Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Cancer Res ; 14(1): 274-299, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38323292

RESUMO

The microbiota residing in the gut environment is essential for host homeostasis. Increasing evidence suggests that microbial perturbation (dysbiosis) regulates cancer initiation and progression at local and distant sites. Here, we have identified microbial dysbiosis with the depletion of commensal bacteria as a host-intrinsic factor associated with metastatic dissemination to the bone. Using a mouse model of triple-negative mammary cancer, we demonstrate that a pre-established disruption of microbial homeostasis using an antibiotic cocktail increases tumor growth, enhanced circulating tumor cells, and subsequent dissemination to the bone. We found that the presence of pathogenic bacteria and loss of commensal bacteria in an antibiotic-induced gut environment is associated with sustained inflammation. Increased secretion of G-CSF and MMP-9 in intestinal tissues, followed by increased neutrophil infiltration and severe systemic inflammation in tumor-bearing mice, indicates the direct consequence of a dysbiotic microbiome. Increased neutrophil infiltration to the bone metastatic niche facilitates extravasation and transendothelial migration of tumor cells. It provides a novel, pre-established, and favorable environment to form an immunosuppressive pre-metastatic niche. The presence of tumor cells in immunosuppressive metastatic tumor niche disrupts the balance between osteoblasts and osteoclasts, promotes osteoclast differentiation, and remodels the bone structure. Excessive bone resorption by osteoclasts causes bone degradation and ultimately causes extreme pain in a bone metastatic mouse model. In clinical settings, bone metastasis is associated with intractable severe pain that severely compromises the quality of life in these patients.

2.
Gut Microbes ; 16(1): 2310291, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38329115

RESUMO

Opioids, such as morphine and oxycodone, are widely used for pain management associated with chronic pancreatitis (CP); however, their impact on the progression and pain sensitivity of CP has never been evaluated. This report investigates the impact of opioid use on the severity of CP, pain sensitivity, and the gut microbiome. C57BL/6 mice were divided into control, CP, CP with morphine/oxycodone, and either morphine or oxycodone alone groups. CP was induced by administration of caerulein (50ug/kg/h, i.p. hourly x7, twice a week for 10 weeks). The mouse-to-pancreas weight ratio, histology, and Sirius red staining were performed to measure CP severity. Tail flick and paw pressure assays were used to measure thermal and mechanical pain. DNA was extracted from the fecal samples and subjected to whole-genome shotgun sequencing. Germ-free mice were used to validate the role of gut microbiome in sensitizing acute pancreatic inflammation. Opioid treatment exacerbates CP by increasing pancreatic necrosis, fibrosis, and immune-cell infiltration. Opioid-treated CP mice exhibited enhanced pain hypersensitivity and showed distinct clustering of the gut microbiome compared to untreated CP mice, with severely compromised gut barrier integrity. Fecal microbiota transplantation (FMT) from opioid-treated CP mice into germ-free mice resulted in pancreatic inflammation in response to a suboptimal caerulein dose. Together, these analyses revealed that opioids worsen the severity of CP and induce significant alterations in pain sensitivity and the gut microbiome in a caerulein CP mouse model. Microbial dysbiosis plays an important role in sensitizing the host to pancreatic inflammation.


Assuntos
Microbioma Gastrointestinal , Pancreatite Crônica , Animais , Camundongos , Analgésicos Opioides/efeitos adversos , Oxicodona/efeitos adversos , Disbiose/induzido quimicamente , Disbiose/tratamento farmacológico , Ceruletídeo/efeitos adversos , Microbioma Gastrointestinal/fisiologia , Camundongos Endogâmicos C57BL , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/tratamento farmacológico , Pancreatite Crônica/patologia , Morfina/efeitos adversos , Dor/tratamento farmacológico , Inflamação
3.
Gut Microbes ; 16(1): 2292224, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38108125

RESUMO

There has been a rapid increase in neonates born with a history of prenatal opioid exposure. How prenatal opioid exposure affects pain sensitivity in offspring is of interest, as this may perpetuate the opioid epidemic. While few studies have reported hypersensitivity to thermal pain, potential mechanisms have not been described. This study posits that alterations in the gut microbiome may underly hypersensitivity to pain in prenatally methadone-exposed 3-week-old male offspring, which were generated using a mouse model of prenatal methadone exposure. Fecal samples collected from dams and their offspring were subjected to 16s rRNA sequencing. Thermal and mechanical pain were assessed using the tail flick and Von Frey assays. Transcriptomic changes in whole brain samples of opioid or saline-exposed offspring were investigated using RNA-sequencing, and midbrain sections from these animals were subjected to qPCR profiling of genes related to neuropathic and inflammatory pain pathways. Prenatal methadone exposure increased sensitivity to thermal and mechanical pain and elevated serum levels of IL-17a. Taxonomical analysis revealed that prenatal methadone exposure resulted in significant alterations in fecal gut microbiota composition, including depletion of Lactobacillus, Bifidobacterium, and Lachnospiracea sp and increased relative abundance of Akkermansia, Clostridium sensu stricto 1, and Lachnoclostridium. Supplementation of the probiotic VSL#3 in dams rescued hypersensitivity to thermal and mechanical pain in prenatally methadone-exposed offspring. Similarly, cross-fostering prenatally methadone-exposed offspring to control dams also attenuated hypersensitivity to thermal pain in opioid-exposed offspring. Modulation of the maternal and neonatal gut microbiome with probiotics resulted in transcriptional changes in genes related to neuropathic and immune-related signaling in whole brain and midbrain samples of prenatally methadone-exposed offspring. Together, our work provides compelling evidence of the gut-brain-axis in mediating pain sensitivity in prenatally opioid-exposed offspring.


Assuntos
Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Humanos , Gravidez , Animais , Feminino , Masculino , Analgésicos Opioides/efeitos adversos , Microbioma Gastrointestinal/genética , Disbiose/induzido quimicamente , Disbiose/microbiologia , RNA Ribossômico 16S/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Metadona , Dor
4.
iScience ; 26(11): 108166, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37915593

RESUMO

The cycle of substance use disorder (SUD) leading to dependence is a complex process involving multiple neurocircuitries and brain regions. The amygdala is the core brain region that is involved in processing withdrawal and anxiety and depressive-like behaviors. However, the transcriptional changes in each cell type within the amygdala during SUD remains unknown. Here, we performed single-cell RNA sequencing and classified all cell types in the mouse amygdala. We particularly focused on gene expression changes in glial cells under dependent state and compared to either naive or withdrawal state. Our data revealed distinct changes in key biological processes, such as gene expression, immune response, inflammation, synaptic transmission, and mitochondrial respiration. Significant differences were unraveled in the transcriptional profiles between dependence and withdrawal states. This report is the first single-cell RNA sequencing dataset from the amygdala under opioid dependence and withdrawal conditions, providing unique insights in understanding brain alterations during SUD.

5.
Gut Microbes ; 15(2): 2246184, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37610102

RESUMO

Opioid crisis is an ongoing epidemic since the past several decades in the United States. Opioid use-associated microbial dysbiosis is emerging as a key regulator of intestinal homeostasis and behavioral responses to opioid. However, the mechanistic insight into the role of microbial community in modulating host response is unavailable. To uncover the role of opioid-induced dysbiosis in disrupting intestinal homeostasis we utilized whole genome sequencing, untargeted metabolomics, and mRNA sequencing to identify changes in microbiome, metabolome, and host transcriptome respectively. Morphine treatment resulted in significant expansion of Parasuterella excrementihominis, Burkholderiales bacterium 1_1_47, Enterococcus faecalis, Enterorhabdus caecimuris and depletion of Lactobacillus johnsonii. These changes correlated with alterations in lipid metabolites and flavonoids. Significant alteration in microbial metabolism (metabolism of lipids, amino acids, vitamins and cofactors) and increased expression of virulence factors and biosynthesis of lipopolysaccharides (LPS) and lipoteichoic acid (LTA) were observed in microbiome of morphine-treated animals. In concurrence with changes in microbiome and metabolome extensive changes in innate and adaptive immune response, lipid metabolism, and gut barrier dysfunction were observed in the host transcriptome. Microbiome depleted mice displayed lower levels of inflammation, immune response and tissue destruction compared to mice harboring a dysbiotic microbiome in response to morphine treatment, thus establishing dysbiotic microbiome as mediator of morphine gut pathophysiology. Integrative analysis of multi-omics data highlighted the associations between Parasutterella excrementihominis, Burkholderiales bacterium 1_1_47, Enterococcus faecalis, Enterorhabdus caecimuris and altered levels of riboflavin, flavonoids, and lipid metabolites including phosphocholines, carnitines, bile acids, and ethanolamines with host gene expression changes involved in inflammation and barrier integrity of intestine. Omic analysis also highlighted the role of probiotic bacteria Lactobacillus johnsonii, metabolites flavonoids and riboflavin that were depleted with morphine as important factors for intestinal homeostasis. This study presents for the first time ever an interactive view of morphine-induced changes in microbial metabolism, strain level gut microbiome analysis and comprehensive view of changes in gut transcriptome. We also identified areas of potential therapeutic interventions to limit microbial dysbiosis and present a unique resource to the opioid research community.


Assuntos
Microbioma Gastrointestinal , Transtornos Relacionados ao Uso de Opioides , Animais , Camundongos , Analgésicos Opioides , Disbiose/induzido quimicamente , Multiômica , Riboflavina , Derivados da Morfina , Lipídeos
6.
Gut Microbes ; 15(1): 2242610, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37589387

RESUMO

The ongoing opioid epidemic has left millions of people suffering from opioid use disorder due to the over-prescription of highly addictive substances. Chronic opioid exposure leads to dependence, where the absence of the drug results in negative symptoms of withdrawal, often driving patients to continue drug use; however, few therapeutic strategies are currently available to combat the cycle of addiction and the severity of morphine withdrawal. This study investigates the microbiome as a potential therapeutic target for morphine withdrawal, as gut dysbiosis caused by morphine use has been proven to contribute to other aspects of opioid use disorders, such as tolerance. Results show that although the microbiome during morphine withdrawal trends toward recovery from morphine-induced dysbiosis, there continues to be a disruption in the alpha and beta diversity as well as the abundance of gram-positive bacteria that may still contribute to the severity of morphine withdrawal symptoms. Germ-free mice lacking the microbiome did not develop somatic withdrawal symptoms, indicating that the microbiome is necessary for the development of somatic withdrawal behavior. Notably, only TLR2 but not TLR4 whole-body knockout models display less withdrawal severity, implicating that the microbiome, through a gram-positive, TLR2 mediated mechanism, drives opioid-induced somatic withdrawal behavior.


Assuntos
Microbioma Gastrointestinal , Transtornos Relacionados ao Uso de Opioides , Síndrome de Abstinência a Substâncias , Humanos , Camundongos , Animais , Morfina/efeitos adversos , Analgésicos Opioides/efeitos adversos , Receptor 2 Toll-Like/genética , Disbiose/induzido quimicamente , Disbiose/complicações , Síndrome de Abstinência a Substâncias/etiologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-37498760

RESUMO

Diagnosis, treatment planning, surveillance, and the monitoring of clinical trials for brain diseases all benefit greatly from neuroimaging-based tumor segmentation. Recently, Convolutional Neural Networks (CNNs) have demonstrated promising results in enhancing the efficiency of image-based brain tumor segmentation. Most current work on CNNs, however, is devoted to creating increasingly complicated convolution modules to improve performance, which in turn raises the computing cost of the model. This work proposes a simple and effective feed-forward CNN, LightNet (Light Network). Based on multi-path and multi-level, it replaces traditional convolutional methods with light operations, which reduces network parameters and redundant feature maps. In the up-sampling stage, a light channel attention module is added to achieve richer multi-scale and spatial semantic feature information extraction of brain tumor. The performance of the network is evaluated in the Multimodal Brain Tumor Segmentation Challenge (BraTS 2015) dataset, and results are presented here alongside other high-performing CNNs. Results show comparable accuracy with other methods but with increased efficiency, segmentation performance, and reduced redundancy and computational complexity. The result is a high-performing network with a balance between efficiency and accuracy, allowing, for example, better energy performance on mobile devices.

8.
Br J Pharmacol ; 180(10): 1362-1378, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36562107

RESUMO

BACKGROUND AND PURPOSE: Opioids are commonly used for the management of cancer-associated pain and chemotherapy-induced diarrhoea. The chemotherapeutic irinotecan (CPT-11) causes severe gastrointestinal (GI) toxicity due to deconjugation of inactive metabolite SN-38 glucuronide (SN-38G) by bacterial ß-glucuronidases to the active 7-ethyl-10-hydroxycamptothecin (SN-38). Opioids are known to cause gut microbial dysbiosis, this study evaluated whether CPT-11 anti-tumour efficacy and GI toxicity are exacerbated by opioid co-administration. EXPERIMENTAL APPROACH: Eight-week-old C57BL/6 male mice were co-administration with CPT-11 ± opioid. 16S rRNA sequencing was used for gut microbiome analysis. LC-MS analyses of plasma and intestinal extracts were performed to investigate the pharmacokinetic profile of CPT-11. Histological analysis and quantitative real-time polymerase chain reaction were used to determine the severity of intestinal tissue damage. Human liver microsome In vitro assay was performed to confirm the effects of opioids on CPT-11 metabolism. KEY RESULTS: Gut microbiome analysis showed that morphine treatment induced enrichment of ß-glucuronidase-producing bacteria in the intestines of CPT-11-treated mice, resulting in SN-38 accumulation and exacerbation of GI toxicity in the small intestine. Oral administration of both antibiotics and glucuronidase inhibitor protected mice against GI toxicity induced with CPT-11 and morphine co-administration, implicating a microbiome-dependent mechanism. Additionally, morphine and loperamide decreased the plasma concentration of SN-38 and compromised CPT-11 anti-tumour efficacy, this seemed to be microbiome independent. CONCLUSION AND IMPLICATIONS: Gut microbiota play a significant role in opioid and chemotherapeutic agent drug-drug interactions. Inhibition of gut microbial glucuronidase may also prevent adverse GI effects of CPT-11 in patients on opioids.


Assuntos
Antineoplásicos Fitogênicos , Neoplasias , Humanos , Camundongos , Masculino , Animais , Irinotecano , Analgésicos Opioides/farmacologia , Disbiose , Modelos Animais de Doenças , RNA Ribossômico 16S , Antineoplásicos Fitogênicos/toxicidade , Camundongos Endogâmicos C57BL , Camptotecina/toxicidade , Bactérias , Glucuronidase/metabolismo , Glucuronidase/farmacologia , Derivados da Morfina/farmacologia
9.
Microbiol Spectr ; 11(1): e0247222, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36511710

RESUMO

This study characterized compositional and functional shifts in the intestinal and oral microbiome in HIV-positive patients on antiretroviral therapy compared to HIV-negative individuals. Seventy-nine specimens were collected from 5 HIV-positive and 12 control subjects from five locations (colon brush, colon wash, terminal ileum [TI] brush, TI wash, and saliva) during colonoscopy and at patient visits. Microbiome composition was characterized using 16S rRNA sequencing, and microbiome function was predicted using bioinformatics tools (PICRUSt and BugBase). Our analysis indicated that the ß-diversity of all intestinal samples (colon brush, colon wash, TI brush, and TI wash) from patients with HIV was significantly different from patients without HIV. Specifically, bacteria from genera Prevotella, Fusobacterium, and Megasphaera were more abundant in samples from HIV-positive patients. On the other hand, bacteria from genera Ruminococcus, Blautia, and Clostridium were more abundant in samples from HIV-negative patients. Additionally, HIV-positive patients had higher abundances of biofilm-forming and pathogenic bacteria. Furthermore, pathways related to translation and nucleotide metabolism were elevated in HIV-positive patients, whereas pathways related to lipid and carbohydrate metabolism were positively correlated with samples from HIV-negative patients. Our analyses further showed variations in microbiome composition in HIV-positive and negative patients by sampling site. Samples from colon wash, colon brush, and TI wash were significant between groups, while samples from TI brush and saliva were not significant. Taken together, here, we report altered intestinal microbiome composition and predicted function in patients with HIV compared to uninfected patients, though we found no changes in the oral microbiome. IMPORTANCE Over 37 million people worldwide are living with HIV. Although the availability of antiretroviral therapy has significantly reduced the number of AIDS-related deaths, individuals living with HIV are at increased risk for opportunistic infections. We now know that HIV interacts with the trillions of bacteria, fungi, and viruses in the human body termed the microbiome. Only a limited number of previous studies have compared variations in the oral and gastrointestinal microbiome with HIV infection. Here, we detail how the oral and gastrointestinal microbiome changes with HIV infection, having used 5 different sampling sites to gain a more comprehensive view of these changes by location. Our results show site-specific changes in the intestinal microbiome associated with HIV infection. Additionally, we show that while there were significant changes in the intestinal microbiome, there were no significant changes in the oral microbiome.


Assuntos
Microbioma Gastrointestinal , Infecções por HIV , Microbiota , Humanos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/microbiologia , RNA Ribossômico 16S/genética , Microbioma Gastrointestinal/genética , Mucosa Intestinal/microbiologia , Bactérias/genética
10.
Gut Microbes ; 14(1): 2143225, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36409161

RESUMO

The gut microbial ecosystem exhibits a complex bidirectional communication with the host and is one of the key contributing factors in determining mucosal immune homeostasis or an inflammatory state. Opioid use has been established to induce gut microbial dysbiosis consistent with increased intestinal tissue inflammation. In this study, we investigated the role of infiltrated immune cells in morphine-induced intestinal tissue damage and gut microbial dysbiosis in mice. Results reveal a significant increase in chemokine expression in intestinal tissues followed by increased neutrophil infiltration post morphine treatment which is direct consequence of a dysbiotic microbiome since the effect is attenuated in antibiotics treated animals and in germ-free mice. Neutrophil neutralization using anti-Ly6G monoclonal antibody showed a significant decrease in tissue damage and an increase in tight junction protein organization. 16S rRNA sequencing on intestinal samples highlighted the role of infiltrated neutrophils in modulating microbial community structure by providing a growth benefit for pathogenic bacteria, such as Enterococcus, and simultaneously causing a significant depletion of commensal bacteria, such as Lactobacillus. Taken together, we provide the first direct evidence that neutrophil infiltration contributes to morphine-induced intestinal tissue damage and gut microbial dysbiosis. Our findings implicate that inhibition of neutrophil infiltration may provide therapeutic benefits against gastrointestinal dysfunctions associated with opioid use.


Assuntos
Microbioma Gastrointestinal , Microbiota , Transtornos Relacionados ao Uso de Opioides , Animais , Camundongos , Infiltração de Neutrófilos , Disbiose/induzido quimicamente , Morfina , Analgésicos Opioides , RNA Ribossômico 16S/genética
11.
J Environ Manage ; 323: 116206, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36115244

RESUMO

Due to anthropogenic disturbances, the karst region in southern China is vulnerable to ecological problems such as soil erosion and surface exposure. However, limited studies on variations in large-scale ecological risk (ER) and their influencing factors, particularly the coupling/decoupling relationship with an exposed surface fraction (ESF), make ER regulations and ecological restoration challenging. The present study evaluates the ER of eight typical karst provinces in Southern China from 1990 to 2020 using the technique for order preference by similarity to an ideal solution (TOPSIS) model and ecosystem services (habitat quality, water yield, carbon storage, soil conservation, and food production), and extracts the contemporaneous ESF using Landsat satellite data in Google Earth Engine (GEE). The spatiotemporal change of ER and ESF are analyzed, and their coupling/decoupling relationship and driving mechanism are explored using coupling coordination degree (CCD) and multi-scale geographically weighted regression (MGWR) models. The results show that: (1) Over the past 30 years, the ER has increased until 2010 and subsequently declined, with an increasing mean value (0.463-0.503), except in Chongqing municipality. The ESF decreased significantly (the mean value dropped from 44.7% to 38.7%), except that in Sichuan province. (2) The average CCD between ER and ESF decreased with fluctuation of -0.017, with a decoupling relationship (58.18%). The coupling area is larger than the decoupling area in the Sichuan area, while other provinces are opposite. (3) The coupling/decoupling relationship in the study area is mainly driven by terrain (elevation, slope) and socio-economic (population density, per capita GDP) factors. More attention should be paid to the role of these factors in the continuous reduction and control of ESF and ER. This study can serve as a reference for similar studies in karst regions, such as risk assessment and surface monitoring, rocky desertification control, ecological engineering layout, and territorial planning.


Assuntos
Conservação dos Recursos Naturais , Ecossistema , Carbono , China , Solo
12.
Pharmaceutics ; 14(9)2022 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-36145627

RESUMO

Despite the many advancements in the field of pain management, the use of intravenous opioids, such as morphine, in neonates is still a challenge for clinicians and researchers, as the available evidence concerning the long-term consequences of such an early exposure is limited. In particular, little is known concerning the long-term consequences of neonatal morphine exposure on the gut microbiome, which has been identified as a key modulator of health and diseases. Consequently, the purpose of this study was to investigate those long-term consequences of neonatal morphine on the gut microbiome. Newborn mice were exposed to either morphine (5 mg/kg/day) or saline for a duration of 7 ± 2 days. Fecal samples were collected during adolescence and adulthood to longitudinally assess the gut microbiome. DNA extracted from the stool samples were sent out for 16s rRNA sequencing. During adolescence, neonatal morphine resulted in a significant increase of α-diversity and an overall decrease in the abundance of several commensal genera. During adulthood, ß-diversity revealed a significantly different microbial composition of the neonatally morphine-exposed mice than that of the controls. The results demonstrate that morphine exposure during this critical developmental period resulted in long-lasting changes, particularly a reduction in several commensal bacteria. Thus, an adjunct therapeutic intervention with probiotics could potentially be used along with opioids to manage pain while attenuating the long-term co-morbidities of neonatal morphine later in life.

13.
J Exp Clin Cancer Res ; 41(1): 258, 2022 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-36002889

RESUMO

BACKGROUND: Pancreatic cancer is one of the most difficult cancers to detect early and most patients die from complications arising due to distant organ metastases. The lack of bona fide early biomarkers is one of the primary reasons for late diagnosis of pancreatic cancer. It is a multifactorial disease and warrants a novel approach to identify early biomarkers. METHODS: In order to characterize the proteome, Extracellular vesicles (EVs) isolated from different in vitro conditions mimicking tumor-microenvironment interactions between pancreatic cancer epithelial and stromal cells were analyzed using high throughput mass spectrometry. The biological activity of the secreted EVome was analyzed by investigating changes in distant organ metastases and associated early changes in the microbiome. Candidate biomarkers (KIF5B, SFRP2, LOXL2, and MMP3) were selected and validated on a mouse-human hybrid Tissue Microarray (TMA) that was specifically generated for this study. Additionally, a human TMA was used to analyze the expression of KIF5B and SFRP2 in progressive stages of pancreatic cancer. RESULTS: The EVome of co-cultured epithelial and stromal cells is different from individual cells with distinct protein compositions. EVs secreted from stromal and cancer cells cultures could not induce significant changes in Pre-Metastatic Niche (PMN) modulation, which was assessed by changes in the distant organ metastases. However, they did induce significant changes in the early microbiome, as indicated by differences in α and ß-diversities. KIF5B and SFRP2 show promise for early detection and investigation in progressive pancreatic cancer. These markers are expressed in all stages of pancreatic cancer such as low grade PanINs, advanced cancer, and in liver and soft tissue metastases. CONCLUSIONS: Proteomic characterization of EVs derived from mimicking conditions of epithelial and stromal cells in the tumor-microenvironment resulted in the identification of several proteins, some for the first time in EVs. These secreted EVs cannot induce changes in distant organ metastases in in vivo models of EV education, but modulate changes in the early murine microbiome. Among all the proteins that were analyzed (MMP3, KIF5B, SFRP2, and LOXL2), KIF5B and SFRP2 show promise as bona fide early pancreatic cancer biomarkers expressed in progressive stages of pancreatic cancer.


Assuntos
Cinesinas , Proteínas de Membrana , Neoplasias Pancreáticas , Microambiente Tumoral , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Metaloproteinase 3 da Matriz , Camundongos , Neoplasias Pancreáticas/patologia , Proteoma/metabolismo , Proteômica/métodos , Neoplasias Pancreáticas
14.
J Adv Res ; 37: 19-31, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35499050

RESUMO

Introduction: The functional relevance of intra-species diversity in natural microbial communities remains largely unexplored. The guts of two closely related honey bee species, Apis cerana and A. mellifera, are colonised by a similar set of core bacterial species composed of host-specific strains, thereby providing a good model for an intra-species diversity study. Objectives: We aim to assess the functional relevance of intra-species diversity of A. cerana and A. mellifera gut microbiota. Methods: Honey bee workers were collected from four regions of China. Their gut microbiomes were investigated by shotgun metagenomic sequencing, and the bacterial compositions were compared at the species level. A cross-species colonisation assay was conducted, with the gut metabolomes being characterised by LC-MS/MS. Results: Comparative analysis showed that the strain composition of the core bacterial species was host-specific. These core bacterial species presented distinctive functional profiles between the hosts. However, the overall functional profiles of the A. cerana and A. mellifera gut microbiomes were similar; this was further supported by the consistency of the honey bees' gut metabolome, as the gut microbiota of different honey bee species showed rather similar metabolic profiles in the cross-species colonisation assay. Moreover, this experiment also demonstrated that the gut microbiota of A. cerana and A. mellifera could cross colonise between the two honey bee species. Conclusion: Our findings revealed functional differences in most core gut bacteria between the guts of A. cerana and A. mellifera, which may be associated with their inter-species diversity. However, the functional profiles of the overall gut microbiomes between the two honey bee species converge, probably as a result of the overlapping ecological niches of the two species. Our findings provide critical insights into the evolution and functional roles of the mutualistic microbiota of honey bees and reveal that functional redundancy could stabilise the gene content diversity at the strain-level within the gut community.


Assuntos
Microbioma Gastrointestinal , Animais , Bactérias/genética , Abelhas/genética , Cromatografia Líquida , Microbioma Gastrointestinal/genética , Metagenoma , Espectrometria de Massas em Tandem
15.
J Neuroimmune Pharmacol ; 17(1-2): 76-93, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34993905

RESUMO

Opioid use disorder (OUD) is defined as the chronic use or misuse of prescribed or illicitly obtained opioids and is characterized by clinically significant impairment. The etiology of OUD is multifactorial as it is influenced by genetics, environmental factors, stress response and behavior. Given the profound role of the gut microbiome in health and disease states, in recent years there has been a growing interest to explore interactions between the gut microbiome and the central nervous system as a causal link and potential therapeutic source for OUD. This review describes the role of the gut microbiome and opioid-induced immunopathological disturbances at the gut epithelial surface, which collectively contribute to OUD and perpetuate the vicious cycle of addiction and relapse.


Assuntos
Analgésicos Opioides , Transtornos Relacionados ao Uso de Opioides , Humanos , Analgésicos Opioides/efeitos adversos , Sistema Nervoso
16.
J Neuroimmune Pharmacol ; 17(1-2): 367-375, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34562195

RESUMO

Prenatal opioid exposure is associated with significantly adverse medical, developmental, and behavioral outcomes in offspring, though the underlying mechanisms driving these impairments are still unclear. Accumulating evidence implicates gut microbial dysbiosis as a potential modulator of these adverse effects. However, how opioid exposure during pregnancy alters the maternal and neonatal microbiome remain to be elucidated. Here, we utilize a murine model of brief hydromorphone exposure during pregnancy (gestation day 11-13; i.p.; 10 mg/kg) to examine its impact on the maternal and neonatal microbiome. Fecal samples were collected at various timepoints in dams (4 days post hydromorphone exposure, birth, and weaning) and offspring (2, 3, and 5 weeks) to interrogate longitudinal changes in the microbiome. Stomach contents at 2 weeks were also collected as a surrogate for breastmilk and microbial analysis was performed using 16S rRNA sequencing. Alongside alterations in the maternal gut microbial composition, offspring gut microbiota exhibited distinct communities at 2 and 3 weeks. Furthermore, functional profiling of microbial communities revealed significant differences in microbial community-level phenotypes gram-negative, gram-positive, and potentially pathogenic in maternal and/or neonatal hydromorphone exposed groups compared with controls. We also observed differences in stomach microbiota in opioid-exposed vs non-exposed offspring, which suggests breast milk may also play a role in shaping the development of the neonatal gut microbiota. Together, we provide evidence of maternal and neonatal microbial dysbiosis provoked even with brief hydromorphone exposure during pregnancy.


Assuntos
Analgésicos Opioides , Hidromorfona , Animais , Camundongos , Gravidez , Feminino , Analgésicos Opioides/toxicidade , RNA Ribossômico 16S
17.
Appl Environ Microbiol ; 85(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30709823

RESUMO

Few characteristics are more important to a bacterium than the substrates it consumes. It is hard to identify what substrates are consumed by bacteria in natural communities, however, because most bacteria have not been cultured. In this study, we developed a method that uses fluorescent substrate analogs, cell sorting, and DNA sequencing to identify substrates taken up by bacteria. We deployed this method using 2[N-(7-nitrobenz-2-oxa-1,2-diaxol-4-yl)amino]-2-deoxyglucose (2-NBDG), a fluorescent glucose analog, and bacteria of the bovine rumen. This method revealed over 40 different bacteria (amplicon sequence variants [ASVs]) from the rumen that take up glucose. Nearly half of these ASVs represent previously uncultured bacteria. We attempted to grow these ASVs on agar media, and we confirmed that nearly two-thirds resisted culture. In coculture experiments, the fluorescent label of 2-NBDG was not transferred to nontarget bacteria by cross-feeding. Because it is not affected by cross-feeding, our method has an advantage over stable isotope probing. Though we focus on glucose, many substrates can be labeled with the fluorophore NBD. Our method represents a new paradigm for identifying substrates used by uncultured bacteria. It will help delineate the niche of bacteria in their environment.IMPORTANCE We introduce a method for identifying what substrates are consumed by bacteria in natural communities. Our method offers significant improvement over existing methods for studying this characteristic. Our method uses a fluorescently labeled substrate which clearly labels target bacteria (glucose consumers in our case). Previous methods use isotope-labeled substrates, which are notorious for off-target labeling (due to cross-feeding of labeled metabolites). Our method can be deployed with a variety of substrates and microbial communities. It represents a major advance in connecting bacteria to the substrates they take up.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Desoxiglucose/análogos & derivados , Glucose/análogos & derivados , Rúmen/microbiologia , Animais , Transporte Biológico , Bovinos , Citometria de Fluxo , Corantes Fluorescentes , Glucose/metabolismo , Marcação por Isótopo , RNA Ribossômico 16S/genética
18.
Environ Microbiol ; 19(11): 4670-4683, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28892251

RESUMO

Bacteria have been thought to follow only a few well-recognized biochemical pathways when fermenting glucose or other hexoses. These pathways have been chiseled in the stone of textbooks for decades, with most sources rendering them as they appear in the classic 1986 text by Gottschalk. Still, it is unclear how broadly these pathways apply, given that they were established and delineated biochemically with only a few model organisms. Here, we show that well-recognized pathways often cannot explain fermentation products formed by bacteria. In the most extensive analysis of its kind, we reconstructed pathways for glucose fermentation from genomes of 48 species and subspecies of bacteria from one environment (the rumen). In total, 44% of these bacteria had atypical pathways, including several that are completely unprecedented for bacteria or any organism. In detail, 8% of bacteria had an atypical pathway for acetate formation; 21% of bacteria had an atypical pathway for propionate or succinate formation; 6% of bacteria had an atypical pathway for butyrate formation and 33% of bacteria had an atypical or incomplete Embden-Meyerhof-Parnas pathway. This study shows that reconstruction of metabolic pathways - a common goal of omics studies - could be incorrect if well-recognized pathways are used for reference. Furthermore, it calls for renewed efforts to delineate fermentation pathways biochemically.


Assuntos
Bactérias/genética , Bactérias/metabolismo , Fermentação/genética , Glucose/metabolismo , Glicólise/genética , Rúmen/microbiologia , Acetatos/metabolismo , Animais , Bactérias/classificação , Butiratos/metabolismo , Ácidos Graxos Voláteis/metabolismo , Fermentação/fisiologia , Genoma Bacteriano/genética , Glicólise/fisiologia , Propionatos/metabolismo , Ácido Succínico/metabolismo
19.
FEMS Microbiol Ecol ; 93(6)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28486619

RESUMO

When given excess carbohydrate, certain microbial species respond by storing energy (synthesizing reserve carbohydrate), but other species respond by dissipating the energy as heat (spilling energy). To determine the importance of these responses in the rumen microbial community, this study quantified the responses of mixed ciliate protozoa vs bacteria to glucose. We hypothesized that ciliates would direct more glucose to synthesis of reserve carbohydrate (and less to energy spilling) than would bacteria. Ciliates and bacteria were isolated from rumen fluid using filtration and centrifugation, resuspended in nitrogen-free buffer to limit growth, and dosed with 5 mM glucose. Compared with bacteria, ciliates consumed glucose >3-fold faster and synthesized reserve carbohydrate 4-fold faster. They incorporated 53% of glucose carbon into reserve carbohydrate-nearly double the value (27%) for bacteria. Energy spilling was not detected for ciliates, as all heat production (104%) was accounted by synthesis of reserve carbohydrate and endogenous metabolism. For bacteria, reserve carbohydrate and endogenous metabolism accounted for only 68% of heat production, and spilling was detected within 11 min of dosing glucose. These results suggest that ciliates alter the course of ruminal carbohydrate metabolism by outcompeting bacteria for excess carbohydrate, maximizing reserve carbohydrate synthesis, and minimizing energy spilling.


Assuntos
Bactérias/metabolismo , Metabolismo dos Carboidratos/fisiologia , Cilióforos/metabolismo , Rúmen/microbiologia , Rúmen/parasitologia , Animais , Carboidratos , Bovinos , Feminino , Glucose/metabolismo , Nitrogênio/metabolismo
20.
J Med Syst ; 40(11): 228, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27628729

RESUMO

Nowadays people can get many services including health-care services from distributed information systems remotely via public network. By considering that these systems are built on public network, they are vulnerable to many malicious attacks. Hence it is necessary to introduce an effective mechanism to protect both users and severs. Recently many two-factor authentication schemes have been proposed to achieve this goal. In 2016, Li et al. demonstrated that Lee et al.'s scheme was not satisfactory to be deployed in practice because of its security weaknesses and then proposed a security enhanced scheme to overcome these drawbacks. In this paper, we analyze Li et al.'s scheme is still not satisfactory to be applied in telecare medicine information systems (TMIS) because it fails to withstand off-line dictionary attack and known session-specific temporary information attack. Moreover, their scheme cannot provide card revocation services for lost smart card. In order to solve these security problems, we propose an improved scheme. Then we analyze our scheme by using BAN-logic model and compare the improved scheme with related schemes to prove that our scheme is advantageous to be applied in practice.


Assuntos
Segurança Computacional/instrumentação , Cartões Inteligentes de Saúde , Sistemas de Informação/instrumentação , Telemedicina/instrumentação , Confidencialidade , Troca de Informação em Saúde/normas , Humanos , Dinâmica não Linear
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...