), elevated mid-trimester maternal serum human chorionic gonadotropin (hCG) levels (RR = 14.3, 95% CI = 7.0-16.6, p < 0.0001), SGA <3rd percentile (RR = 13.9, 95% CI = 7.4-21.1, p < 0.0001), pre-term delivery (<32 weeks) (RR = 12.0, 95% CI = 4.9-21.6, p < 0.0001), pre-eclampsia (RR = 4.0, 95% CI = 1.6-7.4, p = 0.006), and decreased fetal movement (RR = 3.3, 95% CI = 1.6-5.2, p = 0.0018). Abnormal placental development is an underlying mechanism that may explain the constellation of observed complications in our study. Thus, we hypothesize that TTD DNA repair and transcription genes play an important role in normal human placental development.


Subject(s)
DNA Repair/genetics , Fetal Development/genetics , Transcription, Genetic , Trichothiodystrophy Syndromes/embryology , Trichothiodystrophy Syndromes/genetics , Adult , Demography , Family , Female , Humans , Live Birth , Middle Aged , Pregnancy , Pregnancy Outcome , Reference Values , Young Adult
6.
J Med Genet ; 45(10): 609-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18603627

ABSTRACT

Trichothiodystrophy (TTD) is a rare, autosomal recessive disease, characterised by brittle, sulfur deficient hair and multisystem abnormalities. A systematic literature review identified 112 patients ranging from 12 weeks to 47 years of age (median 6 years). In addition to hair abnormalities, common features reported were developmental delay/intellectual impairment (86%), short stature (73%), ichthyosis (65%), abnormal characteristics at birth (55%), ocular abnormalities (51%), infections (46%), photosensitivity (42%), maternal pregnancy complications (28%) and defective DNA repair (37%). There was high mortality, with 19 deaths under the age of 10 years (13 infection related), which is 20-fold higher compared to the US population. The spectrum of clinical features varied from mild disease with only hair involvement to severe disease with profound developmental defects, recurrent infections and a high mortality at a young age. Abnormal characteristics at birth and pregnancy complications, unrecognised but common features of TTD, suggest a role for DNA repair genes in normal fetal development.


Subject(s)
Hair/abnormalities , Trichothiodystrophy Syndromes/pathology , Adolescent , Adult , Birth Weight , Body Height , Child , Child, Preschool , DNA Repair/physiology , Developmental Disabilities/epidemiology , Eye Diseases/epidemiology , Eye Diseases/microbiology , Female , Genes, Recessive , Gonadal Dysgenesis/epidemiology , Hair/chemistry , Humans , Ichthyosis/epidemiology , Infant , Male , Middle Aged , Photosensitivity Disorders/epidemiology , Prevalence , Trichothiodystrophy Syndromes/genetics , Trichothiodystrophy Syndromes/microbiology
8.
Neuroscience ; 145(4): 1388-96, 2007 Apr 14.
Article in English | MEDLINE | ID: mdl-17276014

ABSTRACT

Patients with the rare genetic disorders, xeroderma pigmentosum (XP), trichothiodystrophy (TTD) and Cockayne syndrome (CS) have defects in DNA nucleotide excision repair (NER). The NER pathway involves at least 28 genes. Three NER genes are also part of the basal transcription factor, TFIIH. Mutations in 11 NER genes have been associated with clinical diseases with at least eight overlapping phenotypes. The clinical features of these patients have some similarities but also have marked differences. NER is involved in protection against sunlight-induced DNA damage. While XP patients have 1000-fold increase in susceptibility to skin cancer, TTD and CS patients have normal skin cancer risk. Several of the genes involved in NER also affect somatic growth and development. Some patients have short stature and immature sexual development. TTD patients have sulfur deficient brittle hair. Progressive sensorineural deafness is an early feature of XP and CS. Many of these clinical diseases are associated with developmental delay and progressive neurological degeneration. The main neuropathology of XP is a primary neuronal degeneration. In contrast, CS and TTD patients have reduced myelination of the brain. These complex neurological abnormalities are not related to sunlight exposure but may be caused by developmental defects as well as faulty repair of DNA damage to neuronal cells induced by oxidative metabolism or other endogenous processes.


Subject(s)
Cockayne Syndrome/genetics , DNA Damage/genetics , DNA Repair/genetics , Mutation/genetics , Xeroderma Pigmentosum/genetics , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/physiopathology , Cockayne Syndrome/metabolism , Cockayne Syndrome/physiopathology , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/physiopathology , Humans , Phenotype , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/physiopathology , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/physiopathology
9.
J Leukoc Biol ; 65(2): 211-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10088604

ABSTRACT

The importance of reactive oxygen species (ROS) in neutrophil (PMN)-mediated injury to host tissues has been strongly implicated in a number of animal models. Peculiarities of the laboratory rat PMN, including an apparent paucity of superoxide release, prompted us to examine disparities in the respiratory burst between human and rat PMNs. Using isolated PMNs, we examined oxygen consumption, superoxide release, nitrate/nitrite release, and dihydrorhodamine (DHR) oxidation in response to an array of soluble stimuli. Our findings confirm that intact rat PMNs release little superoxide in comparison to human PMNs when primed and activated by soluble stimuli. For example, PMA-activated human PMNs released superoxide at 10.1 +/- 2.7 times the rate of rat PMNs (P < 0.01). However, measurements of oxygen consumption, cell-associated oxidant production (by DHR oxidation) and release of superoxide from electroporated cells suggests that rat PMNs generate oxidants at rates equivalent to human PMNs but preferentially release them in an intracellular compartment. Implications for the study of PMN-mediated oxidant injury in animal models are discussed.


Subject(s)
Neutrophils/physiology , Respiratory Burst/physiology , Animals , Humans , Nitrates/metabolism , Nitrites/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Rhodamines/metabolism , Species Specificity , Superoxides/metabolism
10.
Shock ; 11(2): 77-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10030791

ABSTRACT

UNLABELLED: Postinjury neutrophil (PMN) dysfunction is a well recognized event that may be responsible for increased infections. PMN cytokine production is an important component of their bactericidal capacity. When PMNs are stimulated, inhibitory factor kappaB (IkappaB) is degraded, allowing nuclear factor kappaB (NFkappaB) to translocate to the nucleus and promotes genes for the transcription of the interleukin-8 (IL-8) and tumor necrosis factor (TNF) genes. We hypothesize that similar to their late postinjury depressed superoxide production, postinjury PMNs manifest suppressed cytokine production, which is mediated by stabilization of IkappaB levels. METHODS: Twelve severely injured patients with an injury severity score (ISS) of 24 (+/-4.6) were studied as well as 10 elective surgical patients as a control. PMNs were isolated and incubated for 24 h in RPMI. PMNs were stimulated with lipopolysaccharide (LPS; 100 ng) or PAF (200 nm) and fMLP (1 microM) and release of IL-8, TNF, and interleukin-1 receptor antagonist (IL-1ra) were measured. Postinjury PMNs were also stimulated with LPS (100 ng), and IkappaB breakdown was measured at 0, 30, and 60 min using gel electrophoresis. RESULTS: Postinjury PMNs displayed a significant suppression of both IL-8 and TNF on postinjury Days 1-3, while the release of IL-1ra was preserved throughout the entire study period. In contrast, elective surgical patients demonstrated no decrease in IL-8 or TNF. Furthermore, IkappaB levels were preserved in the postinjury PMNs as compared with normal control PMNs. CONCLUSION: Postinjury PMNs have a suppressed release of both IL-8 and TNF following injury that did not occur in elective surgical patients. Furthermore, the NFkappaB/IkappaB-independent IL-1ra did not show suppression of release. In addition, stabilization of IkappaB following severe injury leads to decreased PMN IL-8 and TNF production. This genetic reprogramming may help explain PMN dysfunction and subsequent infections seen in severely injured patients.


Subject(s)
Cytokines/blood , DNA-Binding Proteins/blood , Neutrophils/metabolism , Wounds, Nonpenetrating/blood , Wounds, Penetrating/blood , Adult , Humans , I-kappa B Proteins , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein , Interleukin-8/blood , Lipopolysaccharides/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Postoperative Complications/blood , Sialoglycoproteins/blood , Tumor Necrosis Factor-alpha/metabolism
11.
Shock ; 9(3): 171-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525323

ABSTRACT

In a recent clinical trial, liposomal prostaglandin E1 (PGE1) improved oxygenation, increased compliance, and decreased ventilator dependency in patients with adult respiratory distress syndrome (ARDS), thus renewing interest in PGE1 as a potential modulator of inflammation. The neutrophil (PMN) is believed to play a key role in the development of ARDS. Consequently, we investigated the effects of PGE1 on three components of the neutrophil inflammatory response: reactive oxygen species (ROS) generation, protease release, and surface expression of adhesion molecules. Human neutrophils were incubated with PGE1 and then primed with platelet-activating factor (PAF) and activation with N-formyl-methionyl-leucylphenylalanine (fMLP). PGE1 at a dose range of (10[-8] to 10[-5] M) attenuated primed/activated (PAF/fMLP) PMN superoxide anion generation and elastase release. In contrast, PGE1 doses > or =10[-5] M were required to attenuate PAF-stimulated neutrophil upregulation of CD11b/CD18 adhesion molecules. PGE1 also diminished the duration of the PAF-induced cytosolic calcium (Ca2+) flux. Our results suggest that plasma levels of PGE1 attained in patients with ARDS may attenuate ROS and protease neutrophil cytotoxicity but may not effectively block PMN-endothelial cell (EC) adhesion. This attenuation may occur through abrogation of the Ca2+ influx.


Subject(s)
Alprostadil/pharmacology , Macrophage-1 Antigen/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Pancreatic Elastase/metabolism , CD18 Antigens/drug effects , CD18 Antigens/metabolism , Calcium/metabolism , Humans , Macrophage-1 Antigen/drug effects , Superoxides/metabolism , Up-Regulation
12.
Surgery ; 124(2): 403-7; discussion 408, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9706165

ABSTRACT

BACKGROUND: Increased expression of pulmonary endothelial intercellular adhesion molecule-1 (ICAM-1) is obligatory to neutrophil adherence culminating in adult respiratory distress syndrome (ARDS). The p38 mitogen-activated protein kinases (MAPKs) have been established as crucial in leukocyte proinflammatory signaling, but their role in the endothelial cells remains ill defined. We hypothesized that p38 MAPK activity is integral to ICAM-1 up-regulation on pulmonary endothelium. METHODS: Human pulmonary microvascular endothelial cells (HMVECs) were grown to confluence and pretreated with either the tyrosine phosphorylation inhibitor herbimycin A (1 mumol/L or the p38 MAPK inhibitor SB 203580 (10(-7) to 10(-5) mol /L) for 6 hours. ICAM-1 expression was quantified by flow cytometry. Data expressed as mean fluorescence intensity. Western blotting was used to show p38 MAPK activity after stimulation with lipopolysaccharide (LOS) or tumor necrosis factor-alpha (TNF-alpha). RESULTS: Tyrosine phosphorylation inhibition with herbimycin A attenuated both LPS and TNF-alpha stimulated ICAM-1 up-regulation. Similarly, specific inhibition of p38 MAPK attenuated both LPS (10(-6) to 10(-5) mol/L SB203580) and TNF-alpha (10(-7) to 10(-5) mol/L SB203580) stimulated expression of ICAM-1 on HMVECs. Both LPS and TNF-alpha induced activation of p38 in HMVECs. CONCLUSIONS: Signaling through p 38 MAPKs contributes to LP and TNF-alpha stimulated ICAM-1 surface expression on HMVECs. Thus p38 MAPKs appear integral to both neutrophil and endothelial cell proinflammatory signaling and may be a potential therapeutic target in the treatment of ARDS.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Endothelium, Vascular/enzymology , Intercellular Adhesion Molecule-1/metabolism , Mitogen-Activated Protein Kinases , Up-Regulation/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Capillaries/chemistry , Capillaries/cytology , Capillaries/enzymology , Cells, Cultured , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Lung/blood supply , Phosphorylation , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases
13.
Metabolism ; 49(10): 1255-60, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11079812

ABSTRACT

A high proportion of patients with depression develop glucose intolerance accompanied by hyperinsulinemia, suggestive of reduced insulin sensitivity (insulin resistance). The aim of this study was to evaluate insulin sensitivity in patients with depression and its changes during the clinical course of depression. Twenty nondiabetic patients with depression (13 males and 7 females aged 44+/-14 years; body mass index [BMI] 23.2+/-2.8 kg/m2) were prospectively studied by the frequently sampled intravenous glucose tolerance test (FSIGT) and the oral glucose tolerance test (OGTT) before and after treatment of depression, and an age-, sex-, and BMI-matched control group (n = 13) was examined once by the FSIGT. Metabolic indices measuring glucose effectiveness at basal insulin (SG) and insulin sensitivity (SI) were derived from minimal model analysis. Each patient was treated by cyclic antidepressants with an 1,800 to 2,200 kcal/d food intake and underwent no exercise therapy. SI was significantly lower in patients before treatment versus control subjects (6.0+/-2.5 v 13.8+/-8.6 x 10(-5) min(-1) x mol(-1) x L, P < .01). After treatment of depression, a significant increase in SI (10.7+/-7.5 x 10(-5) min(-1) x mol(-1) x 1, P

Subject(s)
Depression/metabolism , Insulin Resistance , Adult , Aged , Blood Glucose/analysis , Body Mass Index , Female , Glucose Tolerance Test , Humans , Male , Middle Aged , Serotonin/physiology
14.
Arch Surg ; 133(11): 1229-33, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9820355

ABSTRACT

BACKGROUND: Secretory phospholipase A2 (sPLA2) is a potent proinflammatory enzyme that stimulates inflammation through the production of reactive lipids. However, enzymatic inhibitors have been disappointing in their effectiveness in halting hyperinflammation. OBJECTIVE: To determine whether sPLA2 acts directly on neutrophil plasma membrane lipids or via a nonenzymatic mechanism. DESIGN: Isolated neutrophils (PMNs) were incubated with 3 types of sPLA2, and elastase and superoxide release from PMNs was measured. Ethyleneglycotetraacetic acid was used as a selective enzymatic inhibitor. The PMNs were exposed to sPLA2 in the presence and absence of ethyleneglycotetraacetic acid and the release of elastase was measured. SETTING: Urban trauma research laboratory. PATIENTS: Normal healthy donors of PMNs. MAIN OUTCOME MEASURES: Stimulated release of superoxide and elastase. RESULTS: The sPLA2 acted directly on plasma membrane lipids to stimulate the PMN to produce superoxide and release elastase. This mechanism is blocked with enzymatic inhibition of sPLA2. The sPLA2 also provokes elastase release from PMNs independently of its enzymatic function. This mechanism is not blocked with traditional enzymatic inhibitors. CONCLUSIONS: These data indicate that the sPLA2 can act directly on PMNs to stimulate the release of inflammatory mediators via enzymatic degradation of plasma membrane lipids. In addition, sPLA2 can act as a ligand and stimulate the PMN independently of its enzymatic activity.


Subject(s)
Inflammation/immunology , Neutrophil Activation/immunology , Neutrophils/metabolism , Pancreatic Elastase/immunology , Phospholipases A/physiology , Reactive Oxygen Species/immunology , Superoxides/immunology , Animals , Egtazic Acid/pharmacology , Elapidae/immunology , Group II Phospholipases A2 , Humans , Multiple Organ Failure/immunology , Phospholipases A2 , Respiratory Burst/immunology
15.
Arch Surg ; 134(10): 1074-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10522849

ABSTRACT

BACKGROUND: Inflammatory stimuli rapidly activate mitogen-activated protein kinases (MAPKs) in neutrophils (PMNs). However, their role in cytotoxic function remains unknown. Elucidating the signals involved in release of cytotoxic agents from PMNs may provide new avenues for therapy in diseases of diminished or excessive PMN function. HYPOTHESIS: The p38 MAPK and extracellular signal-related kinase 1/2 (ERK1/2) modulate superoxide generation and elastase release in activated human PMNs. STUDY DESIGN: Isolated human PMNs were incubated with specific inhibitors of MAPK pathways, or vehicle control solution, before activation with the bacterial peptide f-Met-Leu-Phe. MAIN OUTCOME MEASURES: The rate of superoxide release from activated PMNs was measured by the superoxide dismutase-inhibitable reduction of cytochrome-c. Elastase release from PMNs was determined by cleavage of the substrate Ala-Ala-Pro-Val-pNA. RESULTS: Superoxide release from activated PMNs was inhibited by blockade of p38 MAPK activation but unaffected by blockade of ERK1/2. Conversely, elastase release was unaffected by p38 MAPK inhibition and increased by ERK1/2 inhibition. CONCLUSIONS: Activation of p38 MAPK promotes superoxide release from PMNs activated by f-Met-Leu-Phe. The ERK1/2 pathway may serve as a negative feedback mechanism for granule exocytosis.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Neutrophils/immunology , Cytotoxicity, Immunologic , Humans , Mitogen-Activated Protein Kinase 3 , Pancreatic Elastase/metabolism , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases
16.
Arch Surg ; 135(2): 219-25, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10668885

ABSTRACT

HYPOTHESIS: Neutrophil priming has been implicated in the development of multiple organ failure, although the precise intracellular mechanisms that regulate neutrophil priming remain unclear. Our previous work characterized platelet-activating factor (PAF) priming of human neutrophils for concordant superoxide anion (O2-) generation and elastase degranulation. The p38 mitogen-activated protein kinase (MAPK) is activated by PAF stimulation. We hypothesized that PAF-induced human neutrophil priming for O2- and elastase release is mediated via the p38 MAPK pathway. DESIGN: Isolated neutrophils from 6 human donors were preincubated with the specific p38 MAPK inhibitor SB 203580 (1 micromol/L) or buffer (control) for 30 minutes. Cells were then primed with PAF (200 nmol/L), followed by receptor-dependent (N-formyl-methionyl-leucyl-phenylalanine, 1 micromol/L) or receptor-independent phorbol myristate acetate (PMA, 100 ng/mL) activation. SETTING: Urban trauma research laboratory. PATIENTS: Healthy volunteer donors of neutrophils. MAIN OUTCOME MEASURES: Maximal rate of O2- generation was measured by superoxide dismutase-inhibitable reduction of cytochrome c and elastase release by the cleavage of N-methoxysuccinyl-Ala-Ala-Pro-Val-p-nitroanilide. RESULTS: SB 203580 significantly attenuated the generation of O2- and release of elastase from neutrophils activated with N-formyl-methionyl-leucyl-phenylalanine but not with PMA. Independent of the activator receptor status, SB 203580 almost completely blocked the exaggerated neutrophil cytotoxic response due to PAF priming. CONCLUSIONS: The p38 MAPK pathway is required for maximal PAF-induced neutrophil priming for O2- production and elastase degranulation. Therefore, the MAPK signaling cascade may offer a potential therapeutic strategy to preempt global neutrophil hyperactivity rather than attempt to nullify the end products independently.


Subject(s)
Leukocyte Elastase/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neutrophil Activation/physiology , Superoxides/metabolism , Enzyme Activation , Humans , Platelet Activating Factor/pharmacology
17.
Am J Surg ; 176(6): 612-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9926800

ABSTRACT

BACKGROUND: Hemoglobin-based blood substitutes appear poised to deliver the promise of a universally compatible, disease-free alternative to banked blood. However, vasoconstriction following administration of tetrameric hemoglobins has been problematic, likely because of nitric oxide binding. Polymerized hemoglobin is effectively excluded from the abluminal space because of its size, and is thus less likely to perturb vasorelaxation. We therefore hypothesized that hemodynamic responses would be no different in injured patients receiving polymerized hemoglobin versus banked blood. METHODS: Injured patients requiring urgent transfusion were randomized to receive either polymerized hemoglobin or banked blood. Systemic arterial pressure, pulmonary arterial pressure, cardiac index, pulmonary capillary wedge pressure, systemic vascular resistance, and pulmonary vascular resistance were measured serially. RESULTS: There was no difference in any of the measured hemodynamic parameters between patients resuscitated with polymerized hemoglobin versus blood. CONCLUSIONS: Polymerized hemoglobin given in large doses to injured patients lacks the vasoconstrictive effects reported in the use of other hemoglobin-based blood substitutes. This supports the continued investigation of polymerized hemoglobin in injured patients requiring urgent transfusion.


Subject(s)
Blood Substitutes , Hemoglobins , Hypertension, Pulmonary/etiology , Polymers , Wounds and Injuries/therapy , Adult , Female , Hemodynamics/drug effects , Humans , Male , Resuscitation/methods , Shock, Hemorrhagic/therapy , Vasoconstriction
18.
Can J Cardiol ; 8(9): 954-60, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1486546

ABSTRACT

OBJECTIVE: To investigate vascular responses to the endothelin-1 (ET-1) precursor, human big endothelin 1-38 (big ET), in the peripheral vascular bed of the cat. DESIGN: These studies were designed to investigate the hypothesis that bit ET is converted to an active peptide with properties similar to ET-1. SETTING: Hindquarters vascular bed of the cat under conditions of controlled bloodflow; changes in perfusion pressure reflect changes in vascular resistance. ANIMALS: Fifty-four adult mongrel cats. INTERVENTIONS: Big ET, ET-1, the peptidases chymotrypsin, pepsin and cathepsin-D, and the metalloprotease inhibitor phosphoramidon. MAIN RESULTS: Intra-arterial injections of big ET induced a slow-developing and sustained increase in hindquarters perfusion pressure which could be blocked by phosphoramidon. ET-1 (0.3 nmol), administered as a slow infusion over a 10-min period, produced a slowly developing increase in hindquarters perfusion pressure in a manner similar to that observed in response to injection of big ET. A bolus injection of ET-1 produced a biphasic response characterized by a transient decrease in pressure followed by an increase which was significantly greater in magnitude and more rapid in onset than the pressor response to big ET (0.3 nmol). After incubation of big ET with chymotrypsin, pepsin and cathepsin-D (each 5% weight/weight) for 30 mins at 37 degrees C, injection of activated big ET produced a biphasic response characteristic of the response to ET-1 with an initial transient decrease in pressure followed by a secondary increase in hindquarters perfusion pressure. CONCLUSIONS: Big ET produces a phosphoramidon-sensitive pressor response which is similar to that produced by an infusion of ET-1. These data suggest that chymotrypsin, pepsin and cathepsin-D can convert big ET to an active peptide which elicits a biphasic response similar to that produced by ET-1.


Subject(s)
Blood Vessels/drug effects , Endothelins/pharmacology , Protein Precursors/pharmacology , Animals , Blood Pressure/drug effects , Blood Vessels/physiology , Cathepsin D/pharmacology , Cats , Chymotrypsin/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Endothelin-1 , Glycopeptides/pharmacology , Hindlimb/blood supply , Infusions, Intra-Arterial , Injections, Intra-Arterial , Neprilysin/antagonists & inhibitors , Pepsin A/pharmacology , Regional Blood Flow/drug effects
19.
J Surg Res ; 83(2): 83-8, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10329099

ABSTRACT

Mesenteric lymph has recently been invoked as an avenue for gut-derived factors that may result in distant organ injury following hemorrhagic shock. We demonstrate that posthemorrhagic shock mesenteric lymph primes neutrophils (PMNs) and causes lung injury. Methods. Mesenteric lymph was collected from Sprague-Dawley rats from their mesenteric lymph duct prior to, during, and following hemorrhagic shock (MAP 40 for 90 min). The rats were then resuscitated with shed blood plus lactated Ringers (2X shed blood) over 3 h. Lung leak was assessed by transudation of Evan's blue dye into the alveolus as measured by bronchoalveolar lavage. Isolated human PMNs were incubated with 1 and 10% lymph; priming was measured by the fMLP (1 microM)-stimulated production of superoxide and surface expression of CD11b determined by flow cytometry. Results. Mesenteric lymph flow increased significantly during resuscitation: preshock 144.4 microl/h, shock 44.5 microl/h, resuscitation 566.6 microl/h. Furthermore, diversion of this lymph abrogated lung injury as compared to rats without lymph diversion. Finally, mesenteric lymph from postshock animals primed PMNs for superoxide production (nearly three times control cells) as well as increased surface expression of CD11b (2-fold over control). Conclusion. Mesenteric lymph primes PMNs and causes lung injury following hemorrhagic shock. Mesenteric lymph provides a conduit for proinflammatory mediators that may participate in the pathogenesis of MOF.


Subject(s)
Lung/physiopathology , Lymph/physiology , Neutrophils/physiology , Shock, Hemorrhagic/physiopathology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Flow Cytometry , Gene Expression Regulation , Humans , Lung Injury , Macrophage-1 Antigen/blood , Macrophage-1 Antigen/genetics , Male , Mesentery , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Rats , Rats, Sprague-Dawley , Superoxides/blood
20.
J Trauma ; 44(4): 592-7; discussion 598, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9555828

ABSTRACT

BACKGROUND: Ongoing clinical trials have revived interest in hypertonic saline (HTS) for postinjury resuscitation; these studies have documented serum Na+ concentrations > or = 170 mmol/L. Recent animal studies have shown that HTS enhances T-cell and monocyte function, but effects on the polymorphonuclear neutrophil (PMN) remain unclear. The postinjury lipid mediators platelet-activating factor (PAF) and leukotriene B4 (LTB4) have been implicated in PMN priming for cytotoxicity, which is believed to be important in the pathogenesis of multiple organ failure. We hypothesized that HTS would stimulate PMN superoxide (O2-) and elastase release from PAF- and LTB4-primed PMNs. METHODS: Isolated PMNs from five donors were primed for 5 minutes with 200 nmol/L PAF or 1 micromol/L LTB4 in Kreb's-Ringer's phosphate with dextrose at a Na+ concentration of 140 mmol/L (normal serum Na+ concentration), pelleted, and resuspended in Kreb's-Ringer's phosphate with dextrose for 10 minutes at a Na+ concentration of 130 to 170 mmol/L. O2- generation was measured by superoxide dismutase-inhibitable reduction of cytochrome c and elastase release by cleavage of N-methoxysuccinyl-Ala-Ala-Pro-Val p-nitroanilide. RESULTS: HTS with Na+ concentration up to 170 mmol/L had no significant effect on O2- production or elastase release from quiescent cells. Na+ concentration of 160 and 170 mmol/L, however, activated PAF- and LTB4-primed PMNs for enhanced elastase release with no effect on O2- production. CONCLUSION: In clinically relevant concentrations, elevated Na+ activates lipid-primed neutrophils for enhanced elastase degranulation. Consequently, the administration of HTS in the early postinjury resuscitation period, when PMNs are maximally primed, may activate PMN elastase release and thereby promote the development of multiple organ failure.


Subject(s)
Leukotriene B4/physiology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Platelet Activating Factor/physiology , Saline Solution, Hypertonic/therapeutic use , Humans , Multiple Organ Failure/etiology , Multiple Trauma/complications , Multiple Trauma/immunology , Multiple Trauma/therapy , Neutrophils/metabolism , Osmolar Concentration , Resuscitation , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL