Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Drug Resist Updat ; 73: 101052, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38262246

RESUMEN

AIMS: This investigation aims to elucidate the mechanism underlying sorafenib-induced ferroptosis in hepatocellular carcinoma (HCC). METHODS: The role of dual specificity phosphatase 4 (DUSP4) in sorafenib-treated HCC was investigated using comprehensive assessments both in vitro and in vivo, including Western blotting, qRT-PCR, cell viability assay, lipid reactive oxygen species (ROS) assay, immunohistochemistry, and xenograft tumor mouse model. Additionally, label-free quantitative proteomics was employed to identify potential proteins associated with DUSP4. RESULTS: Our study revealed that suppression of DUSP4 expression heightens the susceptibility of HCC cells to ferroptosis inducers, specifically sorafenib and erastin, in both in vitro and in vivo settings. Furthermore, we identified DUSP4-mediated regulation of key ferroptosis-related markers, such as ferritin light chain (FTL) and ferritin heavy chain 1 (FTH1). Notably, label-free quantitative proteomics unveiled the phosphorylation of threonine residue T148 on YTH Domain Containing 1 (YTHDC1) by DUSP4. Further investigations unraveled that YTHDC1, functioning as an mRNA nuclear export regulator, is a direct target of DUSP4, orchestrating the subcellular localization of FTL and FTH1 mRNAs. Significantly, our study highlights a strong correlation between elevated DUSP4 expression and sorafenib resistance in HCC. CONCLUSIONS: Our findings introduce DUSP4 as a negative regulator of sorafenib-induced ferroptosis. This discovery opens new avenues for the development of ferroptosis-based therapeutic strategies tailored for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Humanos , Animales , Ratones , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Sorafenib/farmacología , Sorafenib/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Monoéster Fosfórico Hidrolasas/uso terapéutico , Ferroptosis/genética , Línea Celular Tumoral
2.
J Toxicol Environ Health A ; 87(6): 227-244, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38095149

RESUMEN

Docetaxel (DTX) is one of the chemotherapeutic drugs indicated as a first-line treatment against metastatic prostate cancer (mPCa). This study aimed to compare the impact of DTX on mPCa (DU-145) tumor cells cultured as 2D monolayers and 3D multicellular tumor spheroids (MCTS) in vitro. The cells were treated with DTX (1-96 µM) at 24, 48, or 72 hr in cell viability assays (resazurin, phosphatase acid, and lactate dehydrogenase). Cell death was assessed with fluorescent markers and proliferation by clonogenic assay (2D) and morphology, volume, and integrity assay (3D). The cell invasion was determined using transwell (2D) and extracellular matrix (ECM) (3D). Results showed that DTX decreased cell viability in both culture models. In 2D, the IC50 (72 hr) values were 11.06 µM and 14.23 µM for resazurin and phosphatase assays, respectively. In MCTS, the IC50 values for the same assays were 114.9 µM and 163.7 µM, approximately 10-fold higher than in the 2D model. The % of viable cells decreased, while the apoptotic cell number was elevated compared to the control in 2D. In 3D spheroids, only DTX 24 µM induced apoptosis. DTX (≥24 µM at 216 hr) lowered the volume, and DTX 96 µM completely disintegrated the MCTS. DTX reduced the invasion of mPCa cells to matrigel (2D) and migration from MCTS to the ECM. Data demonstrated significant differences in drug response between 2D and 3D cell culture models using mPCa DU-145 tumor cells. MCTS resembles the early stages of solid tumors in vivo and needs to be considered in conjunction with 2D cultures when searching for new therapeutic targets.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata , Masculino , Humanos , Docetaxel/farmacología , Docetaxel/uso terapéutico , Próstata , Línea Celular Tumoral , Esferoides Celulares , Neoplasias de la Próstata/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
3.
J Transl Med ; 21(1): 819, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974250

RESUMEN

BACKGROUND: The protein tyrosine phosphatase H receptor (PTPRH) is known to regulate the occurrence and development of pancreatic and colorectal cancer. However, its association with glycolysis in non-small cell lung cancer (NSCLC) is still unclear. In this study, we aimed to investigate the relationship between PTPRH expression and glucose metabolism and the underlying mechanism of action. METHODS: The expression of PTPRH in NSCLC cells was evaluated by IHC staining, qRT‒PCR and Western blotting. The effect of PTPRH on cell biological behavior was evaluated by colony assays, EdU experiments, Transwell assays, wound healing assays and flow cytometry. Changes in F-18-fluorodeoxyglucose (18F-FDG) uptake and glucose metabolite levels after altering PTPRH expression were detected via a gamma counter and lactic acid tests. The expression of glycolysis-related proteins in NSCLC cells was detected by Western blotting after altering PTPRH expression. RESULTS: The results showed that PTPRH was highly expressed in clinical patient tissue samples and closely related to tumor diameter and clinical stage. In addition, PTPRH expression was associated with glycometabolism indexes on 18F-FDG positron emission tomography/computed tomography (PET/CT) imaging, the expression level of Ki67 and the expression levels of glycolysis-related proteins. PTPRH altered cell behavior, inhibited apoptosis, and promoted 18F-FDG uptake, lactate production, and the expression of glycolysis-related proteins. In addition, PTPRH modulated the glycometabolism of NSCLC cells via the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway, as assessed using LY294002 and 740Y-P (an inhibitor and agonist of PI3K, respectively). The same results were validated in vivo using a xenograft tumor model in nude mice. Protein expression levels of PTPRH, glycolysis-related proteins, p-PI3K/PI3K and p-AKT/AKT were measured by IHC staining using a subcutaneous xenograft model in nude mice. CONCLUSIONS: In summary, we report that PTPRH promotes glycolysis, proliferation, migration, and invasion via the PI3K/AKT/mTOR signaling pathway in NSCLC and ultimately promotes tumor progression, which can be regulated by LY294002 and 740Y-P. These results suggest that PTPRH is a potential therapeutic target for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Ratones , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratones Desnudos , Neoplasias Pulmonares/patología , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/farmacología , Monoéster Fosfórico Hidrolasas/uso terapéutico , Fluorodesoxiglucosa F18 , Tomografía Computarizada por Tomografía de Emisión de Positrones , Proliferación Celular , Línea Celular Tumoral , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Glucólisis , Mamíferos/metabolismo
5.
ACS Appl Bio Mater ; 6(1): 164-170, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36525564

RESUMEN

Drug-resistant breast cancers such as Triple negative breast cancer (TNBC) do not respond successfully to chemotherapy treatments because they lack the expression of receptor targets. Drug-resistant anti-cancer treatments require innovative approaches to target these cells without relying on the receptors. Intracellular self-assembly of small molecules induced by enzymes is a nanotechnology approach for inhibiting cancer cell growth. In this approach, enzymes will induce the self-assembly of small molecules to nanofibers, which leads to cell death. Here, we investigate the self-assembly of a modified small peptide induced by two different phosphatases: alkaline phosphatase (ALP) and eye absent tyrosine phosphatase (EYA). ALPs are expressed in many adult human tissues and are critical for many cellular functions. EYAs are embryonic enzymes that are over-expressed in drug-resistant breast cancers. We synthesized a small diphenylalanine-based peptide with a tyrosine phosphate end group as the substrate of phosphatase enzymes. Peptides were synthesized with solid phase techniques and were characterized by HPLC and MALDI-TOF. To characterize the self-assembly of peptides exposed to enzymes, different techniques were used such as scattering light intensity, microscopes, and phosphate detection kit. We then determined the toxicity effect of the peptide against normal breast cancer cells, MCF-7, and drug-resistant breast cancer cells, MDA-MB-231. The results showed that the EYA enzyme is able to initiate self-assembly at lower peptide concentration with higher self-assembling intensity compared to ALP. A significant decrease in the TNBC cell number was observed even with a low peptide concentration of 60 µM. These results collectively support the exploration of enzyme self-assembly to treat TNBC.


Asunto(s)
Nanofibras , Neoplasias de la Mama Triple Negativas , Humanos , Fosfatasa Alcalina , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Nanofibras/química , Péptidos/farmacología , Monoéster Fosfórico Hidrolasas/farmacología , Monoéster Fosfórico Hidrolasas/uso terapéutico , Proliferación Celular
6.
Cell Mol Biol (Noisy-le-grand) ; 68(7): 75-84, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-36495515

RESUMEN

Protein tyrosine phosphatase-1B (PTP-1B) is a well-known therapeutic target for diabetes and obesity as it suppresses insulin and leptin signaling. PTP-1B deletion or pharmacological suppression boosted glucose homeostasis and insulin signaling without altering hepatic fat storage. Inhibitors of PTP-1B may be useful in the treatment of type 2 diabetes, and shikonin, a naturally occurring naphthoquinone dye pigment, is reported to inhibit PTP-1B and possess antidiabetic properties. Since the cell contains a large number of phosphatases, PTP-1B inhibitors must be effective and selective. To explore more about the mechanism underlying the inhibitor's efficacy and selectivity, we investigated its top four pharmacophores and used site-directed mutagenesis to insert amino acid mutations into PTP-1B as an extension of our previous study where we identified 4 pharmacophores of shikonin. The study aimed to examine the site-directed mutations like R24Y, S215E, and S216C influence the binding of shikonin pharmacophores, which act as selective inhibitors of PTP-1B. To achieve this purpose, docking and molecular dynamics simulations of wild-type (WT) and mutant PTP-1B with antidiabetic compounds were undertaken. The simulation results revealed that site-directed mutations can change the hydrogen bond and hydrophobic interactions between shikonin pharmacophores and many residues in PTP-1B's active site, influencing the drug's binding affinity. These findings could aid researchers in better understanding PTP-1B inhibitors' selective binding mechanism and pave the path for the creation of effective PTP-1B inhibitors.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Monoéster Fosfórico Hidrolasas/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Unión Proteica , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico
7.
J Nat Prod ; 85(12): 2836-2844, 2022 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-36399709

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) is highly validated as a therapeutic target for type 2 diabetes. However, active site-directed PTP1B inhibitors generally suffer from poor selectivity and bioavailability. Inspired by the identification of a unique anthraquinone-coumarin hybrid from Knoxia valerianoides exhibiting good specificity for PTP1B over the highly homologous T-cell protein tyrosine phosphatase (TCPTP), further chemical investigation of this plant species led to the isolation of nine new anthraquinone glycosides (1-9) and two known ones (10 and 11). Structures were characterized by a combination of spectroscopic analyses and chemical methods. All compounds showed PTP1B inhibitory activities with IC50 values ranging from 1.05 to 13.74 µM. Compounds 4 and 8 exhibited greater than 64-fold selectivity over TCPTP. Enzyme kinetic studies revealed that compounds 4 and 7 behaved as mixed-type inhibitors. Docking studies predicted similar binding modes of these compounds at the allosteric site positioned between helices α3 and α6.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/uso terapéutico , Cinética , Inhibidores Enzimáticos/farmacología , Antraquinonas/química , Glicósidos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Simulación del Acoplamiento Molecular
8.
Exp Mol Med ; 54(8): 1290-1305, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36042375

RESUMEN

The functional role of microRNA-375 (miR-375) in the development of prostate cancer (PCa) remains controversial. Previously, we found that plasma exosomal miR-375 is significantly elevated in castration-resistant PCa (CRPC) patients compared with castration-sensitive PCa patients. Here, we aimed to determine how miR-375 modulates CRPC progression and thereafter to evaluate the therapeutic potential of human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes loaded with miR-375 antisense oligonucleotides (e-375i). We used miRNA in situ hybridization technique to evaluate miR-375 expression in PCa tissues, gain- and loss-of-function experiments to determine miR-375 function, and bioinformatic methods, dual-luciferase reporter assay, qPCR, IHC and western blotting to determine and validate the target as well as the effects of miR-375 at the molecular level. Then, e-375i complexes were assessed for their antagonizing effects against miR-375. We found that the expression of miR-375 was elevated in PCa tissues and cancer exosomes, correlating with the Gleason score. Forced expression of miR-375 enhanced the expression of EMT markers and AR but suppressed apoptosis markers, leading to enhanced proliferation, migration, invasion, and enzalutamide resistance and decreased apoptosis of PCa cells. These effects could be reversed by miR-375 silencing. Mechanistically, miR-375 directly interfered with the expression of phosphatase nonreceptor type 4 (PTPN4), which in turn stabilized phosphorylated STAT3. Application of e-375i could inhibit miR-375, upregulate PTPN4 and downregulate p-STAT3, eventually repressing the growth of PCa. Collectively, we identified a novel miR-375 target, PTPN4, that functions upstream of STAT3, and targeting miR-375 may be an alternative therapeutic for PCa, especially for CRPC with high AR levels.


Asunto(s)
MicroARNs , Neoplasias de la Próstata Resistentes a la Castración , Proteína Tirosina Fosfatasa no Receptora Tipo 4 , Factor de Transcripción STAT3 , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 4/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 4/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
9.
Inflammopharmacology ; 30(5): 1871-1890, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35922737

RESUMEN

BACKGROUND AND OBJECTIVE: Tauopathy is a group of neurodegenerative diseases in which the pathogenesis processes are related to tau protein. The imbalances between the activities of kinases and phosphatases of tau protein lead to tau hyperphosphorylation and subsequent neurodegeneration. Numerous studies suggest a strong linkage between type 2 diabetes mellitus (T2D) and neurodegenerative diseases. Therefore, finding a drug with a dual therapeutic activity against T2D and neuroprotective will be a promising idea. Hence, the potential neuroprotective effect of Glimepiride (GPD) against tauopathy was evaluated in the current study. METHODS: P301S mice model was employed for tauopathy and C57BL/6 wild type mice (WT) was used as control. Phosphorylated and acetylated tau protein levels was assessed in cortex and hippocampus by western blot. Effect of GPD on tauopathy related enzymes, neuroinflammation, apoptotic markers were evaluated. Furthermore, the neuroprotective effects against anxiety like behavior and motor impairment was analyzed using Parallel rod floor and Open field tests. RESULTS: GPD significantly ameliorates motor impairment, anxiety like behavior and neurodegeneration in P301S mice. Phosphorylated tau and acetylated tau were significantly decreased in both cortex and hippocampus of P301S mice via decreasing GSK3ß, increasing ratio of phosphorylated-AKT to total-AKT, increasing PP2A and normalization of CDK5 levels. Furthermore, GPD treatment also decreased neuroinflammation and apoptosis by reducing NF-kB, TNF-α and caspase 3 levels. CONCLUSION: The current data suggests that GPD exerts a protective effect against tauopathy, behavioural consequences, neurodegeneration, neuroinflammation and apoptosis. GPD is therefore a promising agent for the treatment of neurodegenerative diseases associated with tauopathy.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Tauopatías , Animales , Caspasa 3 , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B , Enfermedades Neuroinflamatorias , Fármacos Neuroprotectores/farmacología , Monoéster Fosfórico Hidrolasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Compuestos de Sulfonilurea , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Tauopatías/patología , Factor de Necrosis Tumoral alfa , Proteínas tau/metabolismo , Proteínas tau/uso terapéutico
10.
Appl Biochem Biotechnol ; 194(10): 4683-4701, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35819691

RESUMEN

Insulin resistance (IR) is a condition of impaired response of cells towards insulin. It is marked by excessive blood glucose, dysregulated insulin signalling, altered pathways, damaged pancreatic ß-cells, metabolic disorders, etc. Chronic hyperglycemic conditions leads to type 2 diabetes mellitus (T2DM) which causes excess generation of highly reactive free radicals, causing oxidative stress, further leading to development and progression of complications like vascular dysfunction, damaged cellular proteins, and DNA. One of the causes for IR is dysregulation of protein tyrosine phosphatase 1B (PTP1B). Advancements in drug therapeutics have helped people manage IR by regulating PTP1B, however have been reported to cause side effects. Therefore, there is a growing interest on usage of phytochemical constituents having IR therapeutic properties and aiding to minimize these complications. Medicinal plants have not been utilized to their full potential as a therapeutic drug due to lack of knowledge of their active and effective chemical constituents, mode of action, regulation of IR parameters, and dosage of administration. This review highlights phytochemical constituents present in medicinal plants or spices, their potential effectiveness on proteins (PTP1B) regulating IR, and reported possible mechanism of action studied on in vitro models. The study gives current knowledge and future recommendations on the above aspects and is expected to be beneficial in developing herbal drug using these phytochemical constituents, either alone or in combination, for medication of IR and diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Drogas Sintéticas , Glucemia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Insulina/metabolismo , Monoéster Fosfórico Hidrolasas/uso terapéutico , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Drogas Sintéticas/uso terapéutico
11.
Blood Adv ; 6(5): 1371-1380, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-34847232

RESUMEN

Prognostic factors associated with chemotherapy outcomes in patients with acute myeloid leukemia (AML) are extensively reported, and one gene whose mutation is recognized as conferring resistance to several newer targeted therapies is protein tyrosine phosphatase non-receptor type 11 (PTPN11). The broader clinical implications of PTPN11 mutations in AML are still not well understood. The objective of this study was to determine which cytogenetic abnormalities and gene mutations co-occur with PTPN11 mutations and how PTPN11 mutations affect outcomes of patients treated with intensive chemotherapy. We studied 1725 patients newly diagnosed with AML (excluding acute promyelocytic leukemia) enrolled onto the Cancer and Leukemia Group B/Alliance for Clinical Trials in Oncology trials. In 140 PTPN11-mutated patient samples, PTPN11 most commonly co-occurred with mutations in NPM1, DNMT3A, and TET2. PTPN11 mutations were relatively common in patients with an inv(3)(q21q26)/t(3;3)(q21;q26) and a normal karyotype but were very rare in patients with typical complex karyotype and core-binding factor AML. Mutations in the N-terminal SH2 domain of PTPN11 were associated with a higher early death rate than those in the phosphatase domain. PTPN11 mutations did not affect outcomes of NPM1-mutated patients, but these patients were less likely to have co-occurring kinase mutations (ie, FLT3-ITD), suggesting activation of overlapping signaling pathways. However, in AML patients with wild-type NPM1, PTPN11 mutations were associated with adverse patient outcomes, providing a rationale to study the biology and treatment approaches in this molecular group. This trial was registered at www.clinicaltrials.gov as #NCT00048958 (CALGB 8461), #NCT00899223 (CALGB 9665), and #NCT00900224 (CALGB 20202).


Asunto(s)
Leucemia Mieloide Aguda , Monoéster Fosfórico Hidrolasas , Ensayos Clínicos como Asunto , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación , Nucleofosmina , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/uso terapéutico , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética
12.
Crit Rev Food Sci Nutr ; 62(15): 4095-4151, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33554619

RESUMEN

Type 2 diabetes (T2D) is an expanding global health problem, resulting from defects in insulin secretion and/or insulin resistance. In the past few years, both protein tyrosine phosphatase 1B (PTP1B) and dipeptidyl peptidase-4 (DPP-4), as well as their role in T2D, have attracted the attention of the scientific community. PTP1B plays an important role in insulin resistance and is currently one of the most promising targets for the treatment of T2D, since no available PTP1B inhibitors were still approved. DPP-4 inhibitors are among the most recent agents used in the treatment of T2D (although its use has been associated with possible cardiovascular adverse events). The antidiabetic properties of flavonoids are well-recognized, and include inhibitory effects on the above enzymes, although hitherto not therapeutically explored. In the present study, a comprehensive review of the literature of both synthetic and natural isolated flavonoids as inhibitors of PTP1B and DPP-4 activities is made, including their type of inhibition and experimental conditions, and structure-activity relationship, covering a total of 351 compounds. We intend to provide the most favorable chemical features of flavonoids for the inhibition of PTP1B and DPP-4, gathering information for the future development of compounds with improved potential as T2D therapeutic agents.


Asunto(s)
Diabetes Mellitus Tipo 2 , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV , Resistencia a la Insulina , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Inhibidores Enzimáticos/química , Flavonoides/farmacología , Flavonoides/uso terapéutico , Humanos , Hipoglucemiantes/química , Monoéster Fosfórico Hidrolasas/uso terapéutico , Relación Estructura-Actividad
13.
BMC Med ; 19(1): 239, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34615542

RESUMEN

BACKGROUND: With the revolutionary progress of immune checkpoint inhibitors (ICIs) achieved in non-small cell lung cancers (NSCLC), identifying patients benefiting from ICIs becomes critical and urgent. The associations of genomic alterations in protein tyrosine phosphatase receptor-type (PTPRs) and ICIs responses are unknown. METHODS: Whole-exome sequencing (WES) of 73 advanced NSCLC tumors sampled before anti-PD-(L)1 therapy was carried out with corresponding clinical data collected as a discovery cohort to find the associations of PTPR mutations and ICI responses. Three validation cohorts consolidated by 7 public cohorts of 1920 NSCLC patients with WES or target sequencing data of tumor tissue-derived DNA or circulating tumor DNA (ctDNA) and relevant clinical data were applied as validation cohorts. The lung adenocarcinoma (LUAD) cohort (n=586) in The Cancer Genome Atlas (TCGA) database was used for analyzing the potential anti-tumor immunologic mechanisms. RESULTS: With the highest mutation frequency among all PTPRs, PTPRD mutations in non-squamous NSCLC (ns-NSCLC) were linked to longer progression-free survivals (PFS, 324 vs 63 days, hazard ratio (HR)=0.36, p= 0.0152) and higher objective response rate (ORR, p=0.0099). In validation cohort 1 (n=377), ns-NSCLC patients with tissue PTPRD mutations had favorable PFS (9.10 vs 4.33 months, HR=0.62, p=0.0184) and ORR (p=0.013). In validation cohort 2 (n=406), ns-NSCLC patients with tissue PTPRD mutations had favorable overall survivals (OS, over 40 vs 11.94 months, HR=0.57, p=0.011). In validation cohort 3 (n=1137), ns-NSCLC patients with ctDNA PTPRD mutations had longer PFS (6.97 vs 2.73 months, HR=0.63, p=0.028) and higher ORR (p=0.047). Moreover, it was deleterious mutations in phosphatase domains (phosphatase-mut), rather than other mutations (other-mut), that were responsible of PTPRD's prediction efficiency. In addition, in validation cohort 3, ctDNA phosphatase-mut also functioned as a predictive biomarker helping identify patients benefiting more from ICIs than chemotherapy (interaction P for PFS=0.0506, for OS=0.04). Univariate and multivariate regression analysis revealed that phosphatase-mut was independent on PD-L1 expression and tumor mutation burden (TMB) to predict. In silico analysis based on TCGA LUAD cohort discovered enhanced anti-tumor immunity in phosphatase-mut patients. CONCLUSIONS: Tissue or ctDNA PTPRD phosphatase domain deleterious mutations might function as a both prognostic and predictive biomarker predicting clinical outcomes of ICIs in ns-NSCLC patients, independent on TMB or PD-L1 expression.


Asunto(s)
ADN Tumoral Circulante , Neoplasias Pulmonares , Biomarcadores de Tumor/genética , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación , Monoéster Fosfórico Hidrolasas/uso terapéutico , Pronóstico , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores
14.
Toxicol Lett ; 339: 60-69, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359019

RESUMEN

The progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) correlates with rupture of lysosome in Parkinson's disease (PD). It has been found that TP53-induced glycolysis and apoptosis regulator (TIGAR) has been attributed to the regulation of metabolic pathways and neuroprotective effect. In the present study, we showed in a mouse model that 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) caused lysosomal damage and DA neurons loss in the SNpc. MPTP only induced SP1-mediated TIGAR upregulation in the early stage of neurotoxin-induced pathology, and this compensatory mechanism was not enough to maintain normal lysosomal function. MPTP significantly decreased the levels of NADPH and GSH, and the effects were ameliorated by the expression of exogenous TIGAR but execerbated by knockdown of TIAGR. TIGAR or NADPH alleviated oxidative stress, rescued lysosomal dysfunction and attenuated DA neurons degeneration. Overexpression of TIGAR or NADPH supplement inhibited MPP+-mediated reactive oxygen species (ROS), lysosomal membrane permeabilization (LMP) and autophagic flux impairment in PC12 cells. Together, these findings suggest that TIGAR reduces MPTP-mediated oxidative stress, lysosomal depletion and DA neuron damage.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/uso terapéutico , Glucólisis/efectos de los fármacos , Lisosomas/efectos de los fármacos , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/fisiopatología , Enfermedad de Parkinson/fisiopatología , Monoéster Fosfórico Hidrolasas/uso terapéutico , Proteína p53 Supresora de Tumor/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Modelos Animales
15.
J Cell Biochem ; 119(12): 9899-9909, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30132953

RESUMEN

Osteosarcoma (OS) is an aggressive malignant tumor of bone, which often occurs in children and adolescents. Currently, the effective method for the treatment of OS is still limited. The study aimed to investigate the synergistic antitumor effect of inositol polyphosphate-4-phosphatase, type-II (INPP4B) and rucaparib on OS cells. The expression levels of INPP4B in OS tissues and OS cell lines were examined by quantitative real-time polymerase chain reaction and Western blot analysis. SaOS2 and U2OS cells were then transfected with overexpression vector of INPP4B or were treated with different concentrations of rucaparib, and cell viability, cell cycle, and apoptosis were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometry. Western blot assay uncovered the combined effects of INPP4B and rucaparib on cell cycle, apoptosis and phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signal pathway. Further, the tumor formation was examined in vivo. Results showed that INPP4B was low expressed in OS tissues and in OS cell lines. INPP4B overexpression significantly decreased cell viability and induced apoptosis in SaOS2 and U2OS cells. Additionally, rucaparib remarkably reduced cell viability in a dose-dependent and time-dependent manner. Meanwhile, rucaparib suppressed cell cycle progression in the S phase and promoted apoptosis in a dose-dependent manner. Further, combination of INPP4B overexpression and rucaparib declined Myc, cyclin E1 and cyclin D1 expressions, enhanced Bad, Bax, and cleaved-caspase-3 expressions, and blocked PI3K/AKT signal pathway in SaOS2 and U2OS cells. Finally, combination of INPP4B overexpression and rucaparib inhibited tumor formation in vivo. The study demonstrated that INPP4B and rucaparib exhibited synergistic antitumor effect by regulating PI3K/AKT pathway in OS cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Óseas/tratamiento farmacológico , Indoles/farmacología , Osteosarcoma/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/farmacología , Transducción de Señal , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/fisiopatología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/uso terapéutico , Masculino , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/fisiopatología , Fosfatidilinositol 3-Quinasa/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adulto Joven
16.
Neuropediatrics ; 45(1): 64-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297574

RESUMEN

Pyridoxal-5'-phosphate oxidase (PNPO) deficiency is a rare autosomal recessive, vitamin-responsive metabolic disorder causing refractory neonatal seizures that respond to the administration of pyridoxal-5'-phosphate (PLP). There are currently few case studies that have documented the functional outcome in PNPO deficiency, which remains poor in the majority of cases. We present the case of a male infant born at 35 weeks gestation who promptly responded to oral administration of PLP, following resistance to common anticonvulsive therapy and to a pyridoxine trial. Neurological outcome at 21 months is favorable and illustrates the importance of standardized vitamin trials in the acute setting of "therapy-resistant" neonatal seizures. Early recognition of PNPO deficiency and appropriate intervention might be associated with a more favorable outcome than initially considered.


Asunto(s)
Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/diagnóstico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/uso terapéutico , Piridoxaminafosfato Oxidasa/deficiencia , Convulsiones/diagnóstico , Convulsiones/tratamiento farmacológico , Humanos , Recien Nacido Prematuro , Masculino , Resultado del Tratamiento
17.
Neurochem Int ; 61(6): 899-906, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22342821

RESUMEN

Phosphorylation is a key post-translational modification for cellular signaling, and abnormalities in this process are observed in several neurodegenerative disorders. Among these disorders, Parkinson's disease (PD) is particularly intriguing as there are both genetic causes of disease that involve phosphorylation, and pathological hallmarks of disease composed of a hyperphosphorylated protein. Two of the major genes linked to PD are themselves kinases - leucine rich repeat kinase 2 (LRRK2) and phosphatase and tensin induced homolog kinase 1 (PINK1). Mutations in LRRK2 lead to its increased kinase activity and dominantly inherited PD, while mutations in PINK1 lead to loss of function and recessive PD. A third genetic linkage to disease is α-synuclein, a protein that is heavily phosphorylated in Lewy bodies and Lewy neurites, the pathological hallmarks of PD. The phosphorylation of α-synuclein at various residues influences its aggregation, either positively or negatively, thereby impacting its central role in disease pathogenesis. Given these associations of phosphorylation with PD, modulation of this modification is an attractive therapeutic strategy. The kinases that act in these disease relevant pathways have been the primary target for such approaches. But, the development of kinase inhibitors has been complicated by the necessary specificity to retain safety, the redundancy of kinases leading to lack of efficacy, and the difficulties in overcoming the blood-brain barrier. The field of modulating phosphatases has the potential to overcome some of these issues and provide the next generation of therapeutic targets for PD. In this review, we address the phosphorylation pathways involved in PD, the kinases and issues related to their inhibition, and the evolving field of the phosphatases relevant in PD and how they may be targeted pharmacologically.


Asunto(s)
Enfermedad de Parkinson/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mutación , Monoéster Fosfórico Hidrolasas/uso terapéutico , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
18.
Front Biosci (Schol Ed) ; 2(1): 209-20, 2010 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-20036941

RESUMEN

Organophosphorus (OP) pesticide poisoning causes significant morbidity and mortality, particularly in the developing world, with upwards of 3 million people poisoned each year. Although OP poisoning is not common in developed countries, recently greater attention has been given to these chemicals because of their similarity to chemical warfare agents. Despite the agricultural use of OP pesticides for roughly 60 years, no new therapies have been developed since the 1960s. A promising field of novel antidotes for OP poisoning, OP hydrolases, has recently garnered increased support. These bacterial enzymes have demonstrated tremendous prophylactic and antidotal efficacy against a few different OP classes in animal models. These studies, as well as the limitations and challenges of therapeutic development of these enzymes, are discussed.


Asunto(s)
Antídotos/uso terapéutico , Intoxicación por Organofosfatos , Plaguicidas/envenenamiento , Monoéster Fosfórico Hidrolasas/uso terapéutico , Intoxicación/tratamiento farmacológico , Antídotos/farmacocinética , Catálisis , Glutatión Transferasa/uso terapéutico , Humanos , Monoéster Fosfórico Hidrolasas/farmacocinética , Hidrolasas de Triéster Fosfórico/farmacocinética , Hidrolasas de Triéster Fosfórico/uso terapéutico
19.
Scand J Surg ; 93(4): 288-97, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15658670

RESUMEN

Prevention and treatment of secondary hyperparathyroidism (SHPT) in patients on chronic maintenance hemodialysis and of tertiary hyperparathyroidism (THPT) in patients after kidney transplantation is a challenge for the nephrologist and for the surgeon. Indication and results of medical and surgical therapy for SHPT and THPT have remained under discussion during the last decades. This review resumes the current medical and surgical strategies for patients with SHPT and THPT.


Asunto(s)
Hiperparatiroidismo Secundario/terapia , Fallo Renal Crónico/complicaciones , Huesos/metabolismo , Huesos/patología , Calcio/sangre , Calcio/metabolismo , Calcio/uso terapéutico , Dieta , Humanos , Hiperparatiroidismo Secundario/diagnóstico , Hiperparatiroidismo Secundario/etiología , Hipocalcemia/etiología , Trasplante de Riñón , Hormona Paratiroidea/sangre , Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/uso terapéutico , Paratiroidectomía/métodos , Monoéster Fosfórico Hidrolasas/sangre , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/uso terapéutico , Cuidados Posoperatorios , Complicaciones Posoperatorias , Recurrencia , Diálisis Renal/efectos adversos , Vitamina D/sangre , Vitamina D/metabolismo , Vitamina D/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...