Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 426
Filtrar
1.
Parasit Vectors ; 17(1): 279, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38943214

RESUMEN

BACKGROUND: Reliance on praziquantel for the treatment and control of schistosomiasis is likely to facilitate the emergence of drug resistance. Combination therapy targeting adult and juvenile schistosome worms is urgently needed to improve praziquantel efficacy and delay the potential development of drug resistance. We assessed the efficacy and safety of single-dose praziquantel combined with single-dose artesunate plus sulfalene-pyrimethamine in the treatment of Kenyan children with schistosomiasis. METHODS: This was an open-label, randomised clinical trial involving 426 school-aged children (7-15 years old) diagnosed with Schistosoma mansoni (by Kato-Katz) or S. haematobium (by urine filtration). They were randomly assigned (1:1:1) to receive a single dose of praziquantel (40 mg/kg), a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate) or combination therapy using a single dose of praziquantel (40 mg/kg) combined with a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate). The primary outcome was cure and egg reduction rates at 6 weeks post-treatment in the available case population. Adverse events were assessed within 3 h after treatment. RESULTS: Of the 426 children enrolled, 135 received praziquantel, 150 received artesunate plus sulfalene-pyrimethamine, and 141 received combination therapy. Outcome data were available for 348 (81.7%) children. For S. mansoni-infected children (n = 335), the cure rates were 75.6%, 60.7%, and 77.8%, and the egg reduction rates were 80.1%, 85.0%, and 88.4% for praziquantel, artesunate plus sulfalene-pyrimethamine, and combination therapy, respectively. For S. haematobium-infected children (n = 145), the corresponding cure rates were 81.4%, 71.1%, and 82.2%, and the egg reduction rates were 95.6%, 97.1%, and 97.7%, respectively. Seventy-one (16.7%) children reported mild-intensity adverse events. The drugs were well tolerated and no serious adverse events were reported. CONCLUSIONS: A single oral dose of praziquantel combined with artesunate plus sulfalene-pyrimethamine cured a high proportion of children with S. haematobium but did not significantly improve the treatment efficacy for either urinary or intestinal schistosomiasis. Sequential administration of praziquantel and artesunate plus sulfalene-pyrimethamine may enhance the efficacy and safety outcomes.


Asunto(s)
Antihelmínticos , Artemisininas , Artesunato , Quimioterapia Combinada , Praziquantel , Pirimetamina , Schistosoma haematobium , Schistosoma mansoni , Esquistosomiasis Urinaria , Esquistosomiasis mansoni , Humanos , Niño , Praziquantel/administración & dosificación , Praziquantel/efectos adversos , Praziquantel/uso terapéutico , Pirimetamina/administración & dosificación , Pirimetamina/uso terapéutico , Pirimetamina/efectos adversos , Animales , Adolescente , Artesunato/administración & dosificación , Artesunato/uso terapéutico , Femenino , Masculino , Esquistosomiasis mansoni/tratamiento farmacológico , Schistosoma haematobium/efectos de los fármacos , Esquistosomiasis Urinaria/tratamiento farmacológico , Schistosoma mansoni/efectos de los fármacos , Kenia , Artemisininas/administración & dosificación , Artemisininas/uso terapéutico , Artemisininas/efectos adversos , Resultado del Tratamiento , Antihelmínticos/administración & dosificación , Antihelmínticos/efectos adversos , Antihelmínticos/uso terapéutico , Sulfaleno/administración & dosificación , Sulfaleno/uso terapéutico , Sulfaleno/efectos adversos , Combinación de Medicamentos , Recuento de Huevos de Parásitos
2.
Oncol Res ; 32(6): 1093-1107, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827320

RESUMEN

Breast cancer is the leading cause of cancer-related deaths in women worldwide, with Hormone Receptor (HR)+ being the predominant subtype. Tamoxifen (TAM) serves as the primary treatment for HR+ breast cancer. However, drug resistance often leads to recurrence, underscoring the need to develop new therapies to enhance patient quality of life and reduce recurrence rates. Artemisinin (ART) has demonstrated efficacy in inhibiting the growth of drug-resistant cells, positioning art as a viable option for counteracting endocrine resistance. This study explored the interaction between artemisinin and tamoxifen through a combined approach of bioinformatics analysis and experimental validation. Five characterized genes (ar, cdkn1a, erbb2, esr1, hsp90aa1) and seven drug-disease crossover genes (cyp2e1, rorc, mapk10, glp1r, egfr, pgr, mgll) were identified using WGCNA crossover analysis. Subsequent functional enrichment analyses were conducted. Our findings confirm a significant correlation between key cluster gene expression and immune cell infiltration in tamoxifen-resistant and -sensitized patients. scRNA-seq analysis revealed high expression of key cluster genes in epithelial cells, suggesting artemisinin's specific impact on tumor cells in estrogen receptor (ER)-positive BC tissues. Molecular target docking and in vitro experiments with artemisinin on LCC9 cells demonstrated a reversal effect in reducing migratory and drug resistance of drug-resistant cells by modulating relevant drug resistance genes. These results indicate that artemisinin could potentially reverse tamoxifen resistance in ER-positive breast cancer.


Asunto(s)
Artemisininas , Neoplasias de la Mama , Biología Computacional , Resistencia a Antineoplásicos , Receptores de Estrógenos , Tamoxifeno , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Humanos , Artemisininas/farmacología , Artemisininas/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Resistencia a Antineoplásicos/genética , Biología Computacional/métodos , Receptores de Estrógenos/metabolismo , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Línea Celular Tumoral , Simulación del Acoplamiento Molecular , Proliferación Celular/efectos de los fármacos
3.
Science ; 384(6701): eadk5382, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38870290

RESUMEN

Polycystic ovary syndrome (PCOS), a prevalent reproductive disorder in women of reproductive age, features androgen excess, ovulatory dysfunction, and polycystic ovaries. Despite its high prevalence, specific pharmacologic intervention for PCOS is challenging. In this study, we identified artemisinins as anti-PCOS agents. Our finding demonstrated the efficacy of artemisinin derivatives in alleviating PCOS symptoms in both rodent models and human patients, curbing hyperandrogenemia through suppression of ovarian androgen synthesis. Artemisinins promoted cytochrome P450 family 11 subfamily A member 1 (CYP11A1) protein degradation to block androgen overproduction. Mechanistically, artemisinins directly targeted lon peptidase 1 (LONP1), enhanced LONP1-CYP11A1 interaction, and facilitated LONP1-catalyzed CYP11A1 degradation. Overexpression of LONP1 replicated the androgen-lowering effect of artemisinins. Our data suggest that artemisinin application is a promising approach for treating PCOS and highlight the crucial role of the LONP1-CYP11A1 interaction in controlling hyperandrogenism and PCOS occurrence.


Asunto(s)
Proteasas ATP-Dependientes , Artemisininas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol , Proteínas Mitocondriales , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratones , Ratas , Andrógenos/metabolismo , Artemisininas/uso terapéutico , Artemisininas/farmacología , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Modelos Animales de Enfermedad , Hiperandrogenismo/tratamiento farmacológico , Hiperandrogenismo/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Ovario/efectos de los fármacos , Ovario/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Proteolisis , Ratones Endogámicos C57BL , Adulto Joven , Adulto , Ratas Sprague-Dawley , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo
4.
Am J Trop Med Hyg ; 111(1): 43-47, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38806022

RESUMEN

Increasing antimicrobial resistance (AMR) is a global public health emergency. Although chemoprevention has improved malaria-related pregnancy outcomes, the downstream effects on AMR have not been characterized. We compared the abundance of 10 AMR genes in stool samples from pregnant women receiving sulfadoxine-pyrimethamine (SP) as intermittent preventive treatment against malaria in pregnancy (IPTp) to that in samples from women receiving dihydroartemisinin-piperaquine (DP) for IPTp. All participants had at least one AMR gene at baseline. Mean quantities of the antifolate gene dfrA17 were increased after two or more doses of SP (mean difference = 1.6, 95% CI: 0.4-2.7, P = 0.008). Antimicrobial resistance gene abundance tended to increase from baseline in SP recipients compared with a downward trend in the DP group. Overall, IPTp-SP had minimal effects on the abundance of antifolate resistance genes (except for dfrA17), potentially owing to a high starting prevalence. However, the trend toward increasing AMR in SP recipients warrants further studies.


Asunto(s)
Antimaláricos , Artemisininas , Combinación de Medicamentos , Heces , Pirimetamina , Quinolinas , Sulfadoxina , Humanos , Femenino , Pirimetamina/uso terapéutico , Pirimetamina/administración & dosificación , Pirimetamina/farmacología , Sulfadoxina/uso terapéutico , Sulfadoxina/administración & dosificación , Sulfadoxina/farmacología , Embarazo , Antimaláricos/uso terapéutico , Antimaláricos/farmacología , Antimaláricos/administración & dosificación , Quinolinas/uso terapéutico , Quinolinas/administración & dosificación , Artemisininas/uso terapéutico , Artemisininas/farmacología , Artemisininas/administración & dosificación , Adulto , Heces/microbiología , Adulto Joven , Complicaciones Parasitarias del Embarazo/prevención & control , Complicaciones Parasitarias del Embarazo/tratamiento farmacológico , Resistencia a Medicamentos/genética , Malaria Falciparum/prevención & control , Malaria Falciparum/epidemiología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Piperazinas
5.
Lancet Microbe ; 5(7): 633-644, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38705163

RESUMEN

BACKGROUND: Artemether-lumefantrine is widely used for uncomplicated Plasmodium falciparum malaria; sulfadoxine-pyrimethamine plus amodiaquine is used for seasonal malaria chemoprevention. We aimed to determine the efficacy of artemether-lumefantrine with and without primaquine and sulfadoxine-pyrimethamine plus amodiaquine with and without tafenoquine for reducing gametocyte carriage and transmission to mosquitoes. METHODS: In this phase 2, single-blind, randomised clinical trial conducted in Ouelessebougou, Mali, asymptomatic individuals aged 10-50 years with P falciparum gametocytaemia were recruited from the community and randomly assigned (1:1:1:1) to receive either artemether-lumefantrine, artemether-lumefantrine with a single dose of 0·25 mg/kg primaquine, sulfadoxine-pyrimethamine plus amodiaquine, or sulfadoxine-pyrimethamine plus amodiaquine with a single dose of 1·66 mg/kg tafenoquine. All trial staff other than the pharmacist were masked to group allocation. Participants were not masked to group allocation. Randomisation was done with a computer-generated randomisation list and concealed with sealed, opaque envelopes. The primary outcome was the median within-person percent change in mosquito infection rate in infectious individuals from baseline to day 2 (artemether-lumefantrine groups) or day 7 (sulfadoxine-pyrimethamine plus amodiaquine groups) after treatment, assessed by direct membrane feeding assay. All participants who received any trial drug were included in the safety analysis. This study is registered with ClinicalTrials.gov, NCT05081089. FINDINGS: Between Oct 13 and Dec 16, 2021, 1290 individuals were screened and 80 were enrolled and randomly assigned to one of the four treatment groups (20 per group). The median age of participants was 13 (IQR 11-20); 37 (46%) of 80 participants were female and 43 (54%) were male. In individuals who were infectious before treatment, the median percentage reduction in mosquito infection rate 2 days after treatment was 100·0% (IQR 100·0-100·0; n=19; p=0·0011) with artemether-lumefantrine and 100·0% (100·0-100·0; n=19; p=0·0001) with artemether-lumefantrine with primaquine. Only two individuals who were infectious at baseline infected mosquitoes on day 2 after artemether-lumefantrine and none at day 5. By contrast, the median percentage reduction in mosquito infection rate 7 days after treatment was 63·6% (IQR 0·0-100·0; n=20; p=0·013) with sulfadoxine-pyrimethamine plus amodiaquine and 100% (100·0-100·0; n=19; p<0·0001) with sulfadoxine-pyrimethamine plus amodiaquine with tafenoquine. No grade 3-4 or serious adverse events occurred. INTERPRETATION: These data support the effectiveness of artemether-lumefantrine alone for preventing nearly all mosquito infections. By contrast, there was considerable post-treatment transmission after sulfadoxine-pyrimethamine plus amodiaquine; therefore, the addition of a transmission-blocking drug might be beneficial in maximising its community impact. FUNDING: Bill & Melinda Gates Foundation.


Asunto(s)
Amodiaquina , Antimaláricos , Combinación Arteméter y Lumefantrina , Combinación de Medicamentos , Fluorenos , Malaria Falciparum , Plasmodium falciparum , Primaquina , Pirimetamina , Sulfadoxina , Humanos , Antimaláricos/uso terapéutico , Antimaláricos/administración & dosificación , Pirimetamina/uso terapéutico , Pirimetamina/administración & dosificación , Amodiaquina/uso terapéutico , Amodiaquina/administración & dosificación , Sulfadoxina/uso terapéutico , Sulfadoxina/administración & dosificación , Masculino , Adulto , Femenino , Adolescente , Niño , Malaria Falciparum/transmisión , Malaria Falciparum/prevención & control , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/epidemiología , Método Simple Ciego , Persona de Mediana Edad , Primaquina/uso terapéutico , Primaquina/administración & dosificación , Combinación Arteméter y Lumefantrina/uso terapéutico , Combinación Arteméter y Lumefantrina/administración & dosificación , Adulto Joven , Fluorenos/administración & dosificación , Fluorenos/uso terapéutico , Malí/epidemiología , Plasmodium falciparum/efectos de los fármacos , Artemisininas/administración & dosificación , Artemisininas/uso terapéutico , Aminoquinolinas/administración & dosificación , Aminoquinolinas/uso terapéutico , Aminoquinolinas/efectos adversos , Etanolaminas/administración & dosificación , Etanolaminas/uso terapéutico , Animales , Quimioterapia Combinada
6.
J Mol Neurosci ; 74(2): 52, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38724832

RESUMEN

Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism-based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease.


Asunto(s)
Artemisininas , Neoplasias Encefálicas , Glioblastoma , Glutamina , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Humanos , Glutamina/metabolismo , Línea Celular Tumoral , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Artemisininas/uso terapéutico , Artemisininas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Glutaminasa/metabolismo , Glutaminasa/antagonistas & inhibidores , Microambiente Tumoral , Apoptosis , Tiadiazoles/farmacología , Tiadiazoles/uso terapéutico , Movimiento Celular , Bencenoacetamidas/farmacología , Bencenoacetamidas/uso terapéutico , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología
7.
Eur J Med Res ; 29(1): 293, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773551

RESUMEN

Artesunate (ART), an effective antimalarial semisynthetic derivative of artemisinin, exhibits antitumour properties, but the mechanism(s) involved remain elusive. In this study, we investigated the antitumour effects of ART on human oesophageal squamous cell carcinoma (ESCC) cell lines. Treatment of ESCC cell lines with ART resulted in the production of excessive reactive oxygen species (ROS) that induced DNA damage, reduced cell proliferation and inhibited clonogenicity via G1-S cell cycle arrest and/or apoptosis in vitro. The administration of ART to nude mice with ESCC cell xenografts inhibited tumour formation in vivo. However, the cytotoxicity of ART strongly differed among the ESCC cell lines tested. Transcriptomic profiling revealed that although the expression of large numbers of genes in ESCC cell lines was affected by ART treatment, these genes could be functionally clustered into pathways involved in regulating cell cycle progression, DNA metabolism and apoptosis. We revealed that p53 and Cdk4/6-p16-Rb cell cycle checkpoint controls were critical determinants required for mediating ART cytotoxicity in ESCC cell lines. Specifically, KYSE30 cells with p53Mut/p16Mut were the most sensitive to ART, KYSE150 and KYSE180 cells with p53Mut/p16Nor exhibited intermediate responses to ART, and Eca109 cells with p53Nor/p16Nor exhibited the most resistance to ATR. Consistently, perturbation of p53 expression using RNA interference (RNAi) and/or Cdk4/6 activity using the inhibitor palbociclib altered ART cytotoxicity in KYSE30 cells. Given that the p53 and Cdk4/6-cyclin D1-p16-Rb genes are commonly mutated in ESCC, our results potentially shed new light on neoadjuvant chemotherapy strategies for ESCC.


Asunto(s)
Apoptosis , Artesunato , Puntos de Control del Ciclo Celular , Proliferación Celular , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Artesunato/farmacología , Artesunato/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Animales , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/genética , Ratones , Línea Celular Tumoral , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Ratones Desnudos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Daño del ADN/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Artemisininas/farmacología , Artemisininas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología
8.
Sci Rep ; 14(1): 12556, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821986

RESUMEN

Diabetic patients are at high risk of developing lacrimal gland dysfunction, and the antimalarial drug artesunate (ART) was recently used to induce experimental-induced diabetes mellitus. This study's objective is to investigate the lacrimal gland alteration and the effect of ART on experimentally induced diabetes rat models and its related mechanisms. Forty rats were divided into five groups (8 rats/group): healthy control group (HC), diabetic group (DM), 50 mg/kg ART intervention diabetic group [DM + ART (50 mg/kg)], 100 mg/kg ART intervention diabetic group [DM + ART (100 mg/kg)] and 6 U/kg Insulin intervention diabetic group (DM + INS). The morphology of the eyeball and lacrimal gland tissues was determined using hematoxylin and eosin staining. In addition, external lacrimal glands were harvested for electronic microscopic examination, NFκB1, and TNF-α protein expression evaluation by immunohistochemistry and mRNA expression analysis by RT-PCR. Histopathological and ultrastructural changes suggest ART intervention has an improved structural effect. Protein expression of NFκB1 in the DM + ART (100 mg/kg) group was decreased. TNF-α significantly decreased in the DM + ART (50 mg/kg) and insulin groups. We concluded that ART improves structural changes in a lacrimal gland in diabetic rats. The present study provides further evidence of the therapeutic effect of ART on the lacrimal gland of diabetic rats by decreasing the expression of NFκB1 and TNF-α.


Asunto(s)
Artesunato , Diabetes Mellitus Experimental , Aparato Lagrimal , Animales , Artesunato/farmacología , Artesunato/uso terapéutico , Aparato Lagrimal/efectos de los fármacos , Aparato Lagrimal/metabolismo , Aparato Lagrimal/patología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Ratas , Masculino , Factor de Necrosis Tumoral alfa/metabolismo , Artemisininas/farmacología , Artemisininas/uso terapéutico
9.
Int Immunopharmacol ; 133: 112157, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38678671

RESUMEN

In non-small cell lung cancer (NSCLC), identifying a component with certain molecular targets can aid research on cancer treatment. Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin which induced the anti-cancer effects via the STAT3 signaling pathway, but the underlying molecular mechanism is still elusive. In this study, we first proved that DHA prohibits the growth of tumors both in vitro and in vivo. Data from transcriptomics showed that DHA reduced the expression level of the genes involved in cell cycle-promoting and anti-apoptosis, and most importantly, DHA restricted the expression level of receptor tyrosine kinase-like orphan receptor 1 (ROR1) which has been reported to have abnormal expression on tumor cells and had close interaction with STAT3 signaling. Then, we performed comprehensive experiments and found that DHA remarkably decreased the expression of ROR1 at both mRNA and protein levels and it also diminished the phosphorylation level of STAT3 in NSCLC cell lines. In addition, our data showed that exogenously introduced ROR1 could significantly enhance the phosphorylation of STAT3 while blocking ROR1 had the opposite effects indicating that ROR1 plays a critical role in promoting the activity of STAT3 signaling. Finally, we found that ROR1 overexpression could partially reverse the decreased activity of STAT3 induced by DHA which indicates that DHA-induced anti-growth signaling is conferred, at least in part, through blocking ROR1-mediated STAT3 activation. In summary, our study indicates that in NSCLC, ROR1 could be one of the critical molecular targets mediating DHA-induced STAT3 retardation.


Asunto(s)
Artemisininas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Factor de Transcripción STAT3 , Artemisininas/farmacología , Artemisininas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Animales , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos , Apoptosis/efectos de los fármacos , Ratones , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células A549 , Ratones Endogámicos BALB C
10.
J Cell Mol Med ; 28(8): e18335, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38652216

RESUMEN

Management of hepatocellular carcinoma (HCC) remains challenging due to population growth, frequent recurrence and drug resistance. Targeting of genes involved with the ferroptosis is a promising alternative treatment strategy for HCC. The present study aimed to investigate the effect of dihydroartemisinin (DHA) against HCC and explore the underlying mechanisms. The effects of DHA on induction of ferroptosis were investigated with the measurement of malondialdehyde concentrations, oxidised C11 BODIPY 581/591 staining, as well as subcutaneous xenograft experiments. Activated transcription factor 4 (ATF4) and solute carrier family 7 member 11 (SLC7A11 or xCT) were overexpressed with lentiviruses to verify the target of DHA. Here, we confirmed the anticancer effect of DHA in inducing ferroptosis is related to ATF4. High expression of ATF4 is related to worse clinicopathological prognosis of HCC. Mechanistically, DHA inhibited the expression of ATF4, thereby promoting lipid peroxidation and ferroptosis of HCC cells. Overexpression of ATF4 rescued DHA-induced ferroptosis. Moreover, ATF4 could directly bound to the SLC7A11 promoter and increase its transcription. In addition, DHA enhances the chemosensitivity of sorafenib on HCC in vivo and in vitro. These findings confirm that DHA induces ferroptosis of HCC via inhibiting ATF4-xCT pathway, thereby providing new drug options for the treatment of HCC.


Asunto(s)
Factor de Transcripción Activador 4 , Sistema de Transporte de Aminoácidos y+ , Artemisininas , Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Ferroptosis/efectos de los fármacos , Artemisininas/farmacología , Artemisininas/uso terapéutico , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Humanos , Animales , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Ratones , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino , Ratones Desnudos , Sorafenib/farmacología , Sorafenib/uso terapéutico , Femenino , Ratones Endogámicos BALB C
11.
Mol Immunol ; 170: 144-155, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38669759

RESUMEN

OBJECTIVE: Dihydroartemisinin (DHA) plays a very important role in various diseases. However, the precise involvement of DHA in systemic lupus erythematosus (SLE), relation to the equilibrium between M1 and M2 cells, remains uncertain. Therefore, we aimed to investigate the role of DHA in SLE and its effect on the M1/M2 cells balance. METHODS: SLE mice model was established by pristane induction. Flow cytometry was employed to measure the abundance of M1 and M2 cells within the peripheral blood of individuals diagnosed with SLE. The concentrations of various cytokines, namely TNF-α, IL-1ß, IL-4, IL-6, and IL-10, within the serum of SLE patients or SLE mice were assessed via ELISA. Immunofluorescence staining was utilized to detect the deposition of IgG and complement C3 in renal tissues of the mice. We conducted immunohistochemistry analysis to assess the expression levels of Collagen-I, a collagen protein, and α-SMA, a fibrosis marker protein, in the renal tissues of mice. Hematoxylin-eosin staining, Masson's trichrome staining, and Periodic acid Schiff staining were used to examine histological alterations. In this study, we employed qPCR and western blot techniques to assess the expression levels of key molecular markers, namely CD80 and CD86 for M1 cells, as well as CD206 and Arg-1 for M2 cells, within kidney tissue. Additionally, we investigated the involvement of the MAPK signaling pathway. The Venny 2.1 online software tool was employed to identify shared drug-disease targets, and subsequently, the Cytoscape 3.9.2 software was utilized to construct the "disease-target-ingredient" network diagram. Protein-protein interactions of the target proteins were analyzed using the String database, and the network proteins underwent enrichment analysis for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways. RESULTS: The results showed that an increase in M1 cells and a decrease in M2 cells within the peripheral blood of individuals diagnosed with SLE. Further analysis revealed that prednisone (PDN) combined with DHA can alleviate kidney damage and regulate the balance of M1 and M2 cells in both glomerular mesangial cells (GMC) and kidney. The MAPK signaling pathway was found to be involved in SLE kidney damage and M1/M2 balance in the kidney. Furthermore, PDN and/or DHA were found to inhibit the MAPK signaling pathway in GMC and kidney. CONCLUSION: We demonstrated that PDN combined with DHA attenuates SLE by regulating M1/M2 balance through MAPK signaling pathway. These findings propose that the combination of PDN and DHA could serve as a promising therapeutic strategy for SLE, as it has the potential to mitigate kidney damage and reinstate the equilibrium of M1 and M2 cells.


Asunto(s)
Artemisininas , Lupus Eritematoso Sistémico , Sistema de Señalización de MAP Quinasas , Prednisona , Animales , Humanos , Ratones , Artemisininas/farmacología , Artemisininas/uso terapéutico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Lupus Eritematoso Sistémico/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Prednisona/farmacología , Prednisona/uso terapéutico
12.
Neoplasia ; 51: 100991, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38507887

RESUMEN

Dihydroartemisinin (DHA) exerts an anti-tumor effect in multiple cancers, however, the molecular mechanism of DHA and whether DHA facilitates the anti-tumor efficacy of cisplatin in non-small cell lung cancer (NSCLC) are unclear. Here, we found that DHA potentiated the anti-tumor effects of cisplatin in NSCLC cells by stimulating reactive oxygen species (ROS)-mediated endoplasmic reticulum (ER) stress, C-Jun-amino-terminal kinase (JNK) and p38 MAPK signaling pathways both in vitro and in vivo. Of note, we demonstrated for the first time that DHA inhibits prostaglandin G/H synthase 1 (PTGS1) expression, resulting in enhanced ROS production. Importantly, silencing PTGS1 sensitized DHA-induced cell death by increasing ROS production and activating ER-stress, JNK and p38 MAPK signaling pathways. In summary, our findings provided new experimental basis and therapeutic prospect for the combined therapy with DHA and cisplatin in some NSCLC patients.


Asunto(s)
Artemisininas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Especies Reactivas de Oxígeno , Humanos , Apoptosis , Artemisininas/farmacología , Artemisininas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Muerte Celular , Línea Celular Tumoral , Cisplatino/farmacología , Ciclooxigenasa 1/metabolismo , Neoplasias Pulmonares/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Inhibidores de la Ciclooxigenasa/farmacología
13.
Int J Parasitol Drugs Drug Resist ; 24: 100532, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38520842

RESUMEN

Artemether-lumefantrine (AL) is the most widely used antimalarial drug for treating uncomplicated falciparum malaria. This study evaluated whether the K65Q mutation in the Plasmodium falciparum cysteine desulfurase IscS (Pfnfs1) gene was associated with alternated susceptibility to lumefantrine using clinical parasite samples from Ghana and the China-Myanmar border area. Parasite isolates from the China-Myanmar border had significantly higher IC50 values to lumefantrine than parasites from Ghana. In addition, the K65 allele was significantly more prevalent in the Ghanaian parasites (34.5%) than in the China-Myanmar border samples (6.8%). However, no difference was observed in the lumefantrine IC50 value between the Pfnfs1 reference K65 allele and the non reference 65Q allele in parasites from the two regions. These data suggest that the Pfnfs1 K65Q mutation may not be a reliable marker for reduced susceptibility to lumefantrine.


Asunto(s)
Antimaláricos , Artemisininas , Malaria Falciparum , Humanos , Lumefantrina/farmacología , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Plasmodium falciparum , Combinación Arteméter y Lumefantrina/uso terapéutico , Ghana , Artemisininas/farmacología , Artemisininas/uso terapéutico , Arteméter/uso terapéutico , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Mutación , Etanolaminas/farmacología , Etanolaminas/uso terapéutico , Resistencia a Medicamentos/genética
14.
J Nat Med ; 78(2): 439-454, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38351420

RESUMEN

Dihydroartemisinin (DHA), a derivative of artemisinin which is primarily used to treat malaria in clinic, also confers protective effect on lipopolysaccharide-induced nephrotoxicity. While, the activities of DHA in cisplatin (CDDP)-caused nephrotoxicity are elusive. To investigate the role and underlying mechanism of DHA in CDDP-induced nephrotoxicity. Mice were randomly separated into four groups: normal, CDDP, and DHA (25 and 50 mg/kg were orally injected 1 h before CDDP for consecutive 10 days). All mice except the normal were single injected intraperitoneally with CDDP (22 mg/kg) for once on the 7th day. Combined with quantitative proteomics and bioinformatics analysis, the impact of DHA on renal cell apoptosis, oxidative stress, biochemical indexes, and inflammation in mice were investigated. Moreover, a human hepatocellular carcinoma cells xenograft model was established to elucidate the impact of DHA on tumor-related effects of CDDP. DHA reduced the levels of creatinine (CREA) (p < 0.01) and blood urea nitrogen (BUN) (p < 0.01), reversed CDDP-induced oxidative, inflammatory, and apoptosis indexes (p < 0.01). Mechanistically, DHA attenuated CDDP-induced inflammation by inhibiting nuclear factor κB p65 (NFκB p65) expression, and suppressed CDDP-induced renal cell apoptosis by inhibiting p63-mediated endogenous and exogenous apoptosis pathways. Additionally, DHA alone significantly decreased the tumor weight and did not destroy the antitumor effect of CDDP, and did not impact AST and ALT. In conclusion, DHA prevents CDDP-triggered nephrotoxicity via reducing inflammation, oxidative stress, and apoptosis. The mechanisms refer to inhibiting NFκB p65-regulated inflammation and alleviating p63-mediated mitochondrial endogenous and Fas death receptor exogenous apoptosis pathway.


Asunto(s)
Antineoplásicos , Artemisininas , Humanos , Ratones , Animales , Cisplatino/toxicidad , Artemisininas/farmacología , Artemisininas/uso terapéutico , Artemisininas/metabolismo , Riñón/metabolismo , Riñón/patología , Estrés Oxidativo , Inflamación/metabolismo , Apoptosis , Antineoplásicos/toxicidad
15.
Am J Chin Med ; 52(1): 161-181, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38328829

RESUMEN

Ferroptosis, an iron-dependent cell death mechanism driven by an accumulation of lipid peroxides on cellular membranes, has emerged as a promising strategy to treat various diseases, including cancer. Ferroptosis inducers not only exhibit cytotoxic effects on multiple cancer cells, including drug-resistant cancer variants, but also hold potential as adjuncts to enhance the efficacy of other anti-cancer therapies, such as immunotherapy. In addition to synthetic inducers, natural compounds, such as artemisinin, can be considered ferroptosis inducers. Artemisinin, extracted from Artemisia annua L., is a poorly water-soluble antimalarial drug. For clinical applications, researchers have synthesized various water-soluble artemisinin derivatives such as dihydroartemisinin, artesunate, and artemether. Artemisinin and artemisinin derivatives (ARTEs) upregulate intracellular free iron levels and promote the accumulation of intracellular lipid peroxides to induce cancer cell ferroptosis, alleviating cancer development and resulting in strong anti-cancer effects in vitro and in vivo. In this review, we introduce the mechanisms of ferroptosis, summarize the research on ARTEs-induced ferroptosis in cancer cells, and discuss the clinical research progress and current challenges of ARTEs in anti-cancer treatment. This review deepens the current understanding of the relationship between ARTEs and ferroptosis and provides a theoretical basis for the clinical anti-cancer application of ARTEs in the future.


Asunto(s)
Artemisininas , Ferroptosis , Neoplasias , Humanos , Artemisininas/farmacología , Artemisininas/uso terapéutico , Hierro , Peróxidos Lipídicos , Neoplasias/tratamiento farmacológico , Agua
16.
Int J Infect Dis ; 139: 41-49, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38016502

RESUMEN

OBJECTIVES: Partial artemisinin resistance, mediated by Plasmodium falciparum K13 (PfK13) mutations, has been confirmed in certain areas of East Africa that are historically associated with high-level antimalarial resistance. The Democratic Republic of Congo (DRC) borders these areas in the East. This study aimed to determine the prevalence of resistance markers in six National Malaria Control Program surveillance sites; Boende, Kabondo, Kapolowe, Kimpese, Mikalayi, and Rutshuru. METHODS: The single nucleotide polymorphisms (SNPs) in P. falciparum genes PfK13, Pfdhfr, Pfdhps, Pfmdr1, and Pfcrt were assessed using targeted next-generation sequencing of isolates collected at enrollment in therapeutic efficacy studies. RESULTS: PfK13 SNPs were detected in two samples: in Kabondo (R561H) and in Rutshuru (P441L), both areas near Uganda and Rwanda. The Pfdhps ISGEGA haplotype, associated with reduced sulfadoxine-pyrimethamine chemoprevention efficacy, ranged from 0.8% in Mikalayi (central DRC) to 42.2% in Rutshuru (East DRC). CONCLUSIONS: R561H and P441L observed in eastern DRC are a concern, as they are associated with delayed artemisinin-based combination therapies-clearance and candidate marker of resistance, respectively. This is consistent with previous observations of shared drug resistance profiles in parasites of that region with bordering areas of Rwanda and Uganda. The likely circulation of parasites has important implications for the ongoing surveillance of partial artemisinin-resistant P. falciparum and for future efforts to mitigate its dispersal.


Asunto(s)
Antimaláricos , Artemisininas , Malaria Falciparum , Humanos , Plasmodium falciparum/genética , República Democrática del Congo/epidemiología , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Artemisininas/farmacología , Artemisininas/uso terapéutico , Mutación , Uganda , Proteínas Protozoarias/genética
17.
Exp Parasitol ; 255: 108655, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37981259

RESUMEN

In previous studies, the artemisinin derivatives artemisone, its pro-drug artemiside and the bumped-kinase inhibitor BKI-1748 were effective against T. gondii via different modes of action. This suggests that they may act synergistically resulting in improved efficacies in vitro and in vivo. To test this hypothesis, the compounds were applied alone and in combination to T. gondii infected human fibroblast host cells in order to determine their inhibition constants and effects on cellular ultrastructure. In addition, the efficacy of either single- or combined treatments were assessed in an acute TgShSp1-oocyst infection model based on CD1 outbred mice. Whereas the IC50 of the compounds in combination (42 nM) was close to the IC50 of BKI-1748 alone (46 nM) and half of the IC50 of artemisone alone (92 nM), the IC90 of the combination was half of the values found with the single compounds (138 nM vs. ca. 270 nM). Another indication for synergistic effects in vitro were distinct alterations of the cellular ultrastructure of tachyzoites observed in combination, but not with the single compounds. These promising results could not be reproduced in vivo. There was no decrease in number of T. gondii positive brains by either treatment. However, the levels of infection in these brains, i. e. the number of tachyzoites, was significantly decreased upon BKI-1748 treatment alone, and the combination with artemiside did not produce any further decrease. The treatment with artemiside alone had no significant effects. A vertical transmission model could not be established since artemiside strongly interfered with pregnancy and caused abortion. These results show that is difficult to extrapolate from promising in vitro results to the situation in vivo.


Asunto(s)
Antineoplásicos , Artemisininas , Toxoplasma , Toxoplasmosis , Embarazo , Femenino , Ratones , Humanos , Animales , Toxoplasmosis/tratamiento farmacológico , Artemisininas/farmacología , Artemisininas/uso terapéutico , Transmisión Vertical de Enfermedad Infecciosa , Antineoplásicos/farmacología
18.
J Oral Pathol Med ; 52(9): 849-859, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37573872

RESUMEN

BACKGROUND: Translationally controlled tumour protein (TCTP) is a multifunctional protein elevated in multiple cancers. However, studies on its role in oral carcinogenesis and prognosis are rare. We recently reported the role of its interacting partner, MCL1, in oral cancer progression and outcome. Hence, the present study aimed to assess TCTP expression in oral tumorigenesis and its association with patient outcomes alone and in combination with MCL1. METHODS: TCTP expression was assessed by immunohistochemistry and immunoblotting in oral tissues and cells, respectively. Cell viability post siRNA/dihydroartemisinin treatment was analysed by tetrazolium salt assay. Cell survival, invasion and tumorigenic potential post TCTP knockdown were assessed by clonogenic, Matrigel and soft-agar assays, respectively. The association of TCTP with patient outcome was analysed by Kaplan-Meier and Cox regression. RESULTS: TCTP was significantly overexpressed in oral premalignant lesions (p < 0.0001), oral tumours (p < 0.0001) and oral dysplastic and cancer cells versus normal oral mucosa and also in recurrent (p < 0.05) versus non-recurrent oral tumours. Further, elevated TCTP was significantly (p < 0.05) associated with poor recurrence free survival (RFS) and poor overall survival (OS; hazard ratio = 2.29; p < 0.05). Intriguingly, the high co-expression of TCTP and MCL1 further reduced the RFS (p < 0.05) and OS (p < 0.05; hazard-ratio = 3.49; p < 0.05). Additionally, TCTP knockdown decreased survival (p < 0.05), invasion (p < 0.01) and in vitro tumorigenic potential (p < 0.0001). Dihydroartemisinin treatment reduced TCTP levels and viability of oral cancer cells. CONCLUSION: Our studies demonstrate an oncogenic role of TCTP in oral cancer progression and poor outcome. Thus, TCTP may be a potential prognostic marker and therapeutic target in oral cancers.


Asunto(s)
Artemisininas , Neoplasias de la Boca , Humanos , Artemisininas/farmacología , Artemisininas/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Boca/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteína Tumoral Controlada Traslacionalmente 1
19.
J Med Chem ; 66(16): 11335-11350, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37552639

RESUMEN

Artemisinin, a prominent anti-malaria drug, is being investigated for its potential as a repurposed cancer treatment. However, its effectiveness in tumor cell lines remains limited, and its mechanism of action is unclear. To make more progress, the PROteolysis-TArgeting chimera (PROTAC) technique has been applied to design and synthesize novel artemisinin derivatives in this study. Among them, AD4, the most potent compound, exhibited an IC50 value of 50.6 nM in RS4;11 cells, over 12-fold better than that of its parent compound, SM1044. This was supported by prolonged survival of RS4;11-transplanted NOD/SCID mice. Meanwhile, AD4 effectively degraded PCLAF in RS4;11 cells and thus activated the p21/Rb axis to exert antitumor activity by directly targeting PCLAF. The discovery of AD4 highlights the great potential of using PROTACs to improve the efficacy of natural products, identify therapeutic targets, and facilitate drug repurposing. This opens a promising avenue for transforming other natural products into effective therapies.


Asunto(s)
Artemisininas , Quimera Dirigida a la Proteólisis , Animales , Ratones , Artemisininas/farmacología , Artemisininas/uso terapéutico , Reposicionamiento de Medicamentos , Ratones Endogámicos NOD , Ratones SCID , Proteolisis
20.
Phytochemistry ; 214: 113798, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37517615

RESUMEN

Plant-based drugs have been used for centuries for treating different ailments. Malaria, one of the prevalent threats in many parts of the world, is treated mainly by artemisinin-based drugs derived from plants of genus Artemisia. However, the distribution of artemisinin is restricted to a few species of the genus; besides, its yield depends on ontogeny and the plant's geographical location. Here, we review the studies focusing on biosynthesis and distributional pattern of artemisinin production in species of the genus Artemisia. We also discussed various agronomic and in vitro methods and molecular approaches to increase the yield of artemisinin. We have summarized different mechanisms of artemisinin involved in its anti-malarial, anti-cancer, anti-inflammatory and anti-viral activities (like against Covid-19). Overall the current review provides a synopsis of a global view of the distribution of artemisinin, its biosynthesis, and pharmacological potential in treating various diseases like malaria, cancer, and coronavirus, which may provoke future research efforts in drug development. Nevertheless, long-term trials and molecular approaches, like CRISPR-Cas, are required for in-depth research.


Asunto(s)
Antimaláricos , Artemisia annua , Artemisia , Artemisininas , COVID-19 , Malaria , Artemisininas/farmacología , Artemisininas/uso terapéutico , Antimaláricos/farmacología , Malaria/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA