Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
PLoS Pathog ; 19(7): e1011018, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37428793

RESUMO

Human schistosomiasis is a neglected tropical disease caused by Schistosoma mansoni, S. haematobium, and S. japonicum. Praziquantel (PZQ) is the method of choice for treatment. Due to constant selection pressure, there is an urgent need for new therapies for schistosomiasis. Previous treatment of S. mansoni included the use of oxamniquine (OXA), a drug that is activated by a schistosome sulfotransferase (SULT). Guided by data from X-ray crystallography and Schistosoma killing assays more than 350 OXA derivatives were designed, synthesized, and tested. We were able to identify CIDD-0150610 and CIDD-0150303 as potent derivatives in vitro that kill (100%) of all three Schistosoma species at a final concentration of 71.5 µM. We evaluated the efficacy of the best OXA derivates in an in vivo model after treatment with a single dose of 100 mg/kg by oral gavage. The highest rate of worm burden reduction was achieved by CIDD -150303 (81.8%) against S. mansoni, CIDD-0149830 (80.2%) against S. haematobium and CIDD-066790 (86.7%) against S. japonicum. We have also evaluated the ability of the derivatives to kill immature stages since PZQ does not kill immature schistosomes. CIDD-0150303 demonstrated (100%) killing for all life stages at a final concentration of 143 µM in vitro and effective reduction in worm burden in vivo against S. mansoni. To understand how OXA derivatives fit in the SULT binding pocket, X-ray crystal structures of CIDD-0150303 and CIDD-0150610 demonstrate that the SULT active site will accommodate further modifications to our most active compounds as we fine tune them to increase favorable pharmacokinetic properties. Treatment with a single dose of 100 mg/kg by oral gavage with co-dose of PZQ + CIDD-0150303 reduced the worm burden of PZQ resistant parasites in an animal model by 90.8%. Therefore, we conclude that CIDD-0150303, CIDD-0149830 and CIDD-066790 are novel drugs that overcome some of PZQ limitations, and CIDD-0150303 can be used with PZQ in combination therapy.


Assuntos
Anti-Helmínticos , Esquistossomose mansoni , Esquistossomose , Animais , Humanos , Praziquantel/farmacologia , Praziquantel/química , Oxamniquine/farmacologia , Esquistossomose/tratamento farmacológico , Esquistossomose/parasitologia , Schistosoma mansoni , Terapia Combinada , Doenças Negligenciadas/tratamento farmacológico , Esquistossomose mansoni/tratamento farmacológico , Esquistossomose mansoni/parasitologia
2.
J Org Chem ; 89(7): 5134-5141, 2024 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-38489762

RESUMO

CIDD-0072424 is a novel small molecule developed in silico with remarkable activity for the inhibition of protein kinase C (PKC)-epsilon to treat alcohol use disorder. We developed a concise synthesis of (S)-2 that is highly enantioselective, scalable, and amenable for 3-point structure-activity relationship (SAR) studies for compound optimization. The highly enantioselective nitro-Mannich reaction was achieved through a dual-reagent catalysis system. The overall utility and the efficiency of the enantioselective route provided a scalable synthesis of both PKCε inhibitors 1 and 2.


Assuntos
Proteína Quinase C-épsilon , Estereoisomerismo , Catálise
3.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33593898

RESUMO

Tethered photoswitches are molecules with two photo-dependent isomeric forms, each with different actions on their biological targets. They include reactive chemical groups capable of covalently binding to their target. Our aim was to develop a ß-subunit-tethered propofol photoswitch (MAP20), as a tool to better study the mechanism of anesthesia through the GABAA α1ß3γ2 receptor. We used short spacers between the tether (methanethiosulfonate), the photosensitive moiety (azobenzene), and the ligand (propofol), to allow a precise tethering adjacent to the putative propofol binding site at the ß+α- interface of the receptor transmembrane helices (TMs). First, we used molecular modeling to identify possible tethering sites in ß3TM3 and α1TM1, and then introduced cysteines in the candidate positions. Two mutant subunits [ß3(M283C) and α1(V227C)] showed photomodulation of GABA responses after incubation with MAP20 and illumination with lights at specific wavelengths. The α1ß3(M283C)γ2 receptor showed the greatest photomodulation, which decreased as GABA concentration increased. The location of the mutations that produced photomodulation confirmed that the propofol binding site is located in the ß+α- interface close to the extracellular side of the transmembrane helices. Tethering the photoswitch to cysteines introduced in the positions homologous to ß3M283 in two other subunits (α1W288 and γ2L298) also produced photomodulation, which was not entirely reversible, probably reflecting the different nature of each interface. The results are in agreement with a binding site in the ß+α- interface for the anesthetic propofol.


Assuntos
Anestésicos Intravenosos/farmacologia , Membrana Celular/metabolismo , Luz , Oócitos/metabolismo , Propofol/farmacologia , Receptores de GABA-A/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/efeitos da radiação , Humanos , Oócitos/efeitos dos fármacos , Oócitos/efeitos da radiação , Conformação Proteica , Domínios Proteicos , Receptores de GABA-A/química , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/efeitos da radiação , Xenopus laevis , Ácido gama-Aminobutírico
4.
J Biol Chem ; 294(41): 15172-15175, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511327

RESUMO

Proteolysis targeting chimeras (PROTACs) are bivalent molecules that bring a cellular protein to a ubiquitin ligase E3 for ubiquitination and subsequent degradation. Although PROTAC has emerged as a promising therapeutic means for cancers as it rewires the ubiquitin pathway to destroy key cancer regulators, the degradation signals/pathways for PROTACs remain underdeveloped. Here we append single amino acids, the simplest degradation signal, to a ligand specific for estrogen-related receptor α (ERRα) and demonstrate their utility in ERRα knockdown via the N-end rule pathway and also their efficiency in the growth inhibition of breast cancer cells. The modular design described offers unique advantages including smaller molecular size with shortest degradation sequences and degradation speed modulation with different amino acids. Our study expands the repertoire of limited ubiquitin pathways currently available for PROTACs and could be easily adapted for broad use in targeted protein degradation.


Assuntos
Proteólise/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Receptores de Estrogênio/química , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor ERRalfa Relacionado ao Estrogênio
5.
Anal Chem ; 92(5): 3860-3866, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32040916

RESUMO

The four glucosyl esters were synthesized and tested for the determination of infection enzyme leukocyte esterase (LE) in human synovial (joint) fluid and urine. The esters acted as LE substrates releasing glucose in a direct proportion to the activity of LE in a sample. The freed glucose was then detected by a coupled-enzyme assay at either a nitrogen-doped carbon nanotube (N-CNT) electrode or a commercial glucose test strip. The assays at the N-CNT electrode detected LE down to 0.81 nM (25 µg L-1) and showed the fastest kinetics (2.1 × 105 M-1 s-1) for esters with the least crowded space around their carbonyl group. When used with glucose strips, the esters discerned clinically relevant levels of LE up to at least 26 nM (800 µg L-1) in the microliter-sized samples of bodily fluids. The reading of glucose strips with a potentiostat, instead of a personal glucose meter (blood glucometer), shortened the time of required sample incubation from 3 h to 5 min. Correcting the signal of incubated sample for that of original sample eliminated matrix effects and accounted for the presence of native glucose. The new esters have a potential to extend the use of glucose strips (already used by millions for diabetes monitoring) to the quantification of the severity of urinary tract and periprosthetic joint infections.


Assuntos
Hidrolases de Éster Carboxílico/análise , Técnicas Eletroquímicas/métodos , Líquido Sinovial/enzimologia , Hidrolases de Éster Carboxílico/metabolismo , Hidrolases de Éster Carboxílico/urina , Técnicas Eletroquímicas/instrumentação , Eletrodos , Glucose/química , Glucose/metabolismo , Humanos , Cinética , Limite de Detecção , Nanotubos de Carbono/química , Nitrogênio/química
6.
Bioorg Med Chem ; 27(1): 208-215, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30528162

RESUMO

We previously demonstrated that capsazepine (CPZ), a synthetic transient receptor potential Vanilloid subtype 1 (TRPV1) antagonist, has significant anti-cancer effects in vivo. The purpose of this study was to develop more potent analogs based upon CPZ pharmacophore and structure-activity relationships (SAR) across analogs. We generated 30 novel compounds and screened for their anti-proliferative effects in cultured HeLa cervical cancer cells. Cell viability assays identified multiple compounds with IC50s < 15 µM and one compound, 29 with an IC50 < 5 µM; six fold more potent than CPZ. We validated the anti-proliferative efficacy of two lead compounds, 17 and 29, in vivo using HeLa-derived xenografts in athymic nude mice. Both analogs significantly reduced tumor volumes by day 8 compared to control treated animals (p < 0.001) with no observable adverse effects. Calcium imaging determined that compound 17 activates TRPV1 whereas 29 neither activates nor inhibits TRPV1; indicating a unique mechanism-of-action that does not involve TRPV1 signaling. Cell viability assays using a panel of additional tumor types including oral squamous cell carcinoma, non-small cell lung cancer (NSCLC), breast cancer, and prostate cancer cell lines (HSC-3, H460, MDA-231, and PC-3 respectively) demonstrated that both lead compounds were efficacious against every cancer type tested. Compounds 29 displayed IC50s of 1-2.5 µM in HSC-3and PC-3cells. Thus, we propose that these novel CPZ analogs may serve as efficacious therapeutic agents against multiple tumor types that warrant further development for clinical application.


Assuntos
Antineoplásicos/uso terapêutico , Capsaicina/análogos & derivados , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Capsaicina/síntese química , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos Nus , Estrutura Molecular , Relação Estrutura-Atividade , Canais de Cátion TRPV/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Oral Pathol Med ; 48(5): 389-399, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30825343

RESUMO

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a deadly disease with a mere 40% five-year survival rate for patients with advanced disease. Previously, we discovered that capsazepine (CPZ), a transient receptor potential channel, Vanilloid subtype 1 (TRPV1) antagonist, has significant anti-tumor effects against OSCC via a unique mechanism-of-action that is independent of TRPV1. Thus, we developed novel CPZ analogs with more potent anti-proliferative effects (CIDD-24, CIDD-99, and CIDD-111). METHODS: Using OSCC xenograft models, we determined the efficacy of these analogs in vivo. TRPV1 interactions were evaluated using calcium imaging and a rat model of orofacial pain. Anti-cancer mechanism(s)-of-action were assessed by cell cycle analysis and mitochondrial depolarization assays. RESULTS: CIDD-99 was the most potent analog demonstrating significant anti-tumor effects in vivo (P < 0.001). CIDD-24 was equipotent to the parent compound CPZ, but less potent than CIDD-99. CIDD-111 was the least efficacious analog. Calcium imaging studies confirmed that CIDD-99 neither activates nor inhibits TRPV1 confirming that TRPV1 activity is not involved in its anti-cancer effects. All analogs induced an S-phase block, dose-dependent mitochondrial depolarization, and apoptosis. Histological analyses revealed increased apoptosis and reduced cell proliferation in tumors treated with these analogs. Importantly, CIDD-99 had the most dramatic anti-tumor effects with 85% of tumors resolving leaving only minute traces of viable tissue. Additionally, CIDD-99 was non-noxious and demonstrated no observable adverse reactions CONCLUSION: This study describes a novel, highly efficacious, CPZ analog, CIDD-99, with dramatic anti-tumor effects against OSCC that may be efficacious as a lone therapy or in combination with standard therapies.


Assuntos
Apoptose , Capsaicina/análogos & derivados , Carcinoma de Células Escamosas/tratamento farmacológico , Estresse do Retículo Endoplasmático , Isoquinolinas/farmacologia , Mitocôndrias/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Capsaicina/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/patologia , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Biol Chem ; 292(27): 11154-11164, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28536265

RESUMO

The antischistosomal prodrug oxamniquine is activated by a sulfotransferase (SULT) in the parasitic flatworm Schistosoma mansoni. Of the three main human schistosome species, only S. mansoni is sensitive to oxamniquine therapy despite the presence of SULT orthologs in Schistosoma hematobium and Schistosoma japonicum The reason for this species-specific drug action has remained a mystery for decades. Here we present the crystal structures of S. hematobium and S. japonicum SULTs, including S. hematobium SULT in complex with oxamniquine. We also examined the activity of the three enzymes in vitro; surprisingly, all three are active toward oxamniquine, yet we observed differences in catalytic efficiency that implicate kinetics as the determinant for species-specific toxicity. These results provide guidance for designing oxamniquine derivatives to treat infection caused by all species of schistosome to combat emerging resistance to current therapy.


Assuntos
Resistência a Medicamentos , Proteínas de Helminto/química , Oxamniquine , Schistosoma haematobium/enzimologia , Schistosoma japonicum/enzimologia , Sulfotransferases/química , Animais , Cristalografia por Raios X , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Schistosoma haematobium/genética , Schistosoma japonicum/genética , Sulfotransferases/genética
9.
Chembiochem ; 19(14): 1488-1491, 2018 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-29679431

RESUMO

We introduce a new class of substrates (compounds I-III) for leukocyte esterase (LE) that react with LE yielding anodic current in direct proportion to LE activity. The kinetic constants Km and kcat for the enzymatic reactions were determined by amperometry at a glassy carbon electrode. The binding affinity of I-III for LE was two orders of magnitude better than that of existing optical LE substrates. The specificity constant kcat /Km was equal to 2.7, 3.8, and 5.8×105 m-1 s-1 for compounds containing the pyridine (I), methoxypyridine (II), and (methoxycarbonyl)pyridine (III), respectively, thus showing an increase in catalytic efficiency in this order. Compound III had the lowest octanol/water partition coefficient (log p=0.33) along with the highest topological surface area (tPSA=222 Å2 ) and the best aqueous solubility (4.0 mg mL-1 ). The average enzymatic activity of LE released from a single leukocyte was equal to 4.5 nU when measured with compound III.

10.
Biochemistry ; 56(6): 824-832, 2017 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-28121423

RESUMO

Nicotinamide N-methyltransferase (NNMT) is an important biotransforming enzyme that catalyzes the transfer of a labile methyl group from the ubiquitous cofactor S-5'-adenosyl-l-methionine (SAM) to endogenous and exogenous small molecules to form methylated end products. NNMT has been implicated in a number of chronic disease conditions, including metabolic disorders, cardiovascular disease, cancer, osteoarthritis, kidney disease, and Parkinson's disease. We have developed a novel noncoupled fluorescence-based methyltransferase assay that allows direct ultrasensitive real-time detection of the NNMT reaction product 1-methylquinolinium. This is the first assay reported to date to utilize fluorescence spectroscopy to directly monitor NNMT product formation and activity in real time. This assay provided accurate kinetic data that allowed detailed comparative analysis of the NNMT reaction mechanism and kinetic parameters. A reaction model based on a random bireactant mechanism produced global curve fits that were most consistent with steady-state initial velocity data collected across an array of substrate concentrations. On the basis of the reaction mechanism, each substrate could independently bind to the NNMT apoenzyme; however, both substrates bound to the complementary binary complexes with an affinity ∼20-fold stronger compared to their binding to the apoenzyme. This reaction mechanism implies either substrate-induced conformational changes or bireactant intermolecular interactions may stabilize the binding of the substrate to the binary complex and formation of the ternary complex. Importantly, this assay could rapidly generate concentration response curves for known NNMT inhibitors, suggesting its applicability for high-throughput screening of chemical libraries to identify novel NNMT inhibitors. Furthermore, our novel assay potentially offers a robust detection technology for use in SAM substrate competition assays for the discovery and development of SAM-dependent methyltransferase inhibitors.


Assuntos
Modelos Moleculares , Nicotinamida N-Metiltransferase/metabolismo , Apoenzimas/antagonistas & inibidores , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Biocatálise/efeitos dos fármacos , Calibragem , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Limite de Detecção , Metilação/efeitos dos fármacos , Nicotinamida N-Metiltransferase/antagonistas & inibidores , Nicotinamida N-Metiltransferase/química , Nicotinamida N-Metiltransferase/genética , Conformação Proteica , Redobramento de Proteína/efeitos dos fármacos , Compostos de Quinolínio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , S-Adenosilmetionina/metabolismo , Espectrometria de Fluorescência
11.
Anal Chem ; 89(14): 7781-7787, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28608672

RESUMO

The ester 4-((tosyl-l-alanyl)oxy)phenyl tosyl-l-alaninate (TAPTA) was synthesized and tested as a substrate for leukocyte esterase (LE), an enzyme produced by leukocytes (white blood cells). In the presence of LE, TAPTA released a redox-active fragment whose oxidation at an electrode provided a direct numerical measure of LE activity. The assays showed that LE recognized TAPTA as its substrate with the Michaelis constant Km and Imax equal to 0.24 mM and 0.13 mA cm-1, respectively. The esterolytic activity of leukocyte suspensions was determined by using the internally calibrated electrochemical continuous enzyme assay (ICECEA). One activity unit (U) of LE catalyzed the hydrolysis of 1.0 µmol of TAPTA per minute in a pH 7.40 phosphate buffer saline solution containing 10% dimethyl sulfoxide (DMSO) at 21 °C. The measured units were directly proportional to the number of leukocytes in the range of 0.028-4.2 U L-1 (9-690 µg L-1 LE protein). One white blood cell displayed the average esterolytic activity of 0.86 and 1.4 nU when the ultrasonic and chemical cytolysis were used, respectively. The ICECEA is an electrochemical alternative to optical assays for the determination of LE activity as an inflammatory biomarker and proxy for the presence of leukocytes.


Assuntos
Alanina/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Técnicas Eletroquímicas , Ensaios Enzimáticos , Ésteres/metabolismo , Leucócitos/química , Alanina/análogos & derivados , Alanina/química , Biocatálise , Hidrolases de Éster Carboxílico/química , Ésteres/química , Humanos , Hidrólise , Leucócitos/metabolismo , Estrutura Molecular , Especificidade por Substrato
12.
Bioorg Med Chem Lett ; 24(7): 1711-4, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24630558

RESUMO

The goal of this research was to identify structurally novel, non-quaternarypyridinium reactivators of GF (cyclosarin)-inhibited hAChE that possess the capacity to mediate in vitro reactivation of GF-inhibited human acetylcholinesterase (hAChE). New compounds were designed, synthesized and assessed in GF-inhibited hAChE assays. Structure activity relationships for AChE binding and reactivation of GF-inhibited hAChE were developed. Lead compounds from two different chemical series, represented by compounds 17 and 38, displayed proficient in vitro reactivation of GF-inhibited hAChE, while also possessing low inhibition of native enzyme.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Desenho de Fármacos , Compostos Organofosforados/farmacologia , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Compostos Organofosforados/síntese química , Compostos Organofosforados/química , Relação Estrutura-Atividade
14.
Artigo em Inglês | MEDLINE | ID: mdl-36758271

RESUMO

The antischistosomal drug oxamniquine, OXA, requires activation by a sulfotransferase within the parasitic worm to enable killing. Examination of the pharmacokinetic/pharmacodynamic (PK/PD) relationship for OXA identified an in vitro-in vivo paradox with the maximal clinical plasma concentrations five-to ten-times lower than the efficacious concentration for in vitro schistosomal killing. The parasite resides in the vasculature between the intestine and the liver, and modeling the PK data to determine portal concentrations fits with in vitro studies and explains the required human dose. In silico models were used to predict murine dosing to recapitulate human conditions for OXA portal concentration and time course. Follow-up PK studies verified in mice that a 50-100 mg/kg oral gavage dose of OXA formulated in acetate buffer recapitulates the 20-40 mg/kg dose common in patients. OXA was rapidly cleared through a combination of metabolism and excretion into bile. OXA absorbance and tissue distribution were similar in wild-type and P-gp efflux transporter knockout mice. The incorporation of in vitro efficacy data and portal concentration was demonstrated for an improved OXA-inspired analog that has been shown to kill S. mansoni, S. haematobium, and S. japonicum, whereas OXA is only effective against S. mansoni. Second-generation OXA analogs should optimize both in vitro killing and physiochemical properties to achieve high portal concentration via rapid oral absorption, facilitated by favorable solubility, permeability, and minimal intestinal metabolism.


Assuntos
Oxamniquine , Esquistossomicidas , Humanos , Camundongos , Animais , Oxamniquine/farmacologia , Schistosoma , Esquistossomicidas/farmacologia , Schistosoma mansoni
15.
Mol Cancer Ther ; 22(11): 1248-1260, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37493258

RESUMO

Glioblastoma (GBM) is the most prevalent and aggressive type of adult brain tumors with low 5-year overall survival rates. Epidemiologic data suggest that estrogen may decrease brain tumor growth, and estrogen receptor beta (ERß) has been demonstrated to exert antitumor functions in GBM. The lack of potent, selective, and brain permeable ERß agonist to promote its antitumor action is limiting the therapeutic promise of ERß. In this study, we discovered that Indanone and tetralone-keto or hydroxyl oximes are a new class of ERß agonists. Because of its high activity in ERß reporter assays, specific binding to ERß in polar screen assays, and potent growth inhibitory activity in GBM cells, CIDD-0149897 was discovered as a possible hit by screening a library of compounds. CIDD-0149897 is more selective for ERß than ERα (40-fold). Treatment with CIDD-0149897 markedly reduced GBM cell viability with an IC50 of ∼7 to 15 µmol/L, while having little to no effect on ERß-KO cells and normal human astrocytes. Further, CIDD-0149897 treatment enhanced expression of known ERß target genes and promoted apoptosis in established and patient-derived GSC models. Pharmacokinetic studies confirmed that CIDD-0149897 has systemic exposure, and good bioavailability in the brain. Mice tolerated daily intraperitoneal treatment of CIDD-0149897 (50 mg/kg) with a 7-day repeat dosage with no toxicity. In addition, CIDD-0149897 treatment significantly decreased tumor growth in U251 xenograft model and extended the survival of orthotopic GBM tumor-bearing mice. Collectively, these findings pointed to CIDD-0149897 as a new class of ERß agonist, offering patients with GBM a potential means of improving survival.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Camundongos , Animais , Glioblastoma/patologia , Receptor beta de Estrogênio/genética , Linhagem Celular Tumoral , Encéfalo/metabolismo , Estrogênios , Neoplasias Encefálicas/patologia
16.
Pediatr Blood Cancer ; 59(3): 485-92, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22238194

RESUMO

BACKGROUND: Alveolar rhabdomyosarcoma (ARMS) and embryonal rhabdomyosarcoma (ERMS) are among the most common and most treatment resistant soft tissue sarcomas of childhood. Here, we evaluated the potential of (18)F-Fluorodeoxyglucose (FDG) as a marker of therapeutic response to picropodophyllin (PPP), an IGF1R inhibitor, in a conditional mouse model of ARMS and a conditional model of ERMS/undifferentiated pleomorphic sarcoma (UPS). PROCEDURE: Primary tumor cell cultures from Myf6Cre,Pax3:Fkhr,p53 and Pax7CreER,Ptch1,p53 conditional models of ARMS and ERMS/UPS were found to be highly sensitive to PPP (IC(50) values 150 and 200 nM, respectively). Animals of each model were then treated with 80 mg/kg/day PPP by intraperitoneal injection for 12 days and imaged by (18)F-FDG microPET. RESULTS: Tumor volumes on day 4 for PPP-treated ARMS and ERMS mice were lower than untreated control mouse tumor volumes, although treated tumors were larger than day 0. However, tumor FDG uptake was significantly reduced on day 4 for PPP-treated mice compared to pretreatment baseline or untreated control mice on day 4 (P < 0.05). Nevertheless, by day 12 tumor volumes and FDG uptake for treated mice had increased significantly, indicating rapidly evolving resistance to therapy. CONCLUSIONS: (18)F-FDG PET imaging is a potential imaging biomarker of molecular susceptibility to targeted agents early in treatment for this aggressive form of sarcoma, but may find best use serially for Phase I/II studies where chemotherapy and targeted agents are combined to cytoreduce tumors and abrogate Igf1r inhibitor resistance.


Assuntos
Fluordesoxiglucose F18 , Podofilotoxina/análogos & derivados , Tomografia por Emissão de Pósitrons/métodos , Receptor IGF Tipo 1/antagonistas & inibidores , Rabdomiossarcoma/diagnóstico por imagem , Rabdomiossarcoma/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Camundongos , Podofilotoxina/uso terapêutico
17.
Pharmaceutics ; 14(7)2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35890311

RESUMO

Oxamniquine (OXA) is a prodrug activated by a sulfotransferase (SULT) that was only active against Schistosoma mansoni. We have reengineered OXA to be effective against S. haematobium and S. japonicum. Three derivatives stand out, CIDD-0066790, CIDD-0072229, and CIDD-0149830 as they kill all three major human schistosome species. However, questions remain. Is the OXA mode of action conserved in derivatives? RNA-interference experiments demonstrate that knockdown of the SmSULT, ShSULT, and SjSULT results in resistance to CIDD-0066790. Confirming that the OXA-derivative mode of action is conserved. Next is the level of expression of the schistosome SULTs in each species, as well as changes in SULT expression throughout development in S. mansoni. Using multiple tools, our data show that SmSULT has higher expression compared to ShSULT and SjSULT. Third, is the localization of SULT in the adult, multicellular eucaryotic schistosome species. We utilized fluorescence in situ hybridization and uptake of radiolabeled OXA to determine that multiple cell types throughout the adult schistosome worm express SULT. Thus, we hypothesize the ability of many cells to express the sulfotransferase accounts for the ability of the OXA derivatives to kill adult worms. Our studies demonstrate that the OXA derivatives are able to kill all three human schistosome species and thus will be a useful complement to PZQ.

18.
Front Physiol ; 13: 969000, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36187775

RESUMO

Oral cancer patients have a poor prognosis, with approximately 66% of patients surviving 5-years after diagnosis. Treatments for oral cancer are limited and have many adverse side effects; thus, further studies are needed to develop drugs that are more efficacious. To achieve this objective, we developed CIDD-99, which produces cytotoxic effects in multiple oral squamous cell carcinoma (OSCC) cell lines. While we demonstrated that CIDD-99 induces ER stress and apoptosis in OSCC, the mechanism was unclear. Investigation of the Bcl-family of proteins showed that OSCC cells treated with CIDD-99 undergo downregulation of Bcl-XL and Bcl-2 anti-apoptotic proteins and upregulation of Bax (pro-apoptotic). Importantly, OSCC cells treated with CIDD-99 displayed decreased calcium signaling in a dose and time-dependent manner, suggesting that blockage of calcium signaling is the key mechanism that induces cell death in OSCC. Indeed, CIDD-99 anti-proliferative effects were reversed by the addition of exogenous calcium. Moreover, electrophysiological properties further established that calcium entry was via the non-selective TRPC1 channel and prolonged CIDD-99 incubation inhibited STIM1 expression. CIDD-99 inhibition of calcium signaling also led to ER stress and inhibited mitochondrial complexes II and V in vitro. Taken together, these findings suggest that inhibition of TRPC mediates induction of ER stress and mitochondrial dysfunction as a part of the cellular response to CIDD-99 in OSCC.

19.
Chem Sci ; 12(11): 3984-3998, 2021 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-34163669

RESUMO

SfmD is a heme-dependent enzyme in the biosynthetic pathway of saframycin A. Here, we present a 1.78 Šresolution de novo crystal structure of SfmD, which unveils a novel heme cofactor attached to the protein with an unusual Hx n HxxxC motif (n ∼ 38). This heme cofactor is unique in two respects. It contains a single thioether bond in a cysteine-vinyl link with Cys317, and the ferric heme has two axial protein ligands, i.e., His274 and His313. We demonstrated that SfmD heme is catalytically active and can utilize dioxygen and ascorbate for a single-oxygen insertion into 3-methyl-l-tyrosine. Catalytic assays using ascorbate derivatives revealed the functional groups of ascorbate essential to its function as a cosubstrate. Abolishing the thioether linkage through mutation of Cys317 resulted in catalytically inactive SfmD variants. EPR and optical data revealed that the heme center undergoes a substantial conformational change with one axial histidine ligand dissociating from the iron ion in response to substrate 3-methyl-l-tyrosine binding or chemical reduction by a reducing agent, such as the cosubstrate ascorbate. The labile axial ligand was identified as His274 through redox-linked structural determinations. Together, identifying an unusual heme cofactor with a previously unknown heme-binding motif for a monooxygenase activity and the structural similarity of SfmD to the members of the heme-based tryptophan dioxygenase superfamily will broaden understanding of heme chemistry.

20.
Toxins (Basel) ; 13(6)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34203879

RESUMO

Shiga toxin 1 and 2 (STx1 and STx2) undergo retrograde trafficking to reach the cytosol of cells where they target ribosomes. As retrograde trafficking is essential for disease, inhibiting STx1/STx2 trafficking is therapeutically promising. Recently, we discovered that the chemotherapeutic drug tamoxifen potently inhibits the trafficking of STx1/STx2 at the critical early endosome-to-Golgi step. We further reported that the activity of tamoxifen against STx1/STx2 is independent of its selective estrogen receptor modulator (SERM) property and instead depends on its weakly basic chemical nature, which allows tamoxifen to increase endolysosomal pH and alter the recruitment of retromer to endosomes. The goal of the current work was to obtain a better understanding of the mechanism of action of tamoxifen against the more disease-relevant toxin STx2, and to differentiate between the roles of changes in endolysosomal pH and retromer function. Structure activity relationship (SAR) analyses revealed that a weakly basic amine group was essential for anti-STx2 activity. However, ability to deacidify endolysosomes was not obligatorily necessary because a tamoxifen derivative that did not increase endolysosomal pH exerted reduced, but measurable, activity. Additional assays demonstrated that protective derivatives inhibited the formation of retromer-dependent, Golgi-directed, endosomal tubules, which mediate endosome-to-Golgi transport, and the sorting of STx2 into these tubules. These results identify retromer-mediated endosomal tubulation and sorting to be fundamental processes impacted by tamoxifen; provide an explanation for the inhibitory effect of tamoxifen on STx2; and have important implications for the therapeutic use of tamoxifen, including its development for treating Shiga toxicosis.


Assuntos
Antineoplásicos Hormonais/farmacologia , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Tamoxifeno/farmacologia , Endossomos/efeitos dos fármacos , Células HeLa , Humanos , Transporte Proteico/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA