Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(23): e2118979119, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35658075

RESUMO

Dynamic motions of enzymes occurring on a broad range of timescales play a pivotal role in all steps of the reaction pathway, including substrate binding, catalysis, and product release. However, it is unknown whether structural information related to conformational flexibility can be exploited for the directed evolution of enzymes with higher catalytic activity. Here, we show that mutagenesis of residues exclusively located at flexible regions distal to the active site of Homo sapiens kynureninase (HsKYNase) resulted in the isolation of a variant (BF-HsKYNase) in which the rate of the chemical step toward kynurenine was increased by 45-fold. Mechanistic pre­steady-state kinetic analysis of the wild type and the evolved enzyme shed light on the underlying effects of distal mutations (>10 Å from the active site) on the rate-limiting step of the catalytic cycle. Hydrogen-deuterium exchange coupled to mass spectrometry and molecular dynamics simulations revealed that the amino acid substitutions in BF-HsKYNase allosterically affect the flexibility of the pyridoxal-5'-phosphate (PLP) binding pocket, thereby impacting the rate of chemistry, presumably by altering the conformational ensemble and sampling states more favorable to the catalyzed reaction.


Assuntos
Catálise , Enzimas , Evolução Molecular , Substituição de Aminoácidos , Domínio Catalítico , Enzimas/genética , Enzimas/metabolismo , Humanos , Hidrolases/genética , Hidrolases/metabolismo , Imunoterapia , Cinética , Neoplasias/terapia
2.
Anticancer Drugs ; 23(1): 51-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21955999

RESUMO

Human recombinant arginase I cobalt [HuArgI (Co)] coupled with polyethylene glycol 5000 [HuArgI (Co)-PEG5000] has shown potent in-vitro depletion of arginine from tissue culture medium. We now show that HuArgI (Co)-PEG5000 is toxic to almost all cancer cell lines and to some normal primary cells examined. In contrast, HuArgI (Co)-PEG5000 in combination with supplemental L-citrulline is selectively cytotoxic to a fraction of human cancer cell lines in tissue culture, including some melanomas, mesotheliomas, acute myeloid leukemias, hepatocellular carcinomas, pancreas adenocarcinomas, prostate adenocarcinomas, lung adenocarcinomas, osteosarcomas, and small cell lung carcinomas. Unfortunately, a subset of normal human tissues is also sensitive to HuArgI (Co)-PEG5000 with L-citrulline supplementation, including umbilical endothelial cells, bronchial epithelium, neurons, and renal epithelial cells. We further show that cell sensitivity is predicted by the level of cellular argininosuccinate synthetase protein expression measured by immunoblots. By comparing a 3-day and 7-day exposure to HuArgI (Co)-PEG5000 with supplemental L-citrulline, some tumor cells sensitive on short-term assay are resistant in the 7-day assay consistent with the induction of argininosuccinate synthetase expression. On the basis of these results, we hypothesize that HuArgI (Co)-PEG5000 in combination with L-citrulline supplementation may be an attractive therapeutic agent for some argininosuccinate synthetase-deficient tumors. These in-vitro findings stimulate further development of this molecule and may aid in the identification of tissue toxicities and better selection of patients who will potentially respond to this combination therapy.


Assuntos
Antineoplásicos/farmacologia , Arginase/farmacologia , Argininossuccinato Sintase/metabolismo , Citrulina/farmacologia , Polietilenoglicóis/farmacologia , Arginina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Meios de Cultura , Ensaios de Seleção de Medicamentos Antitumorais , Células Epiteliais/efeitos dos fármacos , Humanos , Masculino , Ornitina Carbamoiltransferase/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Recombinantes/farmacologia
3.
Mol Cancer Ther ; 21(3): 419-426, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35086957

RESUMO

Renal cell carcinomas associated with hereditary leiomyomatosis and renal cell cancer (HLRCC) are notoriously aggressive and represent the leading cause of death among patients with HLRCC. To date, a safe and effective standardized therapy for this tumor type is lacking. Here we show that the engineered synthetic therapeutic enzyme, Cyst(e)inase, when combined with rapamycin, can effectively induce ferroptosis in HLRCC cells in vivo. The drug combination promotes lipid peroxidation to a greater degree than cysteine deprivation or Cyst(e)inase treatment alone, while rapamycin treatment alone does not induce ferroptosis. Mechanistically, Cyst(e)inase induces ferroptosis by depleting the exogenous cysteine/cystine supply, while rapamycin reduces cellular ferritin level by promoting ferritins' destruction via ferritinophagy. Since both Cyst(e)inase and rapamycin are well tolerated clinically, the combination represents an opportunity to exploit ferroptosis induction as a cancer management strategy. Accordingly, using a xenograft mouse model, we showed that the combination treatment resulted in tumor growth suppression without any notable side effects. In contrast, both Cyst(e)inase only and rapamycin only treatment groups failed to induce a significant change when compared with the vehicle control group. Our results demonstrated the effectiveness of Cyst(e)inase-rapamycin combination in inducing ferroptotic cell death in vivo, supporting the potential translation of the combination therapy into clinical HLRCC management.


Assuntos
Carcinoma de Células Renais , Cistos , Ferroptose , Neoplasias Renais , Animais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Cisteína/metabolismo , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Leiomiomatose , Masculino , Camundongos , Síndromes Neoplásicas Hereditárias , Sirolimo/farmacologia , Neoplasias Cutâneas , Neoplasias Uterinas
4.
Biochemistry ; 50(14): 3025-33, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21375249

RESUMO

The enzymatic deamidation of N-terminal L-Asn by N-terminal asparagine amidohydrolase (NTAN1) is a feature of the ubiquitin-dependent N-end rule pathway of protein degradation, which relates the in vivo half-life of a protein to the identity of its N-terminal residue. Herein, we report the bacterial expression, purification, and biochemical characterization of human NTAN1 (hNTAN1). We show here that hNTAN1 is highly selective for the hydrolysis of N-terminal peptidyl L-Asn but fails to deamidate free L-Asn or L-Gln, N-terminal peptidyl L-Gln, or acetylated N-terminal peptidyl L-Asn. Similar to other N-terminal deamidases, hNTAN1 is shown to possess a critical Cys residue that is absolutely required for catalysis, corroborated in part by abolishment of activity through the Cys75Ala point mutation. We also present evidence that the exposure of a conserved L-Pro at the N-terminus of hNTAN1 following removal of the initiating L-Met is important for the function of the enzyme. The results presented here should assist in the elucidation of molecular mechanisms underlying the neurological defects of NTAN1-deficient mice observed in other studies, and in the discovery of potential physiological substrates targeted by the enzyme in the modulation of protein turnover via the N-end rule pathway.


Assuntos
Amidoidrolases/metabolismo , Asparagina/metabolismo , Ácido Aspártico/metabolismo , Proteínas Recombinantes/metabolismo , Amidoidrolases/química , Amidoidrolases/genética , Animais , Asparagina/química , Ácido Aspártico/química , Biocatálise/efeitos dos fármacos , Dicroísmo Circular , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Concentração de Íons de Hidrogênio , Cinética , Metais/farmacologia , Camundongos , Modelos Químicos , Estrutura Molecular , Mutação , Proteínas Recombinantes/química , Espectrometria de Massas por Ionização por Electrospray , Especificidade por Substrato
5.
Biochemistry ; 49(49): 10582-8, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21053939

RESUMO

The active sites of eukaryotic arginase enzymes are strictly conserved, especially the first- and second-shell ligands that coordinate the two divalent metal cations that generate a hydroxide molecule for nucleophilic attack on the guanidinium carbon of l-arginine and the subsequent production of urea and l-ornithine. Here by using comprehensive pairwise saturation mutagenesis of the first- and second-shell metal ligands in human arginase I, we demonstrate that several metal binding ligands are actually quite tolerant to amino acid substitutions. Of >2800 double mutants of first- and second-shell residues analyzed, we found more than 80 unique amino acid substitutions, of which four were in first-shell residues. Remarkably, certain second-shell mutations could modulate the binding of both the nucleophilic water/hydroxide molecule and substrate or product ligands, resulting in activity greater than that of the wild-type enzyme. The data presented here constitute the first comprehensive saturation mutagenesis analysis of a metallohydrolase active site and reveal that the strict conservation of the second-shell metal binding residues in eukaryotic arginases does not reflect kinetic optimization of the enzyme during the course of evolution.


Assuntos
Arginase/metabolismo , Cobalto/química , Manganês/química , Sequência de Aminoácidos , Arginase/antagonistas & inibidores , Arginase/genética , Sítios de Ligação/genética , Cobalto/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Manganês/metabolismo , Dados de Sequência Molecular , Espectrofotometria Ultravioleta , Especificidade por Substrato/genética
6.
ACS Chem Biol ; 15(12): 3159-3166, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33275413

RESUMO

Kynureninases (KYNases) are enzymes that play a key role in tryptophan catabolism through the degradation of intermediate kynurenine and 3'-hydroxy-kynurenine metabolites (KYN and OH-KYN, respectively). Bacterial KYNases exhibit high catalytic efficiency toward KYN and moderate activity toward OH-KYN, whereas animal KYNases are highly selective for OH-KYN, exhibiting only minimal activity toward the smaller KYN substrate. These differences reflect divergent pathways for KYN and OH-KYN utilization in the respective kingdoms. We examined the Homo sapiens and Pseudomonas fluorescens KYNases (HsKYNase and PfKYNase respectively) using pre-steady-state and hydrogen-deuterium exchange mass spectrometry (HDX-MS) methodologies. We discovered that the activity of HsKYNase critically depends on formation of hydrogen bonds with the hydroxyl group of OH-KYN to stabilize the entire active site and allow productive substrate turnover. With the preferred OH-KYN substrate, stabilization is observed at the substrate-binding site and the region surrounding the PLP cofactor. With the nonpreferred KYN substrate, less stabilization occurs, revealing a direct correlation with activity. This correlation holds true for PfKYNases; however there is only a modest stabilization at the substrate-binding site, suggesting that substrate discrimination is simply achieved by steric hindrance. We speculate that eukaryotic KYNases use dynamic mobility as a mechanism of substrate specificity to commit OH-KYN to nicotinamide synthesis and avoid futile hydrolysis of KYN. These findings have important ramifications for the engineering of HsKynase with high KYN activity as required for clinical applications in cancer immunotherapy. Our study shows how homologous enzymes with conserved active sites can use dynamics to discriminate between two highly similar substrates.


Assuntos
Hidrolases/metabolismo , Catálise , Humanos , Hidrolases/química , Cinética , Conformação Proteica , Especificidade por Substrato
7.
Biochemistry ; 48(46): 11026-31, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19839645

RESUMO

Herein we report the bacterial expression, purification, and enzymatic characterization of the human asparaginase-like protein 1 (hASRGL1). We present evidence that hASRGL1 exhibits beta-aspartyl peptidase activity consistent with enzymes designated as plant-type asparaginases, which had thus far been found in only plants and bacteria. Similar to nonmammalian plant-type asparaginases, hASRGL1 is shown to be an Ntn hydrolase for which Thr168 serves as the essential N-terminal nucleophile for intramolecular processing and catalysis, corroborated in part by abolishment of both activities through the Thr168Ala point mutation. In light of the activity profile reported here, ASRGL1s may act synergistically with protein l-isoaspartyl methyl transferase to relieve accumulation of potentially toxic isoaspartyl peptides in mammalian brain and other tissues.


Assuntos
Amidoidrolases/metabolismo , Asparaginase/metabolismo , Autoantígenos/metabolismo , Dipeptidases/química , Amidoidrolases/química , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Asparaginase/química , Asparaginase/genética , Autoantígenos/química , Autoantígenos/genética , Biocatálise , Domínio Catalítico , Dipeptídeos/química , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos
8.
Chem Biol ; 15(5): 467-75, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18482699

RESUMO

Many enzymes in the pentein superfamily use a transient covalent intermediate in their catalytic mechanisms. Here we trap and determine the structure of a stable covalent adduct that mimics this intermediate using a mutant dimethylarginine dimethylaminohydrolase and an alternative substrate. The interactions observed between the enzyme and trapped adduct suggest an altered angle of attack between the nucleophiles of the first and second half-reactions of normal catalysis. The stable covalent adduct is also capable of further reaction. Addition of imidazole rescues the original hydrolytic activity. Notably, addition of other amines instead yields substituted arginine products, which arise from partitioning of the intermediate into the evolutionarily related amidinotransferase reaction pathway. The enzyme provides both selectivity and catalysis for the amidinotransferase reaction, underscoring commonalities among the reaction pathways in this mechanistically diverse enzyme superfamily. The promiscuous partitioning of this intermediate may also help to illuminate the evolutionary history of these enzymes.


Assuntos
Enzimas/metabolismo , Catálise , Cromatografia Líquida de Alta Pressão , Cristalografia por Raios X , Enzimas/química , Hidrólise , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
9.
Cancer Discov ; 9(12): 1673-1685, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31554642

RESUMO

A challenge in oncology is to rationally and effectively integrate immunotherapy with traditional modalities, including radiotherapy. Here, we demonstrate that radiotherapy induces tumor-cell ferroptosis. Ferroptosis agonists augment and ferroptosis antagonists limit radiotherapy efficacy in tumor models. Immunotherapy sensitizes tumors to radiotherapy by promoting tumor-cell ferroptosis. Mechanistically, IFNγ derived from immunotherapy-activated CD8+ T cells and radiotherapy-activated ATM independently, yet synergistically, suppresses SLC7A11, a unit of the glutamate-cystine antiporter xc-, resulting in reduced cystine uptake, enhanced tumor lipid oxidation and ferroptosis, and improved tumor control. Thus, ferroptosis is an unappreciated mechanism and focus for the development of effective combinatorial cancer therapy. SIGNIFICANCE: This article describes ferroptosis as a previously unappreciated mechanism of action for radiotherapy. Further, it shows that ferroptosis is a novel point of synergy between immunotherapy and radiotherapy. Finally, it nominates SLC7A11, a critical regulator of ferroptosis, as a mechanistic determinant of synergy between radiotherapy and immunotherapy.This article is highlighted in the In This Issue feature, p. 1631.


Assuntos
Sistema y+ de Transporte de Aminoácidos/genética , Antineoplásicos Imunológicos/administração & dosagem , Melanoma Experimental/terapia , Sulfassalazina/administração & dosagem , Animais , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Regulação para Baixo , Ferroptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoterapia/métodos , Interferon gama , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos da radiação , Melanoma Experimental/genética , Camundongos , Oxirredução , Sulfassalazina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Melanoma Res ; 24(6): 556-67, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25304236

RESUMO

Metastatic melanoma is a deadly form of cancer with few therapeutic options and the cause of more than 9480 deaths annually in the USA alone. Novel treatment options for this disease are urgently needed. Here we test the efficacy of a novel melanoma drug, the human recombinant Co-arginase (CoArgIPEG), against an aggressive A375 melanoma mouse model. CoArgIPEG is a modification of the naturally occurring human enzyme with improved stability, catalytic activity, and potentially lower immunogenicity compared with current amino acid-depleting drugs. Marked tumor growth reductions (mean P=0.0057) with apoptosis induction and proliferation inhibition are noted with CoArgIPEG treatment, both in the presence and in the absence of supplemental citrulline. Further, improved therapeutic efficacy has been noted against A375 xenografts relative to the naturally occurring human recombinant arginase enzyme at lower doses of CoArgIPEG. Unfortunately, after 1 month, half of the relapsing tumors showed argininosuccinate synthase induction, which correlated with Ser62-phosphorylated cMyc. Although argininosuccinate synthase induction could not be induced in vitro, a drug targeting pathway previously demonstrated to be associated with Ser62 cMyc phosphorylation - U0126 - in combination with CoArgIPEG demonstrated an in-vitro synergistic response (combination indices 0.13±0.10 and 0.14±0.10 with or without citrulline, respectively). Overall, favorable efficacy and potential synergy with other antimelanoma drugs support CoArgIPEG as a potent, novel cancer therapeutic.


Assuntos
Antineoplásicos/uso terapêutico , Arginase/uso terapêutico , Melanoma/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Cobalto/química , Cobalto/uso terapêutico , Feminino , Humanos , Hidrolases/química , Hidrolases/uso terapêutico , Células Jurkat , Melanoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Polietilenoglicóis/química , Polietilenoglicóis/uso terapêutico , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
ACS Chem Biol ; 8(10): 2264-71, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23972067

RESUMO

Taurine, the most abundant free amino acid in mammals, with many critical roles such as neuronal development, had so far only been reported to be synthetized in eukaryotes. Taurine is the major product of cysteine metabolism in mammals, and its biosynthetic pathway consists of cysteine dioxygenase and cysteine sulfinic acid decarboxylase (hCSAD). Sequence, structural, and mutational analyses of the structurally and sequentially related hCSAD and human glutamic acid decarboxylase (hGAD) enzymes revealed a three residue substrate recognition motif (X1aa19X2aaX3), within the active site that is responsible for coordinating their respective preferred amino acid substrates. Introduction of the cysteine sulfinic acid (CSA) motif into hGAD (hGAD-S192F/N212S/F214Y) resulted in an enzyme with a >700 fold switch in selectivity toward the decarboxylation of CSA over its preferred substrate, l-glutamic acid. Surprisingly, we found this CSA recognition motif in the genome sequences of several marine bacteria, prompting us to evaluate the catalytic properties of bacterial amino acid decarboxylases that were predicted by sequence motif to decarboxylate CSA but had been annotated as GAD enzymes. We show that CSAD from Synechococcus sp. PCC 7335 specifically decarboxylated CSA and that the bacteria accumulated intracellular taurine. The fact that CSAD homologues exist in certain bacteria and are frequently found in operons containing the recently discovered bacterial cysteine dioxygenases that oxidize l-cysteine to CSA supports the idea that a bona fide bacterial taurine biosynthetic pathway exists in prokaryotes.


Assuntos
Bactérias/enzimologia , Carboxiliases/química , Carboxiliases/metabolismo , Descoberta de Drogas , Taurina/biossíntese , Motivos de Aminoácidos , Sequência de Aminoácidos , Carboxiliases/genética , Células Cultivadas , Estabilidade Enzimática , Humanos , Modelos Moleculares , Estrutura Molecular , Transdução de Sinais , Especificidade por Substrato , Taurina/química , Taurina/genética
12.
Methods Enzymol ; 502: 291-319, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22208990

RESUMO

Cancer has become the leading cause of death in the developed world and has remained one of the most difficult diseases to treat. One of the difficulties in treating cancer is that conventional chemotherapies often have unacceptable toxicities toward normal cells at the doses required to kill tumor cells. Thus, the demand for new and improved tumor specific therapeutics for the treatment of cancer remains high. Alterations to cellular metabolism constitute a nearly universal feature of many types of cancer cells. In particular, many tumors exhibit deficiencies in one or more amino acid synthesis or salvage pathways forcing a reliance on the extracellular pool of these amino acids to satisfy protein biosynthesis demands. Therefore, one treatment modality that satisfies the objective of developing cancer cell-selective therapeutics is the systemic depletion of that tumor-essential amino acid, which can result in tumor apoptosis with minimal side effects to normal cells. While this strategy was initially suggested over 50 years ago, it has been recently experiencing a renaissance owing to advances in protein engineering technology, and more sophisticated approaches to studying the metabolic differences between tumorigenic and normal cells. Dietary restriction is typically not sufficient to achieve a therapeutically relevant level of amino acid depletion for cancer treatment. Therefore, intravenous administration of enzymes is used to mediate the degradation of such amino acids for therapeutic purposes. Unfortunately, the human genome does not encode enzymes with the requisite catalytic or pharmacological properties necessary for therapeutic purposes. The use of heterologous enzymes has been explored extensively both in animal studies and in clinical trials. However, heterologous enzymes are immunogenic and elicit adverse responses ranging from anaphylactic shock to antibody-mediated enzyme inactivation, and therefore have had limited utility. The one notable exception is Escherichia colil-asparaginase II (EcAII), which has been FDA-approved for the treatment of childhood acute lymphoblastic leukemia. The use of engineered human enzymes, to which natural tolerance is likely to prevent recognition by the adaptive immune system, offers a novel approach for capitalizing on the promising strategy of systemic depletion of tumor-essential amino acids. In this work, we review several strategies that we have developed to: (i) reduce the immunogenicity of a nonhuman enzyme, (ii) engineer human enzymes for novel catalytic specificities, and (iii) improve the pharmacological characteristics of a human enzyme that exhibits the requisite substrate specificity for amino acid degradation but exhibits low activity and stability under physiological conditions.


Assuntos
Aminoácidos/deficiência , Asparaginase/administração & dosagem , Terapia Enzimática/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Animais , Asparaginase/genética , Asparaginase/imunologia , Asparaginase/uso terapêutico , Sítios de Ligação , Clonagem Molecular , Simulação por Computador , Estabilidade Enzimática , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Cinética , Camundongos , Mutagênese Sítio-Dirigida , Polietilenoglicóis/química , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
13.
Expert Opin Biol Ther ; 12(1): 53-61, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22171665

RESUMO

INTRODUCTION: Amino acid auxotrophy or the metabolic defect which renders cancer incapable of surviving under amino acid depleted conditions is being exploited and explored as a therapeutic against cancer. Early clinical data on asparagine- and arginine-depleting drugs have demonstrated low toxicity and efficacy in melanoma, hepatocellular carcinoma and acute lymphoblastic leukemia. Methionine auxotrophy is a novel niche currently under exploration for targeting certain cancers. AREAS COVERED: In this review we explore the discovery of methionine auxotrophy followed by in vitro, in vivo and patient data on targeting cancer with methionine depletion. We end with a small discussion on bioengineering, pegylation and red blood cell encapsulation as mechanisms for decreasing immunogenicity of methionine-depleting drugs. We hope to provide a platform for future pharmacology, toxicology and cytotoxicity studies with methionine depletion therapy and drugs. EXPERT OPINION: Although methionine auxotrophy seems as a viable target, extensive research addressing normal versus cancer cell toxicity needs to be conducted. Further research also needs to be conducted into the molecular mechanism associated with methionine depletion therapy. Finally, novel methods need to be developed to decrease the immunogenicity of methionine-depleting drugs, a current issue with protein therapeutics.


Assuntos
Metionina/metabolismo , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Bioengenharia/métodos , Linhagem Celular Tumoral , Eritrócitos/citologia , Humanos , Sistema Imunitário , Oncologia/métodos , Metionina/uso terapêutico , Camundongos , Modelos Biológicos , Transplante de Neoplasias , Neoplasias/dietoterapia , Polietilenoglicóis/química , Pseudomonas putida/metabolismo , Ratos
14.
ACS Chem Biol ; 7(11): 1840-7, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22891768

RESUMO

The human asparaginase-like protein 1 (hASRGL1) catalyzes the hydrolysis of l-asparagine and isoaspartyl-dipeptides. As an N-terminal nucleophile (Ntn) hydrolase superfamily member, the active form of hASRGL1 is generated by an intramolecular cleavage step with Thr168 as the catalytic residue. However, in vitro, autoprocessing is incomplete (~50%), fettering the biophysical characterization of hASRGL1. We circumvented this obstacle by constructing a circularly permuted hASRGL1 that uncoupled the autoprocessing reaction, allowing us to kinetically and structurally characterize this enzyme and the precursor-like hASRGL1-Thr168Ala variant. Crystallographic and biochemical evidence suggest an activation mechanism where a torsional restraint on the Thr168 side chain helps drive the intramolecular processing reaction. Cleavage and formation of the active site releases the torsional restriction on Thr168, which is facilitated by a small conserved Gly-rich loop near the active site that allows the conformational changes necessary for activation.


Assuntos
Amidoidrolases/química , Amidoidrolases/metabolismo , Asparaginase/química , Asparaginase/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Amidoidrolases/genética , Asparaginase/genética , Asparagina/metabolismo , Autoantígenos/genética , Domínio Catalítico , Cristalografia por Raios X , Ativação Enzimática , Humanos , Hidrólise , Modelos Moleculares , Mutação Puntual , Conformação Proteica
15.
Transl Oncol ; 5(1): 26-31, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22348173

RESUMO

Human recombinant arginase I cobalt coupled to polyethylene glycol 5000 (HuArg I [Co]-PEG5000) achieved potent in vitro depletion of arginine from tissue culture medium and cytotoxicity to many cancer cell lines. The recombinant enzyme also produced tumor growth inhibition of hepatocellular carcinoma and pancreatic carcinoma xenografts. Although these results were promising, the therapeutic index was narrow. Toxicities were seen in normal cells in tissue culture. In vivo normal tissue injury occurred at doses twice the effective dose. The current study was conducted to define, in greater detail, the maximum tolerated dose (MTD), pharmacodynamics, and dose-limiting toxicities (DLTs) of twice-weekly intraperitoneal HuArg I [Co]-PEG5000 in Balb/c mice. Animal weight and survival were monitored, serum arginine levels measured, and complete blood cell counts, chemistries, necropsies, and histologies were performed. In addition, methods to ameliorate the HuArg I [Co]-PEG5000 adverse effects were tested. Supplemental l-citrulline was given concurrently with the arginase drug. The HuArg I [Co]-PEG5000 MTD in mice was 5 mg/kg twice weekly, and DLTs included weight loss and marrow necrosis. No other organ damage or changes in blood cell counts or chemistries were observed. Arginase reduced serum arginine levels from 60 µM to 4 to 6 µM. Supplemental l-citrulline given per os or daily subcutaneously reduced and delayed toxicities, and l-citrulline given twice daily subcutaneously completely prevented animal toxicities. On the basis of these results, we hypothesize that HuArg I [Co]-PEG5000, particularly with supplemental l-citrulline, may be an attractive therapeutic agent for argininosuccinate synthetase-deficient tumors.

16.
Transl Oncol ; 4(3): 138-46, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21633669

RESUMO

Hepatocellular carcinoma (HCC) and pancreatic carcinoma (PC) cells often have inherent urea cycle defects rendering them auxotrophic for the amino acid l-arginine (l-arg). Most HCC and PC require extracellular sources of l-arg and undergo cell cycle arrest and apoptosis when l-arg is restricted. Systemic, enzyme-mediated depletion of l-arg has been investigated in mouse models and human trials. Non-human enzymes elicit neutralizing antibodies, whereas human arginases display poor pharmacological properties in serum. Co(2+) substitution of the Mn(2+) metal cofactor in human arginase I (Co-hArgI) was shown to confer more than 10-fold higher catalytic activity (k(cat)/K(m)) and 5-fold greater stability. We hypothesized that the Co-hArgI enzyme would decrease tumor burden by systemic elimination of l-arg in a murine model. Co-hArgI was conjugated to 5-kDa PEG (Co-hArgI-PEG) to enhance circulation persistence. It was used as monotherapy for HCC and PC in vitro and in vivo murine xenografts. The mechanism of cell death was also investigated. Weekly treatment of 8 mg/kg Co-hArgI-PEG effectively controlled human HepG2 (HCC) and Panc-1 (PC) tumor xenografts (P = .001 and P = .03, respectively). Both cell lines underwent apoptosis in vitro with significant increased expression of activated caspase-3 (P < .001). Furthermore, there was evidence of autophagy in vitro and in vivo. We have demonstrated that Co-hArgI-PEG is effective at controlling two types of l-arg-dependent carcinomas. Being a nonessential amino acid, arginine deprivation therapy through Co-hArgI-PEG holds promise as a new therapy in the treatment of HCC and PC.

17.
ACS Chem Biol ; 5(3): 333-42, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20050660

RESUMO

Replacing the two Mn(2+) ions normally present in human Arginase I with Co(2+) resulted in a significantly lowered K(M) value without a concomitant reduction in k(cat). In addition, the pH dependence of the reaction was shifted from a pK(a) of 8.5 to a pK(a) of 7.5. The combination of these effects led to a 10-fold increase in overall catalytic activity (k(cat)/K(M)) at pH 7.4, close to the pH of human serum. Just as important for therapeutic applications, Co(2+) substitution lead to significantly increased serum stability of the enzyme. Our data can be explained by direct coordination of l-Arg to one of the Co(2+) ions during reaction, consistent with previously reported model studies. In vitro cytotoxicity experiments verified that the Co(2+)-substituted human Arg I displays an approximately 12- to 15-fold lower IC(50) value for the killing of human hepatocellular carcinoma and melanoma cell lines and thus constitutes a promising new candidate for the treatment of l-Arg auxotrophic tumors.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Arginase/metabolismo , Arginase/uso terapêutico , Cobalto/metabolismo , Manganês/metabolismo , Neoplasias/tratamento farmacológico , Antineoplásicos/química , Arginase/química , Arginase/genética , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estabilidade Enzimática , Escherichia coli/genética , Expressão Gênica , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Melanoma/tratamento farmacológico , Espectroscopia por Absorção de Raios X
18.
Biochemistry ; 45(17): 5618-30, 2006 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-16634643

RESUMO

The enzyme dimethylargininase (also known as dimethylarginine dimethylaminohydrolase or DDAH; EC 3.5.3.18) catalyzes the hydrolysis of endogenous nitric oxide synthase inhibitors, N(omega)-methyl-l-arginine and N(omega),N(omega)-dimethyl-l-arginine. Understanding the mechanism and regulation of DDAH activity is important for developing ways to control nitric oxide production during angiogenesis and in many cases of vascular endothelial pathobiology. Several possible physiological regulation mechanisms of DDAH depend upon the presence of an active-site cysteine residue, Cys249 in Pseudomonas aeruginosa (Pa) DDAH, which is proposed to serve as a nucleophile in the catalytic mechanism. Through the use of pH-dependent ultraviolet and visible (UV-vis) difference spectroscopy and inactivation kinetics, the pK(a) of the active-site Cys249 in the resting enzyme was found to be unperturbed from pK(a) values of typical noncatalytic cysteine residues. In contrast, the pH dependence of k(cat) values indicates a much lower apparent pK(a) value. UV-vis difference spectroscopy between wild-type and C249S DDAH shows absorbance changes consistent with Cys249 deprotonation to the anionic thiolate upon binding positively charged ligands. The proton from Cys249 is lost either to the solvent or to an unidentified general base. A mutation of the active-site histidine residue, H162G, does not eliminate cysteine nucleophilicity, further arguing against a pre-formed ion pair with Cys249. Finally, UV-vis and X-ray absorption spectroscopy revealed that inhibitory metal ions can bind at these two active-site residues, Cys249 and His162, and also stabilize the anionic form of Cys249. These results support a proposed substrate-assisted mechanism for Pa DDAH in which ligand binding modulates the reactivity of the active-site cysteine.


Assuntos
Amidoidrolases/metabolismo , Cisteína/metabolismo , Pseudomonas aeruginosa/enzimologia , Amidoidrolases/antagonistas & inibidores , Sítios de Ligação , Citrulina/análogos & derivados , Citrulina/metabolismo , Cobalto/metabolismo , Concentração de Íons de Hidrogênio , Iodoacetamida/farmacologia , Cinética , Lisina/farmacologia , Espectrometria de Massas , Modelos Químicos , Prótons , Espectrofotometria Ultravioleta , Análise Espectral , Tioureia/análogos & derivados , Tioureia/metabolismo , Raios X , Zinco/farmacologia , ômega-N-Metilarginina/metabolismo
19.
Biochemistry ; 44(42): 13744-52, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16229464

RESUMO

The enzymes dimethylargininase [dimethylarginine dimethylaminohydrolase (DDAH); EC 3.5.3.18] and peptidylarginine deiminase (PAD; EC 3.5.3.15) catalyze hydrolysis of substituted arginines. Due to their role in normal physiology and pathophysiology, both enzymes have been identified as potential drug targets, but few useful inhibitors have been reported. Here, we find that 2-chloroacetamidine irreversibly inhibits both DDAH from Pseudomonas aeruginosa and human PAD4 in a time- and concentration-dependent manner, despite the nonoverlapping substrate specificities and low levels of amino acid identity of their catalytic domains. Substrate protection experiments indicate that inactivation occurs by modification at the active site, albeit with modest affinity. Mass spectral analysis demonstrates that irreversible inactivation of DDAH occurs through selective formation of a covalent thioether bond with the active-site Cys249 residue. The mechanism of inactivation by 2-chloroacetamidine is analogous to that of chloromethyl ketones, a set of inhibitors that have found wide application because of their specific covalent modification of active-site residues in serine and cysteine proteases. Likewise, 2-chloroacetamidine may potentially find wide applicability as a general pharmacophore useful in delineating characteristics of the amidinotransferase superfamily.


Assuntos
Amidinas/farmacologia , Amidoidrolases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Hidrolases/antagonistas & inibidores , Amidoidrolases/genética , Amidoidrolases/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Domínio Catalítico , Clonagem Molecular , Primers do DNA , Hidrolases/genética , Hidrolases/metabolismo , Espectrometria de Massas , Desiminases de Arginina em Proteínas
20.
Biochemistry ; 44(20): 7559-69, 2005 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15895999

RESUMO

Lactonases from Bacillus species hydrolyze the N-acylhomoserine lactone (AHL) signaling molecules used in quorum-sensing pathways of many Gram-negative bacteria, including Pseudomonas aeruginosa and Erwinia carotovora, both significant pathogens. Because of sequence similarity, these AHL lactonases have been assigned to the metallo-beta-lactamase superfamily of proteins, which includes metalloenzymes of diverse activity, mechanism, and metal content. However, a recent study claims that AHL lactonase from Bacillus sp. 240B1 is not a metalloprotein [Wang, L. H., et al. (2004) J. Biol. Chem. 279, 13645]. Here, the gene for an AHL lactonase from Bacillus thuringiensis is cloned, and the protein is expressed, purified, and found to bind 2 equiv of zinc. The metal-bound form of AHL lactonase catalyzes the hydrolysis of N-hexanoyl-(S)-homoserine lactone but not the (R) enantiomer. Removal of both zinc ions results in loss of activity, and reconstitution with zinc restores activity, indicating the importance of metal ions for catalytic activity. Metal content, sequence alignments, and X-ray absorption spectroscopy of the zinc-containing lactonase all support a proposed dinuclear zinc binding site similar to that found in glyoxalase II.


Assuntos
Bacillus thuringiensis/enzimologia , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/fisiologia , Metaloproteínas/química , Metaloproteínas/fisiologia , Transdução de Sinais , Sequência de Aminoácidos , Apoenzimas/química , Apoenzimas/metabolismo , Bacillus thuringiensis/genética , Bacillus thuringiensis/patogenicidade , Sítios de Ligação/genética , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/isolamento & purificação , Proteínas de Transporte/metabolismo , Clonagem Molecular , Diálise , Ativação Enzimática , Cinética , Proteínas Ligantes de Maltose , Metaloproteínas/genética , Metaloproteínas/isolamento & purificação , Dados de Sequência Molecular , Análise Espectral , Raios X , Zinco/química , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA