Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
Front Immunol ; 12: 794593, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34956228

RESUMO

The gill of teleost fish is a multifunctional organ involved in many physiological processes, including protection of the mucosal gill surface against pathogens and other environmental antigens by the gill-associated lymphoid tissue (GIALT). Climate change associated phenomena, such as increasing frequency and magnitude of harmful algal blooms (HABs) put extra strain on gill function, contributing to enhanced fish mortality and fish kills. However, the molecular basis of the HAB-induced gill injury remains largely unknown due to the lack of high-throughput transcriptomic studies performed on teleost fish in laboratory conditions. We used juvenile rainbow trout (Oncorhynchus mykiss) to investigate the transcriptomic responses of the gill tissue to two (high and low) sublethal densities of the toxin-producing alga Prymnesium parvum, in relation to non-exposed control fish. The exposure time to P. parvum (4-5 h) was sufficient to identify three different phenotypic responses among the exposed fish, enabling us to focus on the common gill transcriptomic responses to P. parvum that were independent of dose and phenotype. The inspection of common differentially expressed genes (DEGs), canonical pathways, upstream regulators and downstream effects pointed towards P. parvum-induced inflammatory response and gill inflammation driven by alterations of Acute Phase Response Signalling, IL-6 Signalling, IL-10 Signalling, Role of PKR in Interferon Induction and Antiviral Response, IL-8 Signalling and IL-17 Signalling pathways. While we could not determine if the inferred gill inflammation was progressing or resolving, our study clearly suggests that P. parvum blooms may contribute to the serious gill disorders in fish. By providing insights into the gill transcriptomic responses to toxin-producing P. parvum in teleost fish, our research opens new avenues for investigating how to monitor and mitigate toxicity of HABs before they become lethal.


Assuntos
Brânquias/imunologia , Haptófitas/metabolismo , Inflamação/imunologia , Oncorhynchus mykiss/imunologia , Reação de Fase Aguda/genética , Animais , Citocinas/genética , Exposição Ambiental/efeitos adversos , Proteínas de Peixes/genética , Proliferação Nociva de Algas , Ensaios de Triagem em Larga Escala , Hipóxia/genética , Transdução de Sinais , Toxinas Biológicas/efeitos adversos , Transcriptoma
3.
Toxicol Lett ; 350: 133-142, 2021 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-34303789

RESUMO

Non-dialysable protein-bound uremic toxins (PBUTs) contribute to the development of cardiovascular disease (CVD) in chronic kidney disease (CKD) and vice versa. PBUTs have been shown to alter sphingolipid imbalance. Dihydroceramide desaturase 1 (Des1) is an important gatekeeper enzyme which controls the non-reversible conversion of sphingolipids, dihydroceramide, into ceramide. The present study assessed the effect of Des1 inhibition on PBUT-induced cardiac and renal effects in vitro, using a selective Des1 inhibitor (CIN038). Des1 inhibition attenuated hypertrophy in neonatal rat cardiac myocytes and collagen synthesis in neonatal rat cardiac fibroblasts and renal mesangial cells induced by the PBUTs, indoxyl sulfate and p-cresol sulfate. This is at least attributable to modulation of NF-κB signalling and reductions in ß-MHC, Collagen I and TNF-α gene expression. Lipidomic analyses revealed Des1 inhibition restored C16-dihydroceramide levels reduced by indoxyl sulfate. In conclusion, PBUTs play a critical role in mediating sphingolipid imbalance and inflammatory responses in heart and kidney cells, and these effects were attenuated by Des1 inhibition. Therefore, sphingolipid modifying agents may have therapeutic potential for the treatment of CVD and CKD and warrant further investigation.


Assuntos
Doenças Cardiovasculares/induzido quimicamente , Oxirredutases/uso terapêutico , Esfingolipídeos/metabolismo , Toxinas Biológicas/efeitos adversos , Toxinas Biológicas/metabolismo , Uremia/sangue , Uremia/fisiopatologia , Animais , Inibidores Enzimáticos/uso terapêutico , Humanos , Modelos Animais , Ratos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/complicações , Esfingolipídeos/sangue , Toxinas Biológicas/sangue
4.
Int J Mol Sci ; 22(12)2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34200937

RESUMO

The accumulation of uremic toxins (UTs) is a prototypical manifestation of uremic milieu that follows renal function decline (chronic kidney disease, CKD). Frailty as a potential outcome-relevant indicator is also prevalent in CKD. The intertwined relationship between uremic toxins, including small/large solutes (phosphate, asymmetric dimethylarginine) and protein-bound ones like indoxyl sulfate (IS) and p-cresyl sulfate (pCS), and frailty pathogenesis has been documented recently. Uremic toxins were shown in vitro and in vivo to induce noxious effects on many organ systems and likely influenced frailty development through their effects on multiple preceding events and companions of frailty, such as sarcopenia/muscle wasting, cognitive impairment/cognitive frailty, osteoporosis/osteodystrophy, vascular calcification, and cardiopulmonary deconditioning. These organ-specific effects may be mediated through different molecular mechanisms or signal pathways such as peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), mitogen-activated protein kinase (MAPK) signaling, aryl hydrocarbon receptor (AhR)/nuclear factor-κB (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), Runt-related transcription factor 2 (RUNX2), bone morphogenic protein 2 (BMP2), osterix, Notch signaling, autophagy effectors, microRNAs, and reactive oxygen species induction. Anecdotal clinical studies also suggest that frailty may further accelerate renal function decline, thereby augmenting the accumulation of UTs in affected individuals. Judging from these threads of evidence, management strategies aiming for uremic toxin reduction may be a promising approach for frailty amelioration in patients with CKD. Uremic toxin lowering strategies may bear the potential of improving patients' outcomes and restoring their quality of life, through frailty attenuation. Pathogenic molecule-targeted therapeutics potentially disconnect the association between uremic toxins and frailty, additionally serving as an outcome-modifying approach in the future.


Assuntos
Fragilidade/complicações , Insuficiência Renal Crônica/patologia , Toxinas Biológicas/efeitos adversos , Uremia/complicações , Animais , Humanos , Qualidade de Vida , Insuficiência Renal Crônica/etiologia
5.
Int J Mol Sci ; 22(12)2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201270

RESUMO

The presence of toxins is believed to be a major factor in the development of uremia in patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). Uremic toxins have been divided into 3 groups: small substances dissolved in water, medium molecules: peptides and low molecular weight proteins, and protein-bound toxins. One of the earliest known toxins is urea, the concentration of which was considered negligible in CKD patients. However, subsequent studies have shown that it can lead to increased production of reactive oxygen species (ROS), and induce insulin resistance in vitro and in vivo, as well as cause carbamylation of proteins, peptides, and amino acids. Other uremic toxins and their participation in the damage caused by oxidative stress to biological material are also presented. Macromolecules and molecules modified as a result of carbamylation, oxidative stress, and their adducts with uremic toxins, may lead to cardiovascular diseases, and increased risk of mortality in patients with CKD.


Assuntos
Estresse Oxidativo , Insuficiência Renal Crônica/complicações , Toxinas Biológicas/efeitos adversos , Uremia/etiologia , Animais , Humanos , Uremia/patologia
6.
Int J Mol Sci ; 22(9)2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067057

RESUMO

Cancer cells frequently overexpress specific surface receptors providing tumor growth and survival which can be used for precise therapy. Targeting cancer cell receptors with protein toxins is an attractive approach widely used in contemporary experimental oncology and preclinical studies. Methods of targeted delivery of toxins to cancer cells, different drug carriers based on nanosized materials (liposomes, nanoparticles, polymers), the most promising designed light-activated toxins, as well as mechanisms of the cytotoxic action of the main natural toxins used in modern experimental oncology, are discussed in this review. The prospects of the combined therapy of tumors based on multimodal nanostructures are also discussed.


Assuntos
Neoplasias/tratamento farmacológico , Toxinas Biológicas/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Humanos , Estresse Oxidativo/efeitos dos fármacos , Toxinas Biológicas/efeitos adversos
7.
Toxins (Basel) ; 13(2)2021 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-33673020

RESUMO

Animal toxins and venoms have recently been developed as cancer treatments possessing tumor cell growth-inhibitory, antiangiogenesis, and proapoptotic effects. Endometriosis is a common benign gynecological disorder in reproductive-age women, and no definite treatment for this disorder is without severe side effects. As endometriosis and malignant tumors share similar characteristics (progressive, invasive, estrogen-dependent growth, and recurrence), animal toxins and venoms are thought to be effective against endometriosis. The objective of this study was to outline studies using toxic animal-based medicinal materials (TMM) as endometriosis treatment and to explore its clinical applicability. Preclinical and clinical studies using TMM were searched for in four databases from inception to October 2020. A total of 20 studies of TMM on endometriosis were included. In eight clinical studies, herbal medicines containing TMM were effective in relieving symptoms of endometriosis, with no side effects. In twelve experimental studies, the main therapeutic mechanisms of TMM against endometriosis were proapoptotic, antiangiogenesis, estrogen level-reducing, and possible anti-inflammatory effects. TMM are thus considered promising sources for the development of an effective treatment method for endometriosis. Further studies are needed to clarify the therapeutic mechanism of TMM against endometriosis and to provide sufficient grounds for clinical application.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Endometriose/tratamento farmacológico , Medicina Tradicional do Leste Asiático , Extratos de Tecidos/uso terapêutico , Toxinas Biológicas/uso terapêutico , Animais , Medicamentos de Ervas Chinesas/efeitos adversos , Endometriose/metabolismo , Endometriose/patologia , Feminino , Humanos , Medicina Tradicional do Leste Asiático/efeitos adversos , Extratos de Tecidos/efeitos adversos , Toxinas Biológicas/efeitos adversos , Resultado do Tratamento , Peçonhas/uso terapêutico
8.
Clin Chest Med ; 42(1): 19-38, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33541612

RESUMO

Pulmonary arterial hypertension secondary to drugs and toxins is an important subgroup of group 1 pulmonary hypertension associated with significant morbidity and mortality. Many drugs and toxins have emerged as risk factors for pulmonary arterial hypertension, which include anorexigens, illicit agents, and several US Food and Drug Administration-approved therapeutic medications. Drugs and toxins are classified as possible or definite risk factors for pulmonary arterial hypertension. This article reviews agents that have been implicated in the development of pulmonary arterial hypertension, their pathologic mechanisms, and methods to prevent the next deadly outbreak of drug- and toxin-induced pulmonary arterial hypertension.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/complicações , Hipertensão Arterial Pulmonar/fisiopatologia , Toxinas Biológicas/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/fisiopatologia , Humanos , Hipertensão Arterial Pulmonar/induzido quimicamente , Toxinas Biológicas/toxicidade
9.
Sci Rep ; 11(1): 3786, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589722

RESUMO

Protein-bound uremic toxins (Indoxyl sulfate [IS] and p-cresyl sulfate [PCS]) are both associated with cardiovascular (CV) and all-cause mortality in subjects with chronic kidney disease (CKD). Possible mechanisms have not been elucidated. In hemodialysis patients, we investigated the relationship between the free form of IS and PCS and 181 CV-related proteins. First, IS or PCS concentrations were checked, and high levels were associated with an increased risk of acute coronary syndrome (ACS) in 333 stable HD patients. CV proteins were further quantified by a proximity extension assay. We examined associations between the free form protein-bound uremic toxins and the quantified proteins with correction for multiple testing in the discovery process. In the second step, the independent association was evaluated by multivariable-adjusted models. We rank the CV proteins related to protein-bound uremic toxins by bootstrapped confidence intervals and ascending p-value. Six proteins (signaling lymphocytic activation molecule family member 5, complement component C1q receptor, C-C motif chemokine 15 [CCL15], bleomycin hydrolase, perlecan, and cluster of differentiation 166 antigen) were negatively associated with IS. Fibroblast growth factor 23 [FGF23] was the only CV protein positively associated with IS. Three proteins (complement component C1q receptor, CCL15, and interleukin-1 receptor-like 2) were negatively associated with PCS. Similar findings were obtained after adjusting for classical CV risk factors. However, only higher levels of FGF23 was related to increased risk of ACS. In conclusion, IS and PCS were associated with several CV-related proteins involved in endothelial barrier function, complement system, cell adhesion, phosphate homeostasis, and inflammation. Multiplex proteomics seems to be a promising way to discover novel pathophysiology of the uremic toxin.


Assuntos
Cresóis/efeitos adversos , Indicã/efeitos adversos , Insuficiência Renal Crônica/tratamento farmacológico , Ésteres do Ácido Sulfúrico/efeitos adversos , Toxinas Biológicas/química , Síndrome Coronariana Aguda/induzido quimicamente , Síndrome Coronariana Aguda/genética , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Quimiocinas CC/genética , Cresóis/administração & dosagem , Cisteína Endopeptidases/genética , Feminino , Fator de Crescimento de Fibroblastos 23/genética , Proteoglicanas de Heparan Sulfato/genética , Humanos , Indicã/administração & dosagem , Proteínas Inflamatórias de Macrófagos/genética , Masculino , Pessoa de Meia-Idade , Ligação Proteica/efeitos dos fármacos , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/genética , Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Ésteres do Ácido Sulfúrico/administração & dosagem , Toxinas Biológicas/efeitos adversos , Toxinas Biológicas/genética
10.
Sci Rep ; 11(1): 512, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436654

RESUMO

Uraemic toxins increase in serum parallel to a decline in the glomerular filtration rate and the development of sarcopenia in patients with chronic kidney disease (CKD). This study analyses the role of uraemic toxins in sarcopenia at different stages of CKD, evaluating changes in the muscular regeneration process. Cultured C2C12 cells were incubated with a combination of indoxyl sulphate and p-cresol at high doses (100 µg/mL) or low doses (25 µg/mL and 10 µg/mL) resembling late or early CKD stages, respectively. Cell proliferation (analysed by scratch assays and flow cytometry) was inhibited only by high doses of uraemic toxins, which inactivated the cdc2-cyclin B complex, inhibiting mitosis and inducing apoptosis (analysed by annexin V staining). By contrast, low doses of uraemic toxins did not affect proliferation, but reduced myogenic differentiation, primed with 2% horse serum, by inhibiting myogenin expression and promoting fibro-adipogenic differentiation. Finally, to assess the in vivo relevance of these results, studies were performed in gastrocnemii from uraemic rats, which showed higher collagen expression and lower myosin heavy chain expression than those from healthy rats. In conclusion, uraemic toxins impair the skeletal muscular regeneration process, even at low concentrations, suggesting that sarcopenia can progress from the early stages of CKD.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Mioblastos/fisiologia , Regeneração/efeitos dos fármacos , Toxinas Biológicas/efeitos adversos , Uremia/fisiopatologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Camundongos , Músculo Esquelético/fisiologia , Ratos
11.
Methods Mol Biol ; 2248: 19-42, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33185865

RESUMO

The TNF superfamily of proinflammatory and proapoptotic cytokines influence tissue-wide responses to molecular insults such as small molecules, toxins, and viral infections that perturb cellular homeostasis at the level of DNA replication, transcription, and translation. In the context of acute lung injury, for example, TNF superfamily members like TNF-α and TRAIL can severely exacerbate disease pathophysiology. This chapter describes a systematic approach to optimization of mammalian cell viability assays and transcriptional profiling through nCounter® Technology to permit a detailed examination of how TNF-α and TRAIL modulate programmed cell death pathways in concert with ricin toxin, a ribosome-inactivating protein (RIP) and a potent inducer of acute respiratory distress. We compare two widely used luciferase- and colorimetric-based cell viability assays and provide optimization protocols for adherent and non-adherent cell lines. We provide a computational workflow to facilitate downstream analysis of datasets generated from nCounter® gene expression panels. While combined treatment with ricin toxin and TRAIL serves as the exemplar, the methodologies are applicable to any TNF superfamily member in combination with any biological agent of interest.


Assuntos
Citocinas/biossíntese , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Toxinas Biológicas/efeitos adversos , Fatores de Necrose Tumoral/biossíntese , Animais , Apoptose/genética , Biomarcadores , Morte Celular , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Humanos , Família Multigênica , Toxinas Biológicas/imunologia
12.
Neurol Clin ; 38(4): 765-780, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33040860

RESUMO

Channelopathies, neuromuscular junction disorders, and myopathies represent multiple mechanisms by which toxins can affect the peripheral nervous system. These toxins include ciguatoxin, tetrodotoxin, botulinum toxin, metabolic poisons, venomous snake bites, and several medications. These toxins are important to be aware of because they can lead to serious symptoms, disability, or even death, and many can be treated if recognized ear.


Assuntos
Canalopatias/induzido quimicamente , Doenças Musculares/induzido quimicamente , Doenças da Junção Neuromuscular/induzido quimicamente , Síndromes Neurotóxicas/etiologia , Toxinas Biológicas/efeitos adversos , Humanos
13.
Neurol Clin ; 38(4): 881-896, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33040867

RESUMO

Nerve agents and neurobiological weapons are among the most devastating and lethal of weapons. Acetylcholinesterase inhibitors act by increasing the amount of acetylcholine in the neuromuscular junction, resulting in flaccid paralysis. Tabun, VX, soman, and sarin are the major agents in this category. Exposure to nerve agents can be inhalational or through dermal contact. Neurotoxins may have peripheral and central effects on the nervous system. Atropine is an effective antidote to nerve agents. Neurobiological weapons entail using whole organisms or organism-synthesized toxins as agents. Some organisms that can be used as biological weapons include smallpox virus.


Assuntos
Guerra Biológica/métodos , Substâncias para a Guerra Química/efeitos adversos , Síndromes Neurotóxicas/etiologia , Animais , Humanos , Toxinas Biológicas/efeitos adversos
14.
Forensic Sci Int ; 314: 110366, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32683271

RESUMO

Microbial Forensics is a field that continues to grow in interest and application among the forensic community. This review, divided into two sections, covers several topics associated with this new field. The first section presents a historic overview concerning the use of microorganisms (or its product, i.e. toxins) as harmful biological agents in the context of biological warfare (biowarfare), bioterrorism, and biocrime. Each case is illustrated with the examination of case reports that span from prehistory to the present day. The second part of the manuscript is devoted to the role of MF and highlights the necessity to prepare for the pressing threat of the harmful use of biological agents as weapons. Preventative actions, developments within the field to ensure a timely and effective response and are discussed herein.


Assuntos
Guerra Biológica/história , Bioterrorismo/história , Crime/história , Infecções Bacterianas , Ciências Forenses , Infecções por HIV , Ensaios de Triagem em Larga Escala , História do Século XV , História do Século XVI , História do Século XVII , História do Século XVIII , História do Século XIX , História do Século XX , História do Século XXI , História Antiga , História Medieval , Humanos , Aprendizado de Máquina , Técnicas Microbiológicas , Toxinas Biológicas/efeitos adversos
16.
Trends Cardiovasc Med ; 30(5): 310-312, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31477495

RESUMO

Arrhythmogenic ingredients in our diet such as mushrooms, licorice, toxic honey, liquid protein drinks, etc. have long been recognized as rare but important considerations in the differential diagnosis of arrhythmias. Anecdotal reports of torsades de pointes (TdP), arrhythmias and/or sudden death and small studies in normal subjects have suggested that simple ingredients such as grapefruit juice or ingredients in energy drinks marketed as dietary supplements could have direct arrhythmogenic actions, especially in patients with congenital long QT syndrome (cLQTS). Two recent studies that employed the industry-standard "thorough QT" trial design leave no doubt that grapefruit juice and some energy drinks can prolong the QTc interval and to exceed 500 msec. in some patients with cLQTS, a threshold known to signal imminent danger. These reports raise numerous clinically important questions such as which other patients may be at risk of arrhythmias. For example, patients with multiple clinical risk factors for TdP (hypokalemia, bradycardia, female sex, etc.) may be at risk from these and possibly other dietary ingredients ingested by millions of people each day. It is essential that further research evaluate the safety of these and similar food products and that vulnerable patients, especially those with cLQTS, be warned of this serious and emerging threat.


Assuntos
Citrus paradisi/efeitos adversos , Bebidas Energéticas/efeitos adversos , Frutas/efeitos adversos , Sistema de Condução Cardíaco/fisiopatologia , Síndrome do QT Longo/etiologia , Plantas Tóxicas/efeitos adversos , Torsades de Pointes/etiologia , Toxinas Biológicas/efeitos adversos , Potenciais de Ação , Animais , Inocuidade dos Alimentos , Frequência Cardíaca , Humanos , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/fisiopatologia , Período Refratário Eletrofisiológico , Medição de Risco , Fatores de Risco , Torsades de Pointes/diagnóstico , Torsades de Pointes/fisiopatologia
17.
J. venom. anim. toxins incl. trop. dis ; 26: e20190070, 2020. tab, graf
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1484764

RESUMO

Background: Intrathecal injection of voltage-sensitive calcium channel blocker peptide toxins exerts analgesic effect in several animal models of pain. Upon intrathecal administration, recombinant Phα1β exerts the same analgesic effects as the those of the native toxin. However, from a clinical perspective, the intrathecal administration limits the use of anesthetic drugs in patients. Therefore, this study aimed to investigate the possible antinociceptive effect of intravenous recombinant Phα1β in rat models of neuropathic pain, as well as its side effects on motor, cardiac (heart rate and blood pressure), and biochemical parameters. Methods: Male Wistar rats and male Balb-C mice were used in this study. Giotto Biotech® synthesized the recombinant version of Phα1β using Escherichia coli expression. In rats, neuropathic pain was induced by chronic constriction of the sciatic nerve and paclitaxel-induced acute and chronic pain. Mechanical sensitivity was evaluated using von Frey filaments. A radiotelemeter transmitter (TA11PA-C10; Data Sciences, St. Paul, MN, USA) was placed on the left carotid of mice for investigation of cardiovascular side effects. Locomotor activity data were evaluated using the open-field paradigm, and serum CKMB, TGO, TGP, LDH, lactate, creatinine, and urea levels were examined. Results: Intravenous administration of recombinant Phα1β toxin induced analgesia for up to 4 h, with ED50 of 0.02 (0.01-0.03) mg/kg, and reached the maximal effect (Emax = 100% antinociception) at a dose of 0.2 mg/kg. No significant changes were observed in any of the evaluated motor, cardiac or biochemical parameters. Conclusion: Our data suggest that intravenous administration of recombinant Phα1β may be feasible for drug-induced analgesia, without causing any severe side effects.


Assuntos
Masculino , Animais , Ratos , Analgésicos , Neuropatia Ciática/terapia , Paclitaxel , Toxinas Biológicas/administração & dosagem , Toxinas Biológicas/efeitos adversos , Venenos de Aranha/química , Administração Intravenosa , Camundongos Endogâmicos BALB C , Ratos Wistar
18.
Artigo em Inglês | MEDLINE | ID: mdl-31709196

RESUMO

Objective: The human intestinal microbiome plays an important role in inflammatory bowel disease (IBD) and colorectal cancer (CRC) development. One of the first discovered bacterial mediators involves Bacteroides fragilis toxin (BFT, also named as fragilysin), a metalloprotease encoded by enterotoxigenic Bacteroides fragilis (ETBF) that causes barrier disruption and inflammation of the colon, leads to tumorigenesis in susceptible mice, and is enriched in the mucosa of IBD and CRC patients. Thus, targeted inhibition of BFT may benefit ETBF carrying patients. Design: By applying two complementary in silico drug design techniques, drug repositioning and molecular docking, we predicted potential BFT inhibitory compounds. Top candidates were tested in vitro on the CRC epithelial cell line HT29/c1 for their potential to inhibit key aspects of BFT activity, being epithelial morphology changes, E-cadherin cleavage (a marker for barrier function) and increased IL-8 secretion. Results: The primary bile acid and existing drug chenodeoxycholic acid (CDCA), currently used for treating gallstones, cerebrotendinous xanthomatosis, and constipation, was found to significantly inhibit all evaluated cell responses to BFT exposure. The inhibition of BFT resulted from a direct interaction between CDCA and BFT, as confirmed by an increase in the melting temperature of the BFT protein in the presence of CDCA. Conclusion: Together, our results show the potential of in silico drug discovery to combat harmful human and microbiome-derived proteins and more specifically suggests a potential for retargeting CDCA to inhibit the pro-oncogenic toxin BFT.


Assuntos
Carcinógenos/metabolismo , Transformação Celular Neoplásica , Descoberta de Drogas , Reposicionamento de Medicamentos , Microbioma Gastrointestinal , Toxinas Biológicas , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/efeitos dos fármacos , Endotoxinas/química , Endotoxinas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Metaloendopeptidases/química , Metaloendopeptidases/farmacologia , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Relação Estrutura-Atividade , Toxinas Biológicas/efeitos adversos , Toxinas Biológicas/biossíntese
19.
Perit Dial Int ; 39(6): 509-518, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31690700

RESUMO

Background:Protein-bound uremic toxins (PBUTs) are poorly cleared by peritoneal dialysis (PD). This study aimed to enhance PBUT removal in PD by adding a binder to the peritoneal dialysate and to evaluate the feasibility and efficacy of liposome-supported PD (LSPD) to increase the removal of PBUTs compared with albumin PD.Methods:Removal of p-cresyl sulfate (PCS), indoxyl sulfate (IS), and indole-3-acetic acid (3-IAA) was first evaluated in an in vitro PD model using artificial plasma preloaded with test solutes. Male Sprague-Dawley rats (n = 24) were then subjected to 5/6 nephrectomy and fed for 16 weeks to establish end-stage renal failure, after which they were treated with either conventional glucose-based PD, albumin-based PD, or liposome-based PD. Removal of PBUTs and small water-soluble solutes was determined during a 6-hour PD dwell.Results:In vitro experiments showed that adding albumin as a toxin binder to the dialysate markedly increased the removal of PCS, IS, and 3-IAA compared with the control. The uptake capacity of liposomes was comparable with that of albumin for PCS and 3-IAA, though slightly inferior for IS. In vivo PD in uremic rats demonstrated that LSPD resulted in higher intraperitoneal concentrations and more total mass removal for PBUTs than the conventional glucose-based PD, which was comparable with albumin PD.Conclusions:Supplementing conventional glucose-based PD solutions with a binder could efficiently increase the removal of PBUTs. This preliminary study suggested that LSPD may be a promising alternative to albumin PD for increasing PBUT removal in the development of next-generation PD solutions for PD patients.


Assuntos
Soluções para Diálise/farmacologia , Falência Renal Crônica/terapia , Lipossomos/farmacologia , Diálise Peritoneal/métodos , Albumina Sérica/efeitos adversos , Toxinas Biológicas/efeitos adversos , Animais , Modelos Animais de Doenças , Falência Renal Crônica/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Albumina Sérica/metabolismo
20.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-31491842

RESUMO

Sepsis is defined as life-threatening organ dysfunction caused by the dysregulated host response to infection. Despite serious mortality and morbidity, no sepsis-specific drugs exist. Endotoxemia is often used to model the hyperinflammation associated with early sepsis. This model classically uses lipopolysaccharide (LPS) from Gram-negative pathogens to activate the immune system, leading to hyperinflammation, microcirculatory disturbances and death. Other toxins may also be used to activate the immune system including Gram-positive peptidoglycan (PG) and lipoteichoic acid (LTA). In addition to these standard toxins, other bacterial components can induce inflammation. These molecules activate different signaling pathways and produce different physiological responses which can be taken advantage of for sepsis modeling. Endotoxemia modeling can provide information on pathways to inflammation in sepsis and contribute to preclinical drug development.


Assuntos
Inflamação/etiologia , Inflamação/metabolismo , Sepse/etiologia , Toxinas Biológicas/efeitos adversos , Animais , Endotoxemia/diagnóstico , Endotoxemia/etiologia , Endotoxemia/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação/diagnóstico , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/imunologia , Sepse/diagnóstico , Sepse/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...