Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nutr Res Rev ; : 1-9, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38586996

RESUMO

Iron is essential for many physiological functions of the body, and it is required for normal growth and development. Iron deficiency (ID) is the most common form of micronutrient malnutrition and is particularly prevalent in infants and young children in developing countries. Iron supplementation is considered the most effective strategy to combat the risk of ID and ID anaemia (IDA) in infants, although iron supplements cause a range of deleterious gut-related problems in malnourished children. The purpose of this review is to assess the available evidence on the effect of iron supplementation on the gut microbiota during childhood ID and to further assess whether prebiotics offer any benefits for iron supplementation. Prebiotics are well known to improve gut-microbial health in children, and recent reports indicate that prebiotics can mitigate the adverse gut-related effects of iron supplementation in children with ID and IDA. Thus, provision of prebiotics alongside iron supplements has the potential for an enhanced strategy for combatting ID and IDA among children in the developing world. However, further understanding is required before the benefit of such combined treatments of ID in nutritionally deprived children across populations can be fully confirmed. Such enhanced understanding is of high relevance in resource-poor countries where ID, poor sanitation and hygiene, alongside inadequate access to good drinking water and poor health systems, are serious public health concerns.

2.
Angew Chem Int Ed Engl ; 63(16): e202401379, 2024 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-38407997

RESUMO

Ferritins are multimeric cage-forming proteins that play a crucial role in cellular iron homeostasis. All H-chain-type ferritins harbour a diiron site, the ferroxidase centre, at the centre of a 4 α-helical bundle, but bacterioferritins are unique in also binding 12 hemes per 24 meric assembly. The ferroxidase centre is known to be required for the rapid oxidation of Fe2+ during deposition of an immobilised ferric mineral core within the protein's hollow interior. In contrast, the heme of bacterioferritin is required for the efficient reduction of the mineral core during iron release, but has little effect on the rate of either oxidation or mineralisation of iron. Thus, the current view is that these two cofactors function in iron uptake and release, respectively, with no functional overlap. However, rapid electron transfer between the heme and ferroxidase centre of bacterioferritin from Escherichia coli was recently demonstrated, suggesting that the two cofactors may be functionally connected. Here we report absorbance and (magnetic) circular dichroism spectroscopies, together with in vitro assays of iron-release kinetics, which demonstrate that the ferroxidase centre plays an important role in the reductive mobilisation of the bacterioferritin mineral core, which is dependent on the heme-ferroxidase centre electron transfer pathway.


Assuntos
Ceruloplasmina , Ferro , Ferro/química , Ceruloplasmina/química , Escherichia coli/metabolismo , Ferritinas/química , Proteínas de Bactérias/metabolismo , Grupo dos Citocromos b/química , Minerais , Oxirredução , Heme/metabolismo
3.
Microbiology (Reading) ; 169(3)2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36947574

RESUMO

Staphylococcus aureus is a common colonizer of the human gut and in doing so it must be able to resist the actions of the host's innate defences. Bile salts are a class of molecules that possess potent antibacterial activity that control growth. Bacteria that colonize and survive in that niche must be able to resist the action of bile salts, but the mechanisms by which S. aureus does so are poorly understood. Here we show that FadB is a bile-induced oxidoreductase which mediates bile salt resistance and when heterologously expressed in Escherichia coli renders them resistant. Deletion of fadB attenuated survival of S. aureus in a model of the human distal colon.


Assuntos
Colatos , Infecções Estafilocócicas , Humanos , Staphylococcus aureus/genética , Ácidos e Sais Biliares/farmacologia , Oxirredutases
4.
Biometals ; 35(3): 573-589, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35348940

RESUMO

EfeUOB/M has been characterised in Pseudomonas syringae pathovar. syringae as a novel type of ferrous-iron transporter, consisting of an inner-membrane protein (EfeUPsy) and three periplasmic proteins (EfeOPsy, EfeMPsy and EfeBPsy). The role of an iron permease and peroxidase function has been identified for the EfeU and EfeB proteins, respectively, but the role of EfeO/M remains unclear. EfeMPsy is an 'M75-only' EfeO-like protein with a C-terminal peptidase-M75 domain (EfeOII/EfeM family). Herein, we report the 1.6 Å resolution crystal structure of EfeMPsy, the first structural report for an EfeM component of P. syringae pv. syringae. The structure possesses the bi-lobate architecture found in other bacterial periplasmic substrate/solute binding proteins. Metal binding studies, using SRCD and ICP-OES, reveal a preference of EfeMPsy for copper, iron and zinc. This work provides detailed knowledge of the structural scaffold, the metal site geometry, and the divalent metal binding potential of EfeM. This work provides crucial underpinning for a more detailed understanding of the role of EfeM/EfeO proteins and the peptidase-M75 domains in EfeUOB/M iron uptake systems in bacteria.


Assuntos
Ferro , Pseudomonas syringae , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Transporte de Íons , Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Peptídeo Hidrolases/metabolismo , Pseudomonas syringae/metabolismo
5.
J Bacteriol ; 200(24)2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30249704

RESUMO

The RIC (repair of iron clusters) protein of Escherichia coli is a di-iron hemerythrin-like protein that has a proposed function in repairing stress-damaged iron-sulfur clusters. In this work, we performed a bacterial two-hybrid screening to search for RIC-protein interaction partners in E. coli As a result, the DNA-binding protein from starved cells (Dps) was identified, and its potential interaction with RIC was tested by bacterial adenylate cyclase-based two-hybrid (BACTH) system, bimolecular fluorescence complementation, and pulldown assays. Using the activity of two Fe-S-containing enzymes as indicators of cellular Fe-S cluster damage, we observed that strains with single deletions of ric or dps have significantly lower aconitase and fumarase activities. In contrast, the ric dps double mutant strain displayed no loss of aconitase and fumarase activity with respect to that of the wild type. Additionally, while complementation of the ric dps double mutant with ric led to a severe loss of aconitase activity, this effect was no longer observed when a gene encoding a di-iron site variant of the RIC protein was employed. The dps mutant exhibited a large increase in reactive oxygen species (ROS) levels, but this increase was eliminated when ric was also inactivated. Absence of other iron storage proteins, or of peroxidase and catalases, had no impact on RIC-mediated redox stress induction. Hence, we show that RIC interacts with Dps in a manner that serves to protect E. coli from RIC protein-induced ROS.IMPORTANCE The mammalian immune system produces reactive oxygen and nitrogen species that kill bacterial pathogens by damaging key cellular components, such as lipids, DNA, and proteins. However, bacteria possess detoxifying and repair systems that mitigate these deleterious effects. The Escherichia coli RIC (repair of iron clusters) protein is a di-iron hemerythrin-like protein that repairs stress-damaged iron-sulfur clusters. E. coli Dps is an iron storage protein of the ferritin superfamily with DNA-binding capacity that protects cells from oxidative stress. This work shows that the E. coli RIC and Dps proteins interact in a fashion that counters RIC protein-induced reactive oxygen species (ROS). Altogether, we provide evidence for the formation of a new bacterial protein complex and reveal a novel contribution for Dps in bacterial redox stress protection.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Espécies Reativas de Oxigênio/metabolismo , Aconitato Hidratase/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Fumarato Hidratase/metabolismo , Regulação Bacteriana da Expressão Gênica , Mutação , Oxirredução , Técnicas do Sistema de Duplo-Híbrido
6.
Food Microbiol ; 53(Pt B): 82-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26678134

RESUMO

Salmonella enterica serovar Enteritidis is the prevalent egg-product-related food-borne pathogen. The egg-contamination capacity of S. Enteritidis includes its exceptional survival capability within the harsh conditions provided by egg white. Egg white proteins, such as lysozyme and ovotransferrin, are well known to play important roles in defence against bacterial invaders. Indeed, several additional minor proteins and peptides have recently been found to play known or potential roles in protection against bacterial contamination. However, although such antibacterial proteins are well studied, little is known about their efficacy under the environmental conditions prevalent in egg white. Thus, the influence of factors such as temperature, alkalinity, nutrient restriction, viscosity and cooperative interactions on the activities of antibacterial proteins in egg white remains unclear. This review critically assesses the available evidence on the antimicrobial components of egg white. In addition, mechanisms employed by S. Enteritidis to resist egg white exposure are also considered along with various genetic studies that have shed light upon egg white resistance systems. We also consider how multiple, antibacterial proteins operate in association with specific environmental factors within egg white to generate a lethal protective cocktail that preserves sterility.


Assuntos
Clara de Ovo/microbiologia , Salmonella enteritidis/crescimento & desenvolvimento , Animais , Galinhas , Meios de Cultura/metabolismo , Proteínas do Ovo/metabolismo , Salmonella enteritidis/metabolismo
7.
Int J Food Sci Nutr ; 65(1): 79-88, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23941288

RESUMO

Imbalances in gut microbiota composition during ulcerative colitis (UC) indicate a role for the microbiota in propagating the disorder. Such effects were investigated using in vitro batch cultures (with/without mucin, peptone or starch) inoculated with faecal slurries from healthy or UC patients; the growth of five bacterial groups was monitored along with short-chain fatty acid (SCFA) production. Healthy cultures gave two-fold higher growth and SCFA levels with up to ten-fold higher butyrate production. Starch gave the highest growth and SCFA production (particularly butyrate), indicating starch-enhanced saccharolytic activity. Sulphate-reducing bacteria (SRB) were the predominant bacterial group (of five examined) for UC inocula whereas they were the minority group for the healthy inocula. Furthermore, SRB growth was stimulated by peptone presumably due to the presence of sulphur-rich amino acids. The results suggest raised SRB levels in UC, which could contribute to the condition through release of toxic sulphide.


Assuntos
Aminoácidos Sulfúricos/metabolismo , Colite Ulcerativa/microbiologia , Colo/microbiologia , Proteínas Alimentares/administração & dosagem , Ácidos Graxos Voláteis/metabolismo , Mucosa Intestinal/microbiologia , Bactérias Redutoras de Enxofre/crescimento & desenvolvimento , Aminoácidos Sulfúricos/efeitos adversos , Ácido Butírico/metabolismo , Colite Ulcerativa/dietoterapia , Colite Ulcerativa/metabolismo , Colo/metabolismo , Dieta com Restrição de Proteínas , Proteínas Alimentares/efeitos adversos , Proteínas Alimentares/metabolismo , Fezes/microbiologia , Feminino , Fermentação , Bactérias Gram-Negativas/classificação , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/isolamento & purificação , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/crescimento & desenvolvimento , Bactérias Gram-Positivas/isolamento & purificação , Bactérias Gram-Positivas/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Viabilidade Microbiana , Pessoa de Meia-Idade , Tipagem Molecular , Mucinas/metabolismo , Peptonas/metabolismo , Amido/metabolismo , Bactérias Redutoras de Enxofre/classificação , Bactérias Redutoras de Enxofre/isolamento & purificação , Bactérias Redutoras de Enxofre/metabolismo
8.
Data Brief ; 55: 110636, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39234066

RESUMO

The gut microbiota residing in the distal ileum and colon is the most complex, diverse, and densest microbial ecosystem in the human body. Despite its known role in human health and disease, gut microbiome diversity and function are rarely explored in vulnerable populations such as refugees. The current study aimed to explore gut microbiota diversity and sources of variation among adolescent Afghan refugees residing in Peshawar, Pakistan. Stool samples were collected from 10 - 18 years old, healthy adolescents (n=205) for 16S rRNA gene sequence (V4-V5 hypervariable region) analysis on isolated faecal DNA. Bioinformatics analyses were performed using Kraken2, Bracken and Phyloseq. The data presented here will allow researchers to profile the gut microbiota of this rarely explored, vulnerable population who are at high risk of food insecurity and malnutrition. The data can be used to provide insight on the impact of demographic characteristics, dietary intake, nutritional status, and health on gut microbiome diversity, and enables a comparative analysis with similar data sets from other population groups of relevance. The amplicon sequencing data are deposited in the NCBI Sequence Read Archive as BioProject PRJNA1105775.

9.
Front Microbiol ; 14: 1074637, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36910170

RESUMO

Although iron is an essential nutrient for humans, as well as for almost all other organisms, it is poorly absorbed (~15%) from the diet such that most passes through the upper gut into the large intestine. The colonic microbiota is thus exposed to, and potentially influenced by, such residual iron which could have an impact on human health. The aim of the research described here is to determine how the major forms of dietary iron (inorganic iron and haem) influence metabolic activity and composition of the human gut microbiota by utilizing an in vitro parallel, pH-controlled anaerobic batch culture approach. Controlled iron provision was enabled by the design of a 'modified' low-iron gut-model medium whereby background iron content was reduced from 28 to 5 µM. Thus, the impact of both low and high levels of inorganic and haem iron (18-180 µM and 7.7-77 µM, respectively) could be explored. Gut-microbiota composition was determined using next generation sequencing (NGS) based community profiling (16S rRNA gene sequencing) and flow-fluorescent in situ hybridization (FISH). Metabolic-end products (organic acids) were quantified using gas chromatography (GC) and iron incorporation was estimated by inductively coupled plasma optical emission spectroscopy (ICP-OES). Results showed that differences in iron regime induced significant changes in microbiota composition when low (0.1% w/v) fecal inoculation levels were employed. An increase in haem levels from 7.7 to 77 µM (standard levels employed in gut culture studies) resulted in reduced microbial diversity, a significant increase in Enterobacteriaceae and lower short chain fatty acid (SCFA) production. These effects were countered when 18 µM inorganic iron was also included into the growth medium. The results therefore suggest that high-dietary haem may have a detrimental effect on health since the resulting changes in microbiota composition and SCFA production are indicators of an unhealthy gut. The results also demonstrate that employing a low inoculum together with a low-iron gut-model medium facilitated in vitro investigation of the relationship between iron and the gut microbiota.

10.
J Biol Chem ; 286(5): 3473-83, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21106523

RESUMO

Bacterioferritin (BFR) from Escherichia coli is a member of the ferritin family of iron storage proteins and has the capacity to store very large amounts of iron as an Fe(3+) mineral inside its central cavity. The ability of organisms to tap into their cellular stores in times of iron deprivation requires that iron must be released from ferritin mineral stores. Currently, relatively little is known about the mechanisms by which this occurs, particularly in prokaryotic ferritins. Here we show that the bis-Met-coordinated heme groups of E. coli BFR, which are not found in other members of the ferritin family, play an important role in iron release from the BFR iron biomineral: kinetic iron release experiments revealed that the transfer of electrons into the internal cavity is the rate-limiting step of the release reaction and that the rate and extent of iron release were significantly increased in the presence of heme. Despite previous reports that a high affinity Fe(2+) chelator is required for iron release, we show that a large proportion of BFR core iron is released in the absence of such a chelator and further that chelators are not passive participants in iron release reactions. Finally, we show that the catalytic ferroxidase center, which is central to the mechanism of mineralization, is not involved in iron release; thus, core mineralization and release processes utilize distinct pathways.


Assuntos
Proteínas de Bactérias/metabolismo , Grupo dos Citocromos b/metabolismo , Ferritinas/metabolismo , Heme/fisiologia , Ferro/metabolismo , Ceruloplasmina , Elétrons , Proteínas de Escherichia coli , Cinética , Minerais
11.
Mol Microbiol ; 80(2): 286-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21371140

RESUMO

Ribonucleotide reductases supply cells with their deoxyribonucleotides. Three enzyme types are known, classes I, II and III. Class II enzymes are anaerobic whereas class I enzymes are aerobic, and so class I and II enzymes are often produced by the same organism under opposing oxygen regimes. Escherichia coli contains two types of class I enzyme (Ia and Ib) with the Fe-dependent Ia enzyme (NrdAB) performing the major role aerobically, leaving the purpose of the Ib enzyme (NrdEF) unclear. Several papers have recently focused on the class Ib enzymes showing that they are Mn (rather than Fe) dependent and suggesting that the E. coli NrdEF may function under redox-stress conditions. A paper published in this issue of Molecular Microbiology from James Imlay's group confirms that this unexplained NrdEF Ib enzyme is Mn-dependent, but shows that it does not substitute for NrdAB during redox stress. Instead, a role during iron restriction is demonstrated. Thus, the purpose of NrdEF (and possibly other class Ib enzymes) is to enhance growth under aerobic, low-iron conditions, and to functionally replace the Fe-dependent NrdAB when iron is unavailable. This finding reveals a new mechanism by which bacteria adjust to life under iron deprivation.


Assuntos
Proteínas de Bactérias/metabolismo , DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Ferro/metabolismo , Manganês/metabolismo , Ribonucleotídeo Redutases/metabolismo , Aerobiose , Desoxirribonucleotídeos/metabolismo , Modelos Biológicos , Ribonucleosídeo Difosfato Redutase/metabolismo
12.
Nutr Cancer ; 64(8): 1251-60, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23163853

RESUMO

The health benefits of garlic have been proven by epidemiological and experimental studies. Diallyl disulphide (DADS), the major organosulfur compound found in garlic oil, is known to lower the incidence of breast cancer both in vitro and in vivo. The studies reported here demonstrate that DADS induces apoptosis in the MCF-7 breast-cancer cell line through interfering with cell-cycle growth phases in a way that increases the sub-G(0) population and substantially halts DNA synthesis. DADS also induces phosphatidylserine translocation from the inner to the outer leaflet of the plasma membrane and activates caspase-3. Further studies revealed that DADS modulates the cellular levels of Bax, Bcl-2, Bcl-xL, and Bcl-w in a dose-dependent manner, suggesting the involvement of Bcl-2 family proteins in DADS induced apoptosis. Histone deacetylation inhibitors (HDACi) are known to suppress cancer growth and induce apoptosis in cancer cells. Here it is shown that DADS has HDACi properties in MCF-7 cells as it lowers the removal of an acetyl group from an acetylated substrate and induces histone-4 (H4) hyper-acetylation. The data thus indicate that the HDACi properties of DADS may be responsible for the induction of apoptosis in breast cancer cells.


Assuntos
Compostos Alílicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Dissulfetos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Neoplasias da Mama/metabolismo , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Núcleo Celular/química , Ativação Enzimática/efeitos dos fármacos , Alho , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas c-bcl-2/análise
13.
BMJ Open ; 12(6): e058463, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672071

RESUMO

INTRODUCTION: Tuberculosis (TB) caused by Mycobacterium tuberculosis is a common infectious disease associated with significant morbidity and mortality, especially in low-income and middle-income countries. Successful treatment of the disease requires prolonged intake (6-8 months) of multiple antibiotics with potentially detrimental consequences on the composition and functional potential of the human microbiome. The protocol described in the current study aims to identify microbiome (oral and gut) signatures associated with TB pathogenesis, treatment response and outcome in humans. METHODS AND ANALYSIS: Four hundred and fifty, newly diagnosed patients with TB from three district levels (Peshawar, Mardan and Swat) TB diagnosis and treatment centres, will be recruited in this non-interventional, prospective cohort study and will be followed and monitored until treatment completion. Demographic and dietary intake data, anthropometric measurement and blood, stool and salivary rinse samples will be collected at baseline, day 15, month-2 and end of the treatment. Additionally, we will recruit age (±3 years) and sex-matched healthy controls (n=30). Blood sampling will allow monitoring of the immune response during the treatment, while salivary rinse and faecal samples will allow monitoring of dynamic changes in oral and gut microbiome diversity. Within this prospective cohort study, a nested case-control study design will be conducted to assess perturbations in oral and gut microbiome diversity (microbial dysbiosis) and immune response and compare between the patients groups (treatment success vs failure). ETHICS AND DISSEMINATION: The study has received ethics approval from the Ethic Board of Khyber Medical University Peshawar, and administrative approval from Provincial TB Control Programme of Khyber Pakhtunkhwa, Pakistan. The study results will be presented in national and international conferences and published in peer-reviewed journals. TRIAL REGISTRATION NUMBER: NCT04985994.


Assuntos
Microbiota , Tuberculose , Estudos de Casos e Controles , Estudos de Coortes , Humanos , Paquistão , Estudos Prospectivos , Tuberculose/tratamento farmacológico
14.
Front Nutr ; 9: 927754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36267902

RESUMO

Iron deficiency is the most prevalent human micronutrient deficiency, disrupting the physiological development of millions of infants and children. Oral iron supplementation is used to address iron-deficiency anemia and reduce associated stunting but can promote infection risk since restriction of iron availability serves as an innate immune mechanism against invading pathogens. Raised iron availability is associated with an increase in enteric pathogens, especially Enterobacteriaceae species, accompanied by reductions in beneficial bacteria such as Bifidobacteria and lactobacilli and may skew the pattern of gut microbiota development. Since the gut microbiota is the primary driver of immune development, deviations from normal patterns of bacterial succession in early life can have long-term implications for immune functionality. There is a paucity of knowledge regarding how both iron deficiency and luminal iron availability affect gut microbiota development, or the subsequent impact on immunity, which are likely to be contributors to the increased risk of infection. Piglets are naturally iron deficient. This is largely due to their low iron endowments at birth (primarily due to large litter sizes), and their rapid growth combined with the low iron levels in sow milk. Thus, piglets consistently become iron deficient within days of birth which rapidly progresses to anemia in the absence of iron supplementation. Moreover, like humans, pigs are omnivorous and share many characteristics of human gut physiology, microbiota and immunity. In addition, their precocial nature permits early maternal separation, individual housing, and tight control of nutritional intake. Here, we highlight the advantages of piglets as valuable and highly relevant models for human infants in promoting understanding of how early iron status impacts physiological development. We also indicate how piglets offer potential to unravel the complexities of microbiota-immune responses during iron deficiency and in response to iron supplementation, and the link between these and increased risk of infectious disease.

15.
Nutrients ; 14(9)2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35565715

RESUMO

A growing body of research evidence suggests that elevated homocysteine level (hyperhomocysteinemia) is an independent risk factor for cardiovascular diseases. The current study aimed to investigate the prevalence and associated risk factors for hyperhomocysteinemia among adolescent Afghan refugees aged 10−19 years. In total, 206 healthy adolescent boys and girls were randomly recruited from a refugee village in Peshawar, Pakistan, in 2020. Socio-demographic data, anthropometric assessment, and blood sample collection were performed following standard methods. Serum homocysteine was assessed using a chemiluminescent microparticle immunoassay, with hyperhomocysteinemia defined as levels ≥ 15 µmol/L. The overall prevalence of hyperhomocysteinemia was 25%, with mean homocysteine levels significantly (p = 0.004) higher among boys (14.1 µmol/L) than girls (11.8 µmol/L). Multivariate logistic regression analysis revealed a significant association between hyperhomocysteineimia and serum levels of vitamin B12 (OR 0.29; 95% CI of 0.14 to 0.62; p < 0.01) and folate (OR 0.1; 95% CI of 0.03 to 0.27; p < 0.001). Overall, our study findings indicate high prevalence of hyperhomocysteinemia among adolescent Afghan refugees who are potentially at high risk of developing cardiovascular diseases in future. There is a dire need to develop and implement nutritional and public health strategies to control hyperhomocysteinemia, protect against related diseases and complications in future, and ensure healthy lives and well-being among these vulnerable populations.


Assuntos
Doenças Cardiovasculares , Hiper-Homocisteinemia , Desnutrição , Refugiados , Adolescente , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Feminino , Ácido Fólico , Homocisteína , Humanos , Masculino , Desnutrição/complicações , Micronutrientes , Prevalência , Vitamina B 12
16.
Biochim Biophys Acta ; 1800(8): 691-705, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20553812

RESUMO

BACKGROUND: The Ferritins are part of the extensive 'Ferritin-like superfamily' which have diverse functions but are linked by the presence of a common four-helical bundle domain. The role performed by Ferritins as the cellular repository of excess iron is unique. In many ways Ferritins act as tiny organelles in their ability to secrete iron away from the delicate machinery of the cell, and then to release it again in a controlled fashion avoiding toxicity. The Ferritins are ancient proteins, being common in all three domains of life. This ubiquity reflects the key contribution that Ferritins provide in achieving iron homeostasis. SCOPE OF THE REVIEW: This review compares the features of the different Ferritins and considers how they, and other members of the Ferritin-like superfamily, have evolved. It also considers relevant features of the eleven other known families within the Ferritin-like superfamily, particularly the highly diverse rubrerythrins. MAJOR CONCLUSIONS: The Ferritins have travelled a considerable evolutionary journey, being derived from far more simplistic rubrerythrin-like molecules which play roles in defence against toxic oxygen species. The forces of evolution have moulded such molecules into three distinct types of iron storing (or detoxifying) protein: the classical and universal 24-meric ferritins; the haem-containing 24-meric bacterioferritins of prokaryotes; and the prokaryotic 12-meric Dps proteins. These three Ferritin types are similar, but also possess unique properties that distinguish them and enable then to achieve their specific physiological purposes. GENERAL SIGNIFICANCE: A wide range of biological functions have evolved from a relatively simple structural unit.


Assuntos
Evolução Molecular , Ferritinas/genética , Ferritinas/metabolismo , Hemeritrina/genética , Ferro/metabolismo , Rubredoxinas/genética , Sequência de Aminoácidos , Animais , Ferritinas/química , Ferritinas/fisiologia , Hemeritrina/química , Humanos , Ferro/química , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica/fisiologia , Filogenia , Conformação Proteica , Rubredoxinas/química , Homologia de Sequência
17.
Mol Microbiol ; 75(3): 637-57, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20015147

RESUMO

FtnA is the major iron-storage protein of Escherichia coli accounting for < or = 50% of total cellular iron. The FtnA gene (ftnA) is induced by iron in an Fe(2+)-Fur-dependent fashion. This effect is reportedly mediated by RyhB, the Fe(2+)-Fur-repressed, small, regulatory RNA. However, results presented here show that ftnA iron induction is independent of RyhB and instead involves direct interaction of Fe(2+)-Fur with an 'extended' Fur binding site (containing five tandem Fur boxes) located upstream (-83) of the ftnA promoter. In addition, H-NS acts as a direct repressor of ftnA transcription by binding at multiple sites (I-VI) within, and upstream of, the ftnA promoter. Fur directly competes with H-NS binding at upstream sites (II-IV) and consequently displaces H-NS from the ftnA promoter (sites V-VI) which in turn leads to derepression of ftnA transcription. It is proposed that H-NS binding within the ftnA promoter is facilitated by H-NS occupation of the upstream sites through H-NS oligomerization-induced DNA looping. Consequently, Fur displacement of H-NS from the upstream sites prevents cooperative H-NS binding at the downstream sites within the promoter, thus allowing access to RNA polymerase. This direct activation of ftnA transcription by Fe(2+)-Fur through H-NS antisilencing represents a new mechanism for iron-induced gene expression.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Ferritinas/genética , Regulação Bacteriana da Expressão Gênica , Ferro/metabolismo , Proteínas Repressoras/metabolismo , Sítios de Ligação , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Inativação Gênica , Genes Bacterianos , Regiões Promotoras Genéticas , RNA não Traduzido/metabolismo , Ribonuclease III/metabolismo
18.
Nutr Cancer ; 63(7): 1104-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21916706

RESUMO

Colon cancer is a leading and expanding cause of death worldwide. A major contributory factor to this disease is diet composition; some components are beneficial (e.g, dietary fiber), whereas others are detrimental (e.g., alcohol). Garlic oil is a prominent dietary constituent that prevents the development of colorectal cancer. This effect is believed to be mainly due to diallyl disulphide (DADS), which selectively induces redox stress in cancerous (rather than normal) cells that leads to apoptotic cell death. However, the detailed mechanism by which DADS causes apoptosis remains unclear. We show that DADS treatment of colonic adenocarcinoma cells (HT-29) initiates a cascade of molecular events characteristic of apoptosis. These include a decrease in cellular proliferation, translocation of phosphatidylserine to the plasma-membrane outer-layer, activation of caspase-3 and -9, genomic DNA fragmentation, and G(2)/M phase cell-cycle arrest. Short-chain fatty acids (SCFAs), particularly butyrate (abundantly produced in the gut by bacterial fermentation of dietary polysaccharides), enhance colonic cell integrity but, in contrast, inhibit colonic cancer cell growth. Combining DADS with butyrate augmented the apoptotic effect of butyrate on HT-29 cells. These results suggest that the anticancerous properties of DADS afford greater benefit when supplied with other favorable dietary factors (short chain fatty acids/polysaccharides) that likewise reduce colonic tumor development.


Assuntos
Compostos Alílicos/farmacologia , Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Dissulfetos/farmacologia , Óleos de Plantas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Western Blotting , Butiratos/farmacologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Ácidos Graxos Voláteis , Alho/química , Células HT29/efeitos dos fármacos , Humanos , Polissacarídeos
19.
J Glob Antimicrob Resist ; 25: 359-362, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33991746

RESUMO

OBJECTIVES: ST11 is a high-risk sequence type associated with carbapenem-resistant Klebsiella pneumoniae strains. Carbapenemase-producing hypervirulent K. pneumoniae (hvKp) are a major concern as they harbour a diverse range of pathogenicity traits. Here we describe the characteristics of K. pneumoniae strain KP75w isolated from a tertiary-care hospital in Pakistan. METHODS: Antimicrobial susceptibility testing was performed by the Kirby-Bauer disk diffusion test and broth microdilution assay. The virulence phenotype was determined by string test as well as biofilm and cell adhesion assays. Genome sequencing was performed using MiSeq and HiSeq 2500 platforms with 30 × coverage. RESULTS: Antimicrobial resistance profiling characterised strain KP75w as a multidrug-resistant carbapenemase-producing strain with a meropenem minimum inhibitory concentration (MIC) of 4 µg/mL, which is above the CLSI susceptible breakpoint (≤1 µg/mL). The annotated contigs indicated a genome size of 5 644 609 bp with 5679 coding regions. KP75w (ST11) was designated as a carbapenemase-producing hvKp strain on the basis of the presence of a carbapenemase gene (blaNDM-1) and hypervirulence genes (rmpA2, iucABCD-iutA, fyuA, irp, mrk, ybt, fep and virB2). KP75w was found to contain a 163-kb virulence region showing 58.8% identity to the large virulence plasmid pLVPK, supporting the hypervirulence of KP75w. CONCLUSION: KP75w is a novel non-hypermucoviscous carbapenemase-producing hvKp ST11 strain that appears to represent the convergence of multidrug resistance with hypervirulence traits in clinical K. pneumoniae strains from the Southeast Asian region.


Assuntos
Infecções por Klebsiella , Klebsiella pneumoniae , Proteínas de Bactérias , Humanos , Klebsiella pneumoniae/genética , Tipagem de Sequências Multilocus , Mutação , Paquistão , Fenótipo , beta-Lactamases
20.
Nutrients ; 13(9)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34578948

RESUMO

Pakistan has hosted millions of Afghan refugees over the last several decades. Due to poor socioeconomic status, food insecurity and inadequate access to health care, these refugees are considered to be at high risk of malnutrition. Previous studies on nutritional assessment of high-risk populations (refugees) have focused mainly on women and children (0-59 months). The current study aims to assess nutritional status of adolescent Afghan refugees; the population who are equally vulnerable to malnutrition and its consequences. In this cross sectional study, the nutritional status of 206 adolescent (10-19 years old) Afghans boys and girls living in a refugee camp in Peshawar, Pakistan was assessed using standard methods. The results indicate a prevalence of stunting, thinness, and overweight and obesity at 35.3%, 4.4% and 14.8%, respectively. Furthermore, there was a significantly high prevalence of micronutrient deficiencies (vitamin D, 80.5%; vitamin B12, 41.9%; and folate, 28.2%); and anemia (10.1%). Together, these findings indicate that this vulnerable population group suffers from the double burden of malnutrition and are thus at serious risk of impaired psychosocial cognitive development, general ill-health and diminished wellbeing. This study therefore highlights the urgent need to include adolescents in regular screening and intervention programs of such at-risk populations.


Assuntos
Desnutrição/epidemiologia , Estado Nutricional , Sobrepeso/epidemiologia , Refugiados/estatística & dados numéricos , Magreza/epidemiologia , Adolescente , Adulto , Afeganistão/etnologia , Criança , Estudos Transversais , Feminino , Humanos , Masculino , Paquistão/epidemiologia , Prevalência , Classe Social , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA