Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2394: 823-835, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35094360

RESUMO

Aminolevulinic acid (ALA) has been clinically used as an intraoperative fluorescence probe for protoporphyrin IX (PpIX) fluorescence-guided tumor resection and a PDT agent for cancer treatment. Although tumor tissues often show increased ALA-PpIX fluorescence compared with normal tissues, which enables the use of ALA for tumor imaging and targeting, weak tumor PpIX fluorescence as well as the heterogeneity in tumor fluorescence severely limits its clinical application. Intracellular PpIX in tumor cells is reduced by two major mechanisms, efflux by ATP-binding cassette (ABC) transporters such as ABCG2 and bioconversion to form heme by ferrochelatase (FECH) in the heme biosynthesis pathway. Targeting these two predominant PpIX-reducing mechanisms for the enhancement of ALA-PpIX have yielded a plethora of promising results and stimulated the clinical exploration of these enhancement strategies. Here we describe our methods of evaluating chemicals for the inhibition of ABCG2 transporter and FECH activity. Our goal is to further encourage research and development of novel ABCG2 and FECH inhibitors and promote a rational use of these inhibitors to optimize ALA-based tumor detection and treatment.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Ácido Aminolevulínico , Inibidores Enzimáticos , Ferroquelatase , Fotoquimioterapia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Ácido Aminolevulínico/farmacologia , Animais , Linhagem Celular Tumoral , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/metabolismo , Fluorescência , Humanos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas
2.
FASEB J ; 34(9): 12419-12435, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32716567

RESUMO

Ferrochelatase (FECH) is the terminal enzyme in heme biosynthesis. We previously showed that FECH is required for endothelial cell growth in vitro and choroidal neovascularization in vivo. But FECH has not been explored in retinal neovascularization, which underlies diseases like proliferative diabetic retinopathy and retinopathy of prematurity. Here, we investigated the inhibition of FECH using genetic and chemical approaches in the oxygen-induced retinopathy (OIR) mouse model. In OIR mice, FECH expression is upregulated and co-localized with neovascular tufts. Partial loss-of-function Fechm1Pas  mutant mice showed reduced retinal neovascularization and endothelial cell proliferation in OIR. An intravitreal injection of the FECH inhibitor N-methyl protoporphyrin had similar effects. Griseofulvin is an antifungal drug that inhibits FECH as an off-target effect. Strikingly, intravitreal griseofulvin decreased both pathological tuft formation and areas of vasoobliteration compared to vehicle, suggesting potential as a FECH-targeting therapy. Ocular toxicity studies revealed that intravitreal injection of griseofulvin in adult mice does not disrupt retinal vasculature, function, or morphology. In sum, mutation and chemical inhibition of Fech reduces retinal neovascularization and promotes physiological angiogenesis, suggesting a dual effect on vascular repair upon FECH inhibition, without ocular toxicity. These findings suggest that FECH inhibitors could be repurposed to treat retinal neovascularization.


Assuntos
Ferroquelatase/fisiologia , Neovascularização Retiniana/etiologia , Animais , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Ferroquelatase/antagonistas & inibidores , Griseofulvina/farmacologia , Griseofulvina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Retina/efeitos dos fármacos , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/patologia
3.
PLoS One ; 13(7): e0200307, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29985945

RESUMO

Ferrochelatase (FECH) is an enzyme necessary for heme synthesis, which is essential for maintaining normal functions of endothelial nitric oxide synthase (eNOS) and soluble guanylyl cyclase (sGC). We tested the hypothesis that inhibition of vascular FECH to attenuate heme synthesis downregulates eNOS and sGC expression, resulting in impaired NO/cGMP-dependent relaxation. To this end, isolated bovine coronary arteries (BCAs) were in vitro incubated without (as controls) or with N-methyl protoporphyrin (NMPP; 10(-5)-10(-7)M; a selective FECH antagonist) for 24 and 72 hours respectively. Tissue FECH activity, heme, nitrite/NO and superoxide levels were sequentially measured. Protein expression of FECH, eNOS and sGC was detected by western blot analysis. Vascular responses to various vasoactive agents were evaluated via isometric tension studies. Treatment of BCAs with NMPP initiated a time- and dose-dependent attenuation of FECH activity without changes in its protein expression, followed by significant reduction in the heme level. Moreover, ACh-induced relaxation and ACh-stimulated release of NO were significant reduced, associated with suppression of eNOS protein expression in NMPP-treated groups. Decreased relaxation to NO donor spermine-NONOate reached the statistical significance in BCAs incubated with NMPP for 72 hours, concomitantly with downregulation of sGCß1 expression that was independent of heat shock protein 90 (HSP90), nor did it significantly affect BCA relaxation caused by BAY 58-2667 that activates sGC in the heme-deficiency. Neither vascular responses to non-NO/sGC-mediators nor production of superoxide was affected by NMPP-treatment. In conclusion, deletion of vascular heme production via inhibiting FECH elicits downregulation of eNOS and sGC expression, leading to an impaired NO-mediated relaxation in an oxidative stress-independent manner.


Assuntos
Vasos Coronários/efeitos dos fármacos , GMP Cíclico/metabolismo , Ferroquelatase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel/metabolismo , Acetilcolina/farmacologia , Animais , Bovinos , Vasos Coronários/metabolismo , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP90/metabolismo , Protoporfirinas/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
4.
Sci Rep ; 7: 41975, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28176804

RESUMO

Griseofulvin, an orally active antifungal drug used to treat dermatophyte infections, has a secondary effect of inducing cytochrome P450-mediated production of N-methyl protoporphyrin IX (N-MPP). N-MPP is a potent competitive inhibitor of the heme biosynthetic-enzyme ferrochelatase, and inhibits the growth of cultured erythrocyte stage Plasmodium falciparum. Novel drugs against Plasmodium are needed to achieve malaria elimination. Thus, we investigated whether griseofulvin shows anti-plasmodial activity. We observed that the intraerythrocytic growth of P. falciparum is inhibited in red blood cells pretreated with griseofulvin in vitro. Treatment with 100 µM griseofulvin was sufficient to prevent parasite growth and induce the production of N-MPP. Inclusion of the ferrochelatase substrate PPIX blocked the inhibitory activity of griseofulvin, suggesting that griseofulvin exerts its activity through the N-MPP-dependent inhibition of ferrochelatase. In an ex-vivo study, red blood cells from griseofulvin-treated subjects were refractory to the growth of cultured P. falciparum. However, in a clinical trial griseofulvin failed to show either therapeutic or prophylactic effect in subjects infected with blood stage P. falciparum. Although the development of griseofulvin as an antimalarial is not warranted, it represents a novel inhibitor of P. falciparum growth and acts via the N-MPP-dependent inhibition of ferrochelatase.


Assuntos
Antifúngicos/uso terapêutico , Eritrócitos/parasitologia , Ferroquelatase/antagonistas & inibidores , Griseofulvina/uso terapêutico , Malária Falciparum/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Adolescente , Adulto , Animais , Antifúngicos/metabolismo , Estudos de Casos e Controles , Estudos de Coortes , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Feminino , Seguimentos , Griseofulvina/metabolismo , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/patologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Plasmodium falciparum/efeitos dos fármacos , Prognóstico , Adulto Jovem
5.
ACS Chem Biol ; 11(5): 1245-54, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-26863403

RESUMO

Many protein kinases are valid drug targets in oncology because they are key components of signal transduction pathways. The number of clinical kinase inhibitors is on the rise, but these molecules often exhibit polypharmacology, potentially eliciting desired and toxic effects. Therefore, a comprehensive assessment of a compound's target space is desirable for a better understanding of its biological effects. The enzyme ferrochelatase (FECH) catalyzes the conversion of protoporphyrin IX into heme and was recently found to be an off-target of the BRAF inhibitor Vemurafenib, likely explaining the phototoxicity associated with this drug in melanoma patients. This raises the question of whether FECH binding is a more general feature of kinase inhibitors. To address this, we applied a chemical proteomics approach using kinobeads to evaluate 226 clinical kinase inhibitors for their ability to bind FECH. Surprisingly, low or submicromolar FECH binding was detected for 29 of all compounds tested and isothermal dose response measurements confirmed target engagement in cells. We also show that Vemurafenib, Linsitinib, Neratinib, and MK-2461 reduce heme levels in K562 cells, verifying that drug binding leads to a loss of FECH activity. Further biochemical and docking experiments identified the protoporphyrin pocket in FECH as one major drug binding site. Since the genetic loss of FECH activity leads to photosensitivity in humans, our data strongly suggest that FECH inhibition by kinase inhibitors is the molecular mechanism triggering photosensitivity in patients. We therefore suggest that a FECH assay should generally be part of the preclinical molecular toxicology package for the development of kinase inhibitors.


Assuntos
Ferroquelatase/antagonistas & inibidores , Ferroquelatase/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Benzocicloeptenos/farmacologia , Linhagem Celular Tumoral , Ferroquelatase/química , Heme/metabolismo , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Simulação de Acoplamento Molecular , Ligação Proteica , Proteômica , Pirazinas/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Sulfonamidas/farmacologia , Vemurafenib
6.
J Antimicrob Chemother ; 71(4): 946-52, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26747101

RESUMO

OBJECTIVES: During recent decades, the number of invasive fungal infections among immunosuppressed patients has increased significantly, whereas the number of effective systemic antifungal drugs remains low and unsatisfactory. The aim of this study was to characterize a novel antifungal compound, CW-8/haemofungin, which we previously identified in a screen for compounds affecting fungal cell wall integrity. METHODS: The in vitro characteristics of haemofungin were investigated by MIC evaluation against a panel of pathogenic and non-pathogenic fungi, bacteria and mammalian cells in culture. Haemofungin mode-of-action studies were performed by screening an Aspergillus nidulans overexpression genomic library for resistance-conferring plasmids and biochemical validation of the target. In vivo efficacy was tested in the Galleria mellonella and Drosophila melanogaster insect models of infection. RESULTS: We demonstrate that haemofungin causes swelling and lysis of growing fungal cells. It inhibits the growth of pathogenic Aspergillus, Candida, Fusarium and Rhizopus isolates at micromolar concentrations, while only weakly affecting the growth of mammalian cell lines. Genetic and biochemical analyses in A. nidulans and Aspergillus fumigatus indicate that haemofungin primarily inhibits ferrochelatase (HemH), the last enzyme in the haem biosynthetic pathway. Haemofungin was non-toxic and significantly reduced mortality rates of G. mellonella and D. melanogaster infected with A. fumigatus and Rhizopus oryzae, respectively. CONCLUSIONS: Further development and in vivo validation of haemofungin is warranted.


Assuntos
Antifúngicos/farmacologia , Heme/antagonistas & inibidores , Heme/biossíntese , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Animais , Aspergillus nidulans/efeitos dos fármacos , Aspergillus nidulans/genética , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Linhagem Celular , Farmacorresistência Fúngica , Sinergismo Farmacológico , Ferroquelatase/antagonistas & inibidores , Fungos/efeitos dos fármacos , Fungos/crescimento & desenvolvimento , Humanos , Insetos , Testes de Sensibilidade Microbiana , Micoses/tratamento farmacológico , Micoses/microbiologia , Protoporfirinas/biossíntese
7.
Sci Rep ; 5: 10488, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25990790

RESUMO

It is well known that haem serves as the prosthetic group of various haemoproteins that function in oxygen transport, respiratory chain, and drug metabolism. However, much less is known about the functions of the catabolites of haem in mammalian cells. Haem is enzymatically degraded to iron, carbon monoxide (CO), and biliverdin, which is then converted to bilirubin. Owing to difficulties in measuring bilirubin, however, the generation and transport of this end product remain unclear despite its clinical importance. Here, we used UnaG, the recently identified bilirubin-binding fluorescent protein, to analyse bilirubin production in a variety of human cell lines. We detected a significant amount of bilirubin with many non-blood cell types, which was sensitive to inhibitors of haem metabolism. These results suggest that there is a basal level of haem synthesis and its conversion into bilirubin. Remarkably, substantial changes were observed in the bilirubin generation when cells were exposed to stress insults. Since the stress-induced cell damage was exacerbated by the pharmacological blockade of haem metabolism but was ameliorated by the addition of biliverdin and bilirubin, it is likely that the de novo synthesis of haem and subsequent conversion to bilirubin play indispensable cytoprotective roles against cell damage.


Assuntos
Bilirrubina/metabolismo , Citoproteção/fisiologia , Heme Oxigenase-1/metabolismo , Heme/metabolismo , Arsenitos/farmacologia , Cloreto de Cádmio/farmacologia , Linhagem Celular Tumoral , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/metabolismo , Corantes Fluorescentes/metabolismo , Células HEK293 , Células HeLa , Heme/biossíntese , Heme Oxigenase-1/antagonistas & inibidores , Células Hep G2 , Humanos , Células MCF-7 , Malatos/farmacologia , Mitocôndrias/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Ligação Proteica , Compostos de Sódio/farmacologia
8.
Blood ; 125(3): 534-41, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25414439

RESUMO

Many red cell polymorphisms are a result of selective pressure by the malarial parasite. Here, we add another red cell disease to the panoply of erythrocytic changes that give rise to resistance to malaria. Erythrocytes from individuals with erythropoietic protoporphyria (EPP) have low levels of the final enzyme in the heme biosynthetic pathway, ferrochelatase. Cells from these patients are resistant to the growth of Plasmodium falciparum malarial parasites. This phenomenon is due to the absence of ferrochelatase and not an accumulation of substrate, as demonstrated by the normal growth of P falciparum parasites in the EPP phenocopy, X-linked dominant protoporphyria, which has elevated substrate, and normal ferrochelatase levels. This observation was replicated in a mouse strain with a hypomorphic mutation in the murine ferrochelatase gene. The parasite enzyme is not essential for parasite growth as Plasmodium berghei parasites carrying a complete deletion of the ferrochelatase gene grow normally in erythrocytes, which confirms previous studies. That ferrochelatase is essential to parasite growth was confirmed by showing that inhibition of ferrochelatase using the specific competitive inhibitor, N-methylprotoporphyrin, produced a potent growth inhibition effect against cultures of P falciparum. This raises the possibility of targeting human ferrochelatase in a host-directed antimalarial strategy.


Assuntos
Eritrócitos/parasitologia , Ferroquelatase/fisiologia , Malária Falciparum/prevenção & controle , Plasmodium berghei/crescimento & desenvolvimento , Protoporfiria Eritropoética/prevenção & controle , Animais , Eritrócitos/enzimologia , Feminino , Ferroquelatase/antagonistas & inibidores , Heme/metabolismo , Humanos , Malária Falciparum/enzimologia , Malária Falciparum/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Protoporfiria Eritropoética/enzimologia , Protoporfiria Eritropoética/parasitologia , Protoporfirinas/farmacologia
9.
Mol Pharmacol ; 84(6): 824-33, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24043703

RESUMO

Salicylic acid is a classic nonsteroidal anti-inflammatory drug. Although salicylic acid also induces mitochondrial injury, the mechanism of its antimitochondrial activity is not well understood. In this study, by using a one-step affinity purification scheme with salicylic acid-immobilized beads, ferrochelatase (FECH), a homodimeric enzyme involved in heme biosynthesis in mitochondria, was identified as a new molecular target of salicylic acid. Moreover, the cocrystal structure of the FECH-salicylic acid complex was determined. Structural and biochemical studies showed that salicylic acid binds to the dimer interface of FECH in two possible orientations and inhibits its enzymatic activity. Mutational analysis confirmed that Trp301 and Leu311, hydrophobic amino acid residues located at the dimer interface, are directly involved in salicylic acid binding. On a gel filtration column, salicylic acid caused a shift in the elution profile of FECH, indicating that its conformational change is induced by salicylic acid binding. In cultured human cells, salicylic acid treatment or FECH knockdown inhibited heme synthesis, whereas salicylic acid did not exert its inhibitory effect in FECH knockdown cells. Concordantly, salicylic acid treatment or FECH knockdown inhibited heme synthesis in zebrafish embryos. Strikingly, the salicylic acid-induced effect in zebrafish was partially rescued by FECH overexpression. Taken together, these findings illustrate that FECH is responsible for salicylic acid-induced inhibition of heme synthesis, which may contribute to its antimitochondrial and anti-inflammatory function. This study establishes a novel aspect of the complex pharmacological effects of salicylic acid.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Ferroquelatase/antagonistas & inibidores , Heme/antagonistas & inibidores , Mitocôndrias/efeitos dos fármacos , Ácido Salicílico/efeitos adversos , Animais , Anti-Inflamatórios não Esteroides/química , Linhagem Celular Tumoral , Cristalografia por Raios X , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Proteínas de Escherichia coli/química , Ferroquelatase/biossíntese , Ferroquelatase/química , Heme/biossíntese , Humanos , Mitocôndrias/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Protoporfirinas/metabolismo , Ácido Salicílico/química , Peixe-Zebra
10.
Acta Med Okayama ; 67(3): 153-64, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23804138

RESUMO

Ever since protoporphyrin IX (PpIX) was discovered to accumulate preferentially in cancer cells after 5-aminolevulinic acid (ALA) treatment, photodynamic treatment or therapy (PDT) has been developed as an exciting new treatment option for cancer patients. However, the level of PpIX accumulation in oral cancer is fairly low and insufficient for PDT. Ferrochelatase (FECH) and ATP-binding cassette transporter G2 (ABCG2) are known to regulate PpIX accumulation. In addition, serum enhances PpIX export by ABCG2. We investigated here whether and how inhibitors of FECH and ABCG2 and their combination could improve PpIX accumulation and PDT efficacy in an oral cancer cell line in serum-containing medium. ABCG2 inhibitor and the combination of ABCG2 and FECH inhibitors increased PpIX in the presence of fetal bovine serum (FBS) in an oral cancer cell line. Analysis of ABCG2 gene silencing also revealed the involvement of ABCG2 in the regulation of PpIX accumulation. Inhibitors of FECH and ABCG2, and their combination increased the efficiency of ALA-PDT even in the presence of FBS. ALA-PDT-induced cell death was accompanied by apoptotic events and lipid peroxidation. These results suggest that accumulation of PpIX is determined by the activities of ABCG2 and FECH and that treatment with a combination of their inhibitors improves the efficacy of PDT for oral cancer, especially in the presence of serum.


Assuntos
Ácido Aminolevulínico/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Bucais/tratamento farmacológico , Fotoquimioterapia/métodos , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Sanguíneas/farmacologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Desferroxamina/farmacologia , Ferroquelatase/antagonistas & inibidores , Inativação Gênica , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Proteínas de Neoplasias/genética , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/metabolismo , Sideróforos/farmacologia
11.
Cancer Sci ; 104(6): 765-72, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23480042

RESUMO

The purpose of the present study was to investigate the mechanism of photodynamic therapy (PDT) supplemented with exogenously added 5-aminolevulinic acid (ALA) on human urothelial cancer (UC). Moreover, we aimed to determine whether the therapeutic effects of ALA-based PDT (ALA-PDT) for UC could be enhanced by deferoxamine (DFX), an inhibitor of ferrochelatase. The efficiency of ALA-PDT on these cells was analyzed using flow cytometry and the type of cell death was also assessed. The ALA-PDT promoting effect of DFX was examined on both UC cells and human umbilical vein endothelial cells (HUVEC). The ALA-PDT decreased levels of mitochondrial membrane potential and induced cell death mainly via apoptosis in these cells. Moreover, inhibition of ferrochelatase by DFX led to an increase of protoporphyrin IX (PpIX) accumulation and enhanced the effect of ALA-PDT on UC cells. We further investigated the effect of DFX on in vivo PDT with a tumor-bearing animal model and found that DFX efficiently enhanced tumor cell apoptosis. ALA-PDT induced death of neovascular endothelial cells in tumors but did not affect small vessel endothelial cells in normal tissues surrounding the tumor. Furthermore, DFX enhanced inhibition of neovascularization. These results demonstrated ALA-PDT dominantly induced apoptosis over necrosis by direct action on UC as well as via antiangiogenic action on neovacular endothelial cells, suggesting that the therapeutic damage by ALA-PDT could be kept to a minimum in the surrounding normal tissues. In addition, increased accumulation of PpIX by DFX could enhance this effectiveness of ALA-PDT.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinoma de Células de Transição/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Ácido Aminolevulínico/farmacologia , Animais , Apoptose , Carcinoma de Células de Transição/enzimologia , Linhagem Celular , Desferroxamina/farmacologia , Inibidores Enzimáticos/farmacologia , Ferroquelatase/antagonistas & inibidores , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Protoporfirinas , Neoplasias da Bexiga Urinária/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cell Biochem ; 358(1-2): 297-307, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21748335

RESUMO

Accumulation of protoporphyrin IX (PpIX) in cancer cells is a basis of 5-aminolevulinic acid (ALA)-induced photodymanic therapy. We studied factors that affect PpIX accumulation in human urothelial carcinoma cell line T24, with particular emphasis on ATP-binding cassette transporter G2 (ABCG2) and serum in the medium. When the medium had no fetal bovine serum (FBS), ALA induced PpIX accumulation in a time- and ALA concentration-dependent manner. Inhibition of heme-synthesizing enzyme, ferrochelatase, by nitric oxide donor (Noc18) or deferoxamine resulted in a substantial increase in the cellular PpIX accumulation, whereas ABCG2 inhibition by fumitremorgin C or verapamil induced a slight PpIX increase. When the medium was added with FBS, cellular accumulation of PpIX stopped at a lower level with an increase of PpIX in the medium, which suggested PpIX efflux. ABCG2 inhibitors restored the cellular PpIX level to that of FBS(-) samples, whereas ferrochelatase inhibitors had little effects. Bovine serum albumin showed similar effects to FBS. Fluorescence microscopic observation revealed that inhibitors of ABC transporter affected the intracellular distribution of PpIX. These results indicated that ABCG2-mediated PpIX efflux was a major factor that prevented PpIX accumulation in cancer cells in the presence of serum. Inhibition of ABCG2 transporter system could be a new target for the improvement of photodynamic therapy.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Neoplasias/metabolismo , Protoporfirinas/metabolismo , Soro/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Ácido Aminolevulínico/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Bovinos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Heme/biossíntese , Humanos , Indóis/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Compostos Nitrosos/farmacologia , Protoporfirinas/biossíntese , Soroalbumina Bovina/metabolismo
13.
Pathobiology ; 76(6): 303-14, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19955842

RESUMO

PURPOSE: The purpose of this study was to clarify the regulatory mechanism of protoporphyrin IX (PpIX) synthesis mediated by 5-aminolevulinic acid (ALA) in human urothelial carcinoma (UC), leading to improved accuracy in photodynamic diagnosis and therapy using ALA. EXPERIMENTAL DESIGN: PpIX accumulation in cultured UC cells after incubation for 1-5 h with 0.5-5 mM ALA was analyzed by fluorescence analysis using fluorescence microscopy and flow cytometry technique. RESULTS: PpIX fluorescence mediated by ALA was increased, and the intensity of PpIX fluorescence was time-dependently increased in UC cells compared to noncancerous cells. The distribution of endogenous PpIX fluorescence primarily coincided with mitochondria, and then increased at a specific perinuclear region in the cells during the time of incubation. The ALA-mediated PpIX synthesis in UC cells was suppressed by beta-alanine, an inhibitor of beta-transporters of cell membrane, and carbonylcyanide p-trifluoromethoxyphenyl hydrazone, an uncoupler of mitochondrial oxidative phosphorylation. In contrast, the ALA-mediated PpIX accumulation was increased by deferoxamine, an iron chelator, manganese and nitric oxide, which is contributed to PpIX metabolism by inhibiting ferrochelatase activity, generated by a nitric oxide-generating reagent NOC-18. As observed above, ALA-mediated PpIX synthesis in human UC cells was regulated by the process of ALA uptake, ALA conversion to PpIX and metabolism of accumulated PpIX to heme. CONCLUSIONS: This shows that the suppression of ferrochelatase increased PpIX accumulation in UC cells using small amount of ALA, thus leading to an improved clinical practicability of photodynamic diagnosis and therapy.


Assuntos
Ácido Aminolevulínico/farmacologia , Carcinoma de Células de Transição/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/biossíntese , Neoplasias da Bexiga Urinária/metabolismo , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Carcinoma de Células de Transição/tratamento farmacológico , Linhagem Celular Tumoral , Desferroxamina/farmacologia , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/metabolismo , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Compostos Nitrosos/farmacologia , Fosforilação Oxidativa/efeitos dos fármacos , Fatores de Tempo , Neoplasias da Bexiga Urinária/tratamento farmacológico , Urotélio/efeitos dos fármacos , Urotélio/metabolismo , beta-Alanina/farmacologia
14.
Cell Biochem Funct ; 27(8): 503-15, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19735078

RESUMO

Photodynamic therapy (PDT) for tumors is based on the tumor-selective accumulation of a photosensitizer, protoporphyrin IX (PpIX), followed by irradiation with visible light. However, the molecular mechanism of cell death caused by PDT has not been fully elucidated. The 5-aminolevulinic acid (ALA)-based photodynamic action (PDA) was dependent on the accumulation of PpIX, the level of which decreased rapidly by eliminating ALA from the incubation medium in human histiocytic lymphoma U937 cells. PDA induced apoptosis characterized by lipid peroxidation, increase in Bak and Bax/Bcl-xL, decrease in Bid, membrane depolarization, cytochrome c release, caspase-3 activation, phosphatidylserine (PS) externalization. PDT-induced cell death seemed to occur predominantly via apoptosis through distribution of PpIX in mitochondria. These cell death events were enhanced by ferrochelatase inhibitors. These results indicated that ALA-based-PDA induced apoptotic cell death through a mitochondrial pathway and that ferrochelatase inhibitors might enhanced the effect of PDT for tumors even at low concentrations of ALA.


Assuntos
Ácido Aminolevulínico/farmacologia , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ferroquelatase/antagonistas & inibidores , Linfoma Difuso de Grandes Células B/fisiopatologia , Fármacos Fotossensibilizantes/farmacologia , Ferroquelatase/metabolismo , Humanos , Luz , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/enzimologia , Fotoquimioterapia , Protoporfirinas/farmacologia , Células U937
15.
J Pediatr Hematol Oncol ; 31(9): 684-6, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19707157

RESUMO

Linezolid (LZD)-induced myelosuppression has been reported in adults; however, LZD-induced pure red cell precursor toxicity rarely occurs. A 2-year-old boy diagnosed with infective endocarditis by Streptococcus mitis received LZD after developing resistance to multiple antibiotics. Although his infective symptoms were improved by LZD, progressive anemia was noticed 2 weeks after LZD therapy. Four weeks after LZD administration, his hemoglobin level was 6.5 g/dL and reticulocytes less than 0.1%. Bone marrow examination revealed markedly decreased erythropoiesis with cytoplasmic vacuolation of erythroblasts. Anemia recovered 19 days after cessation of LZD. Elevated protoporphyrin and a high LZD level in the blood suggested that mitochondrial disturbance by high-dose and long-term treatment with LZD may have been responsible for LZD-induced pure red cell precursor toxicity.


Assuntos
Acetamidas/efeitos adversos , Antibacterianos/efeitos adversos , Endocardite Bacteriana/tratamento farmacológico , Oxazolidinonas/efeitos adversos , Aplasia Pura de Série Vermelha/induzido quimicamente , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus mitis , Acetamidas/farmacologia , Acetamidas/uso terapêutico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Pré-Escolar , Farmacorresistência Bacteriana Múltipla , Transfusão de Eritrócitos , Células Precursoras Eritroides/efeitos dos fármacos , Ferroquelatase/antagonistas & inibidores , Humanos , Linezolida , Masculino , Mitocôndrias/efeitos dos fármacos , Oxazolidinonas/farmacologia , Oxazolidinonas/uso terapêutico , Protoporfirinas/sangue , Aplasia Pura de Série Vermelha/terapia , Ribossomos/efeitos dos fármacos , Streptococcus mitis/efeitos dos fármacos
16.
J Neural Transm (Vienna) ; 116(1): 79-88, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19002554

RESUMO

Decay of mitochondria, energy failure and increased oxidative stress are features commonly detected in brains from Alzheimer's disease (AD) patients. Recent findings indicate that neuronal heme deficiency may contribute to the appearance of those cytopathologies and potentially alter the course of AD. We repressed heme synthesis in cells by inhibiting ferrochelatase enzyme with small interfering RNA and N-methylprotoporphyrin IX. The treatments induced a severe perturbation of mitochondria and energy production, with decrease of the subunit II of Cytochrome c Oxidase, alteration of the membrane potential and a 50% reduction of intracellular ATP. The state and processing of the Amyloid Precursor Protein (APP) was also affected, with the appearance of APP aggregates and a significant decrease (30-40%) of sAPPalpha secretion, associated with perturbation of ADAM10 and TACE, enzymes involved in the alpha-secretase cleavage. The production of sAPPbeta was increased, without augment of Amyloid beta generation. Our findings strengthen the hypothesis that a reduced availability of heme may play a role in AD pathogenesis.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Heme/antagonistas & inibidores , Heme/metabolismo , Fenômenos Fisiológicos/efeitos dos fármacos , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Trifosfato de Adenosina/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Contagem de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/farmacologia , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/genética , Humanos , Proteínas de Membrana/metabolismo , Neuroblastoma , Fragmentos de Peptídeos/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Protoporfirinas/farmacologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transfecção
17.
J Biol Chem ; 283(35): 23685-91, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18593702

RESUMO

Ferrochelatase catalyzes the insertion of ferrous iron into protoporphyrin IX to form heme. Robust kinetic analyses of the reaction mechanism are complicated by the instability of ferrous iron in aqueous solution, particularly at alkaline pH values. At pH 7.00 the half-life for spontaneous oxidation of ferrous ion is approximately 2 min in the absence of metal complexing additives, which is sufficient for direct comparisons of alternative metal ion substrates with iron. These analyses reveal that purified recombinant ferrochelatase from both murine and yeast sources inserts not only ferrous iron but also divalent cobalt, zinc, nickel, and copper into protoporphyrin IX to form the corresponding metalloporphyrins but with considerable mechanistic variability. Ferrous iron is the preferred metal ion substrate in terms of apparent k(cat) and is also the only metal ion substrate not subject to severe substrate inhibition. Substrate inhibition occurs in the order Cu(2+) > Zn(2+) > Co(2+) > Ni(2+) and can be alleviated by the addition of metal complexing agents such as beta-mercaptoethanol or imidazole to the reaction buffer. These data indicate the presence of two catalytically significant metal ion binding sites that may coordinately regulate a selective processivity for the various potential metal ion substrates.


Assuntos
Ferroquelatase/química , Heme/química , Metais Pesados/química , Protoporfirinas/química , Animais , Catálise , Cátions Bivalentes/química , Cátions Bivalentes/metabolismo , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/genética , Ferroquelatase/metabolismo , Heme/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Imidazóis/química , Mercaptoetanol/química , Metais Pesados/metabolismo , Camundongos , Oxirredução , Protoporfirinas/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Especificidade por Substrato/fisiologia
18.
Biochem J ; 399(1): 21-8, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16792525

RESUMO

Protoporhyrin IX ferrochelatase catalyses the terminal step of the haem-biosynthetic pathway by inserting ferrous iron into protoporphyrin IX. NMPP (N-methylprotoporphyrin), a transition-state analogue and potent inhibitor of ferrochelatase, is commonly used to induce haem deficiency in mammalian cell cultures. To create ferrochelatase variants with different extents of tolerance towards NMPP and to understand further the mechanism of ferrochelatase inhibition by NMPP, we isolated variants with increased NMPP resistance, bearing mutations in an active-site loop (murine ferrochelatase residues 248-257), which was previously shown to mediate a protein conformational change triggered by porphyrin binding. The kinetic mechanisms of inhibition of two variants, in which Pro255 was replaced with either arginine (P255R) or glycine (P255G), were investigated and compared with that of wild-type ferrochelatase. While the binding affinity of the P255X variants for NMPP decreased by one order of magnitude in relation to that of wild-type enzyme, the inhibition constant increased by approximately two orders of magnitude (K(i)(app) values of 1 microM and 2.3 microM for P255R and P255G respectively, as against 3 nM for wild-type ferrochelatase). Nonetheless, the drastically reduced inhibition of the variants by NMPP was not paralleled with a decrease in specificity constant (kcat/K(m, protoporhyrin IX)) and/or catalytic activity (kcat). Further, although NMPP binding to either wild-type ferrochelatase or P255R occurred via a similar two-step kinetic mechanism, the forward and reverse rate constants associated with the second and rate-limiting step were comparable for the two enzymes. Collectively, these results suggest that Pro255 has a crucial role in maintaining an appropriate protein conformation and modulating the selectivity and/or regiospecificity of ferrochelatase.


Assuntos
Ferroquelatase/antagonistas & inibidores , Protoporfirinas/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Escherichia coli/metabolismo , Ferroquelatase/metabolismo , Cinética , Camundongos , Ligação Proteica , Conformação Proteica
19.
Mol Microbiol ; 60(3): 563-77, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16629661

RESUMO

Genetic analysis has indicated that the system II pathway for c-type cytochrome biogenesis in Bordetella pertussis requires at least four biogenesis proteins (CcsB, CcsA, DsbD and CcsX). In this study, the eight genes (ccmA-H) associated with the system I pathway in Escherichia coli were deleted. Using B. pertussis cytochrome c4 as a reporter for cytochromes c assembly, it is demonstrated that a single fused ccsBA polypeptide can replace the function of the eight system I genes in E. coli. Thus, the CcsB and CcsA membrane complex of system II is likely to possess the haem delivery and periplasmic cytochrome c-haem ligation functions. Using recombinant system II and system I, both under control of IPTG, we have begun to study the capabilities and characteristics of each system in the same organism (E. coli). The ferrochelatase inhibitor N-methylprotoporphyrin was used to modulate haem levels in vivo and it is shown that system I can use endogenous haem at much lower levels than system II. Additionally, while system I encodes a covalently bound haem chaperone (holo-CcmE), no covalent intermediate has been found in system II. It is shown that this allows system I to use holo-CcmE as a haem reservoir, a capability system II does not possess.


Assuntos
Citocromos c1/biossíntese , Citocromos c2/biossíntese , Escherichia coli/enzimologia , Heme/metabolismo , Proteínas Recombinantes/biossíntese , Sequência de Aminoácidos , Bordetella pertussis/enzimologia , Bordetella pertussis/genética , Grupo dos Citocromos c/química , Grupo dos Citocromos c/genética , Grupo dos Citocromos c/metabolismo , Citocromos c1/genética , Citocromos c2/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Ferroquelatase/antagonistas & inibidores , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Protoporfirinas/farmacologia , Proteínas Recombinantes/genética
20.
J Mol Biol ; 352(5): 1081-90, 2005 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-16140324

RESUMO

Insertion of metals into various tetrapyrroles is catalysed by a group of enzymes called chelatases, e.g. nickel, cobalt, magnesium and ferro-chelatase. It has been proposed that catalytic metallation includes distorting the porphyrin substrate by the enzyme towards a transition state-like geometry in which at least one of the pyrrole rings will be available for metal chelation. Here, we present a study of metal insertion into the transition-state inhibitor of protoporphyrin IX ferrochelatase, N-methyl mesoporphyrin (N-MeMP), by time-resolved crystallography and mass spectrometry with and without the presence of ferrochelatase. The results show that metallation of N-MeMP has a very limited effect on the conformation of the residues that participate in porphyrin and metal binding. These findings support theoretical data, which indicate that product release is controlled largely by the strain created by metal insertion into the distorted porphyrin. The results suggest that, similar to non-catalytic metallation of N-MeMP, the ferrochelatase-assisted metallation depends on the ligand exchange rate for the respective metal. Moreover, ferrochelatase catalyses insertion of Cu(II) and Zn(II) into N-MeMP with a rate that is about 20 times faster than non-enzymatic metallation in solution, suggesting that the catalytic strategy of ferrochelatase includes a stage of acceleration of the rate of ligand exchange for the metal substrate. The greater efficiency of N-MeMP metallation by Cu(II), as compared to Zn(II), contrasts with the K(m) values for Zn(II) (17 microM) and Cu(II) (170 microM) obtained for metallation of protoporphyrin IX. We suggest that this difference in metal specificity depends on the type of distortion imposed by the enzyme on protoporphyrin IX, which is different from the intrinsic non-planar distortion of N-MeMP. A mechanism of control of metal specificity by porphyrin distortion may be general for different chelatases, and may have common features with the mechanism of metal specificity in crown ethers.


Assuntos
Cobre/metabolismo , Ferroquelatase/química , Ferroquelatase/fisiologia , Mesoporfirinas/metabolismo , Bacillus subtilis/enzimologia , Catálise , Cobre/química , Cristalografia por Raios X , Escherichia coli , Ferroquelatase/antagonistas & inibidores , Ferroquelatase/genética , Espectrometria de Massas , Mesoporfirinas/química , Mutação , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...