Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Crit Rev Food Sci Nutr ; : 1-19, 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37129118

RESUMEN

The use of natural bioactive constituents from various food sources for anticancer purposes has become increasingly popular worldwide. Broccoli (Brassica oleracea var. italica) is on the top of the consumed vegetables by the masses. Its raw matrix contains a plethora of phytochemicals, such as glucosinolates and phenolic compounds, along with rich amounts of vitamins, and minerals. Consumption of broccoli-derived phytochemicals provides strong antioxidant effects, particularly due to its sulforaphane content, while modulating numerous molecules involved in cell cycle regulation, control of apoptosis, and tuning enzyme activity. Thus, the inclusion of broccoli in the daily diet lowers the susceptibility to developing cancers. Numerous studies have underlined the undisputable role of broccoli in the diet as a chemopreventive raw food, owing to the content in sulforaphane, an isothiocyanate produced as a result of hydrolysis of precursor glucosinolates called glucoraphanin. This review will provide evidence supporting the specific role of fresh florets and sprouts of broccoli and its key bioactive constituents in the prevention and treatment of different cancers; a number of studies carried out in the in vitro and in vivo conditions as well as clinical trials were analyzed.

2.
Pediatr Res ; 91(2): 447-453, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34952937

RESUMEN

Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. Sepsis has a direct impact on the immune system due to adverse alteration of the production, maturation, function, and apoptosis of immune cells. Dysregulation in both the innate and adaptive immune responses during sepsis leads to a range of phenotypes consisting of both hyperinflammation and immunosuppression that can result in immunoparalysis. In this review, we discuss components of immune dysregulation in sepsis, biomarkers and functional immune assays to aid in immunophenotyping patients, and evolving immunomodulatory therapies. Important research gaps for the future include: (1) Defining how age, host factors including prior exposures, and genetics impact the trajectory of sepsis in children, (2) Developing tools for rapid assessment of immune function in sepsis, and (3) Assessing how evolving pediatric sepsis endotypes respond differently to immunomodulation. Although multiple promising immunomodulatory agents exist or are in development, access to rapid immunophenotyping will be needed to identify which children are most likely to benefit from which therapy. Advancements in the ability to perform multidimensional endotyping will be key to developing a personalized approach to children with sepsis. IMPACT: Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. The resulting spectrum of immunoparalysis that can occur due to sepsis can increase morbidity and mortality in children and adults. This narrative review summarizes the current literature surrounding biomarkers and functional immunologic assays for immune dysregulation in sepsis, with a focus on immunomodulatory therapies that have been evaluated in sepsis. A precision approach toward diagnostic endotyping and therapeutics, including gene expression, will allow for optimal clinical trials to evaluate the efficacy of individualized and targeted treatments for pediatric sepsis.


Asunto(s)
Sepsis/inmunología , Biomarcadores/metabolismo , Niño , Humanos , Factores Inmunológicos/uso terapéutico , Sepsis/tratamiento farmacológico
3.
J Pediatr ; 228: 294-296, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32861693

RESUMEN

We present 4 pediatric patients with trisomy 21 (T21) and associated comorbidities who developed coronavirus disease 2019 requiring hospitalization. A review of the literature revealed that comorbidities associated with T21 may predispose patients to severe disease. Children with T21 should be considered high risk and monitored carefully if infected with severe acute respiratory syndrome coronavirus 2.


Asunto(s)
COVID-19/complicaciones , COVID-19/epidemiología , Comorbilidad , Susceptibilidad a Enfermedades , Síndrome de Down/complicaciones , Síndrome de Down/epidemiología , Adolescente , Hospitalización , Humanos , Lactante , Masculino , Factores de Riesgo , SARS-CoV-2
4.
Biol Blood Marrow Transplant ; 26(11): 2053-2060, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32682948

RESUMEN

Certain anaerobic bacteria are important for maintenance of gut barrier integrity and immune tolerance and may influence the risk of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). We conducted a single-center retrospective cohort study of allogeneic HSCT recipients to evaluate associations between receipt of antibiotics with an anaerobic spectrum of activity and GVHD outcomes. We identified 1214 children and adults who developed febrile neutropenia between 7 days before and 28 days after HSCT and compared GVHD risk and mortality among patients who received anaerobic antibiotics (piperacillin-tazobactam or carbapenems; n = 491) to patients who received only antibiotics with minimal activity against anaerobes (aztreonam, cefepime, or ceftazidime; n = 723). We performed metagenomic sequencing of serial fecal samples from 36 pediatric patients to compare the effects of specific antibiotics on the gut metagenome. Receipt of anaerobic antibiotics was associated with higher hazards of acute gut/liver GVHD (hazard ratio [HR], 1.26; 95% confidence interval [CI], 1.03 to 1.54) and acute GVHD mortality (HR, 1.63; 95% CI, 1.08 to 2.46), but not chronic GVHD diagnosis (HR, 1.04; 95% CI: .84 to 1.28) or chronic GVHD mortality (HR, .88; 95% CI, .53 to 1.45). Anaerobic antibiotics resulted in decreased gut bacterial diversity, reduced abundances of Bifidobacteriales and Clostridiales, and loss of bacterial genes encoding butyrate biosynthesis enzymes from the gut metagenome. Acute gut/liver GVHD was preceded by a sharp decline in bacterial butyrate biosynthesis genes with antibiotic treatment. Our findings demonstrate that exposure to anaerobic antibiotics is associated with increased risks of acute gut/liver GVHD and acute GVHD mortality after allogeneic HSCT. Use of piperacillin-tazobactam or carbapenems should be reserved for febrile neutropenia cases in which anaerobic or multidrug-resistant infections are suspected.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Adulto , Anaerobiosis , Antibacterianos/uso terapéutico , Niño , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Estudios Retrospectivos , Trasplante Homólogo
5.
Artículo en Inglés | MEDLINE | ID: mdl-33077659

RESUMEN

We previously identified a small-molecule inhibitor of capsule biogenesis (designated DU011) and identified its target as MprA, a MarR family transcriptional repressor of multidrug efflux pumps. Unlike other proposed MprA ligands, such as salicylate and 2,4-dinitrophenol (DNP), DU011 does not alter Escherichia coli antibiotic resistance and has significantly enhanced inhibition of capsule expression. We hypothesized that the potency and the unique action of DU011 are due to novel interactions with the MprA binding pocket and the conformation assumed by MprA upon binding DU011 relative to other ligands. To understand the dynamics of MprA-DU011 interaction, we performed hydrogen-deuterium exchange mass spectrometry (HDX-MS); this suggested that four peptide regions undergo conformational changes upon binding DU011. We conducted isothermal calorimetric titration (ITC) to quantitatively characterize MprA binding to DU011 and canonical ligands and observed a distinct two-site binding isotherm associated with the binding reaction of MprA to DU011; however, salicylate and DNP showed a one-site binding isotherm with lower affinity. To elucidate the binding pocket(s) of MprA, we selected single point mutants of MprA that included mutated residues predicted to be within the putative binding pocket (Q51A, F58A, and E65D) as well as on or near the DNA-binding domain (L81A, S83T, and T86A). Our ITC studies suggest that two of the tested MprA mutants had lower affinity for DU011: Q51A and F58A. In addition to elucidating the MprA binding pocket for DU011, we studied the binding of these mutants to salicylate and DNP to reveal the binding pockets of these canonical ligands.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Antibacterianos/farmacología , Sitios de Unión , Farmacorresistencia Microbiana , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Ligandos , Polisacáridos , Unión Proteica
6.
Biol Blood Marrow Transplant ; 25(11): 2274-2280, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31326608

RESUMEN

The gastrointestinal tract is the predicted reservoir for most bloodstream infections (BSIs) after hematopoietic stem cell transplantation (HSCT). Whole-genome sequencing and comparative genomics have the potential to improve our understanding of the dynamics of gut colonization that precede BSI in HSCT recipients. Within a prospective cohort study of children (age <18 years) undergoing HSCT, 9 subjects met criteria for mucosal barrier injury BSI. We performed whole-genome sequencing of the blood culture isolate and weekly fecal samples preceding the BSI to compare the genetic similarity of BSI isolates to fecal strains. We evaluated temporal associations between antibiotic exposures and the abundances of BSI strains in the gut microbiota and correlated the detection of antibiotic resistance genes with the phenotypic antibiotic resistance of these strains. The median patient age was 2.6 years, and 78% were male. BSIs were caused by Escherichia coli (n = 5), Enterococcus faecium (n = 2), Enterobacter cloacae (n = 1), and Rothia mucilaginosa (n = 1). In the 6 BSI episodes with evaluable comparative genomics, the fecal strains were identical to the blood culture isolate (>99.99% genetic similarity). Gut domination by these strains preceded only 4 of 7 E. coli or E. faecium BSIs by a median of 17 days (range, 6 to 21 days). Increasing abundances of the resulting BSI strains in the gut microbiota were frequently associated with specific antibiotic exposures. E. cloacae and R. mucilaginosa were not highly abundant in fecal samples preceding BSIs caused by these species. The detection of antibiotic resistance genes for ß-lactam antibiotics and vancomycin predicted phenotypic resistance in BSI strains. Bacterial strains causing mucosal barrier injury BSI in pediatric HSCT recipients were observed in the gut microbiota before BSI onset, and changes in the abundances of these strains within the gut preceded most BSI episodes. However, frequent sampling of the gut microbiota and sampling of other ecological niches is likely necessary to effectively predict BSI in HSCT recipients.


Asunto(s)
Bacterias , Infecciones Bacterianas , Farmacorresistencia Bacteriana , Microbioma Gastrointestinal , Trasplante de Células Madre Hematopoyéticas , Mucosa Intestinal , Aloinjertos , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/genética , Infecciones Bacterianas/microbiología , Niño , Preescolar , Femenino , Humanos , Mucosa Intestinal/lesiones , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Estudios Prospectivos
7.
Biol Blood Marrow Transplant ; 25(4): 756-763, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30481599

RESUMEN

Infections and graft-versus-host disease (GVHD) have historically resulted in high mortality among children undergoing umbilical cord blood transplantation (UCBT). However, recent advances in clinical practice have likely improved outcomes of these patients. We conducted a retrospective cohort study of children (<18years of age) undergoing UCBT at Duke University between January 1, 1995 and December 31, 2014. We compared 2-year all-cause and cause-specific mortality during 3 time periods based on year of transplantation (1995 to 2001, 2002 to 2007, and 2008 to 2014). We used multivariable Cox regression to identify demographic and UCBT characteristics that were associated with all-cause mortality, transplantation-related mortality, and death from invasive aspergillosis after adjustment for time period. During the 20-year study period 824 children underwent UCBT. Two-year all-cause mortality declined from 48% in 1995 to 2001 to 30% in 2008 to 2014 (P = .0002). White race and nonmalignant UCBT indications were associated with lower mortality. Black children tended to have a higher risk of death for which GVHD (18% versus 11%; P = .06) or graft failure (9% versus 3%; P = .01) were contributory than white children. Comparing 2008 to 2014 with 1995 to 2001, more than half (59%) of the reduced mortality was attributable to a reduction in infectious mortality, with 45% specifically related to reduced mortality from invasive aspergillosis. Antifungal prophylaxis with voriconazole was associated with lower mortality from invasive aspergillosis than low-dose amphotericin B lipid complex (hazard ratio, .09; 95% confidence interval, .01 to .76). With the decline in mortality from invasive aspergillosis, adenovirus and cytomegalovirus have become the most frequentinfectious causes of death in children after UCBT. Advances in clinical practice over the past 20years improved survival of children after UCBT. Reduced mortality from infections, particularly invasive aspergillosis, accounted for the largest improvement in survival and was associated with use of voriconazole for antifungal prophylaxis.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/mortalidad , Estudios de Cohortes , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Masculino , Estudios Retrospectivos , Tasa de Supervivencia , Factores de Tiempo
8.
Biol Blood Marrow Transplant ; 24(9): 1861-1865, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29894730

RESUMEN

Autopsy may confirm clinical diagnoses or identify conditions that were not suspected prior to a patient's death. Previous studies evaluating the utility of autopsy in hematopoietic stem cell transplant (HSCT) recipients yielded conflicting results. We conducted a retrospective cohort study of children (<18 years of age) undergoing allogeneic HSCT at Duke University who died of any cause between January 1, 1995, and December 31, 2016. We evaluated associations between patient characteristics and autopsy performance using chi-square or Fisher exact tests. We reviewed autopsy reports to determine the concordance between preautopsy causes of death and pathological diagnoses identified on autopsy. We classified unexpected diagnoses on autopsy using criteria developed by Goldman et al. We evaluated for temporal changes in the autopsy consent rate and the frequency of unexpected diagnoses on autopsy using Cochran-Armitage tests. During the 22-year study period, 475 patients died and had data available on autopsy performance, and 130 (27%) of these patients underwent autopsy. The autopsy consent rate declined over time (P < .0001), with autopsies being performed for 40% of deaths in 1995 to 1999 and 17% of deaths in 2009 to 2016. White patients were more likely to undergo autopsy than nonwhite patients (P = .03). There were no associations between autopsy performance and patient age, sex, HSCT indication, or HSCT donor. Unexpected diagnoses were identified in 31 (24%) autopsies. The proportion of autopsies with an unexpected diagnosis did not change during the study period (P = .45). However, infectious diagnoses that would have led to a change in management were more frequently identified on autopsies in 1995 to 2003 than in 2004 to 2016 (20% versus 0%; P = .001). The autopsy consent rate for pediatric HSCT recipients at our institution has declined substantially over the past several decades. The utility of autopsy in this patient population remains high despite a reduction in the identification of unexpected infections.


Asunto(s)
Autopsia/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo/métodos , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Estudios Retrospectivos
9.
J Infect Dis ; 213(8): 1330-9, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26671885

RESUMEN

BACKGROUND: Uropathogenic Escherichia coli (UPEC), a leading cause of urinary tract and invasive infections worldwide, is rapidly acquiring multidrug resistance, hastening the need for selective new anti-infective agents. Here we demonstrate the molecular target of DU011, our previously discovered potent, nontoxic, small-molecule inhibitor of UPEC polysaccharide capsule biogenesis and virulence. METHODS: Real-time polymerase chain reaction analysis and a target-overexpression drug-suppressor screen were used to localize the putative inhibitor target. A thermal shift assay quantified interactions between the target protein and the inhibitor, and a novel DNase protection assay measured chemical inhibition of protein-DNA interactions. Virulence of a regulatory target mutant was assessed in a murine sepsis model. RESULTS: MprA, a MarR family transcriptional repressor, was identified as the putative target of the DU011 inhibitor. Thermal shift measurements indicated the formation of a stable DU011-MprA complex, and DU011 abrogated MprA binding to its DNA promoter site. Knockout of mprA had effects similar to that of DU011 treatment of wild-type bacteria: a loss of encapsulation and complete attenuation in a murine sepsis model, without any negative change in antibiotic resistance. CONCLUSIONS: MprA regulates UPEC polysaccharide encapsulation, is essential for UPEC virulence, and can be targeted without inducing antibiotic resistance.


Asunto(s)
Antibacterianos/farmacología , Cápsulas Bacterianas/metabolismo , Descubrimiento de Drogas/métodos , Proteínas de Escherichia coli/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen/métodos , Proteínas Represoras/antagonistas & inhibidores , Escherichia coli Uropatógena/genética , Animales , Antibacterianos/química , Cápsulas Bacterianas/efectos de los fármacos , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Represoras/genética , Escherichia coli Uropatógena/efectos de los fármacos , Virulencia
11.
Curr Med Chem ; 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38251695

RESUMEN

BACKGROUND: The molecular mechanisms regulating coronavirus pathogenesis are complex, including virus-host interactions associated with replication and innate immune control. However, some genetic and epigenetic conditions associated with comorbidities increase the risk of hospitalization and can prove fatal in infected patients. This systematic review will provide insight into host genetic and epigenetic factors that interfere with COVID-19 expression in light of available evidence. METHODS: This study conducted a systematic review to examine the genetic and epigenetic susceptibility to COVID-19 using a comprehensive approach. Through systematic searches and applying relevant keywords across prominent online databases, including Scopus, PubMed, Web of Science, and Science Direct, we compiled all pertinent papers and reports published in English between December 2019 and June 2023. RESULTS: The findings reveal that the host's HLA genotype plays a substantial role in determining how viral protein antigens are showcased and the subsequent immune system reaction to these antigens. Within females, genes responsible for immune system regulation are found on the X chromosome, resulting in reduced viral load and inflammation levels when contrasted with males. Possessing blood group A may contribute to an increased susceptibility to contracting COVID-19 as well as a heightened risk of mortality associated with the disease. The capacity of SARS-CoV-2 involves inhibiting the antiviral interferon (IFN) reactions, resulting in uncontrolled viral multiplication. CONCLUSION: There is a notable absence of research into the gender-related predisposition to infection, necessitating a thorough examination. According to the available literature, a significant portion of individuals affected by the ailment or displaying severe ramifications already had suppressed immune systems, categorizing them as a group with elevated risk.

12.
Gut Microbes ; 16(1): 2333748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38555499

RESUMEN

Antibiotic resistance is a global threat driven primarily by antibiotic use. We evaluated the effects of antibiotic exposures on the gut microbiomes and resistomes of children at high risk of colonization by antibiotic-resistant bacteria. We performed shotgun metagenomic sequencing of 691 serially collected fecal samples from 80 children (<18 years) undergoing hematopoietic cell transplantation. We evaluated the effects of aerobic (cefepime, vancomycin, fluoroquinolones, aminoglycosides, macrolides, and trimethoprim-sulfamethoxazole) and anaerobic (piperacillin-tazobactam, carbapenems, metronidazole, and clindamycin) antibiotic exposures on the diversity and composition of the gut microbiome and resistome. We identified 372 unique antibiotic resistance genes (ARGs); the most frequent ARGs identified encode resistance to tetracyclines (n = 88), beta-lactams (n = 84), and fluoroquinolones (n = 79). Both aerobic and anaerobic antibiotic exposures were associated with a decrease in the number of bacterial species (aerobic, ß = 0.71, 95% CI: 0.64, 0.79; anaerobic, ß = 0.66, 95% CI: 0.53, 0.82) and the number of unique ARGs (aerobic, ß = 0.81, 95% CI: 0.74, 0.90; anaerobic, ß = 0.73, 95% CI: 0.61, 0.88) within the gut metagenome. However, only antibiotic regimens that included anaerobic activity were associated with an increase in acquisition of new ARGs (anaerobic, ß = 1.50; 95% CI: 1.12, 2.01) and an increase in the relative abundance of ARGs in the gut resistome (anaerobic, ß = 1.62; 95% CI: 1.15, 2.27). Specific antibiotic exposures were associated with distinct changes in the number and abundance of ARGs for individual antibiotic classes. Our findings detail the impact of antibiotics on the gut microbiome and resistome and demonstrate that anaerobic antibiotics are particularly likely to promote acquisition and expansion of antibiotic-resistant bacteria.


Asunto(s)
Microbioma Gastrointestinal , Trasplante de Células Madre Hematopoyéticas , Niño , Humanos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias/genética , Fluoroquinolonas/farmacología , Microbioma Gastrointestinal/genética
13.
Health Technol (Berl) ; 12(1): 205-214, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35036281

RESUMEN

Coronavirus disease 2019 (COVID-19) is caused by a novel coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It originated from the Chinese city of Wuhan and very quickly became a challenging public health problem. On 11 March 2020, the World Health Organization termed this potentially deadly disease a pandemic due to its rapid spread in various parts of the world, giving rise to international health emergencies. This virus is transmitted from human to human in the form of respiratory droplets, and in specific circumstances, airborne transmission may occur. Additional sources of exposure for dentists include blood and infected sharps. Due to the contagious nature of COVID-19 many health care providers have also been disproportionately affected, such as physicians, dentists, nurses, and paramedical staff. Dentists and dental staff are at high risk of cross-infection due to their nature of work. Therefore, they face a dual challenge in protecting themselves and their patients from infection transmission while ensuring that patients receive urgent dental care. In this review, the authors highlight the epidemiology, modes of cross-infection, and recent data on SARS-CoV-2 related to dental practice. The primary purpose is to make dental health care providers aware of the pathophysiology of COVID-19 and to increase their preparedness and understanding of this challenge, which will aid in controlling transmission. The information collected will be useful for the dental community in providing effective patient management through evidence-based recommendations for infection control and disinfection protocols.

14.
Microbiol Spectr ; 10(5): e0058222, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36121240

RESUMEN

Extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli can cause invasive infections in infants and immunocompromised children with high associated morbidity and mortality. The gut is a major reservoir of these strains in the community. Current dogma dictates that antimicrobial resistance is associated with a fitness cost. However, recent data show that some contemporary ESBL E. coli strains may be more "fit" compared to nonresistant E. coli strains. Here, we use whole-genome sequencing to first characterize 15 ESBL E. coli strains isolated from infants in a Pakistani community, a clinical extraintestinal pathogenic ESBL E. coli ST131 strain, and a non-ESBL commensal E. coli strain, and then use a novel animal model of early life gut colonization to assess the ability of these strains to colonize the infant mouse gut. We determined that CTX-M-15 was present in all the ESBL strains, as well as additional beta-lactamases and genes conferring resistance to multiple antibiotic classes. In the animal model, 11/16 ESBL E. coli strains had significantly higher burden of colonization at week four of life compared to commensal strains, even in the absence of selective antibiotic pressure, suggesting that these strains may have enhanced fitness despite being highly antimicrobial resistant. IMPORTANCE Antimicrobial resistance is a global public health emergency. Infants, especially preterm infants and those in the neonatal intensive care unit, immunocompromised hosts, and those with chronic illnesses are at highest risk of adverse outcomes from invasive infections with antimicrobial-resistant strains. It has long been thought that resistance is associated with a fitness cost, i.e., antimicrobial-resistant strains are not able to colonize the gut as well as nonresistant strains, and that antibiotic exposure is a key risk factor for persistent colonization with resistant strains. Here, we use a novel infant mouse model to add to the growing body of literature that some highly-resistant contemporary Escherichia coli strains can persist in the gut with a significant burden of colonization despite absence of antibiotic exposure.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Recién Nacido , Humanos , Ratones , Animales , Escherichia coli/genética , Animales Recién Nacidos , Recien Nacido Prematuro , beta-Lactamasas/genética , Antibacterianos/farmacología
15.
Artículo en Inglés | MEDLINE | ID: mdl-36168465

RESUMEN

Measures to prevent coronavirus disease 2019 (COVID-19) spread to household members was assessed by surveying COVID-19-positive physicians and advanced practice providers. Showering and changing were more common than physical distancing. Half of respondents reported a symptomatic household member. Most reported increased stress, worsening of mental health, and concerns about illness and impact on family.

16.
Clin Ther ; 42(9): 1637-1648, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32800382

RESUMEN

The prevalence of multidrug-resistant organisms is increasing worldwide, posing a unique challenge to global health care systems. Novel approaches are needed to combat the spread of infection with these organisms. The enteric microbiome, and in particular the resistome, offers a unique target in both the prevention of infection with these organisms and the acquisition and spread within the community. We highlight a novel approach to combat multidrug-resistant organisms: the use of prebiotics, probiotics, and synbiotics to manipulate the microbiome and resistome. This review summarizes the published literature and clinical trials related to these products to date, with a focus on efficacious trials. It highlights the probable mechanism of action for each product, as well as its safety profile in selective populations. Ultimately, although further research is needed before a definitive statement can be made on the efficacy of any of these 3 interventions, the literature to date offers new hope and a new tool in the arsenal in the fight against bacterial drug resistance.


Asunto(s)
Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Simbióticos/administración & dosificación , Farmacorresistencia Bacteriana Múltiple , Humanos , Probióticos/uso terapéutico
17.
Protein Sci ; 29(3): 647-653, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31682303

RESUMEN

There has been a rapid spread of multidrug-resistant (MDR) bacteria across the world. MDR efflux transporters are an important mechanism of antibiotic resistance in many pathogens among both Gram positive and Gram negative bacteria. These pumps can recognize a variety of chemically and structurally different compounds, including innate and clinically administered antibiotics. Intriguingly, these efflux pumps are often regulated by transcription factors that themselves bind a diverse set of substrates thereby allowing them to regulate the expression of their cognate MDR efflux pumps. One significant family of such transcription factors is the Multiple antibiotic resistance Repressor (MarR) family. Members of this family are well conserved across different bacterial species and in some cases are known to regulate vital bacterial functions. This review focusses on the role of MarR family transcriptional factors in antibiotic resistance within a select group of clinically relevant pathogens.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Antibacterianos/química , Proteínas de Escherichia coli/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Proteínas Represoras/metabolismo
18.
Antibiotics (Basel) ; 9(6)2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32471150

RESUMEN

The recent rapid rise of multi-drug resistant Enterobacteriaceae (MDR-E) is threatening the treatment of common infectious diseases. Infections with such strains lead to increased mortality and morbidity. Using a cross-sectional study, we aimed to estimate the prevalence of gut colonization with extended spectrum beta-lactamase (ESBL) producing Enterobacteriaceae among healthy infants born in Pakistan, a setting with high incidence of MDR-E infections. Stool samples were collected from 104 healthy infants between the ages of 5 and 7 months. Enterobacteriaceae isolates were screened for resistance against several antimicrobial classes. Presence of ESBL and carbapenemase genes was determined using multiplex PCR. Sequence types were assigned to individual strains by multi-locus sequence typing. Phylogenetic analysis of Escherichia coli was done using the triplex PCR method. Forty-three percent of the infants were positive for ESBL-producing Enterobacteriaceae, the majority of which were E. coli. We identified several different ESBL E. coli sequence types most of which belonged to the phylogenetic group B2 (23%) or D (73%). The widespread colonization of infants in a developing country with ESBL-producing Enterobacteriaceae is concerning. The multiple sequence types and reported non-human sources support that multiple non-epidemic MDR lineages are circulating in Pakistan with healthy infants as a common reservoir.

19.
J Pediatric Infect Dis Soc ; 8(2): 152-159, 2019 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-30053044

RESUMEN

Recurrent urinary tract infection (rUTI) continues to challenge pediatric care providers. The diagnosis of an rUTI can be difficult, especially in young febrile children. Antibiotic resistance rates continue to rise, which limits oral treatment options. Prophylactic antibiotics are used commonly to manage rUTI, but their use increases the risk of rUTI with antibiotic-resistant strains without significantly reducing renal scarring. Alternative therapies for rUTI include probiotics and anthocyanidins (eg, cranberry extract) to reduce gut colonization by uropathogens and prevent bacterial adhesion to uroepithelia, but efficacy data for these treatments are sparse. The future of rUTI care rests in addressing the following contemporary issues: best diagnostic practices, risk factors associated with rUTI, and the prevention of recurrent infection. In this review, we summarize the state of the art for each of these issues and highlight future studies that will aim to take an alternative approach to managing rUTI.


Asunto(s)
Profilaxis Antibiótica/métodos , Infecciones Urinarias/diagnóstico , Infecciones Urinarias/tratamiento farmacológico , Antocianinas/uso terapéutico , Antibacterianos/uso terapéutico , Adhesión Bacteriana , Circuncisión Masculina/métodos , Farmacorresistencia Microbiana , Jugos de Frutas y Vegetales , Humanos , Masculino , Probióticos/uso terapéutico , Recurrencia , Factores de Riesgo , Enfermedades de la Vejiga Urinaria , Infecciones Urinarias/epidemiología , Infecciones Urinarias/prevención & control , Vacunas , Vaccinium macrocarpon
20.
Open Forum Infect Dis ; 5(6): ofy107, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29977964

RESUMEN

BACKGROUND: Rates and risk factors for recurrent enterococcal bloodstream infection (R-EBSI) and whether the same genetic lineage causes index EBSI and R-EBSI are unknown in patients with acute leukemia (AL) receiving chemotherapy. METHODS: Ninety-two AL patients with EBSI from 2010 to 2015 were included. Enterococcal bloodstream infection was defined by 31 positive blood cultures for Enterococcus faecium or Enterococcus faecalis and fever, hypotension, or chills. Clearance was defined by 31 negative cultures 324 hours after last positive culture and defervescence. Recurrent enterococcal bloodstream infection was defined by a positive blood culture for Enterococcus 324 hours after clearance. Categorical variables were reported as proportions and compared by the χ2 test. Continuous variables were summarized by median and interquartile range (IQR) and compared by the Wilcoxon-Mann-Whitney Test. P values <.05 were considered significant. Whole-genome sequencing was performed on available paired BSI isolates from 7 patients. RESULTS: Twenty-four patients (26%) had 31 episodes of R-EBSI. Median time to R-EBSI (IQR) was 26 (13-50) days. Patients with R-EBSI had significantly longer durations of fever and metronidazole exposure during their index EBSI. Thirty-nine percent of E. faecium R-EBSI isolates became daptomycin-nonsusceptible Enterococcus (DNSE) following daptomycin therapy for index EBSI. Whole-genome sequencing analysis confirmed high probability of genetic relatedness of index EBSI and R-EBSI isolates for 4/7 patients. CONCLUSIONS: Recurrent enterococcal bloodstream infection and DNSE are common in patients with AL and tend to occur within the first 30 days of index EBSI. Duration of fever and metronidazole exposure may be useful in determining risk for R-EBSI. Whole-genome sequencing analysis demonstrates that the same strain causes both EBSI and R-EBSI in some patients.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda