Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Toxicol Pathol ; 47(5): 612-633, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31409264

RESUMO

To further our understanding of the nonhuman primate kidney anatomy, histology, and incidences of spontaneous pathology, we retrospectively examined kidneys from a total of 505 control Cynomolgus monkeys (Macaca fascicularis; 264 male and 241 females) aged 2 to 6 years, from toxicity studies. Kidney weights, urinalysis, and kidney-related clinical biochemistry parameters were also evaluated. Although the functional anatomy of the monkey kidney is relatively similar to that of other laboratory animals and humans, a few differences and species-specific peculiarities exist. Unlike humans, the macaque kidney is unipapillate, with a relatively underdeveloped papilla, scarce long loops of Henle, and a near-equivalent cortical to medullary ratio. The most common spontaneous microscopic findings were interstitial infiltrates or interstitial nephritis and other tubular lesions, but several forms of glomerulopathy that may be interpreted as drug-induced were occasionally observed. Common incidental findings of little pathological significance included: papillary mineralization, epithelial pigment, multinucleate cells, cuboidal metaplasia of the Bowman's capsule, and urothelial inclusions. Kidney weights, and some clinical chemistry parameters, showed age- and sex-related variations. Taken together, these data will aid the toxicologic pathologist to better evaluate the nonhuman primate kidney and assess the species' suitability as a model for identifying and characterizing drug-induced injury.


Assuntos
Nefropatias/patologia , Rim/anatomia & histologia , Rim/patologia , Animais , Biomarcadores/metabolismo , Feminino , Imuno-Histoquímica , Rim/metabolismo , Nefropatias/metabolismo , Testes de Função Renal , Macaca fascicularis , Masculino , Tamanho do Órgão/fisiologia , Especificidade da Espécie , Urinálise
2.
Biomed Microdevices ; 18(4): 73, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27464497

RESUMO

The vascular endothelium and shear stress are critical determinants of physiological hemostasis and platelet function in vivo, yet current diagnostic and monitoring devices do not fully incorporate endothelial function under flow in their assessment and, therefore, they can be unreliable and inaccurate. It is challenging to include the endothelium in assays for clinical laboratories or point-of-care settings because living cell cultures are not sufficiently robust. Here, we describe a microfluidic device that is lined by a human endothelium that is chemically fixed, but still retains its ability to modulate hemostasis under continuous flow in vitro even after few days of storage. This device lined with a fixed endothelium supports formation of platelet-rich thrombi in the presence of physiological shear, similar to a living arterial vessel. We demonstrate the potential clinical value of this device by showing that thrombus formation and platelet function can be measured within minutes using a small volume (0.5 mL) of whole blood taken from subjects receiving antiplatelet medications. The inclusion of a fixed endothelial microvessel will lead to biomimetic analytical devices that can potentially be used for diagnostics and point-of-care applications.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Dispositivos Lab-On-A-Chip , Trombose/diagnóstico , Plaquetas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fibrina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Sistemas Automatizados de Assistência Junto ao Leito , Estresse Mecânico , Trombose/sangue , Trombose/tratamento farmacológico
3.
Toxicol Pathol ; 43(1): 48-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25398756

RESUMO

Canagliflozin, a sodium glucose co-transporter 2 (SGLT2) inhibitor, has been developed for the treatment of adults with type 2 diabetes mellitus (T2DM). During the phase 3 program, treatment-related pheochromocytomas, renal tubular tumors, and testicular Leydig cell tumors were reported in the 2-year rat toxicology study. Treatment-related tumors were not seen in the 2-year mouse study. A cross-functional, mechanism-based approach was undertaken to determine whether the mechanisms responsible for tumorigenesis in the rat were of relevance to humans. Based on findings from nonclinical and clinical studies, the treatment-related tumors observed in rats were not deemed to be of clinical relevance. Here, we describe the scientific and regulatory journey from learning of the 2-year rat study findings to the approval of canagliflozin for the treatment of T2DM.


Assuntos
Canagliflozina/toxicidade , Hipoglicemiantes/toxicidade , Neoplasias Experimentais/induzido quimicamente , Animais , Canagliflozina/administração & dosagem , Testes de Carcinogenicidade , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos , Hipoglicemiantes/administração & dosagem , Masculino , Camundongos , Ratos , Inibidores do Transportador 2 de Sódio-Glicose , Testosterona
4.
Toxicol Pathol ; 43(7): 915-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25722122

RESUMO

Drug-induced vascular injury (DIVI) is a recurrent challenge in the development of novel pharmaceutical agents. Although DIVI in laboratory animal species has been well characterized for vasoactive small molecules, there is little available information regarding DIVI associated with biotherapeutics such as peptides/proteins or antibodies. Because of the uncertainty about whether DIVI in preclinical studies is predictive of effects in humans and the lack of robust biomarkers of DIVI, preclinical DIVI findings can cause considerable delays in or even halt development of promising new drugs. This review discusses standard terminology, characteristics, and mechanisms of DIVI associated with biotherapeutics. Guidance and points to consider for the toxicologist and pathologist facing preclinical cases of biotherapeutic-related DIVI are outlined, and examples of regulatory feedback for each of the mechanistic types of DIVI are included to provide insight into risk assessment.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Lesões do Sistema Vascular/induzido quimicamente , Animais , Modelos Animais de Doenças , Humanos
5.
Toxicol Pathol ; 43(7): 935-44, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25717082

RESUMO

Drug-induced vascular injury (DIVI) is a recurrent challenge in the development of novel pharmaceutical agents. In recent years, DIVI has been occasionally observed in nonhuman primates given RNA-targeting therapeutics such as antisense oligonucleotide therapies (ASOs) during chronic toxicity studies. While DIVI in laboratory animal species has been well characterized for vasoactive small molecules, and immune-mediated responses against large molecule biotherapeutics have been well described, there is little published information regarding DIVI induced by ASOs to date. Preclinical DIVI findings in monkeys have caused considerable delays in development of promising new ASO therapies, because of the uncertainty about whether DIVI in preclinical studies is predictive of effects in humans, and the lack of robust biomarkers of DIVI. This review of DIVI discusses clinical and microscopic features of vasculitis in monkeys, their pathogenic mechanisms, and points to consider for the toxicologist and pathologist when confronted with ASO-related DIVI. Relevant examples of regulatory feedback are included to provide insight into risk assessment of ASO therapies.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Oligonucleotídeos Antissenso/efeitos adversos , Lesões do Sistema Vascular/induzido quimicamente , Animais , Modelos Animais de Doenças , Humanos
6.
Int J Toxicol ; 34(5): 393-407, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26124191

RESUMO

Interleukin 13 (IL-13) is a type 2 helper T cytokine involved in allergic inflammation and immune responses to parasites. CNTO5825 is an antihuman IL-13 monoclonal antibody that inhibits the pharmacological activity of human, cynomolgus monkey, and rat IL-13. Repeated dose toxicology studies of 1- to 6-month duration were conducted in both rats and monkeys at doses of 20 to 100 mg/kg/wk. A decrease in the T cell-dependent antibody response to Keyhole Limpet Hemocyanin immunization was observed in monkeys but not in rats. In the 6-month rat study, there was a 2.2-fold increase in eosinophils in males at 3 and 6 months that was reversible. At necropsy (main and 4-month recovery), rats from control and CNTO5825-dosed groups were found to have pin worms, which may have contributed to the elevations in eosinophil. Testicular toxicity (dilatation of seminiferous tubules, atrophy, and degeneration of the germinal epithelium) was observed in 2 rats at 20 mg/kg and in 5 rats at 100 mg/kg (main and recovery). Brain lesions (unilateral focal accumulation of cells in the white matter of the cerebral cortex) were observed in 2 rats at 100 mg/kg, and vascular neoplasms (1 fatal multicentric hemangiosarcoma and 1 benign hemangioma) were observed at 100 mg/kg/wk. Overall, these studies show that CNTO5825 was without toxicity when administered to rats for up to 6 weeks and to monkeys for up to 6 months. However, when administered to rats for 6 months, a number of seemingly unrelated events occurred that could not be clearly linked to CNTO5825 administration, inhibition of IL-13, or to the immunological status of the animals.


Assuntos
Anticorpos Monoclonais/toxicidade , Interleucina-13/imunologia , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Feminino , Humanos , Macaca fascicularis , Macaca mulatta , Masculino , Camundongos , Coelhos , Ratos , Ratos Sprague-Dawley , Testículo/efeitos dos fármacos , Testículo/patologia
7.
Toxicol Pathol ; 42(4): 672-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24499802

RESUMO

Pharmacologically, vasoactive agents targeting endothelial and/or smooth muscle cells (SMC) are known to cause acute drug-induced vascular injury (DIVI) and the resulting pathology is due to endothelial cell (EC) perturbation, activation, and/or injury. Alteration in EC structure and/or function may be a critical event in vascular injury and, therefore, evaluation of the circulatory kinetic profile and secretory pattern of EC-specific proteins such as VWF and VWFpp could serve as acute vascular injury biomarkers. In rat and dog models of DIVI, this profile was determined using pharmacologically diverse agents associated with functional stimulation/perturbation (DDAVP), pathological activation (lipopolysaccharide [LPS]/endotoxin), and structural damage (fenoldopam [FD], dopamine [DA], and potassium channel opener (PCO) ZD6169). In rats, FD caused moderate DIVI and time-related increase in plasma VWF levels ∼33% while in control rats VWF increased ∼5%. In dogs, VWF levels transiently increased ∼30% when there was morphologic evidence of DIVI by DA or ZD6169. However, in dogs, VWFpp increased >60-fold (LPS) and >6-fold (DDAVP), respectively. This was in comparison to smaller dynamic 1.38-fold (LPS) and 0.54-fold (DDAVP) increases seen in plasma VWF. Furthermore, DA was associated with a dose-dependent increase in plasma VWFpp. In summary, VWF and VWFpp can discriminate between physiological and pathological perturbation, activation, and injury to ECs.


Assuntos
Células Endoteliais/patologia , Precursores de Proteínas/sangue , Lesões do Sistema Vascular/patologia , Administração Oral , Amidas/efeitos adversos , Animais , Benzofenonas/efeitos adversos , Biomarcadores/sangue , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Cães , Dopamina/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Células Endoteliais/efeitos dos fármacos , Feminino , Fenoldopam/efeitos adversos , Masculino , Flebotomia , Ratos , Ratos Wistar , Lesões do Sistema Vascular/etiologia , Fator de von Willebrand
8.
Int J Toxicol ; 32(4): 251-60, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23788329

RESUMO

Cisplatin (CDDP) is known to produce renal proximal tubule injury. Various renal biomarkers have been related to CDDP nephrotoxicity in previous research, but the temporal and spatial relationship of these biomarkers to injury reversal has not been well defined. In this study, the progression and reversal of renal histopathology findings relative to serum and urinary biomarker changes were examined during a 4-week postdose period following single intraperitoneal administration of CDDP (1 mg/kg) or 0.9% saline. Degeneration, vacuolation, inflammation, and regeneration of the S3 segment of proximal tubules were evident 72 hours following CDDP administration. Tubular degeneration and regeneration were also observed at 1 and 1.5 weeks but at lower incidences and/or severity indicating partial reversal. Complete histologic reversal was observed by 2 weeks following CDDP administration. Urinary kidney injury molecule 1 (KIM-1), α-glutathione-S-transferase (α-GST), and albumin levels increased at 72 hours postdosing, concurrently with the earliest histologic evidence of tubule injury. Changes in urinary KIM-1 correlated with KIM-1 immunostaining in the proximal tubular epithelial cells. No significant changes in serum biomarkers occurred except for a minimal increase in urea nitrogen at 1.5 weeks postdosing. Of the novel renal biomarkers examined, urinary KIM-1, α-GST, and albumin showed excellent concordance with CDDP-induced renal injury progression and reversal; and these biomarkers were more sensitive than traditional serum biomarkers in detecting early, acute renal tubular damage confirmed by histopathology. Furthermore, urinary KIM-1, α-GST, and albumin outperformed other biomarkers in correlating with the time of maximum histologic injury.


Assuntos
Biomarcadores/sangue , Biomarcadores/urina , Cisplatino/administração & dosagem , Túbulos Renais Proximais/efeitos dos fármacos , Albuminas/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Clusterina/urina , Glutationa Transferase/metabolismo , Imuno-Histoquímica , Isoenzimas/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Ratos , Ratos Sprague-Dawley
9.
Toxicol Pathol ; 40(6): 874-86, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22549976

RESUMO

Fenoldopam, a dopaminergic DA1 agonist, induces vasodilatation via nitric oxide (NO), and this may be associated with mesenteric arterial injury. NO is produced from the enzymatic action of nitric oxide synthase (NOS), which is regulated by the shear-stress mediating protein caveolin-1. Profound vasodilatation and accompanied decreased shear are early events that could initiate vascular injury. Therefore, it is of interest to determine the role of caveolin-1 and the NO pathway in fenoldopam-induced vascular injury. At sites of fenoldopam-induced mesenteric arterial injury, decreased caveolin-1 expression and apoptosis were prominent immunohistochemical findings. An additional finding at these sites of injury were loss and/or reduced expression of caveolin-1 regulated structural proteins, connexin-43, (gap junction) ZO-1, and claudin (tight junctions). Because functional loss of caveolin-1 is associated with increased NOS activity and vasodilatation via NO, studies were conducted to show a NO donor produced vascular lesions in the mesenteric arteries morphologically similar to those induced by fenoldopam. Moreover, the incidence and severity of fenoldopam-induced vascular injury were reduced when an NOS inhibitor or a scavenger of NO-generated free radicals were coadministered with fenoldopam. Collectively, these data suggest that caveolin-1 and its regulated NO pathway may play an important role in vasodilatory drug-induced vascular injury.


Assuntos
Fenoldopam/toxicidade , Óxido Nítrico/metabolismo , Lesões do Sistema Vascular/induzido quimicamente , Lesões do Sistema Vascular/metabolismo , Análise de Variância , Animais , Caspase 3/metabolismo , Caveolina 1/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/patologia , Doadores de Óxido Nítrico/toxicidade , Nitroprussiato/toxicidade , Ácido Peroxinitroso/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Lesões do Sistema Vascular/patologia
10.
Int J Toxicol ; 30(4): 385-404, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21832269

RESUMO

RWJ-800088 is a novel, potent polyethylene glycol (PEG)-conjugated thrombopoietin (TPO) mimetic that increases platelet levels and protects against thrombocytopenia. A nonclinical safety program was customized for this peptide that takes into account its protein-like structure, synthetic chemical nature, agonist pharmacologic activity, and mode of administration. In repeat-dose toxicity studies, the salient findings were dose-related increases in circulating platelet counts, mean platelet volume, and megakaryocytes in the bone marrow with no antibody formation. Reversible myelofibrosis and hyperostosis were observed in rats, but not dogs, when the circulating platelet levels exceeded 3× those of vehicle controls. The bone effects were due to the exaggerated pharmacologic effect and excessive stimulation and elevation of megakaryocytes by TPO, which results in intramedullary proliferation of fibroblasts and mesenchymal cells followed by osseous metaplasia. These findings support the use of platelet elevations of >3× as a stopping criterion to prevent potential adverse bone-related effects in humans.


Assuntos
Plaquetas/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Homeostase/efeitos dos fármacos , Peptídeos/farmacocinética , Animais , Plaquetas/citologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Cães , Relação Dose-Resposta a Droga , Feminino , Cobaias , Humanos , Hiperostose/patologia , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Megacariócitos/efeitos dos fármacos , Peptídeos/imunologia , Peptídeos/toxicidade , Mielofibrose Primária/patologia , Coelhos , Ratos , Trombocitopenia/tratamento farmacológico , Testes de Toxicidade
11.
Toxicol Pathol ; 37(1): 28-33, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19171926

RESUMO

Recently, in early clinical development, a few biologics and small molecules intended as antitumor or anti-inflammatory agents have caused a severe adverse pro-inflammatory systemic reaction also known as systemic inflammatory response syndrome (SIRS). This toxicity could result from expected pharmacological effects of a therapeutic antibody and/or from interaction with antigens expressed on cells/tissues other than the intended target. Clinical monitoring of SIRS is challenging because of the narrow diagnostic window to institute a successful intervening therapeutic strategy prior to acute circulatory collapse. Furthermore, for these classes of therapeutic agents, studies in animals have low predictive ability to identify potential human hazards. In vitro screens with human cells, though promising, need further development. Therefore, identification of improved preclinical diagnostic markers of SIRS will enable clinicians to select applicable markers for clinical testing and avoid potentially catastrophic events. There is limited preclinical toxicology data describing the interspecies performance of acute-phase proteins because the response time, type, and duration of major acute-phase proteins vary significantly between species. This review will attempt to address this intellectual gap, as well as the use and applicability of acute-phase proteins as preclinical to clinical translational biomarkers of SIRS.


Assuntos
Proteínas de Fase Aguda/metabolismo , Biomarcadores/metabolismo , Avaliação Pré-Clínica de Medicamentos , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Testes de Toxicidade/métodos , Animais , Modelos Animais de Doenças , Cães , Humanos , Camundongos , Ratos , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Síndrome de Resposta Inflamatória Sistêmica/diagnóstico , Xenobióticos/toxicidade
12.
J Clin Invest ; 114(10): 1457-66, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15545996

RESUMO

Hemoglobin (Hb) A production during red blood cell development is coordinated to minimize the deleterious effects of free alpha- and beta-Hb subunits, which are unstable and cytotoxic. The alpha-Hb-stabilizing protein (AHSP) is an erythroid protein that specifically binds alpha-Hb and prevents its precipitation in vitro, which suggests that it may function to limit free alpha-Hb toxicities in vivo. We investigated this possibility through gene ablation and biochemical studies. AHSP(-/-) erythrocytes contained hemoglobin precipitates and were short-lived. In hematopoietic tissues, erythroid precursors were elevated in number but exhibited increased apoptosis. Consistent with unstable alpha-Hb, AHSP(-/-) erythrocytes contained increased ROS and evidence of oxidative damage. Moreover, purified recombinant AHSP inhibited ROS production by alpha-Hb in solution. Finally, loss of AHSP worsened the phenotype of beta-thalassemia, a common inherited anemia characterized by excess free alpha-Hb. Together, the data support a model in which AHSP binds alpha-Hb transiently to stabilize its conformation and render it biochemically inert prior to Hb A assembly. This function is essential for normal erythropoiesis and, to a greater extent, in beta-thalassemia. Our findings raise the possibility that altered AHSP expression levels could modulate the severity of beta-thalassemia in humans.


Assuntos
Eritrócitos/metabolismo , Eritropoese , Hemoglobinas/química , Hemoglobinas/fisiologia , Talassemia beta/metabolismo , Animais , Apoptose , Eritrócitos/patologia , Corpos de Heinz/química , Corpos de Heinz/metabolismo , Hemoglobinas/genética , Heterozigoto , Cinética , Camundongos , Camundongos Knockout , Modelos Biológicos , Conformação Proteica , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
13.
Chem Biol Interact ; 221: 109-18, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25130857

RESUMO

Canagliflozin is an SGLT2 inhibitor used for the treatment of type 2 diabetes mellitus. Studies were conducted to investigate the mechanism responsible for renal tubular tumors and pheochromocytomas observed at the high dose in a 2-year carcinogenicity study in rats. At the high dose (100mg/kg) in rats, canagliflozin caused carbohydrate malabsorption evidenced by inhibition of intestinal glucose uptake, decreased intestinal pH and increased urinary calcium excretion. In a 6-month mechanistic study utilization of a glucose-free diet prevented carbohydrate malabsorption and its sequelae, including increased calcium absorption and urinary calcium excretion, and hyperostosis. Cell proliferation in the kidney and adrenal medulla was increased in rats maintained on standard diet and administered canagliflozin (100mg/kg), and in addition an increase in the renal injury biomarker KIM-1 was observed. Increased cell proliferation is considered as a proximal event in carcinogenesis. Effects on cell proliferation, KIM-1 and calcium excretion were inhibited in rats maintained on the glucose-free diet, indicating they are secondary to carbohydrate malabsorption and are not direct effects of canagliflozin.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos , Carcinogênese , Glucosídeos/farmacologia , Síndromes de Malabsorção , Inibidores do Transportador 2 de Sódio-Glicose , Tiofenos/farmacologia , Animais , Canagliflozina , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Imuno-Histoquímica , Rim/patologia , Masculino , Ratos , Ratos Sprague-Dawley
14.
Toxicol Sci ; 105(1): 221-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18539914

RESUMO

These studies describe the effect of N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide (AR-M100390), a delta-opioid agonist, on the pancreas and its mechanisms for pancreatic toxicity. Rats were treated with 5, 100, and 600 micromol/kg of AR-M100390 for 3 and/or 7 days; another group of rats treated with 600 micromol/kg of compound were allowed to recover for 14 days. AR-M100390 (600 micromol/kg) caused vacuolation in the beta-cell of the rat pancreas that was associated with depletion of insulin and hyperglycemia after 7 days of dosing. The loss of insulin by AR-M100390 was due to specific inhibition of rat insulin2 mRNA transcription in vivo. Insulin depletion and hyperglycemia were reversible. The effects of AR-M100390 in rats were reproduced in the rat pancreatic beta-cell line RINm5F, where it inhibited intracellular insulin content and secretion without affecting cell survival. Loss of insulin in vitro was also a result of specific inhibition of insulin2 mRNA transcription and was reversible. Pretreatment of cells with the delta-opioid antagonist naltrindole or pertussis toxin did not reverse loss of insulin in AR-M100390-treated cells suggesting that the effects were not mediated by the delta-opioid receptor. AR-M100390 inhibited KCl-mediated calcium mobilization in RINm5F cells, suggesting that L-type calcium channels found in these cells and in pancreatic beta-cells may partially play a role in the inhibition of insulin secretion by this compound. In summary, the in vitro and in vivo studies suggest that inhibition of insulin by AR-M100390 is due to a combination of inhibition of insulin synthesis and/or release.


Assuntos
Benzamidas/toxicidade , Insulina/metabolismo , Pâncreas/efeitos dos fármacos , Piperidinas/toxicidade , Receptores Opioides delta/agonistas , Animais , Glicemia/análise , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Ciclizina/toxicidade , Relação Dose-Resposta a Droga , Insulina/genética , Pâncreas/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Wistar
15.
Toxicol Pathol ; 34(1): 19-26, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16507540

RESUMO

In preclinical safety studies, drug-induced vascular injury can negatively impact candidate-drug selection because there are no obvious diagnostic markers for monitoring this pathology preclinically or clinically. Furthermore, our current understanding of the pathogenesis of this lesion is limited. While vasodilatation and increased shear stress appear to play a role, the exact mechanism(s) of injury to the primary target cells, smooth muscle (SMC) and endothelial cell (EC), are unknown. Evaluation of potential novel markers for clinical monitoring with a mechanistic underpinning would add value in risk assessment and risk management. This mini review focuses on the efforts and progress to identify diagnostic markers as well as understanding the mechanism of action in nonrodent drug-induced vascular injury.


Assuntos
Biomarcadores/metabolismo , Drogas em Investigação/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/metabolismo , Animais , Biomarcadores/análise , Cães , Avaliação Pré-Clínica de Medicamentos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Óxido Nítrico/análise , Óxido Nítrico/metabolismo , Doenças Vasculares/patologia , Fator de von Willebrand/análise , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA