Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
3.
J Pediatric Infect Dis Soc ; 10(5): 556-561, 2021 May 28.
Article En | MEDLINE | ID: mdl-33367801

BACKGROUND: In general, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy is not considered to be an increased risk for severe maternal outcomes but has been associated with an increased risk for fetal distress. Maternal-fetal transmission of SARS-CoV-2 was initially deemed uncertain; however, recently a few cases of vertical transmission have been reported. The intrauterine mechanisms, besides direct vertical transmission, leading to the perinatal adverse outcomes are not well understood. METHODS: Multiple maternal, placental, and neonatal swabs were collected for the detection of SARS-CoV-2 using real-time quantitative polymerase chain reaction (RT-qPCR). Serology of immunoglobulins against SARS-CoV-2 was tested in maternal, umbilical cord, and neonatal blood. Placental examination included immunohistochemical investigation against SARS-CoV-2 antigen expression, with SARS-CoV-2 ribonucleic acid (RNA) in situ hybridization and transmission electron microscopy. RESULTS: RT-qPCRs of the oropharynx, maternal blood, vagina, placenta, and urine were all positive over a period of 6 days, while breast milk, feces, and all neonatal samples tested negative. Placental findings showed the presence of SARS-CoV-2 particles with generalized inflammation characterized by histiocytic intervillositis with diffuse perivillous fibrin depositions with damage to the syncytiotrophoblasts. CONCLUSIONS: Placental infection by SARS-CoV-2 leads to fibrin depositions hampering fetal-maternal gas exchange with resulting fetal distress necessitating a premature emergency cesarean section. Postpartum, the neonate showed a fetal or pediatric inflammatory multisystem-like syndrome with coronary artery ectasia temporarily associated with SARS-CoV-2 for which admittance and care on the neonatal intensive care unit (NICU) were required, despite being negative for SARS-CoV-2. This highlights the need for awareness of adverse fetal and neonatal outcomes during the current coronavirus disease 2019 pandemic, especially considering that the majority of pregnant women appear asymptomatic.


COVID-19/transmission , Fetal Distress/virology , Infectious Disease Transmission, Vertical , Multiple Organ Failure/virology , Placenta/virology , Pneumonia, Viral/transmission , Pregnancy Complications, Infectious/virology , Adult , Cesarean Section , Female , Humans , Infant, Newborn , Infant, Premature , Pneumonia, Viral/virology , Pregnancy , SARS-CoV-2
5.
Mol Med ; 21(1): 758-768, 2016 Jan.
Article En | MEDLINE | ID: mdl-26467707

Spinocerebellar ataxia type 3 (SCA3) is a polyglutamine (polyQ) disorder caused by a CAG repeat expansion in the ataxin-3 (ATXN3) gene resulting in toxic protein aggregation. Inflammation and oxidative stress are considered secondary factors contributing to the progression of this neurodegenerative disease. There is no cure that halts or reverses the progressive neurodegeneration of SCA3. Here we show that overexpression of cystathionine γ-lyase, a central enzyme in cysteine metabolism, is protective in a Drosophila model for SCA3. SCA3 flies show eye degeneration, increased oxidative stress, insoluble protein aggregates, reduced levels of protein persulfidation and increased activation of the innate immune response. Overexpression of Drosophila cystathionine γ-lyase restores protein persulfidation, decreases oxidative stress, dampens the immune response and improves SCA3-associated tissue degeneration. Levels of insoluble protein aggregates are not altered; therefore, the data implicate a modifying role of cystathionine γ-lyase in ameliorating the downstream consequence of protein aggregation leading to protection against SCA3-induced tissue degeneration. The cystathionine γ-lyase expression is decreased in affected brain tissue of SCA3 patients, suggesting that enhancers of cystathionine γ-lyase expression or activity are attractive candidates for future therapies.

6.
Nitric Oxide ; 49: 56-66, 2015 Sep 15.
Article En | MEDLINE | ID: mdl-26192363

Hydrogen sulfide (H2S), carbon monoxide (CO) and nitric oxide (NO) share signaling and vasorelaxant properties and are involved in proliferation and apoptosis. Inhibiting NO production or availability induces hypertension and proteinuria, which is prevented by concomitant blockade of the H2S producing enzyme cystathionine γ-lyase (CSE) by d,l-propargylglycine (PAG). We hypothesized that blocking H2S production ameliorates Angiotensin II (AngII)-induced hypertension and renal injury in a rodent model. Effects of concomitant administration of PAG or saline were therefore studied in healthy (CON) and AngII hypertensive rats. In CON rats, PAG did not affect systolic blood pressure (SBP), but slightly increased proteinuria. In AngII rats PAG reduced SBP, proteinuria and plasma creatinine (180 ± 12 vs. 211 ± 19 mmHg; 66 ± 35 vs. 346 ± 92 mg/24 h; 24 ± 6 vs. 47 ± 15 µmol/L, respectively; p < 0.01). Unexpectedly, kidney to body weight ratio was increased in all groups by PAG (p < 0.05). Renal injury induced by AngII was reduced by PAG (p < 0.001). HO-1 gene expression was increased by PAG alone (p < 0.05). PAG increased inner cortical tubular cell proliferation after 1 week and decreased outer cortical tubular nucleus number/field after 4 weeks. In vitro proximal tubular cell size increased after exposure to PAG. In summary, blocking H2S production with PAG reduced SBP and renal injury in AngII infused rats. Independent of the cardiovascular and renal effects, PAG increased HO-1 gene expression and kidney weight. PAG alone increased tubular cell size and proliferation in-vivo and in-vitro. Our results are indicative of a complex interplay of gasotransmitter signaling/action of mutually compensatory nature in the kidney.


Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Alkynes/pharmacology , Angiotensin II/adverse effects , Blood Pressure/drug effects , Glycine/analogs & derivatives , Hydrogen Sulfide/metabolism , Alkynes/administration & dosage , Animals , Cell Proliferation , Glycine/administration & dosage , Glycine/pharmacology , Heme Oxygenase-1/metabolism , Kidney/drug effects , Kidney/pathology , Male , Nitric Oxide , Organ Size , Rats , Rats, Sprague-Dawley
7.
Ned Tijdschr Geneeskd ; 159: A8886, 2015.
Article Nl | MEDLINE | ID: mdl-26131751

The parents of a 5-month-old boy noticed bowing of his left leg. Radiographic survey showed an anterolateral bowing of the left tibia and fibula with intramedullary sclerosis, typical for neurofibromatosis type 1. As the mother had neurofibromatosis, this boy was diagnosed with neurofibromatosis type 1.


Fibula/pathology , Neurofibromatosis 1/diagnosis , Tibia/pathology , Fibula/diagnostic imaging , Humans , Infant , Leg , Male , Neurofibromatosis 1/genetics , Radiography , Tibia/diagnostic imaging
8.
Nitric Oxide ; 42: 87-98, 2014 Nov 15.
Article En | MEDLINE | ID: mdl-25459997

Hypertension and proteinuria are important mediators of renal damage. Despite therapeutic interventions, the number of patients with end stage renal disease steadily increases. Hydrogen sulfide (H(2)S) is an endogenously produced gasotransmitter with vasodilatory, anti-inflammatory and antioxidant properties. These beneficial characteristics make H(2)S an attractive candidate for pharmacological use in hypertensive renal disease. We investigated the protective properties of H(2)S in angiotensin II (Ang II)-induced hypertensive renal disease in rats. Treatment with the H(2)S donor NaHS and major H(2)S metabolite sodium thiosulfate (STS) during three weeks of Ang II infusion reduced hypertension, proteinuria, oxidative stress and renal functional and structural deterioration. In an ex vivo isolated perfused kidney setup, NaHS, but not STS, reduced intrarenal pressure. The effect of NaHS could partially be explained by its activation of the ATP-sensitive potassium channels. In conclusion, treatment with H(2)S attenuates Ang II-associated functional and structural renal deterioration, suggesting that intervention in H(2)S production pathways has potential therapeutic benefit and might be a valuable addition to the already existing antihypertensive and renoprotective therapies.


Angiotensin II/physiology , Hypertension/chemically induced , Kidney/drug effects , Proteinuria/chemically induced , Thiosulfates/pharmacology , Animals , Base Sequence , DNA Primers , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
9.
PLoS One ; 8(5): e63291, 2013.
Article En | MEDLINE | ID: mdl-23675473

BACKGROUND: Ischemia-reperfusion injury (IRI) is a major cause of cardiac damage following various pathological processes. Gaseous hydrogen sulfide (H2S) is protective during IRI by inducing a hypometabolic state in mice which is associated with anti-apoptotic, anti-inflammatory and antioxidant properties. We investigated whether gaseous H2S administration is protective in cardiac IRI and whether non-hypometabolic concentrations of H2S have similar protective properties. METHODS: Male C57BL/6 mice received a 0, 10, or 100 ppm H2S-N2 mixture starting 30 minutes prior to ischemia until 5 minutes pre-reperfusion. IRI was inflicted by temporary ligation of the left coronary artery for 30 minutes. High-resolution respirometry equipment was used to assess CO2-production and blood pressure was measured using internal transmitters. The effects of H2S were assessed by histological and molecular analysis. RESULTS: Treatment with 100 ppm H2S decreased CO2-production by 72%, blood pressure by 14% and heart rate by 25%, while treatment with 10 ppm H2S had no effects. At day 1 of reperfusion 10 ppm H2S showed no effect on necrosis, while treatment with 100 ppm H2S reduced necrosis by 62% (p<0.05). Seven days post-reperfusion, both 10 ppm (p<0.01) and 100 ppm (p<0.05) H2S showed a reduction in fibrosis compared to IRI animals. Both 10 ppm and 100 ppm H2S reduced granulocyte-influx by 43% (p<0.05) and 60% (p<0.001), respectively. At 7 days post-reperfusion both 10 and 100 ppm H2S reduced expression of fibronectin by 63% (p<0.05) and 67% (p<0.01) and ANP by 84% and 63% (p<0.05), respectively. CONCLUSIONS: Gaseous administration of H2S is protective when administered during a cardiac ischemic insult. Although hypometabolism is restricted to small animals, we now showed that low non-hypometabolic concentrations of H2S also have protective properties in IRI. Since IRI is a frequent cause of myocardial damage during percutaneous coronary intervention and cardiac transplantation, H2S treatment might lead to novel therapeutical modalities.


Hydrogen Sulfide/administration & dosage , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Animals , Atrial Natriuretic Factor/genetics , Blood Pressure/drug effects , Carbon Dioxide/metabolism , Cell Line , Disease Models, Animal , Gene Expression , Heart Rate/drug effects , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Membrane Glycoproteins/genetics , Mice , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Myocardium/pathology , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genetics , Oxidative Stress/drug effects , Rats
10.
J Am Soc Nephrol ; 24(5): 759-70, 2013 Apr.
Article En | MEDLINE | ID: mdl-23449534

Hydrogen sulfide (H2S) is an endogenous gasotransmitter with physiologic functions similar to nitric oxide and carbon monoxide. Exogenous treatment with H2S can induce a reversible hypometabolic state, which can protect organs from ischemia/reperfusion injury, but whether cystathionine γ-lyase (CSE), which produces endogenous H2S, has similar protective effects is unknown. Here, human renal tissue revealed abundant expression of CSE, localized to glomeruli and the tubulointerstitium. Compared with wild-type mice, CSE knockout mice had markedly reduced renal production of H2S, and CSE deficiency associated with increased damage and mortality after renal ischemia/reperfusion injury. Treatment with exogenous H2S rescued CSE knockout mice from the injury and mortality associated with renal ischemia. In addition, overexpression of CSE in vitro reduced the amount of reactive oxygen species produced during stress. Last, the level of renal CSE mRNA at the time of organ procurement positively associated with GFR 14 days after transplantation. In summary, these results suggest that CSE protects against renal ischemia/reperfusion injury, likely by modulating oxidative stress through the production of H2S.


Cystathionine gamma-Lyase/physiology , Kidney/blood supply , Oxidative Stress , Reperfusion Injury/prevention & control , Adolescent , Adult , Aged , Animals , Cell Survival , Cystathionine beta-Synthase/physiology , Cystathionine gamma-Lyase/analysis , Cystathionine gamma-Lyase/genetics , DNA Damage , Female , HEK293 Cells , Humans , Hydrogen Sulfide/metabolism , Kidney/enzymology , Kidney Transplantation , Male , Mice , Mice, Inbred C57BL , Middle Aged , Renin/analysis , Superoxides/metabolism
11.
Transpl Int ; 25(8): 897-908, 2012 Aug.
Article En | MEDLINE | ID: mdl-22716165

Hydrogen sulfide (H2 S) can induce a reversible hypometabolic state, which could protect against hypoxia. In this study we investigated whether H2 S could protect livers from ischemia/reperfusion injury (IRI). Male C57BL/6 mice were subjected to partial hepatic IRI for 60 min. Animals received 0 (IRI) or 100 ppm H2 S (IRI + H2 S) from 30 min prior to ischemia until 5 min before reperfusion. Core body temperature was maintained at 37° C. Animals were sacrificed after 1, 6 or 24 h. Hepatic ischemia caused extensive hepatic necrosis in the IRI animals which coincided with an increase in ALT and AST serum levels. Animals treated with H2 S showed attenuated serum ALT and AST levels and reduced necrotic lesions after 24 h. IRI animals had increased Bcl-2 mRNA expression and increased active Caspase 3 protein, which were both significantly lower in H2 S treated animals. Increased TNFα and IL-6 mRNA in the IRI livers was significantly attenuated by H2 S treatment, as was hepatic influx of Ly-6G positive granulocytes. Hepatic superoxide production after ischemia was attenuated by H2 S treatment. In hepatic ischemia/reperfusion injury, gaseous H2 S treatment is highly protective, substantially reducing necrosis, apoptosis and inflammation. Gaseous H2 S is therefore a very promising treatment for reducing IRI during hepatic transplantation.


Hydrogen Sulfide/therapeutic use , Reperfusion Injury/prevention & control , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Hydrogen Sulfide/pharmacology , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Superoxides/metabolism
12.
J Am Soc Nephrol ; 20(9): 1901-5, 2009 Sep.
Article En | MEDLINE | ID: mdl-19628669

Hydrogen sulfide (H(2)S) can induce a hypometabolic, hibernation-like state in mammals when given in subtoxic concentrations. Pharmacologically reducing the demand for oxygen is a promising strategy to minimize unavoidable hypoxia-induced injury such as ischemia/reperfusion injury during renal transplantation. Here we show that H(2)S reduces metabolism in vivo, ex vivo, and in vitro. Furthermore, we demonstrate the beneficial effects of H(2)S-induced hypometabolism in a model of bilateral renal ischemia/reperfusion injury using three different treatment strategies. The results demonstrate striking protective effects on survival, renal function, apoptosis, and inflammation. A hypometabolic state induced by H(2)S might have therapeutic potential to protect kidneys that suffer from hypoxia.


Hydrogen Sulfide/pharmacology , Kidney Diseases/metabolism , Kidney Diseases/prevention & control , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Adaptation, Physiological/physiology , Air Pollutants/pharmacology , Animals , Disease Models, Animal , Energy Metabolism/drug effects , Hibernation/physiology , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred F344
...