Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
1.
Clin Transl Sci ; 16(9): 1725-1735, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37477356

RESUMEN

Monensin is an ionophore antibiotic that inhibits the growth of cancer cells. The aim of this study was to investigate the apoptosis-mediated anticarcinogenic effects of monensin in SH-SY5Y neuroblastoma cells. The effects of monensin on cell viability, invasion, migration, and colony formation were determined by XTT, matrigel-chamber, wound healing, and colony formation tests, respectively. The effects of monensin on apoptosis were determined by real-time polymerase chain reaction, TUNEL, Western blot, and Annexin V assay. We have shown that monensin suppresses neuroblastoma cell viability, invasion, migration, and colony formation. Moreover, we reported that monensin inhibits cell viability by triggering apoptosis of neuroblastoma cells. Monensin caused apoptosis by increasing caspase-3, 7, 8, and 9 expressions and decreasing Bax and Bcl-2 expressions in neuroblastoma cells. In Annexin V results, the rates of apoptotic cells were found to be 9.66 ± 0.01% (p < 0.001), 29.28 ± 0.88% (p < 0.01), and 62.55 ± 2.36% (p < 0.01) in the 8, 16, and 32 µM monensin groups, respectively. In TUNEL results, these values were, respectively; 35 ± 2% (p < 0.001), 34 ± 0.57% (p < 0.001), and 75 ± 2.51% (p < 0.001). Our results suggest that monensin may be a safe and effective therapeutic candidate for treating pediatric neuroblastoma.


Asunto(s)
Neuroblastoma , Humanos , Niño , Neuroblastoma/tratamiento farmacológico , Monensina/farmacología , Monensina/uso terapéutico , Anexina A5/farmacología , Anexina A5/uso terapéutico , Apoptosis , Línea Celular Tumoral , Proliferación Celular
2.
Artículo en Inglés | MEDLINE | ID: mdl-37247559

RESUMEN

Worldwide distributed coccidiosis is caused by infection of both Eimeria species and Cystoisospora in the host intestine and causes huge economic losses to the livestock industry, especially the poultry industry. The control of such diseases relies mainly on chemoprophylaxis with anticoccidials, which has led to a very common drug resistance in this field. However, the genetic mechanisms underlying resistance to many anticoccidial drugs remain unknown. In this study, strains of E. tenella resistant to 250 mg/kg monensin were generated and characterized. Forward genetic approaches based on pooled genome sequencing, including experimental evolution and linkage group selection, were used to locate candidate targets responsible for resistance to monensin and diclazuril in E. tenella. A total of 16 nonsynonymous mutants in protein-coding genes were identified in monensin-resistant strains, and two genomic regions with strong selection signals were also detected in diclazuril-resistant strains. Our study reveals the genetic characterization of the experimental evolution and linkage group selection in Eimeria species, and also provides important information that contributes to the understanding of the molecular mechanism of drug resistance in coccidia.


Asunto(s)
Coccidiosis , Coccidiostáticos , Eimeria tenella , Eimeria , Enfermedades de las Aves de Corral , Animales , Monensina/uso terapéutico , Eimeria tenella/genética , Coccidiostáticos/farmacología , Coccidiostáticos/uso terapéutico , Pollos , Enfermedades de las Aves de Corral/tratamiento farmacológico , Enfermedades de las Aves de Corral/prevención & control , Coccidiosis/tratamiento farmacológico , Coccidiosis/veterinaria
3.
Commun Biol ; 6(1): 574, 2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37248274

RESUMEN

Pharmacological treatments for advanced hepatocellular carcinoma (HCC) have a partial efficacy. Augmented Na+ content and water retention are observed in human cancers and offer unexplored targets for anticancer therapies. Na+ levels are evaluated upon treatments with the antibiotic cation ionophore Monensin by fluorimetry, ICP-MS, 23Na-MRI, NMR relaxometry, confocal or time-lapse analysis related to energy production, water fluxes and cell death, employing both murine and human HCC cell lines, primary murine hepatocytes, or HCC allografts in NSG mice. Na+ levels of HCC cells and tissue are 8-10 times higher than that of healthy hepatocytes and livers. Monensin further increases Na+ levels in HCC cells and in HCC allografts but not in primary hepatocytes and in normal hepatic and extrahepatic tissue. The Na+ increase is associated with energy depletion, mitochondrial Na+ load and inhibition of O2 consumption. The Na+ increase causes an enhancement of the intracellular water lifetime and death of HCC cells, and a regression and necrosis of allograft tumors, without affecting the proliferating activity of either HCCs or healthy tissues. These observations indicate that HCC cells are, unlike healthy cells, energetically incapable of compensating and surviving a pharmacologically induced Na+ load, highlighting Na+ homeostasis as druggable target for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ratones , Humanos , Animales , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Sodio/metabolismo , Monensina/uso terapéutico , Línea Celular , Agua
4.
Immunopharmacol Immunotoxicol ; 45(1): 35-42, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36043455

RESUMEN

OBJECTIVE: Uveal melanoma (UM) is the common primary cancer of the eye and new treatments are needed. Substantial evidence has shown that an antibiotic monensin is an attractive candidate for the development of anti-cancer drug. In this study, we investigated the potential of repositioning monensin for the treatment of UM in the pre-clinical setting. MATERIALS AND METHODS: Cellular activity assays were performed using multiple cell lines representing UM models with different cellular origins and genetic profiling and normal cells as control. Combination studies were performed using Chou-Talalay method. Mechanism studies were performed using immunoblotting and ELISA. RESULTS: Monensin was effective against all tested UM cell lines and less effective against normal fibroblast cells. Monensin induced G0/G1 arrest and thus decreased S phase, leading to UM cell growth inhibition. It also inhibited migration and induced apoptosis in UM cells. In addition, the combination of monensin and dacarbazine was synergistic in targeting UM cells. Our mechanistic studies showed that monensin specifically decreased activity of RhoA without affecting other small GTPases, such as Ras and Rac1. Consistently, monensin decreased phosphorylation of downstream effectors of RhoA signaling, including ROCK, MYPT1 and MLC. Rescue studies using RhoA activator calpeptin showed that calpeptin significantly abolished the inhibitory effects of monensin on RhoA activity, proliferation, migration and survival, confirming that RhoA is the target of monensin in UM cells. CONCLUSIONS: Our study demonstrates that monensin is a potent inhibitor of UM and synergizes with chemotherapy, via suppressing RhoA activity and RhoA-mediated signaling. Our findings suggest that monensin may be a potential lead compound for further development into a drug for UM treatment.


Asunto(s)
Melanoma , Neoplasias de la Úvea , Humanos , Monensina/farmacología , Monensina/uso terapéutico , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo , Apoptosis , Proliferación Celular , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/farmacología , Proteína de Unión al GTP rhoA/uso terapéutico
5.
Biomed Pharmacother ; 153: 113440, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076555

RESUMEN

Glioblastoma (GBM) remains the most frequently diagnosed primary malignant brain cancer in adults. Despite recent progress in understanding the biology of GBM, the clinical outcome for patients remains poor, with a median survival of approximately one year after diagnosis. One factor contributing to failure in clinical trials is the fact that traditional models used in GBM drug discovery poorly recapitulate patient tumors. Previous studies have shown that monensin (MON) analogs, namely esters and amides on C-26 were potent towards various types of cancer cell lines. In the present study we have investigated the activity of these molecules in GBM organoids, as well as in a host:tumor organoid model. Using a mini-ring cell viability assay we have identified seven analogs (IC50 = 91.5 ± 54.4-291.7 ± 68.8 nM) more potent than parent MON (IC50 = 612.6 ± 184.4 nM). Five of these compounds induced substantial DNA fragmentation in GBM organoids, suggestive of apoptotic cell death. The most active analog, compound 1, significantly reduced GBM cell migration, induced PARP degradation, diminished phosphorylation of STAT3, Akt and GSK3ß, increased É£H2AX signaling and upregulated expression of the autophagy associated marker LC3-II. To investigate the activity of MON and compound 1 in a tumor microenvironment, we developed human cerebral organoids (COs) from human induced pluripotent stem cells (iPSCs). The COs showed features of early developing brain such as multiple neural rosettes with a proliferative zone of neural stem cells (Nestin+), neurons (TUJ1 +), primitive ventricular system (SOX2 +/Ki67 +), intermediate zone (TBR2 +) and cortical plate (MAP2 +). In order to generate host:tumor organoids, we co-cultured RFP-labeled U87MG cells with fully formed COs. Compound 1 and MON reduced U87MG tumor size in the COs after four days of treatment and induced a significant reduction of PARP expression. These findings highlight the therapeutic potential of MON analogs towards GBM and support the application of organoid models in anti-cancer drug discovery.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células Madre Pluripotentes Inducidas , Adulto , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Monensina/farmacología , Monensina/uso terapéutico , Organoides/metabolismo , Organoides/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Microambiente Tumoral
6.
Anticancer Agents Med Chem ; 22(14): 2539-2547, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35168524

RESUMEN

OBJECTIVE: The clinical management of anaplastic thyroid cancer (ATC) remains challenging, and novel treatment methods are needed. Monensin is a carboxyl polyether ionophore that potently inhibits the growth of various cancer types. Our current work investigates whether monensin has selective anti-ATC activity and systematically explores its underlying mechanisms. METHODS: Proliferation and apoptosis assays were performed using a panel of thyroid cancer cell lines. Mitochondrial biogenesis profiles, ATP levels, oxidative stress, AMPK, and mTOR were examined in these cells after monensin treatment. RESULTS: Monensin is effective in inhibiting proliferation and inducing apoptosis in a number of thyroid cancer cell lines. The results are consistent across cell lines of varying cellular origins and genetic mutations. Compared to other thyroid cancer cell types, ATC cell lines are the most sensitive to monensin. Of note, monensin used at our experimental concentration affects less of normal cells. Mechanistic studies reveal that monensin acts on ATC cells by disrupting mitochondrial function, inducing oxidative stress and damage, and AMPK activation-induced mTOR inhibition. We further show that mitochondrial respiration is a critical target for monensin in ATC cells. CONCLUSIONS: Our pre-clinical findings demonstrate the selective anti-ATC activities of monensin. This is supported by increasing evidence that monensin can be repurposed as a potential anti-cancer drug.


Asunto(s)
Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides , Proteínas Quinasas Activadas por AMP , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Mitocondrias , Monensina/farmacología , Monensina/uso terapéutico , Respiración , Serina-Treonina Quinasas TOR , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo
7.
Haematologica ; 107(6): 1438-1447, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34706495

RESUMEN

Sickle cell disease (SCD) is characterized by sickle hemoglobin (HbS) which polymerizes under deoxygenated conditions to form a stiff, sickled erythrocyte. The dehydration of sickle erythrocytes increases intracellular HbS concentration and the propensity of erythrocyte sickling. Prevention of this mechanism may provide a target for potential SCD therapy investigation. Ionophores such as monensin can increase erythrocyte sodium permeability by facilitating its transmembrane transport, leading to osmotic swelling of the erythrocyte and decreased hemoglobin concentration. In this study, we treated 13 blood samples from patients with SCD with 10 nM of monensin ex vivo. We measured changes in cell volume and hemoglobin concentration in response to monensin treatment, and we perfused treated blood samples through a microfluidic device that permits quantification of blood flow under controlled hypoxia. Monensin treatment led to increases in cell volume and reductions in hemoglobin concentration in most blood samples, though the degree of response varied across samples. Monensin-treated samples also demonstrated reduced blood flow impairment under hypoxic conditions relative to untreated controls. Moreover, there was a significant correlation between the improvement in blood flow and the decrease in hemoglobin concentration. Thus, our results demonstrate that a reduction in intracellular HbS concentration by osmotic swelling improves blood flow under hypoxic conditions. Although the toxicity of monensin will likely prevent it from being a viable clinical treatment, these results suggest that osmotic swelling should be investigated further as a potential mechanism for SCD therapy.


Asunto(s)
Anemia de Células Falciformes , Eritrocitos , Ionóforos , Monensina , Anemia de Células Falciformes/tratamiento farmacológico , Eritrocitos/efectos de los fármacos , Hemoglobina Falciforme , Humanos , Hipoxia , Ionóforos/farmacología , Ionóforos/uso terapéutico , Monensina/farmacología , Monensina/uso terapéutico
8.
Front Immunol ; 11: 586572, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324406

RESUMEN

COVID-19 pandemic has infected millions of people with mortality exceeding >1 million. There is an urgent need to find therapeutic agents that can help clear the virus to prevent severe disease and death. Identifying effective and safer drugs can provide more options to treat COVID-19 infections either alone or in combination. Here, we performed a high throughput screening of approximately 1,700 US FDA-approved compounds to identify novel therapeutic agents that can effectively inhibit replication of coronaviruses including SARS-CoV-2. Our two-step screen first used a human coronavirus strain OC43 to identify compounds with anti-coronaviral activities. The effective compounds were then screened for their effectiveness in inhibiting SARS-CoV-2. These screens have identified 20 anti-SARS-CoV-2 drugs including previously reported compounds such as hydroxychloroquine, amlodipine besylate, arbidol hydrochloride, tilorone 2HCl, dronedarone hydrochloride, mefloquine, and thioridazine hydrochloride. Five of the newly identified drugs had a safety index (cytotoxic/effective concentration) of >600, indicating a wide therapeutic window compared to hydroxychloroquine which had a safety index of 22 in similar experiments. Mechanistically, five of the effective compounds (fendiline HCl, monensin sodium salt, vortioxetine, sertraline HCl, and salifungin) were found to block SARS-CoV-2 S protein-mediated cell fusion. These FDA-approved compounds can provide much needed therapeutic options that we urgently need during the midst of the pandemic.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ensayos Analíticos de Alto Rendimiento/métodos , Pandemias/prevención & control , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/epidemiología , COVID-19/virología , Línea Celular , Reposicionamiento de Medicamentos/métodos , Fendilina/uso terapéutico , Células HEK293 , Humanos , Monensina/uso terapéutico , SARS-CoV-2/fisiología , Salicilanilidas/uso terapéutico , Sertralina/uso terapéutico , Vortioxetina/uso terapéutico
9.
Biochem Biophys Res Commun ; 530(2): 479-484, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32595038

RESUMEN

Glioblastoma is characterized by the extensive vascularization with poor prognosis. Targeting both tumor cell and angiogenesis may present an effective therapeutic strategy for glioblastoma. Monensin, a polyether ionophore antibiotic, has been recently recognized as promising anticancer drug candidate due to its potent and selective anti-tumor activities. However, little is known on the effects of monensin on tumor angiogenesis. In this work, we investigated the effects and underlying mechanisms of monensin on glioblastoma angiogenesis and growth. We show that monensin at nanomolar concentrations inhibits early stages of capillary network formation of glioblastoma endothelial cell. Monensin inhibited multiple endothelial cellular events, including migration, growth and survival, without affecting adhesion to Matrigel. We further demonstrate that monensin acts on endothelial cells via suppressing VEGFR- and EGFR-mediated signaling pathways. Monensin also inhibits proliferation and induces apoptosis in a panel of glioblastoma cells. However, monensin is more effective in targeting endothelial cells than tumor cells. Using glioblastoma growth xenograft mice model, we show that monensin at tolerable dose effectively inhibits glioblastoma growth. Of note, there is a significant decreased tumor vascularization from monensin-treated mice. Our work clearly demonstrates the anti-angiogenic activity of monensin and its ability in suppressing multiple tyrosine kinase receptor-mediated pathways. Our findings suggest that is a useful addition to the treatment armamentarium for glioblastoma.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Monensina/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Receptores de Factores de Crecimiento/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Glioblastoma/irrigación sanguínea , Glioblastoma/metabolismo , Humanos , Ratones Desnudos , Monensina/farmacología , Neovascularización Patológica/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Nat Commun ; 11(1): 409, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964869

RESUMEN

The Golgi is a dynamic organelle whose correct assembly is crucial for cellular homeostasis. Perturbations in Golgi structure are associated with numerous disorders from neurodegeneration to cancer. However, whether and how dispersal of the Golgi apparatus is actively regulated under stress, and the consequences of Golgi dispersal, remain unknown. Here we demonstrate that 26S proteasomes are associated with the cytosolic surface of Golgi membranes to facilitate Golgi Apparatus-Related Degradation (GARD) and degradation of GM130 in response to Golgi stress. The degradation of GM130 is dependent on p97/VCP and 26S proteasomes, and required for Golgi dispersal. Finally, we show that perturbation of Golgi homeostasis induces cell death of multiple myeloma in vitro and in vivo, offering a therapeutic strategy for this malignancy. Taken together, this work reveals a mechanism of Golgi-localized proteasomal degradation, providing a functional link between proteostasis control and Golgi architecture, which may be critical in various secretion-related pathologies.


Asunto(s)
Aparato de Golgi/metabolismo , Ionóforos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteostasis/fisiología , Animales , Apoptosis/efectos de los fármacos , Autoantígenos/metabolismo , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Aparato de Golgi/efectos de los fármacos , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Ionóforos/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Monensina/farmacología , Monensina/uso terapéutico , Mieloma Múltiple/patología , Proteolisis/efectos de los fármacos , Proteostasis/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Proteína que Contiene Valosina/metabolismo
11.
J Dairy Sci ; 102(10): 9097-9106, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31400899

RESUMEN

Mycobacterium avium ssp. paratuberculosis (MAP) is the causative agent of Johne's disease, an enteric infection of ruminants that causes significant economic burden for dairy and beef producers. Efforts to control MAP in endemic herds typically focus on herd management practices such as limiting exposure or early culling of infected animals and, occasionally, vaccination. The ionophore monensin sodium may have protective effects against MAP both in vivo and in vitro; however, this has not been thoroughly evaluated experimentally. Using a direct intestinal MAP challenge model, we have observed similarities regarding persistence of MAP in tissues and apparent resilience to infection compared with experimental oral infection or natural disease. Here we sought to investigate the effects of oral monensin supplementation in experimentally MAP-infected calves. We examined the persistence of MAP in the intestinal tissues, MAP-induced intestinal inflammation, fecal MAP shedding, and seroconversion using a commercial serologic assay. Monensin-supplemented MAP-infected calves demonstrated evidence for resilience to MAP infection earlier in this study compared with monensin-free MAP-infected calves. However, statistical modeling did not identify a significant effect of monensin on outcomes of infection, and more work is required to understand how monensin affects early tissue colonization of MAP in calves.


Asunto(s)
Antibacterianos/uso terapéutico , Enfermedades de los Bovinos/tratamiento farmacológico , Monensina/uso terapéutico , Paratuberculosis/tratamiento farmacológico , Administración Oral , Animales , Bovinos , Modelos Animales de Enfermedad , Heces/microbiología , Masculino , Monensina/administración & dosificación , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/microbiología
12.
Bioorg Med Chem Lett ; 29(13): 1549-1554, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31054863

RESUMEN

Chemotherapy is one of the standard methods for the treatment of malignant tumors. It aims to cause lethal damage to cellular structures, mainly DNA. Noteworthy, in recent years discoveries of novel anticancer agents from well-known antibiotics have opened up new treatment pathways for several cancer diseases. The aim of this review article is to describe new applications for the following antibiotics: doxycycline (DOX), salinomycin (SAL), monensin (MON) and ivermectin (IVR) as they are known to show anti-tumor activity, but have not yet been introduced into standard oncological therapy. To date, these agents have been used for the treatment of a broad-spectrum of bacterial and parasitic infectious diseases and are widely available, which is why they were selected. The data presented here clearly show that the antibiotics mentioned above should be recognised in the near future as novel agents able to eradicate cancer cells and cancer stem cells (CSCs) across several cancer types.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxiciclina/uso terapéutico , Ivermectina/uso terapéutico , Monensina/uso terapéutico , Neoplasias/tratamiento farmacológico , Piranos/uso terapéutico , Antineoplásicos/farmacología , Doxiciclina/farmacología , Humanos , Ivermectina/farmacología , Monensina/farmacología , Piranos/farmacología
13.
J Vet Pharmacol Ther ; 41(5): 746-754, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29971788

RESUMEN

Increasing reports of multidrug-resistant bacterial infections in animals has created a need for novel antimicrobial agents that do not promote cross-resistance to critically important antimicrobial classes used in human medicine. In response to the recent emergence of antimicrobial resistance in several bovine mastitis pathogens, in vitro antimicrobial susceptibility was determined for four polyether ionophores (lasalocid, monensin, narasin and salinomycin) against Staphylococcus spp. and Streptococcus spp. isolated from clinical cases. In addition, erythrocyte haemolysis and WST-1 cell proliferation assays were used to assess in vitro mammalian cell cytotoxicity and biofilm susceptibility testing was performed using the minimum biofilm eradication concentration (MBEC™) biofilm assay. Lasalocid, monensin, narasin and salinomycin exhibited bacteriostatic antimicrobial activity against all pathogens tested, including methicillin-resistant staphylococci, with MIC90 values <16 µg/ml. Narasin and monensin displayed the least toxicity against mammalian cell lines and all compounds significantly reduced viable cell numbers in a Staphylococcus aureus biofilm. Based on in vitro characterization, all four ionophores offer potentially novel treatments against bovine mastitis but in vivo studies will be essential to determine whether acceptable safety and efficacy is present following intramammary administration.


Asunto(s)
Antibacterianos/uso terapéutico , Bacterias Grampositivas/efectos de los fármacos , Ionóforos/uso terapéutico , Mastitis Bovina/tratamiento farmacológico , Animales , Biopelículas/efectos de los fármacos , Bovinos , Femenino , Lasalocido/uso terapéutico , Mastitis Bovina/microbiología , Pruebas de Sensibilidad Microbiana/veterinaria , Monensina/uso terapéutico , Piranos/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/veterinaria , Staphylococcus/efectos de los fármacos , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/veterinaria , Streptococcus/efectos de los fármacos
14.
Biomed Pharmacother ; 99: 384-390, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29367107

RESUMEN

Salinomycin (SAL) and monensin (MON) are polyether ionophore antibiotics commonly used in veterinary medicine. They are known from their anti-cancer activity against various types of cancer cells, including those that display multi-drug resistance as well as cancer stem cells. In order to increase the biological activity profile and reduce toxicity against normal cells, while retaining the activities in the micromolar range, a library of ester and amide derivatives of SAL was synthesized and previously reported. In this paper, we examined the activity of SAL, its ten derivatives, and MON on primary acute lymphoblastic leukemia cells. MON and six SAL derivatives were more potent than SAL in cell viability assays. Further, selected active SAL analogs induced characteristics of apoptotic cell death and increased expression of p53. Moreover, SAL acted synergistically with the Bcl-2 inhibitor ABT-263, whereas 2,2,2-trifluoroethyl ester, the most active analog of SAL, antagonized ABT-263, suggesting possible differences in molecular mechanism.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Piranos/uso terapéutico , Amidas/química , Amidas/farmacología , Amidas/uso terapéutico , Compuestos de Anilina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Ésteres/química , Ésteres/farmacología , Ésteres/uso terapéutico , Humanos , Monensina/farmacología , Monensina/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piranos/química , Piranos/farmacología , Sulfonamidas/farmacología , Proteína p53 Supresora de Tumor/metabolismo
15.
Arch Toxicol ; 91(12): 3787-3797, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29063135

RESUMEN

This article reviews the clinical use of the metal chelators sodium 2,3-dimercapto-1-propanesulfonate (DMPS), meso-2,3-dimercaptosuccinic acid (DMSA), and calcium disodium edetate (CaEDTA, calcium EDTA) in overexposure and poisonings with salts of lead (Pb), mercury (Hg), and arsenic (As). DMSA has considerably lower toxicity than the classic heavy metal antagonist BAL (2,3-dimercaptopropanol) and is also less toxic than DMPS. Because of its adverse effects, CaEDTA should be replaced by DMSA as the antidote of choice in treating moderate Pb poisoning. Combination therapy with BAL and CaEDTA was previously recommended in cases of severe acute Pb poisoning with encephalopathy. We suggest that BAL in such cases acted as a shuttling Pb transporter from the intra- to the extracellular space. The present paper discusses if a combination of the extracellularly distributed DMSA with the ionophore, Monensin may provide a less toxic combination for Pb mobilization by increasing both the efflux of intracellularly deposited Pb and the urinary Pb excretion. Anyhow, oral therapy with DMSA should be continued with several intermittent courses. DMPS and DMSA are also promising antidotes in Hg poisoning, whereas DMPS seems to be a more efficient agent against As poisoning. However, new insight indicates that a combination of low-dosed BAL plus DMPS could be a preferred antidotal therapy to obtain mobilization of the intracerebral deposits into the circulation for subsequent rapid urinary excretion.


Asunto(s)
Intoxicación por Arsénico/tratamiento farmacológico , Quelantes/uso terapéutico , Intoxicación del Sistema Nervioso por Plomo/tratamiento farmacológico , Intoxicación del Sistema Nervioso por Mercurio/tratamiento farmacológico , Ácido Edético/uso terapéutico , Humanos , Monensina/uso terapéutico , Succímero/uso terapéutico , Unitiol/uso terapéutico
16.
Can Vet J ; 57(7): 733-40, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27429461

RESUMEN

A partial budget model was developed to evaluate the economic value of Rumensin Controlled Release Capsule (CRC) boluses when administered before calving to reduce disease and increase milk production. After accounting for disease incidences in a herd and the percentage by which Rumensin CRC can reduce them, and the increase in milk production attributable to administration of Rumensin CRC, the return on investment (ROI) per lactation was 4:1. Another partial budget model was developed to estimate the economic value of propylene glycol (PG) to treat ketosis when diagnosed by 3 different cow-side tests or when administered to all cows without using any cow-side testing. After accounting for the sensitivity and specificity of each test, ROI per lactation ranged from 2:1 to 4:1. The ROI was 2:1 when no cow-side testing was used. In conclusion, prevention of diseases that occur in the postpartum period and treatment of ketosis after calving yielded a positive ROI that varies based on disease incidence and method of diagnosis.


Valeur économique des ionophores et du propylèneglycol pour prévenir la maladie et traiter l'acétonémie au Canada. Un modèle de budget partiel a été développé pour évaluer la valeur économique des bolus de capsules à libération contrôlée (CLC) de Rumensin lors de l'administration avant le vêlage afin de réduire les maladies et d'accroître la production de lait. Après avoir tenu compte de l'incidence des maladies dans un troupeau et du pourcentage par lequel la CLC de Rumensin peut les réduire et de l'augmentation de la production de lait attribuable à l'administration de la CLC de Rumensin, le rendement du capital investi (RCI) par lactation était de 4:1. Un autre modèle de budget partiel a été développé pour estimer la valeur économique du propylèneglycol (PG) afin de traiter l'acétonémie lors du diagnostic par 3 tests différents pour les vaches ou lors de l'administration à toutes les vaches sans le recours à des tests auprès des vaches. Après avoir tenu compte de la sensibilité et de la spécificité de chaque test, le RCI par lactation s'échelonnait de 2:1 à 4:1. Le RCI était de 2:1 lorsqu'aucun test auprès des vaches n'était utilisé. En conclusion, la prévention des maladies qui se produit dans la période postpartum et le traitement de l'acétonémie après le vêlage a donné un RCI positif qui varie selon l'incidence de maladies et la méthode de diagnostic.(Traduit par Isabelle Vallières).


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Cetosis/veterinaria , Propilenglicol/uso terapéutico , Animales , Canadá , Bovinos , Enfermedades de los Bovinos/tratamiento farmacológico , Enfermedades de los Bovinos/economía , Industria Lechera/economía , Preparaciones de Acción Retardada/uso terapéutico , Femenino , Ionóforos , Cetosis/tratamiento farmacológico , Cetosis/economía , Modelos Económicos , Monensina/administración & dosificación , Monensina/uso terapéutico , Embarazo , Complicaciones del Embarazo/economía , Complicaciones del Embarazo/prevención & control , Complicaciones del Embarazo/veterinaria , Propilenglicol/administración & dosificación
17.
Exp Parasitol ; 141: 129-33, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24703975

RESUMEN

Coccidiosis is the most important parasitic disease of the poultry production industry. Due to increasing resistance to conventional anticoccidial agents, it is necessary to find new anticoccidial compounds. Herbal compounds such as those from Artemisia species are promising weapons in this regard since preliminary studies have shown its anticoccidial effects. To compare the anticoccidial effect of a granulated extract of Artemisia sieberi (GEAS) versus monensin in experimental broiler coccidiosis, 120 one-day old Ross 308 broiler chickens were divided in four groups, each with three replicates (n=10). Group 1 was separated as an uninfected negative control and received no treatment. At 21days of age, groups 2, 3 and 4 were inoculated with a mixed suspension of 2×10(5) oocysts of Eimeria tenella, Eimeria maxima, Eimeria acervulina and Eimeria necatrix. Group 2 was maintained as an infected positive control and received no treatment while groups 3 and 4 received GEAS (5mg/kg feed), and monensin (110mg/kg feed) from the first day until 42days of age as a feed additive, respectively. Five days after inoculation, the number of oocycts per gram (OPG) of feces for 7 successive days was measured. Also, mean body weight (MBW), weight gain (WG), feed intake (FI) and feed conversion ratio (FCR) were determined weekly in all groups and replicates. The results showed that GEAS and monensin improved performance attributes (FI, MBW, WG, FCR) and significantly (P<0.05) decreased OPG in inoculated broiler chickens.


Asunto(s)
Artemisia/química , Pollos/parasitología , Coccidiosis/veterinaria , Coccidiostáticos/uso terapéutico , Extractos Vegetales/uso terapéutico , Enfermedades de las Aves de Corral/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Coccidiosis/tratamiento farmacológico , Coccidiosis/parasitología , Coccidiostáticos/farmacología , Ingestión de Alimentos/efectos de los fármacos , Heces/parasitología , Monensina/farmacología , Monensina/uso terapéutico , Oocistos/efectos de los fármacos , Recuento de Huevos de Parásitos/veterinaria , Extractos Vegetales/farmacología , Enfermedades de las Aves de Corral/parasitología
18.
Mol Cancer Ther ; 13(4): 812-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24552772

RESUMEN

The Wnt signaling pathway is required during embryonic development and for the maintenance of homeostasis in adult tissues. However, aberrant activation of the pathway is implicated in a number of human disorders, including cancer of the gastrointestinal tract, breast, liver, melanoma, and hematologic malignancies. In this study, we identified monensin, a polyether ionophore antibiotic, as a potent inhibitor of Wnt signaling. The inhibitory effect of monensin on the Wnt/ß-catenin signaling cascade was observed in mammalian cells stimulated with Wnt ligands, glycogen synthase kinase-3 inhibitors, and in cells transfected with ß-catenin expression constructs. Furthermore, monensin suppressed the Wnt-dependent tail fin regeneration in zebrafish and Wnt- or ß-catenin-induced formation of secondary body axis in Xenopus embryos. In Wnt3a-activated HEK293 cells, monensin blocked the phoshorylation of Wnt coreceptor low-density lipoprotein receptor related protein 6 and promoted its degradation. In human colorectal carcinoma cells displaying deregulated Wnt signaling, monensin reduced the intracellular levels of ß-catenin. The reduction attenuated the expression of Wnt signaling target genes such as cyclin D1 and SP5 and decreased the cell proliferation rate. In multiple intestinal neoplasia (Min) mice, daily administration of monensin suppressed progression of the intestinal tumors without any sign of toxicity on normal mucosa. Our data suggest monensin as a prospective anticancer drug for therapy of neoplasia with deregulated Wnt signaling.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Monensina/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Monensina/uso terapéutico , Neoplasias Experimentales , Ensayos Antitumor por Modelo de Xenoinjerto , Xenopus , Pez Cebra , beta Catenina/metabolismo
19.
J Anim Sci ; 91(5): 2219-28, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23422007

RESUMEN

A total of 3,632 crossbred steers were used in 3 separate randomized complete-block designed finishing experiments. Data from Exp. 1 were analyzed separately whereas data were combined for Exp. 2 and 3, based on corn processing method used [Exp.1 = equal combination of dry-rolled and high-moisture (DRC:HMC); Exp. 2 and 3 = steam-flaked (SFC)]. Steers were fed 1 of 5 treatments to evaluate the effects of monensin and tylosin in feedlot diets containing 25% corn wet distillers grains with solubles (WDGS; DM basis). Treatments included: 1) corn-based diet (no WDGS) with 360 mg/d monensin and 90 mg/d tylosin (CORN+MT), 2) 25% wet distillers grains with solubles (WDGS-CON), 3) 25% WDGS with 360 mg/d monensin (WDGS+M), 4) WDGS with monensin and tylosin at same levels as treatment 1, and 5) WDGS with 480 mg/d monensin and 90 mg/d tylosin (WDGS+HIMT). In Exp. 1, WDGS+MT increased (P < 0.01) ADG (6.5%), G:F (6.9%), and HCW (3.0%) compared with CORN+MT. Alternatively, in Exp. 2 and 3, WDGS+MT had no effect on ADG (P = 0.18), decreased (P < 0.01) G:F by 3.2%, and did not affect HCW (P = 0.57) compared with CORN+MT. In Exp. 1, addition of monensin to a WDGS diet increased G:F by 3.1% (P = 0.03) and tended to increase G:F in Exp. 2 and 3 (P = 0.09) compared with WDGS-CON. For all experiments, addition of monensin and tylosin in a diet containing WDGS increased G:F (P < 0.01) and reduced total and severe liver abscesses (P < 0.01) compared with WDGS-CON. Additionally, HCW was increased in Exp. 2 and 3 (P < 0.01) and tended to increase in Exp. 1 (P = 0.09) when monensin and tylosin were fed in a WDGS diet compared with WDGS-CON. Feeding WDGS+MT reduced total liver abscesses by 79.3% (Exp. 1) and 57.6% (Exp. 2 and 3) compared with WDGS-CON. Compared with WDGS+MT, minimal differences were observed when monensin was fed at 480 mg/steer daily with tylosin in diets containing WDGS. Inclusion of WDGS in finishing steer diets did not alter effectiveness of monensin with tylosin in improving G:F and reducing prevalence of liver abscesses, regardless of corn processing method. Corn processing method appears to be an important factor in performance response to WDGS with feeding value of WDGS, measured as an increase in feed efficiency, being greater in a DRC:HMC-based diet compared with a SFC-based diet.


Asunto(s)
Antiinfecciosos/uso terapéutico , Composición Corporal/efectos de los fármacos , Enfermedades de los Bovinos/tratamiento farmacológico , Bovinos/fisiología , Absceso Hepático/veterinaria , Monensina/uso terapéutico , Tilosina/uso terapéutico , Alimentación Animal/análisis , Animales , Bovinos/crecimiento & desarrollo , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/microbiología , Colorado/epidemiología , Dieta/veterinaria , Absceso Hepático/tratamiento farmacológico , Absceso Hepático/epidemiología , Absceso Hepático/microbiología , Masculino , Prevalencia
20.
Br Poult Sci ; 53(3): 325-32, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22978588

RESUMEN

1. A herbal extract containing a blend of three essential oils, derived from oregano, laurel leaf and lavender, was investigated as a feed additive alternative to the conventional anticoccidial sodium monensin. 2. Broilers were infected with a mixture of Eimeria species or left uninfected. Both infected and uninfected broilers were provided with diets containing either herbal extract (HEX), monensin (MON) or without these supplements (CON). The HEX group had 50 mg herbal extract/kg diet and the MON group 100 mg monensin/kg diet. 3. All of the uninfected broilers exhibited higher body weight gain and better feed conversion when compared with their infected counterparts at d 28 and 42 of age. Both HEX and MON supplements caused significant improvements in performance in the infected broilers, but failed to have any effect on uninfected broilers. 4. Faecal oocyst output measured daily by sampling excreta, and expressed on a per bird basis, was lower in the HEX and MON groups than in the CON group. However, the herbal extract was not as effective as monensin in reducing oocyst excretion. Coccidial infection caused a significant increase in total intestinal length and caecal weight, but the dietary treatments did not influence these measurements. 5. These results indicate that providing a herbal extract in the diet was not as effective as monensin in protecting broilers exposed to a coccidial challenge.


Asunto(s)
Pollos , Coccidiosis/veterinaria , Coccidiostáticos/uso terapéutico , Suplementos Dietéticos/análisis , Eimeria/efectos de los fármacos , Aceites de Plantas/uso terapéutico , Enfermedades de las Aves de Corral/tratamiento farmacológico , Alimentación Animal/análisis , Animales , Coccidiosis/tratamiento farmacológico , Coccidiosis/parasitología , Coinfección/tratamiento farmacológico , Coinfección/parasitología , Coinfección/veterinaria , Heces/parasitología , Femenino , Laurus/química , Lavandula/química , Masculino , Monensina/uso terapéutico , Aceites Volátiles/uso terapéutico , Oocistos , Origanum/química , Enfermedades de las Aves de Corral/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...