Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Diabetes Investig ; 14(1): 15-18, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36074333

RESUMO

Insulin therapy using insulin purified from porcine or bovine pancreas revolutionized diabetes therapy in the 1920s. A series of advances including cloning human insulin cDNA enabled the development of recombinant human insulin with improved features. Insulin treatment for diabetes may well be upended by ß-cell replacement therapy in the coming decades.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus , Animais , Bovinos , Humanos , Suínos , Insulina/uso terapêutico , Diabetes Mellitus/tratamento farmacológico , Proteínas Recombinantes
2.
BMC Endocr Disord ; 22(1): 164, 2022 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-35733207

RESUMO

BACKGROUND: Familial hypocalciuric hypercalcemia (FHH) is a rare autosomal dominant disease, which requires differential diagnosis from relatively common primary hyperparathyroidism (PHPT) in order to avoid unnecessary surgery. CASE PRESENTATION: A 16-year-old female had been followed by the department of psychosomatic medicine at our institution. Throughout the follow-up period, her plasma calcium levels were high, plasma Pi levels were relatively low, and plasma intact PTH was relatively high. She was referred to our department to determine the cause of her hypercalcemia. Her 24 h urinary calcium excretion was as low as 100 mg/day, and calcium creatinine clearance ratio was below 0.01. Moreover, she had a family history of hypercalcemia (proband, her brother, and her father). The genetic testing for her family revealed that she, her brother, and her father were definitively diagnosed with FHH type 1 due to the heterozygous calcium-sensing receptor mutation (NM_00388:4:c.164C > T:p.Pro55Leu). CONCLUSION: We experienced a 16-year-old female with FHH, in whom genetic testing identified the heterozygous calcium-sensing receptor mutation (NM_00388:4:c.164C > T:p.Pro55Leu) as pathogenic, permitting a definitive diagnosis of FHH type 1. The genetic testing for calcium sensing receptor is beneficial to distinguish asymptomatic primary hyperparathyroidism from FHH.


Assuntos
Hipercalcemia , Hiperparatireoidismo Primário , Adolescente , Cálcio , Feminino , Humanos , Hipercalcemia/congênito , Hipercalcemia/diagnóstico , Hipercalcemia/genética , Hiperparatireoidismo Primário/diagnóstico , Hiperparatireoidismo Primário/genética , Masculino , Mutação , Receptores de Detecção de Cálcio/genética
3.
Intern Med ; 61(18): 2753-2757, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35228417

RESUMO

We treated a 22-year-old woman suffering from Graves' disease and thymic hyperplasia. She was referred to our institution for a close investigation of thyrotoxicosis and thymic mass. Thyroid tests and magnetic resonance imaging resulted in a diagnosis of Graves' disease and thymic hyperplasia. The thyroid function and thyroid-stimulating hormone receptor antibody (TRAb) were normalized one and five months after thiamazole initiation, respectively. The thymic size began to decrease after 1 month and was further decreased after 5 months; it was normalized after 12 months. The correlation between TRAb titers and the thymic size (R2=0.99) suggested that the patient's autoimmunity might have contributed to the thymic hyperplasia.


Assuntos
Doença de Graves , Hiperplasia do Timo , Adulto , Autoanticorpos , Feminino , Doença de Graves/complicações , Doença de Graves/diagnóstico , Doença de Graves/tratamento farmacológico , Humanos , Metimazol/uso terapêutico , Receptores da Tireotropina , Hiperplasia do Timo/diagnóstico por imagem , Hiperplasia do Timo/tratamento farmacológico , Tireotropina , Adulto Jovem
4.
J Endocrinol ; 248(3): 317-324, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33538705

RESUMO

Carbohydrate response element-binding protein (ChREBP) is critical in the regulation of fatty acid and triglyceride synthesis in the liver. Interestingly, Chrebp-/- mice show reduced levels of plasma cholesterol, which is critical for steroid hormone synthesis in adrenal glands. Furthermore, Chrebp mRNA expression was previously reported in human adrenal glands. Thus, it remains to be investigated whether ChREBP plays a role directly or indirectly in steroid hormone synthesis and release in adrenal glands. In the present study, we find that Chrebp mRNA is expressed in mouse adrenal glands and that ChREBP binds to carbohydrate response elements. Histological analysis of Chrebp-/- mice shows no adrenal hyperplasia and less oil red O staining compared with that in WT mice. In adrenal glands of Chrebp-/- mice, expression of Fasn and Scd1, two enzymes critical for fatty acid synthesis, was substantially lower and triglyceride content was reduced. Expression of Srebf2, a key transcription factor controlling synthesis and uptake of cholesterol and the target genes, was upregulated, while cholesterol content was not significantly altered in the adrenal glands of Chrebp-/- mice. Adrenal corticosterone content and plasma adrenocorticotropic hormone and corticosterone levels were not significantly altered in Chrebp-/- mice. Consistently, expression of genes related to steroid hormone synthesis was not altered. Corticosterone secretion in response to two different stimuli, namely 24-h starvation and cosyntropin administration, was also not altered in Chrebp-/- mice. Taking these results together, corticosterone synthesis and release were not affected in Chrebp-/- mice despite reduced plasma cholesterol levels.


Assuntos
Glândulas Suprarrenais/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/deficiência , Colesterol/sangue , Corticosterona/biossíntese , Lipogênese , Animais , Masculino , Camundongos Endogâmicos C57BL
5.
Intern Med ; 58(2): 259-262, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30146555

RESUMO

A 60-year-old male patient with type 1 diabetes mellitus (T1DM) was admitted for glycemic control. The patient exhibited abdominal adiposity, osteoporosis, and high insulin requirement (>100 U), and we suspected hypogonadism. A physical examination revealed small testes and thin pubic hair, laboratory examination found high luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels and low testosterone levels, and a chromosome analysis (47, XXY) indicated hypogonadism due to Klinefelter syndrome (KS). KS is associated with autoimmune diseases and patients positive for diabetes related auto-antibodies. In male patients with T1DM and abdominal adiposity, the concurrence of KS should be taken into consideration.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Síndrome de Klinefelter/complicações , Síndrome de Klinefelter/diagnóstico , Obesidade Abdominal/complicações , Diabetes Mellitus Tipo 1/tratamento farmacológico , Hormônio Foliculoestimulante/sangue , Humanos , Hipoglicemiantes/uso terapêutico , Hipogonadismo/etiologia , Insulina/uso terapêutico , Hormônio Luteinizante/sangue , Masculino , Pessoa de Meia-Idade , Osteoporose/complicações , Testosterona/sangue
6.
Toxicon ; 48(8): 1002-10, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16973201

RESUMO

Plancitoxin I, the major lethal factor from the crown-of-thorns starfish Acanthaster planci venom, is quite unique not only in exhibiting potent hepatotoxicity but also in sharing high sequence homology with mammalian deoxyribonulease II. In this study, morphological and biochemical changes in rat liver epithelial cells (TRL 1215 cells) treated with the toxin were examined to understand the mechanism by which plancitoxin I displays hepatotoxicity. AlamarBlue assay established that plancitoxin I is cytolethal to TRL 1215 cells. This cytolethalithy was ascribable to apoptotic cell death. Nuclear fragmentation evidenced by either Diff-Quick or Hoechst 33258 staining, DNA fragmentation by TUNEL assay and electrophoretic analysis on agarose gel and phosphatidylserine externalization by flow cytometric analysis of annexin V-FITC stained cells were all characteristics of apoptosis. The observed apoptosis was shown to be independent of the caspase 3 cascade that is generally accepted as the effector of the apoptotic process. Very interestingly, experiments using FITC-labeled plancitoxin I proved that the toxin can enter the nucleus of TRL 1215 cells. Our results suggested that plancitoxin I induces apoptosis of TRL 1215 cells through the following procedure: binding to a specific receptor in the cytoplasmic membrane, entering the cell, entering the nucleus and degrading DNA.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Fígado/efeitos dos fármacos , Toxinas Marinhas/toxicidade , Estrelas-do-Mar/química , Peçonhas/toxicidade , Animais , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Fígado/citologia , Toxinas Marinhas/química , Toxinas Marinhas/metabolismo , Transporte Proteico , Ratos , Peçonhas/química
7.
Environ Sci ; 13(2): 101-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16788561

RESUMO

Although inorganic arsenicals are toxic and carcinogenic in humans, inorganic arsenite has recently emerged as a highly effective chemotherapeutic agent for acute promyelocytic leukemia (APL). Recently, the number of arsenic-related publications has greatly increased, but the effects of arsenicals on the endocrine system have not been well studied. A number of in vivo studies about only inorganic-arsenical-induced endocrine-disruptor-like effects have been conducted using mouse models, and they demonstrated that inorganic arsenicals might act as an estrogen-like chemical in vivo and induce some tumors in mice. In most mammalian species, however, inorganic arsenicals are enzymatically methylated as a detoxifying metabolic response. Thus, studies of endocrine disruptive effects of methylated arsenicals are also needed for a clinical understanding of this attractive metalloid in the near future.


Assuntos
Arsenicais , Disruptores Endócrinos/toxicidade , Animais , Humanos
8.
Biol Pharm Bull ; 29(5): 1022-7, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16651738

RESUMO

The pharmacokinetics of arsenic species in a Japanese patient with relapsed acute promyelocytic leukemia (APL) treated with arsenic trioxide at a daily dose of 0.08 mg/kg was investigated. After achieving complete remission on Day 35 during the induction therapy of arsenic trioxide, we collected the serum and urine samples on Days 4 and 5 during the consolidation therapy of arsenic trioxide. The concentrations of inorganic arsenic and the methylated metabolites in serum and urine were measured by HPLC/ICP-MS. The patient restricted taking the seafood for 3 d before the start of administration and during the sampling period in order to avoid the influence of arsenic derived from seafood. Arsenite (As(III)), methylarsonic acid (MMAs(V)), and dimethylarsinic acid (DMAs(V)) were detected in serum and urine. The total concentration of As(III), MMAs(V) and DMAs(V) in serum ranged from 18 to 41 microg/l (240-547 nM) during 24 h on Day 4. The amount of total arsenic (As(III)+MMAs(V)+DMAs(V)) in urine was 4464 microg/d on Day 4. These results suggest that not the micro-molar but the nano-molar order of arsenic in serum is sufficient to produce the therapeutic effect on APL cells.


Assuntos
Arsenicais/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Óxidos/farmacocinética , Idoso , Antineoplásicos/uso terapêutico , Trióxido de Arsênio , Arsenicais/efeitos adversos , Arsenicais/sangue , Arsenicais/farmacocinética , Arsenicais/urina , Cromatografia Líquida de Alta Pressão , Resistencia a Medicamentos Antineoplásicos , Humanos , Injeções Intravenosas , Masculino , Metilação , Óxidos/efeitos adversos , Alimentos Marinhos/análise , Tretinoína/uso terapêutico
9.
Mol Carcinog ; 45(8): 561-71, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16568437

RESUMO

Prior work has shown that chronic cadmium exposed rat liver epithelial cells (CCE-LE) become malignantly transformed after protracted low level cadmium exposure. Acquisition of apoptotic resistance is common in oncogenesis and the present work explores this possibility in CCE-LE cells. CCE-LE cells were resistant to apoptosis induced by etoposide or an acute high concentration of cadmium as assessed by flow cytometry with annexin/FITC. Three key mitogen-activated protein kinases (MAPKs), namely ERK1/2, JNK1/2, and p38, were phosphorylated in CCE-LE cells after acute cadmium exposure. However, the levels of phosphorylated JNK1/2 were markedly decreased in CCE-LE cells compared to control. JNK kinase activity was also suppressed in CCE-LE cells exposed to cadmium. Epidermal growth factor (EGF), used as a positive control for stimulating JNK phosphorylation, was much less effective in CCE-LE cells than control cells. Ro318220 (Ro), a strong activator of JNK, increased phosphorylated JNK1/2 to levels similar to the cadmium-treated control cells and also enhanced apoptosis in response to cadmium in CCE-LE cells. Metallothionein (MT), which is thought to potentially inhibit apoptosis, was strongly overexpressed in CCE-LE cells. Further, in MT knockout (MT-/-) fibroblasts, JNK1/2 phosphorylation was markedly increased after cadmium exposure compared with similarly treated wild-type (MT+/+) cells. These results indicate cadmium-transformed cells acquired apoptotic resistance, which may be linked to the specific suppression of the JNK pathway and is associated with MT overexpression, which, in turn, may impact this signal transduction pathway. The acquisition of apoptotic resistance may play an important role in cadmium carcinogenesis by contributing to both tumor initiation and malignant progression.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metalotioneína/metabolismo , Animais , Apoptose/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Indóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Fígado/efeitos dos fármacos , Fígado/patologia , Metalotioneína/antagonistas & inibidores , Metalotioneína/genética , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Transdução de Sinais , Regulação para Cima
10.
Int Immunopharmacol ; 6(2): 304-15, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16459422

RESUMO

A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.


Assuntos
Arsenitos/toxicidade , Imunidade Celular/efeitos dos fármacos , Imunotoxinas , Macrófagos/imunologia , Monócitos/imunologia , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/biossíntese , Poluentes Ambientais/toxicidade , Glutationa/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HL-60 , Humanos , Interleucina-12/metabolismo , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
11.
Toxicol Sci ; 91(1): 70-81, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16436460

RESUMO

Although inorganic arsenicals are toxic and carcinogenic in humans, inorganic arsenite has recently emerged as a highly effective chemotherapeutic agent for acute promyelocytic leukemia (APL). Inorganic arsenicals are enzymatically methylated to monomethylarsonic acid (MMAs(V)), dimethylarsinic acid (DMAs(V)), and trimethylarsine oxide (TMAs(V)O) in mammals. We examined the effects of chronic exposure to methylated arsenicals on arsenic tolerance by using rat normal liver TRL 1215 cells. TRL 1215 cells were exposed for 20 weeks to MMAs(V), DMAs(V), or TMAs(V)O at levels that produced submicromolar cellular concentrations of arsenic. On chronic exposure to these methylated arsenicals, the cells acquired tolerance to acute arsenic cytolethality. Cellular arsenic uptake was reduced in these cells compared to passage-matched control cells. The long-term arsenic exposure increased glutathione S-transferase (GST) activity and cellular glutathione (GSH) levels. Glutathione S-transferase, multidrug resistance-associated proteins (Mrps; efflux transporters encoded by Mrp genes), and P-glycoprotein [P-gp; efflux transporter encoded by multidrug resistance gene (MDR)] had also increased in these cells at the transcript and protein levels. The depletion of cellular GSH and the inhibition of Mrps and P-gp functions increased cellular arsenic uptake and reduced arsenic tolerance in these cells. These results indicate that chronic exposure to methylated arsenicals induces a generalized arsenic tolerance that is caused by increased arsenic excretion. Because accumulation of methylated arsenicals may occur in patients with chronic arsenic poisoning and arsenic-treated APL patients, this study may provide important information regarding chronic arsenic poisoning and the latent risk of developing multidrug resistance in APL therapy using inorganic arsenite.


Assuntos
Arsênio/toxicidade , Fígado/efeitos dos fármacos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Arsênio/administração & dosagem , Arsênio/farmacocinética , Glutationa Transferase/metabolismo , Leucemia Promielocítica Aguda/patologia , Fígado/citologia , Fígado/enzimologia , Fígado/metabolismo , Metilação , Ratos , Ratos Endogâmicos F344
12.
Biol Pharm Bull ; 28(10): 1827-32, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16204930

RESUMO

Arsenicals are known to be toxic and carcinogenic in humans. Inorganic arsenicals are enzymatically methylated to monomethylarsonic acid (MMAsV) and dimethylarsinic acid (DMAsV), which are the major pentavalent methyl arsenic metabolites. Recent reports indicate that trivalent methyl arsenicals are produced through methylation of inorganic arsenicals and participate in arsenic poisoning. Trivalent methyl arsenicals may be generated as arsenical-glutathione conjugates, such as monomethylarsonous diglutathione (MMAsIIIDG) and dimethylarsinous glutathione (DMAsIIIG), during the methylation process. It has been well known that reduced glutathione (GSH) reduces MMAsV and DMAsV in vitro, and produces MMAsIIIDG and DMAsIIIG. Some studies have shown that exogenous GSH increased cytolethality of MMAsV and DMAsV in vitro, while other studies have suggested that exogenous GSH decreased them. In this study, we examined the true effects of exogenous GSH on the cytolethality of MMAsV and DMAsV by investigating reactions between various concentrations of MMAsV or DMAsV and GSH. GSH significantly increased the cytolethality and cellular uptake of pentavalent methyl arsenicals when GSH over 25 mM was pre-incubated with mM levels of arsenicals, and this cytolethality might have been caused by arsenical-GSH conjugate generation. However, GSH at less than 25 mM did not affect the cytolethality and cellular uptake of pentavalent methyl arsenicals. These findings suggest that high concentrations of arsenicals and GSH are needed to form arsenical-GSH conjugates and to show significant cytolethality. Furthermore, we speculated that MMAsIIIDG and DMAsIIIG may separate into trivalent methyl arsenicals and glutathione, which are then transported into cells where they show significant cytolethality.


Assuntos
Arsenicais/química , Glutationa/toxicidade , Animais , Arsenicais/farmacocinética , Linhagem Celular , Cromatografia em Camada Fina , Glutationa/química , Glutationa/farmacocinética , Ratos
13.
Toxicol Appl Pharmacol ; 206(1): 54-65, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15963344

RESUMO

Human pentavalent arsenic metabolic intermediate, monomethylarsonic acid (MMAs(V)), is a major arsenic type found in the blood in chronic arsenic poisoning patients, but little information is available on its toxicity potential or mechanisms of action. In this study, we investigated the molecular mechanisms of in vitro cytolethality of MMAs(V) using rat liver TRL 1215 cells. Cellular arsenic concentrations reached the nanomolar range in TRL 1215 cells when cells were exposed to millimolar levels of MMAs(V), and most of the MMAs(V) was not metabolized during the 48-h incubation. Under these conditions, MMAs(V) showed significant cytolethality when cellular reserves of reduced glutathione (GSH) were depleted. Morphological and biochemical evidence confirmed that MMAs(V) induced both necrosis and apoptosis in the cellular GSH-depleted cells. MMAs(V) significantly enhanced cellular caspase 3 activity in the cellular GSH-depleted cells, and a caspase 3 inhibitor blocked MMAs(V)-induced apoptosis. MMAs(V) also enhanced the production of cellular reactive oxygen species (ROS) in the cellular GSH-depleted cells, and addition of a membrane-permeable radical trapping reagent completely prevented both MMAs(V)-induced cellular caspase 3 activation and cytolethality in these cells. These observations suggest that MMAs(V) typically generates harmful ROS in cells, and cellular GSH prevents cytolethality by scavenging these toxic ROS. However, when cellular GSH levels are decreased, MMAs(V) induces oxidative stress in the cells, and this leads to apoptosis and/or necrosis depending on the cellular ROS/GSH ratio.


Assuntos
Arsenicais/efeitos adversos , Glutationa/uso terapêutico , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Arsenicais/antagonistas & inibidores , Células Cultivadas , Herbicidas/antagonistas & inibidores , Marcação In Situ das Extremidades Cortadas , Fígado/metabolismo , Fígado/patologia , Ratos , Ratos Endogâmicos F344
14.
Nitric Oxide ; 12(2): 114-20, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15740985

RESUMO

The liver is an important target tissue of cadmium. The compound O2-vinyl 1-(pyrrolidin-1-yl)diazen-1-ium-1,2 diolate (V-PYRRO/NO) is a liver-selective nitric oxide (NO) prodrug that is metabolized by hepatic P450 enzymes to release NO in hepatocytes. In vivo, V-PYRRO/NO can protect against the toxicity of various hepatotoxicants, including cadmium. Since NO is an effective vasodilator, whether this protective effect against cadmium toxicity is at the level of the hepatic vascular system or actually within the liver cells has not been defined. Thus, we studied the effects of V-PYRRO/NO pretreatment on cadmium-induced toxicity and apoptosis in cultured rat liver epithelial (TRL 1215) cells. Cells were pretreated with V-PYRRO/NO at 500 or 1000 microM for up to 24 h, then exposed to cadmium (as CdCl2) for additional 24 h and cytotoxicity was measured. Cadmium was significantly less cytotoxic in V-PYRRO/NO (1000 microM) pretreated cells (LC50=6.1+/-0.6 microM) compared to control cells (LC50=3.5+/-0.4 microM). TRL 1215 cells acted upon the prodrug to release NO, producing nitrite levels in the extracellular media after 24 h of exposure to 500 or 1000 microM V-PYRRO/NO measured at 87.0+/-4.2 and 324+/-14.8 microM, respectively, compared to basal levels of 7.70+/-0.46 microM. V-PYRRO/NO alone produced small increases in metallothionein (MT), a metal-binding protein associated with cadmium tolerance. However, V-PYRRO/NO pretreatment greatly enhanced cadmium induction of MT. V-PYRRO/NO pretreatment also markedly reduced apoptotic cell death induced by cadmium (5 microM), apparently by blocking cadmium-induced activation of the c-Jun N-terminal kinase (JNK) pathway. Thus, the prodrug, V-PYRRO/NO, protects against the adverse effects of cadmium directly within rat liver cells apparently through generation of NO and, at least in part, by facilitation of cadmium-induced MT synthesis.


Assuntos
Apoptose/efeitos dos fármacos , Cloreto de Cádmio/antagonistas & inibidores , Cloreto de Cádmio/toxicidade , Pirrolidinas/farmacologia , Animais , Apoptose/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metalotioneína/efeitos dos fármacos , Metalotioneína/metabolismo , Óxido Nítrico/biossíntese , Nitritos/metabolismo , Pró-Fármacos/farmacologia , Ratos , Ratos Endogâmicos F344
15.
Toxicol Appl Pharmacol ; 203(2): 145-53, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15710175

RESUMO

Inorganic arsenite has caused severe inflammatory chronic poisoning in humans through the consumption of contaminated well water. In this study, we examined the effects of arsenite at nanomolar concentrations on the in vitro differentiation of human macrophages from peripheral blood monocytes. While arsenite was found to induce cell death in a culture system containing macrophage colony stimulating factor (M-CSF), macrophages induced by granulocyte-macrophage CSF (GM-CSF) survived the treatment, but were morphologically, phenotypically, and functionally altered. In particular, arsenite-induced cells expressed higher levels of a major histocompatibility complex (MHC) class II antigen, HLA-DR, and CD14. They were more effective at inducing allogeneic or autologous T cell responses and responded more strongly to bacterial lipopolysaccharide (LPS) by inflammatory cytokine release as compared to cells induced by GM-CSF alone. On the other hand, arsenite-induced cells expressed lower levels of CD11b and CD54 and phagocytosed latex beads or zymosan particles less efficiently. We also demonstrated that the optimum amount of cellular reactive oxygen species (ROS) induced by nM arsenite might play an important role in this abnormal monocyte differentiation. This work may have implications in chronic arsenic poisoning because the total peripheral blood arsenic concentrations of these patients are at nM levels.


Assuntos
Arsenitos/toxicidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Macrófagos/imunologia , Monócitos/efeitos dos fármacos , Compostos de Sódio/toxicidade , Poluentes Químicos da Água/toxicidade , Arseniatos/toxicidade , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Antígenos HLA-DR/biossíntese , Antígenos de Histocompatibilidade Classe II/biossíntese , Humanos , Interleucina-1/biossíntese , Receptores de Lipopolissacarídeos/biossíntese , Lipopolissacarídeos , Macrófagos/patologia , Monócitos/imunologia , Monócitos/patologia , Fagocitose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/biossíntese
16.
Int Immunopharmacol ; 4(13): 1661-73, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15454118

RESUMO

A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.


Assuntos
Arsenitos/efeitos adversos , Arsenitos/imunologia , Imunotoxinas/efeitos adversos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Compostos de Sódio/efeitos adversos , Compostos de Sódio/imunologia , Arseniatos/efeitos adversos , Arseniatos/imunologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Poluentes Ambientais/efeitos adversos , Poluentes Ambientais/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HL-60 , Humanos , Imunotoxinas/química , Imunotoxinas/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Fator Estimulador de Colônias de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/patologia , Macrófagos/fisiologia , Monócitos/patologia , Monócitos/fisiologia , Testes de Toxicidade/métodos
17.
Toxicol Appl Pharmacol ; 198(3): 354-65, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15276415

RESUMO

Inorganic arsenicals are clearly toxicants and carcinogens in humans. In mammals, including humans, inorganic arsenicals often undergo methylation, forming compounds such as dimethylarsinic acid (DMAs(V)). Recent evidence indicates that DMAs(V) is a complete carcinogen in rodents although evidence for inorganic arsenicals as carcinogens in rodents remains equivocal. Thus, we studied the molecular mechanisms of in vitro cytolethality of DMAs(V) using a rat liver epithelial cell line (TRL 1215). DMAs(V) selectively induced apoptosis in TRL 1215 cells; its LC(50) value after 48 h exposure was 4.5 mM. The addition of a glutathione synthase inhibitor, L-buthionine-[S,R]-sulfoximine (BSO), actually decreased DMAs(V)-induced apoptosis. DMAs(V) exposure temporarily decreased cellular reduced glutathione (GSH) levels and enhanced cellular glutathione S-transferase (GST) activity from 6 h after the exposure when the cells were still alive. Also, DMAs(V) exposure activated cellular caspase 3 activity with a peak at 18 h after the exposure when apoptosis began, and BSO treatment completely inhibited this enzyme activity. The additions of inhibitors of caspase 3, caspase 8, and caspase 9 significantly reduced DMAs(V)-induced apoptosis. Taken together, these data indicate that cellular GSH was required for DMAs(V)-induced apoptosis to occur, and activation of cellular caspases after conjugation of DMAs(V) with cellular GSH appears to be of mechanistic significance. Further research will be required to determine the role of intracellular GSH and methylation in the toxicity of arsenicals in chronic arsenic poisoning or in cases where arsenicals are used as chemotherapeutics.


Assuntos
Apoptose/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Ácido Cacodílico/toxicidade , Inibidores Enzimáticos/farmacologia , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Animais , Ácido Cacodílico/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular , Glutationa Transferase/metabolismo , Herbicidas/metabolismo , Humanos , Fígado/enzimologia , Fígado/metabolismo , Ratos , Ratos Endogâmicos F344
18.
Int Immunopharmacol ; 4(2): 179-84, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14996409

RESUMO

In this study, we observed the in vivo acute immunotoxicity of a trimethyl arsenic compound, arsenobetaine (AsBe), which is present in large quantities in various marine animals that are daily ingested as seafood in many countries. The synthetic pure AsBe was orally administered to CDF(1) mice at a dose of 1.625 g/kg mouse weight once a day on days -6, -4, -2 and 0 (four times, total 6.5 g/kg mouse weight), and its effect on the immune organs and immune effector cells were assessed until day 8. Orally administered AsBe was temporally distributed to the immune organs, such as the spleen and thymus, but was not very toxic both quantitatively and qualitatively on these immune organs and immune effector cells, splenocytes, thymocytes, Peyer's patch lymphocytes and peritoneal macrophages. This finding suggests that the ingestion of AsBe contained in marine animals is relatively safe to the health of people who often consume marine animals in their daily diet.


Assuntos
Arsênio/toxicidade , Arsenicais , Nódulos Linfáticos Agregados/efeitos dos fármacos , Alimentos Marinhos , Baço/efeitos dos fármacos , Timo/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais , Arsenicais/farmacocinética , Contaminação de Alimentos , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Nível de Efeito Adverso não Observado , Nódulos Linfáticos Agregados/citologia , Medição de Risco , Baço/citologia , Timo/citologia , Distribuição Tecidual , Poluentes Químicos da Água/farmacocinética
19.
Toxicol Appl Pharmacol ; 195(2): 129-41, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14998680

RESUMO

Inorganic arsenicals are clearly toxicants and carcinogens in humans. In mammals, including humans, inorganic arsenic often undergoes methylation, forming compounds such as monomethylarsonic acid (MMAs(V)) and dimethylarsinic acid (DMAs(V)). However, much less information is available on the in vitro toxic potential or mechanisms of these methylated arsenicals, especially MMAs(V). We studied the molecular mechanisms of in vitro cytolethality of MMAs(V) using a rat liver epithelial cell line (TRL 1215). MMAs(V) was not cytotoxic in TRL 1215 cells even at concentrations exceeding 10 mM, but it became weakly cytotoxic and induced both necrotic and apoptotic cell death when cellular reduced glutathione (GSH) was depleted with the glutathione synthase inhibitor, l-buthionine-[S,R]-sulfoximine (BSO), or the glutathione reductase inhibitor, carmustine. Similar results were observed in the other mammalian cells, such as human skin TIG-112 cells, chimpanzee skin CRT-1609 cells, and mouse metallothionein (MT) positive and MT negative embryonic cells. Ethacrynic acid (EA), an inhibitor of glutathione S-transferase (GST) that catalyses GSH-substrate conjugation, also enhanced the cytolethality of MMAs(V), but aminooxyacetic acid (AOAA), an inhibitor of beta-lyase that catalyses the final breakdown of GSH-substrate conjugates, had no effect. Both the cellular GSH levels and the cellular GST activity were increased by the exposure to MMAs(V) in TRL 1215 cells. On the other hand, the addition of exogenous extracellular GSH enhanced the cytolethality of MMAs(V), although cellular GSH levels actually prevented the cytolethality of combined MMAs(V) and exogenous GSH. These findings indicate that human arsenic metabolite MMAs(V) is not a highly toxic compound in mammalian cells, and the level of cellular GSH is critical to its eventual toxic effects.


Assuntos
Arsenicais/farmacologia , Glutationa/metabolismo , Ácido Amino-Oxiacético/farmacologia , Animais , Carmustina/farmacologia , Catálise , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácido Etacrínico/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Glutationa/antagonistas & inibidores , Glutationa/farmacologia , Glutationa Transferase/antagonistas & inibidores , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pan troglodytes , Ratos , Ratos Endogâmicos F344 , Pele/citologia , Pele/efeitos dos fármacos , Pele/metabolismo
20.
Chem Res Toxicol ; 15(5): 629-37, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12018983

RESUMO

Inorganic arsenicals are important environmental toxicants and carcinogens in humans. In mammals, including humans, inorganic arsenicals often undergo methylation, forming compounds such as dimethyarsinic acid (DMA). Recent evidence indicates DMA is a complete carcinogen in rodents while evidence for inorganic arsenicals as carcinogens in rodents remains equivocal. Thus, we studied the molecular mechanisms of in vitro cytolethality of DMA compared to that of the trivalent inorganic arsenical, sodium arsenite, using a rat liver epithelial cell line (TRL 1215). Arsenite was very cytotoxic in these cells (LC(50) = 35 microM after 48 h of exposure). With arsenite exposure, most dead cells showed histological and biochemical evidence of necrosis. Arsenite cytotoxicity increased markedly when cellular GSH was depleted with the glutathione synthase inhibitor, L-buthionine-[S,R]-sulfoximine (BSO). In contrast, DMA was nearly 3 orders of magnitude less cytotoxic (LC(50) = 1.5 mM) although evidence showed the predominating form of death was apoptosis. Surprisingly, GSH depletion actually decreased DMA-induced apoptosis. A glutathione scavenger, diethyl maleate (DEM), and a glutathione reductase inhibitor, carmustine, also prevented DMA-induced apoptosis. These data indicate that DMA requires intracellular GSH to induce apoptosis. Ethacrynic acid (EA), an inhibitor of glutathione S-transferase (GST) that catalyzes GSH-substrate conjugation, acivicin, an inhibitor of gamma-glutamyltranspeptidase (GGT) which catalyzes the initial breakdown of GSH-substrate conjugates, and aminooxyacetic acid (AOAA), an inhibitor of beta-lyase which catalyzes the final breakdown of GSH-substrate conjugates, all were effective in suppressing DMA-induced apoptosis. These findings indicate that DMA likely is conjugated in some form with GSH, and that it is this conjugate that induces apoptosis during subsequent metabolic reactions.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/toxicidade , Ácido Cacodílico/toxicidade , Glutationa/farmacologia , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Animais , Arsenitos/metabolismo , Ácido Cacodílico/metabolismo , Linhagem Celular , Cromatografia em Camada Fina , Interações Medicamentosas , Citometria de Fluxo , Glutationa/metabolismo , Herbicidas/metabolismo , Humanos , Fígado/patologia , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA