Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Bioorg Chem ; 138: 106618, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37244231

RESUMEN

Triple-Negative Breast Cancer (TNBC) is found to be one of the life-threatening cancer. Poly (ADP-Ribose) Polymerase-1 (PARP-1) is overexpressed by those tumour cells, which become resistant to chemotherapies. Inhibition of PARP-1 has a considerable effect on treating TNBC. Prodigiosin is a valuable pharmaceutical compound that exhibits anticancer properties. The present study aims to virtually evaluate prodigiosin as a potent PARP-1 inhibitor using Molecular docking and Molecular Dynamics (MD) simulation studies. The PASS (Prediction of Activity Spectra for Substances) prediction tool evaluated the biological properties of prodigiosin. Then the drug-likeness and pharmacokinetic properties of prodigiosin were determined using Swiss-ADME software. It was suggested that prodigiosin obeyed Lipinski's rule of five and thus could act as a drug with good pharmacokinetic properties. Moreover, molecular docking was done with AutoDock 4.2 to identify the critical amino acids of the protein-ligand complex. It was indicated that prodigiosin has a docking score of -8.08 kcal/mol, which showed its effective interaction with crucial amino acid, His201A of PARP-1 protein. Further, MD simulation was performed using Gromacs software to validate the stability of the prodigiosin-PARP-1 complex. Prodigiosin was found to have good structural stability and affinity at the active site of PARP-1 protein. Additionally, PCA and MM-PBSA were calculated for the prodigiosin-PARP-1 complex, which revealed that prodigiosin has an excellent binding affinity towards PARP-1 protein. Prodigiosin can possibly be used as oral drug due to its PARP-1 inhibition through high binding affinity, structural stability, and receptor flexibility towards crucial amino acid residue His201A of PARP-1 protein. In-addition, in-vitro cytotoxicity, and apoptosis analysis of prodigiosin-treated TNBC cell line-MDA-MB-231 revealed that prodigiosin exhibited significant anticancer activity in 101.1 µg/mL concentration, when compared to commercially available synthetic drug cisplatin. Thus, prodigiosin could act as a potential candidate for treatment of TNBC than the commercially available synthetic drugs.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasa-1 , Neoplasias de la Mama Triple Negativas/metabolismo , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Simulación del Acoplamiento Molecular , Línea Celular Tumoral , Antineoplásicos/química
2.
J Physiol Pharmacol ; 74(1)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-37245232

RESUMEN

Prodigiosin (PRO) is a natural pigment that possesses multiple activities, covering anti-tumor, anti-bacteria, and immunosuppression. This study is committed to an investigation into the underlying function and the certain mechanism of PRO in acute lung damage followed by rheumatoid arthritis (RA). Cecal ligation and puncture (CLP) method was implemented to trigger a rat lung injury model, and a rat RA model was constructed with the help of rheumatoid arthritis induced by collagen. Prodigiosin was administered to intervene in the rats' lung tissues post-treatment. The expressions of pro-inflammatory cytokines (interleukin-1beta, interleukin-6, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1 were determined. Western blot was carried out to detect anti-surfactant protein A (SPA), anti-surfactant protein D (SPD), apoptosis-concerned proteins (Bax, cleaved-caspase-3, Bcl-2, and pro-caspase-3), the nuclear factor-kappaB (NF-κB)/nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3)/apoptosis-concerned speckle-like protein (ASC)/caspase-1 signaling pathway. The apoptosis of pulmonary epithelial tissues was checked via TUNEL assay, as corresponding kits were adopted to confirm the activity of lactate dehydrogenase (LDH) and the levels of oxidative stress markers malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px). Prodigiosin ameliorated the pathological damage of CLP rats. Prodigiosin alleviated the production of inflammatory and oxidative stress mediators. In the RA rats with acute lung injury, prodigiosin hampered apoptosis in the lung. Mechanistically, prodigiosin hinders the activation of the NF-κB/NLRP3 signaling axis. In conclusion: prodigiosin relieves acute lung injury in a rat model of rheumatoid arthritis by exerting anti-inflammatory and anti-oxidative effects through downregulating the NF-κB/NLRP3 signaling axis.


Asunto(s)
Lesión Pulmonar Aguda , Artritis Reumatoide , Ratas , Animales , FN-kappa B/metabolismo , Leucina , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dominio Pirina , Transducción de Señal , Lesión Pulmonar Aguda/tratamiento farmacológico , Nucleótidos
3.
Int Immunopharmacol ; 116: 109800, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36780827

RESUMEN

Prodigiosin (PG) is a secondary metabolite of microorganisms with anticancer, antimalarial, antibacterial and immunomodulatory effects. However, the modulatory effects on gut microbiome and intestinal immune microenvironment have never been explored in the ulcerative colitis (UC) mice model. In this study, 2.5% dextran sulfate sodium (DSS) induced UC mice model was constructed to investigate the effects of PG derived from a chromium-resistant Serratia sp. on the intestinal flora and inflammatory response. The results showed that prodigiosin administration attenuated the DSS-induced UC symptoms, including preventing the reduction of colonic length and DSS-induced mortality. Furthermore, prodigiosin ameliorated the DSS-induced gut microbiota community dysbiosis by restoring the abundance of Bacteroidota. At the genus level, the declined abundance of Bifidobacterium, Allobaculum and Akkermannia in UC mice was elevated by the treatment of PG. Pathological results by H&E staining showed that PG prevented the appearance of distortion and atrophy of crypt and neutrophil infiltration in a dose-dependent manner. RT-PCR revealed that the expression levels of the inflammatory factors IL-1ß, IL-6 and IL-10 were significantly suppressed, and the expression of the intestinal tight junction protein Claudin-1, Occludin and ZO-1 were upregulted in PG-treated UC mice. Conclusively, our results revealed that prodigiosin effectively prevented inflammatory response and protected intestinal barrier integrity of DSS-induced colitis mice via modulating gut microbiota community structure, suppressing inflammatory factors' expression, and accelerating the expression of intestinal tight junction protein. These results will provide new insights into the interaction of prodigiosin with intestinal microbiota homeostasis and its application in clinical against inflammatory bowel disease.


Asunto(s)
Colitis Ulcerosa , Colitis , Microbioma Gastrointestinal , Animales , Ratones , Prodigiosina/uso terapéutico , Sulfato de Dextran/farmacología , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Inflamación/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colon/patología , Proteínas de Uniones Estrechas/metabolismo , Homeostasis , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
4.
Probiotics Antimicrob Proteins ; 15(5): 1271-1286, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36030493

RESUMEN

Lactobacillus acidophilus ghosts (LAGs) with the unique safety of a probiotic, inherent tropism for colon cells, and multiple bioactivities offer promise as drug carriers for colon targeting. Our objective was to evaluate LAGs functionalized with prodigiosin (PG), apoptotic secondary bacterial metabolite, as a bioinspired formulation against colorectal cancer (CRC). LAGs were prepared by a chemical method and highly purified by density gradient centrifugation. LAGs were characterized by microscopic and staining techniques as relatively small-sized uniform vesicles (≈1.6 µm), nearly devoid of cytoplasmic and genetic materials and having a negatively charged intact envelope. PG was highly bound to LAGs envelope, generating a physiologically stable bioactive entity (PG-LAGs), as verified by multiple microscopic techniques and lack of PG release under physiological conditions. PG-LAGs were active against HCT116 CRC cells at both the cellular and molecular levels. Cell viability data highlighted the cytotoxicity of PG and LAGs and LAGs-induced enhancement of PG selectivity for HCT116 cells, anticipating dose reduction for PG and LAGs. Molecularly, expression of the apoptotic caspase 3 and P53 biomarkers in HCT116 intracellular proteins was significantly upregulated while that of the anti-apoptotic Bcl-2 (B-cell lymphoma 2) was downregulated by PG-LAGs relative to PG and 5-fluorouracil. PG-LAGs provide a novel bacteria-based combination for anticancer biomedicine.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Probióticos , Humanos , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Antineoplásicos/farmacología , Apoptosis , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología
5.
Environ Toxicol ; 38(2): 266-277, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36447373

RESUMEN

Prodigiosin (PDG) is a bacterial metabolite with numerous biological and pharmaceutical properties. Exposure to aluminium is considered a root etiological factor in the pathological progress of Alzheimer's disease (AD). Here, in this investigation, we explored the neuroprotective potential of PDG against aluminium chloride (AlCl3 )-mediated AD-like neurological alterations in rats. For this purpose, rats were gavaged either AlCl3 (100 mg/kg), PDG (300 mg/kg), or both for 42 days. As a result of the analyzes performed on the hippocampal tissue, it was observed that AlCl3 induced biochemical, molecular, and histopathological changes like those related to AD. PDG pre-treatment significantly decreased acetylcholinesterase activity and restored the levels of brain-derived neurotrophic factor, monoamines (dopamine, norepinephrine, and serotonin), and transmembrane protein (Na+ /K+ -ATPase). Furthermore, PDG boosted the hippocampal antioxidant capacity, as shown by the increased superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, and glutathione contents. These findings were accompanied by decreases in malondialdehyde and nitric oxide levels. The antioxidant effect may promote the upregulation of the expression of antioxidant genes (Nrf2 and HO-1). Moreover, PDG exerted notable anti-inflammatory effects via the lessening of interleukin-1 beta, tumor necrosis factor-alpha, cyclooxygenase-2, nuclear factor kappa B, and decreases in the gene expression of inducible nitric oxide synthase. In addition, noteworthy decreases in pro-apoptotic (Bax and caspase-3) levels and increases in anti-apoptotic (Bcl-2) biomarkers suggested an anti-apoptotic effect of PDG. In support, the hippocampal histological examination validated the aforementioned changes. To summarize, the promising neuromodulatory, antioxidative, anti-inflammatory, and anti-apoptotic activities of PDG establish it as a potent therapeutic option for AD.


Asunto(s)
Enfermedad de Alzheimer , Fármacos Neuroprotectores , Animales , Ratas , Acetilcolinesterasa/metabolismo , Cloruro de Aluminio/toxicidad , Cloruro de Aluminio/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Glutatión/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Prodigiosina/metabolismo , Prodigiosina/farmacología , Prodigiosina/uso terapéutico
6.
Lasers Med Sci ; 37(9): 3631-3638, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36156750

RESUMEN

Prodigiosin (PG) is a secondary metabolite of bacterial origin that is able to absorb the visible light and plays a role as a photosensitizer in photodynamic therapy (PDT). This in vitro study aimed to investigate the cytotoxicity of PG-mediated PDT against the reference strains of Staphylococcus aureus (S. aureus), Escherichia coli (E. coli), and Pseudomonas aeruginosa (P. aeruginosa). The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of PG were determined. Each strain was then allocated into four groups as follows: G1: control (no treatment), G2: PG-treated groups that received different PG concentrations (1000-1.95 µM), G3: laser-treated group (wavelength: 520 nm, radiation dose: 187 J/cm2), and G4: PG-mediated PDT groups that were initially treated with different concentrations of PG and were then exposed to laser irradiation in the same way as the previous group. Finally, the number of colony-forming units per milliliter (CFU/mL) was calculated and analyzed using the SPSS software. PG had both bacteriostatic and bactericidal activities on the tested bacteria, with the maximum antibacterial effect being observed against S. aureus. In all bacterial strains, the maximum number of CFUs was observed in the control group followed by the laser-irradiated and PG-treated groups, but the differences were not statistically significant (p > 0.05). However, the utilization of PG-mediated PDT resulted in a significant decrease in the mean number of CFUs in all the tested bacteria (p < 0.0001). PG-mediated PDT had the potential to kill some bacterial strains in the laboratory. Yet, further studies are warranted to confirm its efficacy and safety to be applied in clinical settings.


Asunto(s)
Antiinfecciosos , Fotoquimioterapia , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus , Pseudomonas aeruginosa , Fotoquimioterapia/métodos , Escherichia coli , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Bacterias
7.
Int Immunopharmacol ; 111: 109068, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35944459

RESUMEN

BACKGROUND: Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS: The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1ß, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS: PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1ß, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION: PDG and/or LDR exhibited obvious anti-RA activity on AIA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Prodigiosina , Animales , Antiinflamatorios/uso terapéutico , Artritis Experimental/terapia , Artritis Reumatoide/terapia , Citocinas/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Metotrexato/uso terapéutico , FN-kappa B/metabolismo , Prodigiosina/uso terapéutico , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Med Chem ; 64(12): 8739-8754, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34111350

RESUMEN

Highly efficient and straightforward synthetic routes toward the first total synthesis of 2-(p-hydroxybenzyl)-prodigiosins (2-5), isoheptylprodigiosin (6), and geometric isomers of tambjamine MYP1 ((E/Z)-7) have been developed. The crucial steps involved in these synthetic routes are the construction of methoxy-bipyrrole-carboxaldehydes (MBCs) and a 20-membered macrocyclic core and a regioselective demethylation of MBC analogues. These new synthetic routes enabled us to generate several natural prodiginines 24-27 in larger quantity. All of the synthesized natural products exhibited potent asexual blood-stage antiplasmodial activity at low nanomolar concentrations against a panel of Plasmodium falciparum parasites, with a great therapeutic index. Notably, prodiginines 6 and 24-27 provided curative in vivo efficacy against erythrocytic Plasmodium yoelii at 25 mg/kg × 4 days via oral route in a murine model. No overt clinical toxicity or behavioral change was observed in any mice treated with prodiginines and tambjamines.


Asunto(s)
Antimaláricos/uso terapéutico , Prodigiosina/análogos & derivados , Prodigiosina/uso terapéutico , Pirroles/uso terapéutico , Animales , Antimaláricos/síntesis química , Antimaláricos/toxicidad , Femenino , Células Hep G2 , Humanos , Ratones , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/efectos de los fármacos , Plasmodium yoelii/efectos de los fármacos , Prodigiosina/toxicidad , Pirroles/síntesis química , Pirroles/toxicidad , Estereoisomerismo , Relación Estructura-Actividad
9.
Eur J Pharmacol ; 888: 173465, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32814079

RESUMEN

Melanoma is a type of skin cancer with an elevated incidence of metastasis and chemoresistance. Such features hamper treatment success of these neoplasms, demanding the search for new therapeutic options. Using a two-step resin-based approach, we recently demonstrated that cytotoxic prodiginines bind to the inhibitor of apoptosis protein, survivin. Herein, we explore the role of survivin in melanoma and whether its modulation is related to the antimelanoma properties of three cytotoxic prodiginines (prodigiosin, cyclononylprodigiosin, and nonylprodigiosin) isolated from marine bacteria. In melanoma patients and cell lines, survivin is overexpressed, and higher levels negatively impact survival. All three prodiginines caused a decrease in cell growth with reduced cytotoxicity after 24 h compared to 72 h treatment, suggesting that low concentrations promote cytostatic effects in SK-Mel-19 (BRAF mutant) and SK-Mel-28 (BRAF mutant), but not in SK-Mel-147 (NRAS mutant). An increase in G1 population was observed after 24 h treatment with prodigiosin and cyclononylprodigiosin in SK-Mel-19. Further studies indicate that prodigiosin induced apoptosis and DNA damage, as detected by increased caspase-3 cleavage and histone H2AX phosphorylation, further arguing for the downregulation of survivin. Computer simulations suggest that prodigiosin and cyclononylprodigiosin bind to the BIR domain of survivin. Moreover, knockdown of survivin increased long-term toxicity of prodigiosin, as observed by reduced clonogenic capacity, but did not alter short-term cytotoxicity. In summary, prodiginine treatment provoked cytostatic rather than cytotoxic effects, cell cycle arrest at G0/G1 phase, induction of apoptosis and DNA damage, downregulation of survivin, and decreased clonogenic capacity in survivin knockdown cells.


Asunto(s)
Melanoma/metabolismo , Prodigiosina/análogos & derivados , Prodigiosina/farmacología , Survivin/antagonistas & inhibidores , Survivin/biosíntesis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Melanoma/tratamiento farmacológico , Prodigiosina/uso terapéutico , Survivin/genética
10.
Mater Sci Eng C Mater Biol Appl ; 112: 110794, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32409024

RESUMEN

This paper presents the results of a combined experimental and analytical study of blended FDA-approved polymers [polylactic-co-glycolic acid (PLGA), polyethylene glycol (PEG) and polycaprolactone (PCL)] with the potential for sustained localized cancer drug release. Porous drug-loaded 3D degradable PLGA-PEG and PLGA-PCL scaffolds were fabricated using a multistage process that involved solvent casting and particulate leaching with lyophilization. The physicochemical properties including the mechanical, thermal and biostructural properties of the drug-loaded microporous scaffolds were characterized. The release of the encapsulated prodigiosin (PG) or paclitaxel (PTX) drug (from the drug-loaded polymer scaffolds) was also studied experimentally at human body temperature (37 °C) and hyperthermic temperatures (41 and 44 °C). These characteristic controlled and localized in vitro drug release from the properties of the microporous scaffold were analyzed using kinetics and thermodynamic models. Subsequently, normal breast cells (MCF-10A) were cultured for a 28-day period on the resulting 3D porous scaffolds in an effort to study the possible regrowth of normal breast tissue, following drug release. The effects of localized cancer drug release on breast cancer cells and normal breast cell proliferation are demonstrated for scenarios that are relevant to palliative breast tumor surgery for 16 weeks under in vivo conditions. Results from the in vitro drug release show a sustained anomalous (non-Fickian) drug release that best fits the Korsmeyer-Peppas (KP) kinetic model with a non-spontaneous thermodynamic process that leads to a massive decrease in breast cancer cell (MDA-MB-231) viability. Our findings from the animal suggest that localized drug release from drug-based 3D resorbable porous scaffolds can be used to eliminate/treat local recurred triple negative breast tumors and promote normal breast tissue regeneration after surgical resection.


Asunto(s)
Antineoplásicos/química , Portadores de Fármacos/química , Paclitaxel/química , Polímeros/química , Prodigiosina/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Femenino , Humanos , Cinética , Ratones , Ratones Desnudos , Paclitaxel/metabolismo , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Polietilenglicoles/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Porosidad , Prodigiosina/metabolismo , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Termodinámica , Andamios del Tejido/química
11.
J. venom. anim. toxins incl. trop. dis ; 25: e20190001, 2019. tab, graf
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1012636

RESUMEN

This work aimed to explore the action of natural prodigiosin on both bacterial organisms and Trypanosoma cruzi cells. Methods: Natural prodigiosin pigment was extracted and purified from cultures of Serratia marcescens. Two media, peanut broth and peptone glycerol broth, both recommended in the literature for prodigiosin production, were compared. The prodigiosin obtained was employed to explore its antimicrobial properties against both bacteria and Trypanosoma cruzi cells. Results: Peanut broth yielded four times more prodigiosin. The prodigiosin showed remarkable activity (minimal inhibitory concentrations in the range of 2-8 µM for bacteria and half maximal inhibitory concentration of 0.6 µM for Trypanosoma cruzi). In fact, the prodigiosin concentration required to inhibit parasite growth was as low as 0.25 mg/l versus 4.9 mg/l of benznidazole required. Furthermore, atomic force microscopy revealed marked morphological alterations in treated epimastigote forms, although no pore-formation activity was detected in protein-free environments. Conclusions: This work demonstrates the potential usefulness of prodigiosin against some gram-positive and gram-negative bacteria and Trypanosoma cruzi although further studies must be done in order to assess its value as a candidate molecule.(AU)


Asunto(s)
Animales , Prodigiosina/uso terapéutico , Trypanosoma cruzi , Enfermedad de Chagas , Bacterias Gramnegativas
12.
Proc Natl Acad Sci U S A ; 113(46): 13150-13155, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799526

RESUMEN

Prodigiosin, a natural red pigment produced by numerous bacterial species, has exhibited promising anticancer activity; however, the molecular mechanisms of action of prodigiosin on malignant cells remain unclear. Aberrant activation of the Wnt/ß-catenin signaling cascade is associated with numerous human cancers. In this study, we identified prodigiosin as a potent inhibitor of the Wnt/ß-catenin pathway. Prodigiosin blocked Wnt/ß-catenin signaling by targeting multiple sites of this pathway, including the low-density lipoprotein-receptor-related protein (LRP) 6, Dishevelled (DVL), and glycogen synthase kinase-3ß (GSK3ß). In breast cancer MDA-MB-231 and MDA-MB-468 cells, nanomolar concentrations of prodigiosin decreased phosphorylation of LRP6, DVL2, and GSK3ß and suppressed ß-catenin-stimulated Wnt target gene expression, including expression of cyclin D1. In MDA-MB-231 breast cancer xenografts and MMTV-Wnt1 transgenic mice, administration of prodigiosin slowed tumor progression and reduced the expression of phosphorylated LRP6, phosphorylated and unphosphorylated DVL2, Ser9 phosphorylated GSK3ß, active ß-catenin, and cyclin D1. Through its ability to inhibit Wnt/ß-catenin signaling and reduce cyclin D1 levels, prodigiosin could have therapeutic activity in advanced breast cancers.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Vía de Señalización Wnt/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D1/genética , Proteínas Dishevelled/genética , Femenino , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Carga Tumoral/efectos de los fármacos , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
13.
Neuroscience ; 305: 26-35, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26247694

RESUMEN

Anhydroexfoliamycin (1) and undecylprodigiosin (2) have been previously described as neuroprotective molecules against oxidative stress in neurons. Since oxidative stress is strongly correlated with neurodegenerative diseases, we have evaluated their effects over the principal hallmarks of Alzheimer's disease (AD). Both compounds were tested in vitro in two different neuroblastoma cellular models, one for amyloid precursor protein metabolism studies (BE(2)-M17) and another one specific for taupathology in AD (SH-SY5Y-TMHT441). Amyloid-beta (Aß) levels, ß-secretase (BACE1) activity, tau phosphorylation, extracellular signal-regulated kinase (ERK) and glycogen synthase kinase-3beta (GSK3ß) expression were analyzed and while undecylprodigiosin (2) produced poor results, anhydroexfoliamycin (1) strongly inhibited GSK3ß, reducing tau phosphorylation in vitro (0.1 µM). A competitive assay of anhydroexfoliamycin (1) and the specific c-Jun N-terminal kinase (JNK) inhibitor, SP600125, showed that the reduction of the phosphorylated tau levels is mediated by the JNK pathway in SH-SY5Y-TMHT441 cells. Thus, this compound was tested in vivo by intraperitoneal administration in 3xTg-AD mice, confirming the positive results registered in the in vitro assays. This work presents anhydroexfoliamycin (1) as a promising candidate for further studies in drug development against neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antracenos/uso terapéutico , Antipsicóticos/uso terapéutico , Encéfalo/metabolismo , Prodigiosina/análogos & derivados , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Antipsicóticos/química , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Neuroblastoma/patología , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Prodigiosina/química , Prodigiosina/uso terapéutico , Proteínas tau/genética
14.
Angew Chem Int Ed Engl ; 52(5): 1374-82, 2013 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23283851

RESUMEN

The development of small-molecule lipid-bilayer anion transporters for potential future use in channel replacement therapy for the treatment of diseases caused by dysregulation of anion transport (such as cystic fibrosis), and in treating cancer by perturbing chemical gradients within cells, thus triggering apoptosis, is an area of intense current interest. This Minireview looks at recent developments in the design of small-molecule transmembrane anion transporters and focuses on the progress so far in employing these compounds in biological systems.


Asunto(s)
Membrana Dobles de Lípidos/química , Aniones/química , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Humanos , Transporte Iónico , Neoplasias/tratamiento farmacológico , Prodigiosina/análogos & derivados , Prodigiosina/uso terapéutico , Prodigiosina/toxicidad
15.
Toxicol Appl Pharmacol ; 257(1): 137-47, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21925195

RESUMEN

This study aimed to explore the mechanisms by which prodigiosin protects against hypoxia-induced oxidative/nitrosative brain injury induced by middle cerebral artery occlusion/reperfusion (MCAo/r) injury in mice. Hypoxia in vitro was modeled using oxygen-glucose deprivation (OGD) followed by reoxygenation of BV-2 microglial cells. Our results showed that treatment of mice that have undergone MCAo/r injury with prodigiosin (10 and 100µg/kg, i.v.) at 1h after hypoxia ameliorated MCAo/r-induced oxidative/nitrosative stress, brain infarction, and neurological deficits in the mice, and enhanced their survival rate. MCAo/r induced a remarkable production in the mouse brains of reactive oxygen species (ROS) and a significant increase in protein nitrosylation; this primarily resulted from enhanced expression of NADPH oxidase 2 (gp91(phox)), inducible nitric oxide synthase (iNOS), and the infiltration of CD11b leukocytes due to breakdown of blood-brain barrier (BBB) by activation of nuclear factor-kappa B (NF-κB). All these changes were significantly diminished by prodigiosin. In BV-2 cells, OGD induced ROS and nitric oxide production by up-regulating gp91(phox) and iNOS via activation of the NF-κB pathway, and these changes were suppressed by prodigiosin. In conclusion, our results indicate that prodigiosin reduces gp91(phox) and iNOS expression possibly by impairing NF-κB activation. This compromises the activation of microglial and/or inflammatory cells, which then, in turn, mediates prodigiosin's protective effect in the MCAo/r mice.


Asunto(s)
Hipoxia-Isquemia Encefálica/prevención & control , Glicoproteínas de Membrana/antagonistas & inhibidores , NADPH Oxidasas/antagonistas & inhibidores , Fármacos Neuroprotectores/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Prodigiosina/uso terapéutico , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Masculino , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos ICR , NADPH Oxidasa 2 , NADPH Oxidasas/biosíntesis , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Nitrosación/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Prodigiosina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tirosina/análogos & derivados , Tirosina/biosíntesis
16.
Curr Med Chem ; 17(21): 2222-31, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20459382

RESUMEN

Apoptosis is involved in the action of several (and perhaps all) cancer-chemotherapeutic agents. Prodiginines are a family of natural red pigmented secondary metabolites, produced by different bacteria and most of them are characterized by a common pyrrolylpyrromethene skeleton. The biosynthesis of prodigiosin and derivatives has been extensively studied in Serratia marcescens. S. marcescens is a Gramnegative bacterium belonging to Enterobacteriaceae. Prodiginines show numerous biological activities pointing out immunosuppressive and anticancer properties. Some prodiginines displayed apoptotic effects in vitro and antitumor activity in vivo. Their cytotoxic effect is attributed to the presence of the C- 6 methoxy substituent. The A-pyrrole ring plays a key role in both the copper nuclease activity and the cytotoxicity of prodiginines. Here we review the main characteristics of prodigiosin and their derivatives as well as the most prominent pharmacological activity of prodiginines and related compounds, including novel synthetic PG-derivatives with lower toxicity like GX15-070 (Obatoclax). The molecular targets of prodiginines are discussed and the mechanism of action for these molecules is a current topic in biomedicine with a real therapeutica potential in the clinic.


Asunto(s)
Antineoplásicos/química , Prodigiosina/análogos & derivados , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Humanos , Indoles , Neoplasias/tratamiento farmacológico , Prodigiosina/química , Prodigiosina/metabolismo , Prodigiosina/uso terapéutico , Prodigiosina/toxicidad , Pirroles/toxicidad , Serratia/química , Transducción de Señal/efectos de los fármacos
17.
Curr Pharm Des ; 15(7): 732-41, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19275639

RESUMEN

Prodigiosins are a family of bright red colored bacterial pigment and derive their name from the miraculous (prodigious) events associated with their occurrence. They indeed seem to be living upto their name as a host of activities such as anti-microbial, anti-malarial, anti-cancer and immunosuppressive have been associated with them. Out of these, immunosuppressive and anti-cancer activity has received more importance as it has a clinical promise. Prodigiosins, isolated mostly from Gram negative bacteria are characterized by a common pyrryldipyrrylmethene structure with varying side chains. The review discusses the mechanisms involved in the anti-cancer activity of this class of compounds. In vitro, prodigiosins have been shown to primarily target the cancer cells independently of the p53 status while little or no effect has been observed on normal cells. In addition, prodigiosins are effective in cancer cells with multidrug resistance phenotype and defects in the apoptotic pathways. These make prodigiosins attractive candidates for further development. Though the molecular targets of prodigiosins have not been clearly defined, they have been found to target different signaling pathways possibly through induction of DNA double strand breaks and/ or neutralization of pH gradients leading to changes in cell cycle proteins and apoptosis. The review will discuss the recent findings related to the mechanism involved in the anti-cancer activity of this class of molecules.


Asunto(s)
Antineoplásicos/farmacología , Prodigiosina/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Bacterias Gramnegativas/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Prodigiosina/química , Prodigiosina/aislamiento & purificación , Prodigiosina/uso terapéutico , Factores de Transcripción/metabolismo
18.
Anticancer Drugs ; 16(4): 393-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15746575

RESUMEN

In the present study, we describe the cytotoxicity of the new drug prodigiosin (PG) in two small cell lung carcinoma (SCLC) cell lines, GLC4 and its derived doxorubicin-resistant GLC4/ADR cell line, which overexpresses multidrug-related protein 1 (MRP-1). We observed through Western blot that PG mediated cytochrome c release, caspase cascade activation and PARP cleavage, thereby leading to apoptosis in a dose-response manner. MRP-1 expression increased after PG treatment, although that does not lead to protein accumulation. The MTT assay showed no difference in sensitivity to PG between the two cell lines. Our results support PG as a potential drug for the treatment of lung cancer as it overcomes the multidrug resistance phenotype produced by MRP-1 overexpression.


Asunto(s)
Antibacterianos/uso terapéutico , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma de Células Pequeñas/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Prodigiosina/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Carcinoma de Células Pequeñas/patología , Caspasas/metabolismo , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células Tumorales Cultivadas
19.
Expert Opin Ther Targets ; 8(6): 613-29, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15584866

RESUMEN

Existing immunosuppressants inhibit lymphocyte activation and T cell cytokine signal transduction pathways, reducing the rate of acute rejection episodes to < 10%. However, the widespread tissue distribution of their molecular targets engenders pleiotropic toxicities. One strategy to address this problem seeks to identify compounds that selectively inhibit a target restricted in distribution to the lymphoid system. Janus kinase (Jak) 3 is such a molecule; it mediates signal transduction via the gamma common chain of lymphokine surface receptors. Disruption of this lymphoid-restricted enzyme would not be predicted to produce collateral damage in other organ systems. Development of selective Jak3 inhibitors has been difficult due to crossreactivity with its homologue, Jak2. In contrast to all other putative antagonists, which are discussed in detail herein, one Jak3 inhibitor, NC1153, shows at least 40-fold greater selective inhibition for Jak3 than for Jak2, is robustly synergistic with calcineurin antagonists, and, either alone or in combination with cyclosporin, produces no adverse effects in rodents preconditioned to be at heightened risk for nephrotoxicity, bone marrow suppression, or altered lipid metabolism.


Asunto(s)
Rechazo de Injerto/prevención & control , Inmunosupresores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/fisiología , Secuencia de Aminoácidos , Animales , Evaluación Preclínica de Medicamentos , Rechazo de Injerto/tratamiento farmacológico , Humanos , Inmunosupresores/química , Inmunosupresores/uso terapéutico , Subunidad gamma Común de Receptores de Interleucina , Janus Quinasa 2 , Janus Quinasa 3 , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Macaca fascicularis , Bases de Mannich/química , Bases de Mannich/farmacología , Bases de Mannich/uso terapéutico , Ratones , Datos de Secuencia Molecular , Estructura Molecular , Fosforilación/efectos de los fármacos , Prodigiosina/análogos & derivados , Prodigiosina/química , Prodigiosina/farmacología , Prodigiosina/uso terapéutico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/fisiología , Pirroles/química , Pirroles/farmacología , Pirroles/uso terapéutico , Ratas , Receptores de Citocinas/antagonistas & inhibidores , Receptores de Interleucina-2/efectos de los fármacos , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato
20.
Blood ; 99(2): 680-9, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11781254

RESUMEN

Janus kinase 3 (Jak3) is a cytoplasmic tyrosine (Tyr) kinase associated with the interleukin-2 (IL-2) receptor common gamma chain (gamma(c)) that is activated by multiple T-cell growth factors (TCGFs) such as IL-2, -4, and -7. Using human T cells, it was found that a recently discovered variant of the undecylprodigiosin family of antibiotics, PNU156804, previously shown to inhibit IL-2-induced cell proliferation, also blocks IL-2-mediated Jak3 auto-tyrosine phosphorylation, activation of Jak3 substrates signal transducers and activators of transcription (Stat) 5a and Stat5b, and extracellular regulated kinase 1 (Erk1) and Erk2 (p44/p42). Although PNU156804 displayed similar efficacy in blocking Jak3-dependent T-cell proliferation by IL-2, -4, -7, or -15, it was more than 2-fold less effective in blocking Jak2-mediated cell growth, its most homologous Jak family member. A 14-day alternate-day oral gavage with 40 to 120 mg/kg PNU156804 extended the survival of heart allografts in a dose-dependent fashion. In vivo, PNU156804 acted synergistically with the signal 1 inhibitor cyclosporine A (CsA) and additively with the signal 3 inhibitor rapamycin to block allograft rejection. It is concluded that inhibition of signal 3 alone by targeting Jak3 in combination with a signal 1 inhibitor provides a unique strategy to achieve potent immunosuppression.


Asunto(s)
Ciclosporina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/uso terapéutico , Proteínas de la Leche , Prodigiosina/análogos & derivados , Prodigiosina/uso terapéutico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas , Pirroles/uso terapéutico , Sirolimus/uso terapéutico , Linfocitos T/efectos de los fármacos , Administración Oral , Animales , Línea Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Cultivadas/enzimología , Ciclosporina/farmacología , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Trasplante de Corazón , Humanos , Inmunosupresores/farmacología , Interleucinas/farmacología , Janus Quinasa 2 , Janus Quinasa 3 , Células Jurkat/efectos de los fármacos , Células Jurkat/enzimología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Prodigiosina/farmacología , Pirroles/farmacología , Ratas , Ratas Endogámicas BUF , Ratas Endogámicas WF , Factor de Transcripción STAT5 , Sirolimus/farmacología , Linfocitos T/enzimología , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA