Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Drug Chem Toxicol ; 46(6): 1193-1202, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36322409

RESUMEN

Antibiotic use, especially fluoroquinolones, has been linked to extensive renal and hepatic injury thus inflicts a considerable health problem. Fifty rats were allocated into five groups (n = 10). Group 1 represented the normal-control group. Group 2 received moxifloxacin only (MOX; 8 mg/kg/day, i.p.) for seven days and represented the MOX-control group. Groups 3, 4, and 5 received MOX for seven days accompanied by royal jelly (RJ; 100 mg/kg/day, p.o.), Echinacea (ECH; 40 mg/kg/day, p.o.), and a combination of both at the aforementioned doses respectively for 30 days. All groups were investigated for renal and hepatic function tests. Renal tissue content of kidney injury molecule-1 (KIM-1) along with renal and hepatic tissue contents of reduced glutathione (GSH) and malondialdehyde (MDA) were assessed for all groups. Histopathological examination was performed followed by immunohistochemical staining for caspase-3 in renal and hepatic tissues. MOX administration resulted in significant renal and hepatic damage. RJ and ECH significantly improved the serum parameters of renal and hepatic functions along with increasing GSH and decreasing MDA in renal and hepatic tissues. Renal contents of KIM-1 were also reduced. Moreover, RJ, ECH, and their combination amended MOX-induced histopathological changes and significantly reduced caspase-3 immunohistochemical staining in both renal and hepatic tissues. The current study is the first to elucidate the effect of RJ, ECH, and their combination against MOX-induced renal and hepatic injury in rats. The study suggests that these protective effects are mainly via the reduction of oxidative stress induced by MOX administration.


Asunto(s)
Antioxidantes , Echinacea , Ratas , Animales , Antioxidantes/farmacología , Moxifloxacino/metabolismo , Moxifloxacino/farmacología , Echinacea/metabolismo , Caspasa 3/metabolismo , Riñón , Estrés Oxidativo , Malondialdehído/metabolismo
2.
Biochem Biophys Res Commun ; 629: 1-5, 2022 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-36087537

RESUMEN

Fluoroquinolones are one of the most frequently prescribed antibiotics. However, their use increases the risk of Aortic aneurysm and dissection (AAD). The mechanism underlying this effect remains unclear. AAD are caused by weakening of the aortic wall and loss of vascular smooth muscle cells. Osteopontin is involved in the occurrence and development of AAD. The aim of the present study was to examine the role of moxifloxacin, a fluoroquinolone, in the occurrence of AAD using a moderate-severity AAD mouse model. Four-week-old male C57BL/6J mice were fed a high-fat diet. At 8 weeks of age, the mice were infused with saline or angiotensin II (1000 ng kg-1 min-1) via osmotic minipumps for 4 weeks, and then orally administered water (vehicle) or moxifloxacin (30 and 100 mg kg-1 day-1) for another 3 weeks. Moxifloxacin (30 and 100 mg kg-1 day-1) induced AAD and elastin degradation in aortic tissues, as revealed by hematoxylin and eosin staining and elastica-van Gieson staining. Additionally, immunohistochemical staining and Western blot analyses showed that moxifloxacin 100 mg kg-1 day-1 decreased the protein expression of smooth muscle protein 22α, one of the markers of the contractile phenotype of vascular smooth muscle cells, in aortic tissues compared to vehicle and moxifloxacin 30 mg kg-1 day-1. Furthermore, moxifloxacin (100 mg kg-1 day-1) increased the protein expression of osteopontin and matrix metalloproteinases-2 in the aortic tissues when compared to control. Moxifloxacin may induce the onset of AAD and weakening of the aortic media by increasing the expression of osteopontin and matrix metalloproteinase-2 and decreasing that of smooth muscle protein 22α in aortic tissue.


Asunto(s)
Aneurisma de la Aorta , Disección Aórtica , Disección Aórtica/inducido químicamente , Disección Aórtica/genética , Angiotensina II/farmacología , Animales , Antibacterianos/farmacología , Aneurisma de la Aorta/inducido químicamente , Modelos Animales de Enfermedad , Elastina/metabolismo , Eosina Amarillenta-(YS)/efectos adversos , Eosina Amarillenta-(YS)/metabolismo , Hematoxilina/metabolismo , Hematoxilina/farmacología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Moxifloxacino/efectos adversos , Moxifloxacino/metabolismo , Proteínas Musculares/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteopontina/metabolismo , Goma/efectos adversos , Goma/metabolismo , Agua/metabolismo
3.
Ecotoxicol Environ Saf ; 240: 113711, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35653971

RESUMEN

The novel fourth-generation fluoroquinolones (FQs) were developed to improve the antimicrobial activity and their utilization has rapidly increased in recent years. However, knowledge of the ecotoxicity and microalgae-mediated biodegradation of these novel FQs is limited. In this research, the toxic effects of moxifloxacin (MOX) and gatifloxacin (GAT) on Chlamydomonas reinhardtii as well as their biodegradation and metabolic fate were investigated. The results showed that the toxicity of MOX to C. reinhardtii was higher than that of GAT, and increased with culture time. Chlorophyll fluorescence and pigment content analyses suggested that the decrease in photosynthetic efficiency was primarily caused by the inhibition of electron transport after QA in PSII complex. These FQs induced oxidative damage in cells, and the antioxidation mechanisms of C. reinhardtii were analyzed. The maximum MOX removal of 77.67% by C. reinhardtii was achieved at 1 mg/L MOX, whereas the maximum GAT removal of 34.04% was attained at 20 mg/L GAT. The different hydrophilicity and lipophilicity of these FQs resulted in distinct findings in biodegradation experiments. Identification of the transformation products suggested that the likely biodegradation pathways of FQs by C. reinhardtii were hydroxylation, demethylation, and ring cleavage.


Asunto(s)
Chlamydomonas reinhardtii , Biodegradación Ambiental , Fluoroquinolonas/metabolismo , Fluoroquinolonas/toxicidad , Gatifloxacina/farmacología , Moxifloxacino/metabolismo , Moxifloxacino/farmacología , Fotosíntesis
4.
IEEE Trans Med Imaging ; 41(8): 2004-2008, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35167445

RESUMEN

Conjunctival goblet cells (CGCs) are mucin-secreting cells in the eye and play essential roles for ocular surface homeostasis. Since various ocular surface pathologies are related to CGC dysfunction, CGC examination is important for the evaluation of ocular surface conditions. Recently we introduced moxifloxacin-based fluorescence microscopy (MBFM) for non-invasive CGC imaging. However, the imaging speed was up to 1 frame per second (fps) and needed to be improved for clinical applications. In this study, we developed a high-speed moxifloxacin-based, extended depth-of-field (EDOF) microscopy system that operates at a maximum imaging speed of 15 fps. The system used a deformable mirror for the high-speed axial sweeping of focal plane during single-frame acquisitions. The acquired images contained both in-focus and out-of-focus information, and deconvolution was used to filter the in-focus information. The system had a DOF of 800 [Formula: see text], field-of-view of 1.2 mm ×1.2 mm, and resolution of [Formula: see text]. Its performance was demonstrated by real-time, breathing-motion-insensitive CGC imaging of mouse and rabbit models, in vivo. High-speed EDOF microscopy has potentials for non-invasive, real-time CGC examinations of human subjects.


Asunto(s)
Conjuntiva , Células Caliciformes , Animales , Conjuntiva/diagnóstico por imagen , Células Caliciformes/metabolismo , Humanos , Microscopía Fluorescente/métodos , Moxifloxacino/metabolismo , Conejos
5.
Microbiol Spectr ; 9(2): e0043421, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34585951

RESUMEN

The therapeutic repertoire for tuberculosis (TB) remains limited despite the existence of many TB drugs that are highly active in in vitro models and possess clinical utility. Underlying the lack of efficacy in vivo is the inability of TB drugs to penetrate microenvironments inhabited by the causative agent, Mycobacterium tuberculosis, including host alveolar macrophages. Here, we determined the ability of the phenoxazine PhX1 previously shown to be active against M. tuberculosis in vitro to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. We also investigated the extent of permeation into uninfected and M. tuberculosis-infected human macrophage-like Tamm-Horsfall protein 1 (THP-1) cells directly and by comparing to results obtained in vitro in synergy assays. Our data indicate that PhX1 (4,750 ± 127.2 ng/ml) penetrates more effectively into THP-1 cells than do the clinically used anti-TB agents, rifampin (3,050 ± 62.9 ng/ml), moxifloxacin (3,374 ± 48.7 ng/ml), bedaquiline (4,410 ± 190.9 ng/ml), and linezolid (770 ± 14.1 ng/ml). Compound efficacy in infected cells correlated with intracellular accumulation, reinforcing the perceived importance of intracellular penetration as a key drug property. Moreover, we detected synergies deriving from redox-stimulatory combinations of PhX1 or clofazimine with the novel prenylated amino-artemisinin WHN296. Finally, we used compound synergies to elucidate the relationship between compound intracellular accumulation and efficacy, with PhX1/WHN296 synergy levels shown to predict drug efficacy. Collectively, our data support the utility of the applied assays in identifying in vitro active compounds with the potential for clinical development. IMPORTANCE This study addresses the development of novel therapeutic compounds for the eventual treatment of drug-resistant tuberculosis. Tuberculosis continues to progress, with cases of Mycobacterium tuberculosis (M. tuberculosis) resistance to first-line medications increasing. We assess new combinations of drugs with both oxidant and redox properties coupled with a third partner drug, with the focus here being on the potentiation of M. tuberculosis-active combinations of compounds in the intracellular macrophage environment. Thus, we determined the ability of the phenoxazine PhX1, previously shown to be active against M. tuberculosis in vitro, to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. In addition, the extent of permeation into human macrophage-like THP-1 cells and H37Rv-infected THP-1 cells was measured via mass spectrometry and compared to in vitro two-dimensional synergy and subsequent intracellular efficacy. Collectively, our data indicate that development of new drugs will be facilitated using the methods described herein.


Asunto(s)
Antituberculosos/metabolismo , Tuberculosis/metabolismo , Animales , Antituberculosos/química , Antituberculosos/farmacología , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Moxifloxacino/química , Moxifloxacino/metabolismo , Moxifloxacino/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Rifampin/química , Rifampin/metabolismo , Rifampin/farmacología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Tuberculosis/fisiopatología
6.
ACS Appl Mater Interfaces ; 12(46): 51225-51235, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-33164509

RESUMEN

In vitro drug release systems have recently received tremendous attention because they allow noninvasive, convenient, and prolonged administration of pharmacological agents. On-demand epidermal drug release systems can improve treatment efficiency, prevent multidrug resistance, and minimize drug toxicity to healthy cells. In addition, real-time monitoring of drug content is also essential for guiding the determination of drug dosage and replacing drug carriers in time. Therefore, it is important to integrate the above properties in one ideal epidermal patch. Herein, photonic crystals (PCs) based on Fe3O4@C nanoparticles were introduced into drug-loaded poly(N-isopropylacrylamide-co-acrylic acid) (P(NIPAM-AAc)) hydrogel-functionalized textiles. Drug loading and release depended on the expansion and contraction of the hydrogels. The lower critical solution temperature (LCST) of the hydrogels was adjusted to 40 °C, which is higher than the skin temperature, by varying the content of hydrophilic comonomer acrylic acid (AAc) to store the drug at room temperature, and on-demand release was achieved by mild thermal stimulation. Moreover, the lattice spacing (d) of PCs varied with the expansion and contraction of the hydrogels, which can cause the color of P(NIPAM-AAc) hydrogel-functionalized textiles to change. These synchronous thermoresponsive chromic drug uptake and release behaviors provided an effective method for visual and real-time monitoring of drug content. Furthermore, in view of the poor mechanical properties of hydrogel wound dressings, textile matrices were composited to prevent holistic breaking during the stretching process. Biological experiments proved that the drug-loaded P(NIPAM-AAc) hydrogel-functionalized textiles had good antibacterial properties and wound-healing effects.


Asunto(s)
Portadores de Fármacos/química , Hidrogeles/química , Textiles , Acrilamidas/química , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Vendajes , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Óxido Ferrosoférrico/química , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Moxifloxacino/química , Moxifloxacino/metabolismo , Moxifloxacino/farmacología , Nanopartículas/química , Polímeros/química , Staphylococcus aureus/efectos de los fármacos , Temperatura , beta-Ciclodextrinas/química
7.
Int J Antimicrob Agents ; 56(6): 106183, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33045345

RESUMEN

This study validates bacterial anionic phospholipids (APs) as a putative molecular target in a novel antibiotic treatment against the Gram-positive bacterium Listeria monocytogenes and the Gram-negative bacterium Escherichia coli. Bacterial AP expression was targeted with its associated protein-ligand partner, annexin A5 (ANXA5). This protein was functionalised with the covalent addition of the antibiotic ampicillin (AMP) and separately with the antibiotic moxifloxacin (MOX). Functionalised ANXA5 serves as a delivery vehicle, directing the antibiotic to bacterial AP expression. The results presented here suggest that this ANXA5-AMP bioconjugate participates in a positive feedback loop where APs, the target of the delivery vehicle ANXA5, are upregulated by the chemotherapeutic payload of the bioconjugate. Importantly, the ANXA5 delivery vehicle is non-toxic to bacterial cells by itself and neither is the ANXA5-antibiotic bioconjugate toxic to human vascular endothelial cells. As measured by the IC50, conjugation to ANXA5 resulted in increasing the antibiotic activity of AMP against L. monocytogenes and E. coli by more than 4 and 3 orders of magnitude, respectively, compared with free AMP, whilst the activity of MOX against L. monocytogenes is increased by 4 orders of magnitude. Given the conservation of AP expression in pathologies such as oncogenesis and other bacterial/viral/parasitic infections, we hypothesise that a therapeutic modality targeting AP expression may be a viable chemotherapeutic strategy in many infectious diseases.


Asunto(s)
Ampicilina/farmacología , Anexina A5/farmacología , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Listeria monocytogenes/efectos de los fármacos , Moxifloxacino/farmacología , Ampicilina/metabolismo , Anexina A5/metabolismo , Células Cultivadas , Escherichia coli/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Listeria monocytogenes/metabolismo , Pruebas de Sensibilidad Microbiana , Moxifloxacino/metabolismo , Fosfatidilserinas/metabolismo , Fosfolípidos/metabolismo
8.
Circ Res ; 126(8): 947-964, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32091972

RESUMEN

RATIONALE: Drug-induced proarrhythmia is so tightly associated with prolongation of the QT interval that QT prolongation is an accepted surrogate marker for arrhythmia. But QT interval is too sensitive a marker and not selective, resulting in many useful drugs eliminated in drug discovery. OBJECTIVE: To predict the impact of a drug from the drug chemistry on the cardiac rhythm. METHODS AND RESULTS: In a new linkage, we connected atomistic scale information to protein, cell, and tissue scales by predicting drug-binding affinities and rates from simulation of ion channel and drug structure interactions and then used these values to model drug effects on the hERG channel. Model components were integrated into predictive models at the cell and tissue scales to expose fundamental arrhythmia vulnerability mechanisms and complex interactions underlying emergent behaviors. Human clinical data were used for model framework validation and showed excellent agreement, demonstrating feasibility of a new approach for cardiotoxicity prediction. CONCLUSIONS: We present a multiscale model framework to predict electrotoxicity in the heart from the atom to the rhythm. Novel mechanistic insights emerged at all scales of the system, from the specific nature of proarrhythmic drug interaction with the hERG channel, to the fundamental cellular and tissue-level arrhythmia mechanisms. Applications of machine learning indicate necessary and sufficient parameters that predict arrhythmia vulnerability. We expect that the model framework may be expanded to make an impact in drug discovery, drug safety screening for a variety of compounds and targets, and in a variety of regulatory processes.


Asunto(s)
Antiarrítmicos/química , Arritmias Cardíacas/tratamiento farmacológico , Cardiotoxinas/química , Simulación por Computador , Descubrimiento de Drogas/métodos , Canal de Potasio ERG1/química , Antiarrítmicos/metabolismo , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/metabolismo , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , Cardiotoxinas/efectos adversos , Cardiotoxinas/metabolismo , Descubrimiento de Drogas/tendencias , Canal de Potasio ERG1/metabolismo , Femenino , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/metabolismo , Aprendizaje Automático , Masculino , Moxifloxacino/química , Moxifloxacino/metabolismo , Moxifloxacino/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Fenetilaminas/química , Fenetilaminas/metabolismo , Fenetilaminas/uso terapéutico , Estructura Secundaria de Proteína , Sulfonamidas/química , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/metabolismo , Inhibidores de Topoisomerasa II/uso terapéutico
9.
Clin Microbiol Infect ; 26(9): 1255.e1-1255.e8, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32088331

RESUMEN

OBJECTIVES: Pharmacokinetic-pharmacodynamic (PK-PD) considerations are at the heart of defining susceptibility breakpoints for antibiotic therapy. However, current approaches follow a fragmented workflow. The aim of this study was to develop an integrative pharmacometric approach to define MIC-based breakpoints for killing and suppression of resistance development for plasma and tissue sites, integrating clinical microdialysis data as well as in vitro time-kill curves and heteroresistance information, exemplified by moxifloxacin against Staphylococcus aureus and Escherichia coli. METHODS: Plasma and target site samples were collected from ten patients receiving 400 mg moxifloxacin/day. In vitro time-kill studies with three S. aureus and two E. coli strains were performed and resistant subpopulations were quantified. Using these data, a hybrid physiologically based (PB) PK model and a PK-PD model were developed, and utilized to predict site-specific breakpoints. RESULTS: For both bacterial species, the predicted MIC breakpoint for stasis at 400 mg/day was 0.25 mg/L. Less reliable killing was predicted for E. coli in subcutaneous tissues where the breakpoint was 0.125 mg/L. The breakpoint for resistance suppression was 0.06 mg/L. Notably, amplification of resistant subpopulations was highest at the clinical breakpoint of 0.25 mg/L. High-dose moxifloxacin (800 mg/day) increased all breakpoints by one MIC tier. CONCLUSIONS: An efficient pharmacometric approach to define susceptibility breakpoints was developed; this has the potential to streamline the process of breakpoint determination. Thereby, the approach provided additional insight into target site PK-PD and resistance development for moxifloxacin. Application of the approach to further drugs is warranted.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Moxifloxacino/farmacología , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/metabolismo , Técnicas Bacteriológicas , Farmacorresistencia Bacteriana , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Moxifloxacino/metabolismo
10.
Helicobacter ; 24(5): e12628, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31282059

RESUMEN

BACKGROUND: Fluoroquinolones hinder bacterial DNA replication by inhibiting DNA gyrase. However, mutations, in the QRDR segment of its A subunit (GyrA), cause antibiotic resistance. Here, the interactions of levofloxacin (LVX), gemifloxacin (GXN), and moxifloxacin (MXN) with Helicobacter pylori GyrA, in LVX-resistant vs -sensitive strains, were studied. METHODS: Levoflixacin-sensitive (n = 4) and -resistant (n = 9) H pylori strains, randomly selected from another antibiotic susceptibility study, underwent PCR amplification of gyrA gene, spanning the QRDR segment. The amplified gene fragments were sequenced and aligned. The homology model of H pylori GyrA was built based on that of Escherichia coli, and energy minimization was done. The interaction patterns of LVX, GXN, and MXN with GyrA were analyzed via molecular docking studies. RESULTS: Sequence alignment of the 13 studied strains, created 5 categories of strains: (A) wild type-like (H pylori ATCC26695), (B) N87K-only, (C) D91N-only, (D) N87K + V94L, and (E) D91N + A97V mutations. The minimum inhibitory concentrations (MIC) for LVX-sensitive (category A) and -resistant (categories B-E) strains were <1 mg/L and ≥32 mg/L, respectively. The binding mode of GyrA in category A with LVX identified G35/N87/Y90/D91/V94/G114/S115/I116/D117/G118/D119, as key residues, some residing outside the QRDR segment. Category B strains lost only one interaction (G35), which led to elevated binding free energy (∆G) and full LVX resistance. Categories C-E lost more contacts, with higher ∆G and again full LVX resistance. GXN bound to GyrA of categories A and B via a different set of key residues, while MXN retained the lost contact (G35) in LVX-resistant, category B strains. CONCLUSION: Using molecular docking tools, we identified the key residues responsible for interaction of GyrA with LVX, GXN, and MXN. In the presence of N87K-only mutation, the loss of one of these contacts (ie, G35) led to full LVX resistance. Yet, GXN and MXN overcame this mutation, by retaining all key contacts with GyrA.


Asunto(s)
Antibacterianos/farmacología , Girasa de ADN/metabolismo , Farmacorresistencia Bacteriana , Gemifloxacina/farmacología , Helicobacter pylori/efectos de los fármacos , Levofloxacino/farmacología , Moxifloxacino/farmacología , Antibacterianos/metabolismo , Girasa de ADN/química , Girasa de ADN/genética , Gemifloxacina/metabolismo , Helicobacter pylori/enzimología , Humanos , Levofloxacino/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Moxifloxacino/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Reacción en Cadena de la Polimerasa , Unión Proteica , Análisis de Secuencia de ADN
11.
Sci Rep ; 8(1): 14174, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242205

RESUMEN

Paneth cells are one of the principal epithelial cell types in the small intestine, located at the base of intestinal crypts. Paneth cells play key roles in intestinal host-microbe homeostasis via granule secretion, and their dysfunction is implicated in pathogenesis of several diseases including Crohn's disease. Despite their physiological importance, study of Paneth cells has been hampered by the limited accessibility and lack of labeling methods. In this study, we developed a simple in vivo imaging method of Paneth cells in the intact mouse small intestine by using moxifloxacin and two-photon microscopy (TPM). Moxifloxacin, an FDA-approved antibiotic, was used for labeling cells and its fluorescence was strongly observed in Paneth cell granules by TPM. Moxifloxacin labeling of Paneth cell granules was confirmed by molecular counterstaining. Comparison of Paneth cells in wild type, genetically obese (ob/ob), and germ-free (GF) mice showed different granule distribution. Furthermore, Paneth cell degranulation was observed in vivo. Our study suggests that TPM with moxifloxacin labeling can serve as a useful tool for studying Paneth cell biology and related diseases.


Asunto(s)
Intestino Delgado/patología , Células de Paneth/patología , Animales , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía/métodos , Moxifloxacino/metabolismo , Células de Paneth/metabolismo , Fotones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA