Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Toxicology ; 502: 153719, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38181850

RESUMEN

Per- and polyfluoroalkyl substances (PFASs), a group of synthetic chemicals that were once widely used for industrial purposes and in consumer products, are widely found in the environment and in human blood due to their extraordinary resistance to degradation. Once inside the body, PFASs can activate nuclear receptors such as PPARα and CAR. The present study aimed to investigate the impact of perfluorooctanoic acid (PFOA) and perfluorodecanoic acid (PFDA) on liver structure and functions, as well as bile acid homeostasis in mice. A single administration of 0.1 mmole/kg of PFDA, not PFOA, elevated serum ALT and bilirubin levels and caused cholestasis in WT mice. PFDA increased total and various bile acid species in serum but decreased them in the liver. Furthermore, in mouse livers, PFDA, not PFOA, down-regulated mRNA expression of uptake transporters (Ntcp, Oatp1a1, 1a4, 1b2, and 2b1) but induced efflux transporters (Bcrp, Mdr2, and Mrp2-4). In addition, PFDA, not PFOA, decreased Cyp7a1, 7b1, 8b1, and 27a1 mRNA expression in mouse livers with concomitant hepatic accumulation of cholesterol. In contrast, in PPARα-null mice, PFDA did not increase serum ALT, bilirubin, or total bile acids, but produced prominent hepatosteatosis; and the observed PFDA-induced expression changes of transporters and Cyps in WT mice were largely attenuated or abolished. In CAR-null mice, the observed PFDA-induced bile acid alterations in WT mice were mostly sustained. These results indicate that, at the dose employed, PFDA has more negative effects than PFOA on liver function. PPARα appears to play a major role in mediating most of PFDA-induced effects, which were absent or attenuated in PPARα-null mice. Lack of PPARα, however, exacerbated hepatic steatosis. Our findings indicate separated roles of PPARα in mediating the adaptive responses to PFDA: protective against hepatosteatosis but exacerbating cholestasis.


Asunto(s)
Caprilatos , Colestasis , Ácidos Decanoicos , Fluorocarburos , Humanos , Ratones , Animales , Ácidos y Sales Biliares/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias , Hígado , Fluorocarburos/metabolismo , Ratones Noqueados , Bilirrubina/toxicidad , Bilirrubina/metabolismo , ARN Mensajero/metabolismo
2.
3.
Neurochem Res ; 48(3): 804-815, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36346495

RESUMEN

Astrocytes play an important role in the pathogenesis of bilirubin neurotoxicity, and activated astrocytes might be potential mediators of neuroinflammation processes contributing to neuronal cell death and tissue injury. Recent studies have reported that activated microglia induce two types of reactive astrocytes. A1 astrocytes could cause neuronal death and synaptic damage, as well as impaired phagocytosis. Therefore, the purpose of this study was to investigate whether unconjugated bilirubin (UCB)-induced A1-like astrocytes take on a neuroinflammation type and the underlying regulatory mechanisms. In this study, primary cortical astrocytes were treated with UCB in vitro. We detected the expression of complement component 3 (C3), S100 calcium binding protein A10 (S100A10), nuclear factor kappa B (NF-κB), NLR family pyrin domain containing 3 (NLRP3), activated caspase-1, gasdermin D N-terminal (GSDMD-N), PSD95, synaptophysin (SYP), the transcription levels of interleukin (IL)-1ß and IL-18, and the survival rate of astrocytes after UCB treatment. The results showed that an increase in C3 was accompanied by a decrease in S100A10, and that A1-like astrocytes were functionally expressed after UCB stimulation. Meanwhile, the NF-κB and caspase-1 pathways were activated after UCB stimulation. After adding the NF-κB-specific inhibitor trans-activator of transcriptional-NEMO-binding domain (TAT-NBD) and caspase-1 specific inhibitor VX-765, the survival rate of astrocytes and neurons increased, whereas the protein expression of C3, NF-κB, NLRP3, activated caspase-1, and GSDMD-N decreased, and the mRNA levels of IL-1ß and IL-18 reduced. Thus, we concluded that UCB stimulates the activation of A1-like astrocytes. Inhibition of NF-κB and caspase-1 alleviated A1-like astrocytes and exerted anti-inflammatory protective effects.


Asunto(s)
Bilirrubina , FN-kappa B , Humanos , Bilirrubina/toxicidad , Bilirrubina/metabolismo , FN-kappa B/metabolismo , Interleucina-18/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias , Caspasa 1/metabolismo
4.
Oxid Med Cell Longev ; 2021: 8869908, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373769

RESUMEN

Delayed ischemic neurological deficit (DIND) is a severe complication after subarachnoid hemorrhage (SAH). Previous studies have suggested that bilirubin oxidation end products (BOXes) are probably associated with the DIND after SAH, but there is a lack of direct evidence yet even on cellular levels. In the present study, we aim to explore the potential role of BOXes and the involved mechanisms in neuronal function. We synthesized high-purity (>97%) BOX A and BOX B isomers. The pharmacokinetics showed they are permeable to the blood-brain barrier. Exposure of a moderate concentration (10 or 30 µM) of BOX A or BOX B to isolated primary cortical neurons increased the production of reactive oxygen species. In the human neuroblastoma SH-SY5Y cells, BOX A and BOX B decreased the mitochondrial membrane potential and enhanced nuclear accumulation of the protein Nrf2 implicated in oxidative injury repair. In addition, both chemicals increased the mRNA and protein expression levels of multiple antioxidant response genes including Hmox1, Gsta3, Blvrb, Gclm, and Srxn1, indicating that the antioxidant response element (ARE) transcriptional cascade driven by Nrf2 is activated. In conclusion, we demonstrated that primary cortical neurons and neuroblastoma cells undergo an adaptive response against BOX A- and BOX B-mediated oxidative stress by activation of multiple antioxidant responses, in part through the Nrf2 pathway, which provides in-depth insights into the pathophysiological mechanism of DIND after SAH or other neurological dysfunctions related to cerebral hemorrhage.


Asunto(s)
Bilirrubina/análogos & derivados , Barrera Hematoencefálica/metabolismo , Neuronas/metabolismo , Oxidantes/toxicidad , Estrés Oxidativo , Animales , Bilirrubina/farmacocinética , Bilirrubina/toxicidad , Línea Celular Tumoral , Células Cultivadas , Glutamato-Cisteína Ligasa/metabolismo , Glutatión Transferasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Masculino , Potencial de la Membrana Mitocondrial , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Oxidantes/síntesis química , Oxidantes/farmacocinética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo
5.
Sci Rep ; 10(1): 11374, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647318

RESUMEN

Newborns with significant neonatal jaundice (SNJ) would admit for evaluation and/or intervention due to an earlier or more rapid increase in bilirubin level. Bilirubin-induced neurological dysfunction in this population might be underestimated. We aimed to investigate the risk of long-term neurodevelopmental sequelae of SNJ in Taiwan. An SNJ 2000-2003 follow-up cohort consisting of 66,983 neonates was extracted from the nationwide, population-based health insurance database in Taiwan to survey the accumulative incidence of long-term (7-year) neurodevelopmental sequelae in comparison to a reference general-population neonate cohort of 12,579 individuals born in 2000. The SNJ follow-up cohort was furtherly categorized into subgroups according to interventions (phototherapy, intensive phototherapy, and exchange transfusion). The SNJ follow-up cohort exhibited significantly higher cumulative rates of long-term neurodevelopmental sequelae than did the reference cohort (P < 0.05). The risks of infantile cerebral palsy, hearing loss, and developmental delay in the SNJ follow-up cohort were between twice and three times of those in the reference cohort after adjusting for gender, comorbid perinatal disorders and urbanization levels. All intervention subgroups demonstrated higher risks for long-term neurodevelopmental sequelae than the reference cohort (P < 0.05) after adjustment. Patients with SNJ are at risk of developing neurodevelopmental disorders during their growth period. A scheduled follow-up protocol of physical and neurodevelopmental assessment during early childhood for these SNJ patients would potentially be helpful for the early detection of and intervention for neurodevelopmental disorders.


Asunto(s)
Eritroblastosis Fetal/epidemiología , Ictericia Neonatal/complicaciones , Trastornos del Neurodesarrollo/epidemiología , Bilirrubina/sangre , Bilirrubina/toxicidad , Niño , Preescolar , Eritroblastosis Fetal/sangre , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Lactante , Recién Nacido , Ictericia Neonatal/sangre , Ictericia Neonatal/epidemiología , Masculino , Trastornos del Neurodesarrollo/etiología , Estudios Retrospectivos , Taiwán/epidemiología
6.
J Alzheimers Dis ; 73(1): 277-295, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31796680

RESUMEN

Hyperbilirubinemia may increase the risk of Alzheimer's disease (AD) but its mechanistic role in AD pathogenesis remains obscure. Here, we used animal models to investigate the short- and long-term effects of neonatal systemic exposure to bilirubin on brain histology and function as well as the acute effect of lateral ventricle injection of bilirubin in adult rats. We found that three days exposure to bilirubin in newborn rats could induce AD-like pathological changes in late life, including tau protein hyperphosphorylation at multiple sites, increased Aß production in brain tissues, and spatial learning and memory injury. Bilirubin activated the activities of several protein kinases (GSK-3ß, CDK5, and JNK), which were positively correlated with hyperphosphorylated tau; simultaneously increased the expression of AßPP γ-secretase PS2 and decreased the expression of α-secretase ADAM17, which were positively correlated with Aß production. The above results were well replicated in primary hippocampal cell cultures. These data demonstrate that bilirubin encephalopathy is an AD-like disease, suggesting a potent role of bilirubin in AD.


Asunto(s)
Enfermedad de Alzheimer/inducido químicamente , Bilirrubina/toxicidad , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Animales , Animales Recién Nacidos , Bilirrubina/administración & dosificación , Encéfalo/patología , Hipocampo/metabolismo , Hipocampo/patología , Inyecciones Intraventriculares , Aprendizaje por Laberinto , Trastornos de la Memoria , Fosforilación , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Proteínas tau/metabolismo
7.
Brain Res Bull ; 154: 102-105, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31733348

RESUMEN

Unconjugated bilirubin, the end product of heme catabolism and antioxidant, induced brain damage in human neonates is a well-recognized clinical syndrome. However, the cellular and molecular mechanisms underlying bilirubin neurotoxicity remain unclear. To characterize the sequence of events leading to bilirubin-induced neurotoxicity, we investigated whether bilirubin-induced glial activation was involved in bilirubin neurotoxicity by exposing co-cultured rat glial cells and cerebellar granule neurons (CGN) to bilirubin. We found that bilirubin could markedly induce the expression of TNF-α and iNOS in glial cells, and even at low concentrations, the co-culture of glial cells with neurons significantly enhances neurotoxicity of bilirubin. Pretreatment of the co-cultured cells with minocycline protected CGN from glia-mediated bilirubin neurotoxicity and inhibited overexpression of TNF-α and iNOS in glia. Furthermore, we found that high doses of bilirubin were able to induce glial injury, and minocycline attenuated bilirubin-induced glial cell death. Our data suggest that glial cells play an important role in brain damage caused by bilirubin, and minocycline blocks bilirubin-induced encephalopathy possibly by directly and indirectly inhibiting neuronal death pathways.


Asunto(s)
Bilirrubina/metabolismo , Minociclina/farmacología , Neuroglía/metabolismo , Animales , Bilirrubina/toxicidad , Muerte Celular/efectos de los fármacos , Cerebelo/citología , Minociclina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Cell Death Dis ; 10(10): 774, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601780

RESUMEN

Neonatal jaundice is prevalent among newborns and can lead to severe neurological deficits, particularly sensorimotor dysfunction. Previous studies have shown that bilirubin (BIL) enhances the intrinsic excitability of central neurons and this can potentially contribute to their overexcitation, Ca2+ overload, and neurotoxicity. However, the cellular mechanisms underlying elevated neuronal excitability remain unknown. By performing patch-clamp recordings from neonatal neurons in the rat medial vestibular nucleus (MVN), a crucial relay station for locomotor and balance control, we found that BIL (3 µM) drastically increases the spontaneous firing rates by upregulating the current-mediated voltage-gated sodium channels (VGSCs), while shifting their voltage-dependent activation toward more hyperpolarized potentials. Immunofluorescence labeling and western immunoblotting with an anti-NaV1.1 antibody, revealed that BIL elevates the expression of VGSCs by promoting their recruitment to the membrane. Furthermore, we found that this VGSC-trafficking process is Ca2+ dependent because preloading MVN neurons with the Ca2+ buffer BAPTA-AM, or exocytosis inhibitor TAT-NSF700, prevents the effects of BIL, indicating the upregulated activity and density of functional VGSCs as the core mechanism accountable for the BIL-induced overexcitation of neonatal neurons. Most importantly, rectification of such overexcitation with a low dose of VGSC blocker lidocaine significantly attenuates BIL-induced cell death. We suggest that this enhancement of VGSC currents directly contributes to the vulnerability of neonatal brain to hyperbilirubinemia, implicating the activity and trafficking of NaV1.1 channels as a potential target for neuroprotection in cases of severe jaundice.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Bilirrubina/toxicidad , Calcio/metabolismo , Neuronas/efectos de los fármacos , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Muerte Celular , Exocitosis/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Núcleos Vestibulares/citología , Núcleos Vestibulares/efectos de los fármacos , Núcleos Vestibulares/metabolismo
9.
Mol Cell Neurosci ; 96: 35-46, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30877033

RESUMEN

Elevated levels of unbound unconjugated bilirubin (UCB) can lead to bilirubin encephalopathy and kernicterus. In spite of a large number of studies demonstrating UCB-induced changes in central neurotransmission, it is still unclear whether these effects involve alterations in the function of specific ion channels. To assess how different UCB concentrations and UCB:albumin (U/A) molar ratios affect neuronal R-type voltage-gated Ca2+ channels, we evaluated their effects on whole-cell currents through recombinant Cav2.3 + ß3 channel complexes and ex-vivo electroretinograms (ERGs) from wildtype and Cav2.3-deficient mice. Our findings show that modestly elevated levels of unbound UCB (U/A = 0.5) produce subtle but significant changes in the voltage-dependence of activation and prepulse inactivation, resulting in a stimulation of currents activated by weak depolarization and inhibition at potentials on the plateau of the activation curve. Saturation of the albumin binding capacity (U/A = 1) produced additional suppression that became significant when albumin was omitted completely and might involve a complete loss of channel function. Acutely administered UCB (U/A = 0.5) has recently been shown to affect transsynaptic signaling in the isolated vertebrate retina. The present report reveals that sustained exposure of the murine retina to UCB significantly suppresses also late responses of the inner retina (b-wave) from wildtype compared to Cav2.3-deficient mice. In addition, recovery during washout was significantly more complete and faster in retinae lacking Cav2.3 channels. Together, these findings show that UCB affects cloned and native Cav2.3 channels at clinically relevant U/A molar ratios and indicate that supersaturation of albumin is not required for modulation but associated with a loss of channel functional that could contribute to chronic neuronal dysfunction.


Asunto(s)
Bilirrubina/farmacología , Canales de Calcio Tipo R/metabolismo , Proteínas de Transporte de Catión/metabolismo , Retina/efectos de los fármacos , Potenciales de Acción , Animales , Bilirrubina/toxicidad , Células HEK293 , Humanos , Masculino , Ratones , Retina/metabolismo , Retina/fisiología
10.
Neonatology ; 115(3): 217-225, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30645995

RESUMEN

BACKGROUND: The physiopathology of bilirubin-induced neurological disorders is not completely understood. OBJECTIVES: The aim of our study was to assess the effect on bilirubin neurotoxicity of the maturity or immaturity of exposed cells, the influence of different unconjugated bilirubin (UCB) and human serum albumin (HSA) concentrations, and time of UCB exposure. METHODS: Organotypic hippocampal slices were exposed for 48 h to different UCB and HSA concentrations after 14 (mature) or 7 (immature) days of in vitro culture. Immature slices were also exposed to UCB and HSA for 72 h. The different effects of exposure time to UCB on neurons and astrocytes were evaluated. RESULTS: We found that 48 h of UCB exposure was neurotoxic for mature rat organotypic hippocampal slices while 72 h of exposure was neurotoxic for immature slices. Forty-eight-hour UCB exposure was toxic for astrocytes but not for neurons, while 72-h exposure was toxic for both astrocytes and neurons. HSA prevented UCB toxicity when the UCB:HSA molar ratio was ≤1 in both mature and immature slices. CONCLUSIONS: We confirmed UCB neurotoxicity in mature and immature rat hippocampal slices, although immature ones were more resistant. HSA was effective in preventing UCB neurotoxicity in both mature and immature rat hippocampal slices.


Asunto(s)
Bilirrubina/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Fármacos Neuroprotectores/farmacología , Albúmina Sérica Humana/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Humanos , Ictericia/fisiopatología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar
11.
Pediatr Res ; 85(2): 183-190, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30518884

RESUMEN

Neonatal hyperbilirubinemia is one of the most frequent diagnoses made in neonates. A high level of unconjugated bilirubin that is unbound to albumin is neurotoxic when the level exceeds age-specific thresholds or at lower levels in neonates with neurotoxic risk factors. Lower range of unbound bilirubin results in apoptosis, while moderate-to-high levels result in neuronal necrosis. Basal ganglia and various brain stem nuclei are more susceptible to bilirubin toxicity. Proposed mechanisms of bilirubin-induced neurotoxicity include excessive release of glutamate, mitochondrial energy failure, release of proinflammatory cytokines, and increased intracellular calcium concentration. These mechanisms are similar to the events that occur following hypoxic-ischemic insult in neonates. Severe hyperbilirubinemia in term neonates has been shown to be associated with increased risk for autism spectrum disorders. The neuropathological finding of bilirubin-induced neurotoxicity also includes cerebellar injury with a decreased number of Purkinje cells, and disruption of multisensory feedback loop between cerebellum and cortical neurons which may explain the clinical characteristics of autism spectrum disorders. Severe hyperbilirubinemia occurs more frequently in infants from low- and middle-income countries (LMIC). Simple devices to measure bilirubin, and timely treatment are essential to reduce neurotoxicity, and improve outcomes for thousands of neonates around the world.


Asunto(s)
Bilirrubina/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Neurotoxinas/toxicidad , Bilirrubina/biosíntesis , Humanos , Hiperbilirrubinemia Neonatal/fisiopatología , Hiperbilirrubinemia Neonatal/terapia , Recién Nacido
12.
Neurotoxicology ; 71: 75-86, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30578813

RESUMEN

Exposure to high levels of bilirubin in hyperbilirubinemia patients and animal models can result in sensorineural deafness. However, the mechanisms underlying bilirubin-induced damage to the inner ear, including the cochlear and vestibular organs, remain unknown. The present analyses of cochlear and vestibular organotypic cultures obtained from postnatal day 3 rats exposed to bilirubin at varying concentrations (0, 10, 50, 100, or 250 µM) for 24 h revealed that auditory nerve fibers (ANFs) and vestibular nerve endings were destroyed even at low doses (10 and 50 µM). Additionally, as the bilirubin dose increased, spiral ganglion neurons (SGNs) and vestibular ganglion neurons (VGNs) exhibited gradual shrinkage in conjunction with nuclei condensation or fragmentation in a dose-dependent manner. The loss of cochlear and vestibular hair cells (HCs) was only evident in explants treated with the highest concentration of bilirubin (250 µM), and bilirubin-induced major apoptosis most likely occurred via the extrinsic apoptotic pathway. Thus, the present results indicate that inner ear neurons and fibers were more sensitive to, and exhibited more severe damage following, bilirubin-induced neurotoxicity than sensory HCs, which illustrates the underlying causes of auditory neuropathy and vestibulopathy in hyperbilirubinemia patients.


Asunto(s)
Bilirrubina/toxicidad , Neuronas/efectos de los fármacos , Ototoxicidad/patología , Ganglio Espiral de la Cóclea/efectos de los fármacos , Vestíbulo del Laberinto/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Nervio Coclear/efectos de los fármacos , Nervio Coclear/patología , Neuronas/patología , Técnicas de Cultivo de Órganos , Ratas Sprague-Dawley , Ganglio Espiral de la Cóclea/patología , Nervio Vestibular/efectos de los fármacos , Nervio Vestibular/patología , Vestíbulo del Laberinto/patología
13.
PLoS One ; 13(8): e0201022, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30106954

RESUMEN

Hyperbilirubinemia (jaundice) is caused by raised levels of unconjugated bilirubin in the blood. When severe, susceptible brain regions including the cerebellum and auditory brainstem are damaged causing neurological sequelae such as ataxia, hearing loss and kernicterus. The mechanism(s) by which bilirubin exerts its toxic effect have not been completely understood to date. In this study we investigated the acute mechanisms by which bilirubin causes the neurotoxicity that contributes to hearing loss. We developed a novel mouse model that exhibits the neurological features seen in human Bilirubin-Induced Neurological Dysfunction (BIND) syndrome that we assessed with a behavioural score and auditory brainstem responses (ABR). Guided by initial experiments applying bilirubin to cultured cells in vitro, we performed whole genome gene expression measurements on mouse brain tissue (cerebellum and auditory brainstem) following bilirubin exposure to gain mechanistic insights into biochemical processes affected, and investigated further using immunoblotting. We then compared the gene changes induced by bilirubin to bacterial lipopolysaccharide (LPS), a well characterized inducer of neuroinflammation, to assess the degree of similarity between them. Finally, we examined the extent to which genetic perturbation of inflammation and both known and novel anti-inflammatory drugs could protect hearing from bilirubin-induced toxicity. The in vitro results indicated that bilirubin induces changes in gene expression consistent with endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR). These gene changes were similar to the gene expression signature of thapsigargin-a known ER stress inducer. It also induced gene expression changes associated with inflammation and NF-κB activation. The in vivo model showed behavioural impairment and a raised auditory threshold. Whole genome gene expression analysis confirmed inflammation as a key mechanism of bilirubin neurotoxicity in the auditory pathway and shared gene expression hallmarks induced by exposure to bacterial lipopolysaccharide (LPS) a well-characterized inducer of neuroinflammation. Interestingly, bilirubin caused more severe damage to the auditory system than LPS in this model, but consistent with our hypothesis of neuroinflammation being a primary part of bilirubin toxicity, the hearing loss was protected by perturbing the inflammatory response. This was carried out genetically using lipocalin-2 (LCN2)-null mice, which is an inflammatory cytokine highly upregulated in response to bilirubin. Finally, we tested known and novel anti-inflammatory compounds (interfering with NF-κB and TNFα signalling), and also demonstrated protection of the auditory system from bilirubin toxicity. We have developed a novel, reversible, model for jaundice that shows movement impairment and auditory loss consistent with human symptoms. We used this model to establish ER-stress and inflammation as major contributors to bilirubin toxicity. Because of the rapid and reversible onset of toxicity in this novel model it represents a system to screen therapeutic compounds. We have demonstrated this by targeting inflammation genetically and with anti-inflammatory small molecules that offered protection against bilirubin toxicity. This also suggests that anti-inflammatory drugs could be of therapeutic use in hyperbilirubinemia.


Asunto(s)
Bilirrubina/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Pérdida Auditiva/etiología , Kernicterus/etiología , Síndromes de Neurotoxicidad/etiología , Enfermedad Aguda , Animales , Antiinflamatorios/farmacología , Ataxia/etiología , Ataxia/metabolismo , Bilirrubina/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Pérdida Auditiva/metabolismo , Pérdida Auditiva/prevención & control , Humanos , Hiperbilirrubinemia/complicaciones , Hiperbilirrubinemia/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Kernicterus/metabolismo , Lipocalina 2/deficiencia , Lipocalina 2/genética , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos CBA , Ratones Noqueados , FN-kappa B/metabolismo , Síndromes de Neurotoxicidad/metabolismo
14.
Int J Nanomedicine ; 13: 2997-3010, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872292

RESUMEN

BACKGROUND: Activated carbon (AC) is a common adsorbent that is used in both artificial and bioartificial liver devices. METHODS: Three natural materials - date pits of Phoenix dactylifera (fruit), Simmondsia chinensis (jojoba) seeds, and Scenedesmus spp. (microalgae) - were used in the present investigation as precursors for the synthesis of AC using physical activation. The chemical structures and morphology of AC were analyzed. Then, AC's bilirubin adsorption capacity and its cytotoxicity on normal liver (THLE2) and liver cancer (HepG2) cells were characterized. RESULTS: Compared with the other raw materials examined, date-pit AC was highly selective and showed the most effective capacity of bilirubin adsorption, as judged by isotherm-modeling analysis. MTT in vitro analysis indicated that date-pit AC had the least effect on the viability of both THLE2 and HepG2 cells compared to jojoba seeds and microalgae. All three biomaterials under investigation were used, along with collagen and Matrigel, to grow cells in 3D culture. Fluorescent microscopy confirmed date-pit AC as the best to preserve liver cell integrity. CONCLUSION: The findings of this study introduce date-pit-based AC as a novel alternative biomaterial for the removal of protein-bound toxins in bioartificial liver devices.


Asunto(s)
Bilirrubina/farmacocinética , Carbón Orgánico/química , Magnoliopsida/química , Phoeniceae/química , Scenedesmus/química , Adsorción , Albúminas/química , Bilirrubina/química , Bilirrubina/toxicidad , Línea Celular , Células Hep G2 , Humanos , Inactivación Metabólica , Hígado/citología , Hígado Artificial , Semillas/química
15.
J Neuroinflammation ; 15(1): 23, 2018 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-29357878

RESUMEN

BACKGROUND: Bilirubin-induced neurological dysfunction (BIND), a severe complication of extreme neonatal hyperbilirubinemia, could develop into permanent neurodevelopmental impairments. Several studies have demonstrated that inflammation and nerve cell death play important roles in bilirubin-induced neurotoxicity; however, the underlying mechanism remains unidentified. METHODS: The present study was intended to investigate whether pyroptosis, a highly inflammatory form of programmed cell death, participated in the bilirubin-mediated toxicity on cultured rat cortical astrocytes. Further, VX-765, a potent and selective competitive drug, was used to inhibit the activation of caspase-1. The effects of VX-765 on astrocytes treated with bilirubin, including the cell viability, morphological changes of the cell membrane and nucleus, and the production of pro-inflammation cytokines, were observed. RESULTS: Stimulation of the astrocytes with unconjugated bilirubin (UCB) at the conditions mimicking those of jaundiced newborns significantly increased the activation of caspase-1. Further, caspase-1 activation was inhibited by treatment with VX-765. Compared with UCB-treated astrocytes, the relative cell viability of VX-765-pretreated astrocytes was improved; meanwhile, the formation of plasma membrane pores was prevented, as measured by lactate dehydrogenase release, trypan blue staining, and ethidium bromide (EtBr) uptake. Moreover, DNA fragmentation was partly attenuated and the release of IL-1ß and IL-18 was apparently decreased. CONCLUSION: Pyroptosis is involved in the process of UCB-induced rat cortical astrocytes' injury in vitro and may be the missing link of cell death and inflammatory response exacerbating UCB-related neurotoxicity. More importantly, the depression of caspase-1 activation, the core link of pyroptosis, attenuated UCB-induced cellular dysfunction and cytokine release, which might shed light on a new therapeutic approach to BIND.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Bilirrubina/toxicidad , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Piroptosis/efectos de los fármacos , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Piroptosis/fisiología , Ratas , Ratas Sprague-Dawley
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt B): 1356-1366, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28851656

RESUMEN

Acute kidney injury is common in patients with liver disease and associated with significant morbidity and mortality. Besides bacterial infections, fluid loss, and use of nephrotoxic drugs AKI in liver disease may be triggered by tubular toxicity of cholephiles. Cholemic nephropathy, also known as bile cast nephropathy, supposedly represents a widely underestimated but important cause of renal dysfunction in cholestasic or advanced liver diseases with jaundice. Cholemic nephropathy describes impaired renal function along with characteristic histomorphological changes consisting of intratubular cast formation and tubular epithelial cell injury directed towards distal nephron segments. The underlying pathophysiologic mechanisms are not entirely understood and clear defined diagnostic criteria are still missing. This review aims to summarize (i) the present knowledge on clinical and morphological characteristics of cholemic nephropathy, (ii) available preclinical models, (iii) potential pathomechanisms especially the potential role of bile acids, and (iv) future diagnostic and therapeutic strategies for cholemic nephropathy. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.


Asunto(s)
Lesión Renal Aguda/etiología , Ácidos y Sales Biliares/toxicidad , Colestasis/complicaciones , Ictericia Obstructiva/complicaciones , Túbulos Renales/patología , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Animales , Ácidos y Sales Biliares/metabolismo , Conductos Biliares/citología , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Conductos Biliares/patología , Bilirrubina/metabolismo , Bilirrubina/toxicidad , Colagogos y Coleréticos/farmacología , Colagogos y Coleréticos/uso terapéutico , Colestasis/tratamiento farmacológico , Colestasis/patología , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ictericia Obstructiva/tratamiento farmacológico , Ictericia Obstructiva/patología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Eliminación Renal , Ácido Ursodesoxicólico/análogos & derivados , Ácido Ursodesoxicólico/farmacología , Ácido Ursodesoxicólico/uso terapéutico
17.
Toxicol Mech Methods ; 28(1): 55-61, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28805483

RESUMEN

The aim of this study is to identify the role of Tet1-mediated DNA demethylation in the neurotoxicity caused by unconjugated bilirubin (UCB) in vitro. Primary neuronal cells after cultured for 72 h were exposed to UCB (0-100 µmol/L) for 24 h. Following exposure to UCB cytotoxicity was determined with the methyl tetrazolium (MTT) assay, reactive oxygen species (ROS) and caspase-3 activity in neuron cells were measured with the corresponding assay kits. The expression of Tet1 and Klotho was determined with RT-PCR at mRNA level and western blot at protein level. Our results showed that UCB can cause time-dependent and dose-dependent reduction of cell viability of neuronal cells, induce oxidative stress through increasing the production of ROS and increase caspase-3 activity. Quantitative real-time PCR and western blot analysis showed that UCB can inhibit Tet1 and Klotho expression in cultured neuronal cells at both the mRNA and protein level, respectively. These results are first to suggest UCB may, in part, exert its neurotoxicity through alteration of the neuronal antioxidant status and inhibition of Klotho and Tet1 gene expression. The elevation of DNA methylation in global genome through inhibition of Tet1 gene expression may, in part, play an important role in the neurotoxicity caused by UCB in vitro.


Asunto(s)
Bilirrubina/toxicidad , Corteza Cerebral/efectos de los fármacos , Desmetilación del ADN/efectos de los fármacos , Dioxigenasas/metabolismo , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Dioxigenasas/genética , Relación Dosis-Respuesta a Droga , Femenino , Glucuronidasa/genética , Glucuronidasa/metabolismo , Proteínas Klotho , Masculino , Neuronas/enzimología , Neuronas/patología , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo/efectos de los fármacos , Cultivo Primario de Células , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
18.
Chem Biol Interact ; 278: 129-134, 2017 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-29079291

RESUMEN

Toxic effects of unconjugated bilirubin (BR) in neonatal hyperbilirubinemia have been related to redox and/or coordinate interactions with Cu2+. However, the development and mechanisms of such interactions at physiological pH have not been resolved. This study shows that BR reduces Cu2+ to Cu1+ in 1:1 stoichiometry. Apparently, BR undergoes degradation, i.e. BR and Cu2+ do not form stable complexes. The binding of Cu2+ to inorganic phosphates, liposomal phosphate groups, or to chelating drug penicillamine, impedes redox interactions with BR. Cu1+ undergoes spontaneous oxidation by O2 resulting in hydrogen peroxide accumulation and hydroxyl radical production. In relation to this, copper and BR induced synergistic oxidative/damaging effects on erythrocytes membrane, which were alleviated by penicillamine. The production of reactive oxygen species by BR and copper represents a plausible cause of BR toxic effects and cell damage in hyperbilirubinemia. Further examination of therapeutic potentials of copper chelators in the treatment of severe neonatal hyperbilirubinemia is needed.


Asunto(s)
Bilirrubina/química , Cobre/química , Penicilamina/química , Bilirrubina/toxicidad , Células Cultivadas , Cobre/toxicidad , Espectroscopía de Resonancia por Spin del Electrón , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Humanos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Radical Hidroxilo/química , Radical Hidroxilo/metabolismo , Oxidación-Reducción , Fosfatos/química , Espectrofotometría Ultravioleta
19.
Sci Rep ; 7: 41032, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-28102362

RESUMEN

The neurologic manifestations of neonatal hyperbilirubinemia in the central nervous system (CNS) exhibit high variations in the severity and appearance of motor, auditory and cognitive symptoms, which is suggestive of a still unexplained selective topography of bilirubin-induced damage. By applying the organotypic brain culture (OBC: preserving in vitro the cellular complexity, connection and architecture of the in vivo brain) technique to study hyperbilirubinemia, we mapped the regional target of bilirubin-induced damage, demonstrated a multifactorial toxic action of bilirubin, and used this information to evaluate the efficacy of drugs applicable to newborns to protect the brain. OBCs from 8-day-old rat pups showed a 2-13 fold higher sensitivity to bilirubin damage than 2-day-old preparations. The hippocampus, inferior colliculus and cerebral cortex were the only brain regions affected, presenting a mixed inflammatory-oxidative mechanism. Glutamate excitotoxicity was appreciable in only the hippocampus and inferior colliculus. Single drug treatment (indomethacin, curcumin, MgCl2) significantly improved cell viability in all regions, while the combined (cocktail) administration of the three drugs almost completely prevented damage in the most affected area (hippocampus). Our data may supports an innovative (complementary to phototherapy) approach for directly protecting the newborn brain from bilirubin neurotoxicity.


Asunto(s)
Bilirrubina/toxicidad , Encefalopatías Metabólicas/patología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encefalitis/patología , Hiperbilirrubinemia/complicaciones , Animales , Supervivencia Celular/efectos de los fármacos , Curcumina/farmacología , Indometacina/farmacología , Inflamación/patología , Cloruro de Magnesio/farmacología , Modelos Biológicos , Fármacos Neuroprotectores/farmacología , Técnicas de Cultivo de Órganos , Estrés Oxidativo , Ratas
20.
Hum Mol Genet ; 26(1): 145-157, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28025333

RESUMEN

Moderate neonatal jaundice is the most common clinical condition during newborn life. However, a combination of factors may result in acute hyperbilirubinemia, placing infants at risk of developing bilirubin encephalopathy and death by kernicterus. While most risk factors are known, the mechanisms acting to reduce susceptibility to bilirubin neurotoxicity remain unclear. The presence of modifier genes modulating the risk of developing bilirubin-induced brain damage is increasingly being recognised. The Abcb1 and Abcc1 members of the ABC family of transporters have been suggested to have an active role in exporting unconjugated bilirubin from the central nervous system into plasma. However, their role in reducing the risk of developing neurological damage and death during neonatal development is still unknown.To this end, we mated Abcb1a/b-/- and Abcc1-/- strains with Ugt1-/- mice, which develop severe neonatal hyperbilirubinemia. While about 60% of Ugt1-/- mice survived after temporary phototherapy, all Abcb1a/b-/-/Ugt1-/- mice died before postnatal day 21, showing higher cerebellar levels of unconjugated bilirubin. Interestingly, Abcc1 role appeared to be less important.In the cerebellum of Ugt1-/- mice, hyperbilirubinemia induced the expression of Car and Pxr nuclear receptors, known regulators of genes involved in the genotoxic response.We demonstrated a critical role of Abcb1 in protecting the cerebellum from bilirubin toxicity during neonatal development, the most clinically relevant phase for human babies, providing further understanding of the mechanisms regulating bilirubin neurotoxicity in vivo. Pharmacological treatments aimed to increase Abcb1 and Abcc1 expression, could represent a therapeutic option to reduce the risk of bilirubin neurotoxicity.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Bilirrubina/toxicidad , Cerebelo/patología , Modelos Animales de Enfermedad , Glucuronosiltransferasa/fisiología , Hiperbilirrubinemia Neonatal/complicaciones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Síndromes de Neurotoxicidad/etiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Animales Recién Nacidos , Supervivencia Celular , Cerebelo/efectos de los fármacos , Femenino , Humanos , Hiperbilirrubinemia Neonatal/metabolismo , Hiperbilirrubinemia Neonatal/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...