Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 182(6): 1441-1459.e21, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32888430

ABSTRACT

Throughout a 24-h period, the small intestine (SI) is exposed to diurnally varying food- and microbiome-derived antigenic burdens but maintains a strict immune homeostasis, which when perturbed in genetically susceptible individuals, may lead to Crohn disease. Herein, we demonstrate that dietary content and rhythmicity regulate the diurnally shifting SI epithelial cell (SIEC) transcriptional landscape through modulation of the SI microbiome. We exemplify this concept with SIEC major histocompatibility complex (MHC) class II, which is diurnally modulated by distinct mucosal-adherent SI commensals, while supporting downstream diurnal activity of intra-epithelial IL-10+ lymphocytes regulating the SI barrier function. Disruption of this diurnally regulated diet-microbiome-MHC class II-IL-10-epithelial barrier axis by circadian clock disarrangement, alterations in feeding time or content, or epithelial-specific MHC class II depletion leads to an extensive microbial product influx, driving Crohn-like enteritis. Collectively, we highlight nutritional features that modulate SI microbiome, immunity, and barrier function and identify dietary, epithelial, and immune checkpoints along this axis to be potentially exploitable in future Crohn disease interventions.


Subject(s)
Crohn Disease/microbiology , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Histocompatibility Antigens Class II/metabolism , Intestine, Small/immunology , Intestine, Small/microbiology , Transcriptome/genetics , Animals , Anti-Bacterial Agents/pharmacology , Circadian Clocks/physiology , Crohn Disease/immunology , Crohn Disease/metabolism , Diet , Epithelial Cells/cytology , Epithelial Cells/immunology , Flow Cytometry , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Histocompatibility Antigens Class II/genetics , Homeostasis , In Situ Hybridization, Fluorescence , Interleukin-10/metabolism , Interleukin-10/pharmacology , Intestine, Small/physiology , Lymphocytes , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodicity , T-Lymphocytes/immunology , Transcriptome/physiology
2.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193112

ABSTRACT

Empiric probiotics are commonly consumed by healthy individuals as means of life quality improvement and disease prevention. However, evidence of probiotic gut mucosal colonization efficacy remains sparse and controversial. We metagenomically characterized the murine and human mucosal-associated gastrointestinal microbiome and found it to only partially correlate with stool microbiome. A sequential invasive multi-omics measurement at baseline and during consumption of an 11-strain probiotic combination or placebo demonstrated that probiotics remain viable upon gastrointestinal passage. In colonized, but not germ-free mice, probiotics encountered a marked mucosal colonization resistance. In contrast, humans featured person-, region- and strain-specific mucosal colonization patterns, hallmarked by predictive baseline host and microbiome features, but indistinguishable by probiotics presence in stool. Consequently, probiotics induced a transient, individualized impact on mucosal community structure and gut transcriptome. Collectively, empiric probiotics supplementation may be limited in universally and persistently impacting the gut mucosa, meriting development of new personalized probiotic approaches.


Subject(s)
Gastrointestinal Microbiome , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Bacteria/genetics , Bacteria/isolation & purification , Feces/microbiology , Female , Gastric Mucosa/microbiology , Humans , Intestinal Mucosa/microbiology , Male , Metagenomics , Mice , Mice, Inbred C57BL , Middle Aged , Placebo Effect , Principal Component Analysis , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Transcriptome , Young Adult
3.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193113

ABSTRACT

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Fecal Microbiota Transplantation , Feces/microbiology , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lactobacillus/drug effects , Lactobacillus/genetics , Lactobacillus/isolation & purification , Lactococcus/genetics , Lactococcus/isolation & purification , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Young Adult
4.
Nature ; 572(7770): 474-480, 2019 08.
Article in English | MEDLINE | ID: mdl-31330533

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disorder, in which the clinical manifestations may be influenced by genetic and unknown environmental factors. Here we show that ALS-prone Sod1 transgenic (Sod1-Tg) mice have a pre-symptomatic, vivarium-dependent dysbiosis and altered metabolite configuration, coupled with an exacerbated disease under germ-free conditions or after treatment with broad-spectrum antibiotics. We correlate eleven distinct commensal bacteria at our vivarium with the severity of ALS in mice, and by their individual supplementation into antibiotic-treated Sod1-Tg mice we demonstrate that Akkermansia muciniphila (AM) ameliorates whereas Ruminococcus torques and Parabacteroides distasonis exacerbate the symptoms of ALS. Furthermore, Sod1-Tg mice that are administered AM are found to accumulate AM-associated nicotinamide in the central nervous system, and systemic supplementation of nicotinamide improves motor symptoms and gene expression patterns in the spinal cord of Sod1-Tg mice. In humans, we identify distinct microbiome and metabolite configurations-including reduced levels of nicotinamide systemically and in the cerebrospinal fluid-in a small preliminary study that compares patients with ALS with household controls. We suggest that environmentally driven microbiome-brain interactions may modulate ALS in mice, and we call for similar investigations in the human form of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/microbiology , Amyotrophic Lateral Sclerosis/physiopathology , Gastrointestinal Microbiome/physiology , Niacinamide/metabolism , Akkermansia , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Dysbiosis , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Longevity , Male , Mice , Mice, Transgenic , Niacinamide/biosynthesis , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Survival Rate , Symbiosis/drug effects , Verrucomicrobia/metabolism , Verrucomicrobia/physiology
5.
Semin Immunol ; 32: 74-81, 2017 08.
Article in English | MEDLINE | ID: mdl-28431920

ABSTRACT

The commensal microbiome constitutes an important modulator of host physiology and risk of disease, including cancer development and progression. Lately, the microbiome has been suggested to modulate the efficacy of anti-cancer treatment. Examples include chemotherapy and total body irradiation-induced barrier function disruption, leading to microbial efflux that drives activation of anti-tumorigenic T cells; Microbiome-driven release of reactive oxygen species contributing to the efficacy of platinum salts; and microbiome-induced immune priming promoting the anti-tumor effects of alkylating chemotherapy and immune checkpoint inhibitors. Furthermore, selected commensals are able to colonize solid tumors. This 'tumor microbiome' may further impact local tumor responses to treatment and potentially be harnessed for tumor-specific targeting and therapeutic delivery. In this review, we present recent advances in understanding of the intricate role of microbiome in modulating efficacy of a number of anti-cancer treatments, and discuss how anti-cancer treatment approaches utilizing the tumor microbiome may enhance oncological treatment efficacy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Immunomodulation , Immunotherapy/methods , Microbiota/immunology , Neoplasms/microbiology , Neoplasms/therapy , Animals , Carcinogenesis , Costimulatory and Inhibitory T-Cell Receptors/immunology , Humans , Neoplasms/immunology , Symbiosis , Treatment Outcome
6.
J Pediatr Gastroenterol Nutr ; 69(6): 633-638, 2019 12.
Article in English | MEDLINE | ID: mdl-31765333

ABSTRACT

The human genome has been proposed to contribute to interpersonal variability in the way we respond to nutritional intake. However, personalized diets solely based on gene-nutrient interactions have not lived up to their expectations to date. Advances in microbiome research have indicated that a science-based generation of a personalized diet based on a combination of clinical and microbial features may constitute a promising new approach enabling accurate prediction of dietary responses. In addition, scientific advances in our understanding of defined dietary components and their effects on human physiology led to the incorporation and testing of defined diets as preventive and treatment approaches for diseases, such as epilepsy, ulcerative colitis, Crohn disease, and type 1 diabetes mellitus. Additionally, exciting new studies show that tailored diet regiments have the potential to modulate pharmaceutical treatment efficacy in cancer treatment. Overall, the true therapeutic potential of nutritional interventions is coming to light but is also facing substantial challenges in understanding mechanisms of activity, optimization of dietary interventions for specific human subpopulations, and elucidation of adverse effects potentially stemming from some dietary components in a number of individuals.


Subject(s)
Microbiota , Nutrigenomics/methods , Precision Medicine/methods , Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 1/microbiology , Epilepsy/diet therapy , Epilepsy/microbiology , Humans , Inflammatory Bowel Diseases/diet therapy , Inflammatory Bowel Diseases/microbiology , Neoplasms/diet therapy , Neoplasms/microbiology , Nutrition Therapy/methods , Pediatric Obesity/diet therapy , Pediatric Obesity/microbiology
7.
Proc Natl Acad Sci U S A ; 113(17): E2421-9, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27035961

ABSTRACT

Inherited demyelinating peripheral neuropathies are progressive incurable diseases without effective treatment. To develop a gene therapy approach targeting myelinating Schwann cells that can be translatable, we delivered a lentiviral vector using a single lumbar intrathecal injection and a myelin-specific promoter. The human gene of interest, GJB1, which is mutated in X-linked Charcot-Marie-Tooth Disease (CMT1X), was delivered intrathecally into adult Gjb1-null mice, a genetically authentic model of CMT1X that develops a demyelinating peripheral neuropathy. We obtained widespread, stable, and cell-specific expression of connexin32 in up to 50% of Schwann cells in multiple lumbar spinal roots and peripheral nerves. Behavioral and electrophysiological analysis revealed significantly improved motor performance, quadriceps muscle contractility, and sciatic nerve conduction velocities. Furthermore, treated mice exhibited reduced numbers of demyelinated and remyelinated fibers and fewer inflammatory cells in lumbar motor roots, as well as in the femoral motor and sciatic nerves. This study demonstrates that a single intrathecal lentiviral gene delivery can lead to Schwann cell-specific expression in spinal roots extending to multiple peripheral nerves. This clinically relevant approach improves the phenotype of an inherited neuropathy mouse model and provides proof of principle for treating inherited demyelinating neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Demyelinating Diseases/genetics , Animals , Genetic Therapy , Humans , Myelin Sheath/metabolism , Schwann Cells/metabolism
8.
BMC Med ; 14(1): 83, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27256449

ABSTRACT

HIV/AIDS causes severe dysfunction of the immune system through CD4+ T cell depletion, leading to dysregulation of both the adaptive and innate immune arms. A primary target for viral infection is the gastrointestinal tract, which is a reservoir of CD4+ T cells. In addition to being a major immune hub, the human gastrointestinal tract harbors trillions of commensal microorganisms, the microbiota, which have recently been shown to play critical roles in health. Alterations in the composition and function of microbiota have been implicated in a variety of 'multi-factorial' disorders, including infectious, autoimmune, metabolic, and neoplastic disorders. It is widely accepted that, in addition to its direct role in altering the gastrointestinal CD4+ T cell compartment, HIV infection is characterized by gut microbiota compositional and functional changes. Herein, we review such alterations and discuss their potential local and systemic effects on the HIV-positive host, as well as potential roles of novel microbiota-targeting treatments in modulating HIV progression and associated adverse systemic manifestations.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , HIV Infections/microbiology , CD4-Positive T-Lymphocytes/microbiology , HIV Infections/virology , Humans
9.
Ann Neurol ; 78(2): 303-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26010264

ABSTRACT

OBJECTIVE: X-linked Charcot-Marie-Tooth disease (CMT1X) is a common inherited neuropathy caused by mutations in the GJB1 gene encoding the gap junction protein connexin32 (Cx32). Clinical studies and disease models indicate that neuropathy mainly results from Schwann cell autonomous, loss-of-function mechanisms; therefore, CMT1X may be treatable by gene replacement. METHODS: A lentiviral vector LV.Mpz-GJB1 carrying the GJB1 gene under the Schwann cell-specific myelin protein zero (Mpz) promoter was generated and delivered into the mouse sciatic nerve by a single injection immediately distal to the sciatic notch. Enhanced green fluorescent protein (EGFP) reporter gene expression was quantified and Cx32 expression was examined on a Cx32 knockout (KO) background. A gene therapy trial was performed in a Cx32 KO model of CMT1X. RESULTS: EGFP was expressed throughout the length of the sciatic nerve in up to 50% of Schwann cells starting 2 weeks after injection and remaining stable for up to 16 weeks. Following LV.Mpz-GJB1 injection into Cx32 KO nerves, we detected Cx32 expression and correct localization in non-compact myelin areas where gap junctions are normally formed. Gene therapy trial by intraneural injection in groups of 2-month-old Cx32 KO mice, before demyelination onset, significantly reduced the ratio of abnormally myelinated fibers (p = 0.00148) and secondary inflammation (p = 0.0178) at 6 months of age compared to mock-treated animals. INTERPRETATION: Gene delivery using a lentiviral vector leads to efficient gene expression specifically in Schwann cells. Restoration of Cx32 expression ameliorates nerve pathology in a disease model and provides a promising approach for future treatments of CMT1X and other inherited neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Connexins/genetics , RNA, Messenger/metabolism , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Charcot-Marie-Tooth Disease/metabolism , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Lentivirus , Mice , Mice, Knockout , Gap Junction beta-1 Protein
10.
J Gene Med ; 16(11-12): 364-73, 2014.
Article in English | MEDLINE | ID: mdl-25394283

ABSTRACT

BACKGROUND: Most leukodystrophies result from mutations in genes expressed in oligodendrocytes that may cause autonomous loss of function of cell structural proteins. Therefore, effective gene delivery to oligodendrocytes is necessary to develop future treatments. MATERIALS: To achieve this, we cloned a lentiviral vector in which the enhanced green fluorescent protein (EGFP) expression was driven by the oligodendrocyte specific 2,3-cyclic nucleotide 3-phosphodiesterase promoter. The vector was inserted into C57BL/6 neonatal mouse brain by combined intraventricular and parenchymal injections. RESULTS: Assessment of EGFP expression revealed a widespread distribution, specifically in cells of the oligodendrocyte linage, starting from postnatal day 6 (P6) in the subventricular zone and spreading through migrating oligodendrocyte precursors. By P30, it was detectable throughout the brain and persisted for at least 3 months, showing an increase both in the number of expressing cells and in intensity over time. EGFP expression was restricted to oligodendrocyte linage cells. On average, 20.3 ± 2.56% of all oligodendrocytes in different central nervous system areas were EGFP-positive, with regional variations. CONCLUSIONS: Lentiviral gene delivery using an oligodendrocyte-specific promoter may achieve widespread and long-lasting expression selectively in oligodendrocytes, offering a possibility for gene therapy in certain leukodystrophies, although the relatively low rates of oligodendrocyte transduction are a limitation that remains to be overcome.


Subject(s)
Genetic Therapy , Lentivirus/genetics , Oligodendroglia/metabolism , Animals , Brain/metabolism , Coculture Techniques , Female , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice, Inbred C57BL , PC12 Cells , Rats , Sciatic Nerve/metabolism
11.
BMC Pediatr ; 14: 6, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24410941

ABSTRACT

BACKGROUND: Erythema infectiosum is the most common clinical manifestation of Parvovirus B19 infection although it has also been associated with rheumatologic diseases and various types of systemic vasculitides. Acute hepatitis and benign myositis however are rarely reported in association with Parvovirus B19 infection. CASE PRESENTATION: Here we report a 14-year old male, who developed acute hepatitis and benign myositis associated with erythema infectiosum following Parvovirus B19 infection. CONCLUSION: Parvovirus B19 infection has rarely been associated with acute hepatitis and exceptionally rarely with benign myositis. Parvovirus B19 should be considered in the differential diagnosis of acute non-A to E hepatitis and in the case of acute benign myositis presenting with a rash especially in children.


Subject(s)
Erythema Infectiosum , Hepatitis/virology , Myositis/virology , Acute Disease , Adolescent , Hepatitis/complications , Humans , Male , Myositis/complications , Parvovirus B19, Human
12.
Microorganisms ; 12(2)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38399646

ABSTRACT

Orally administered compounds represent the great majority of all pharmaceutical compounds produced for human use and are the most popular among patients since they are practical and easy to self-administer. Following ingestion, orally administered drugs begin a "perilous" journey down the gastrointestinal tract and their bioavailability is modulated by numerous factors. The gastrointestinal (GI) tract anatomy can modulate drug bioavailability and accounts for interpatient drug response heterogeneity. Furthermore, host genetics is a contributor to drug bioavailability modulation. Importantly, a component of the GI tract that has been gaining notoriety with regard to drug treatment interactions is the gut microbiota, which shares a two-way interaction with pharmaceutical compounds in that they can be influenced by and are able to influence administered drugs. Overall, orally administered drugs are a patient-friendly treatment option. However, during their journey down the GI tract, there are numerous host factors that can modulate drug bioavailability in a patient-specific manner.

13.
J Neurosci ; 31(12): 4535-43, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21430154

ABSTRACT

Sensory hair cells of the inner ear are the mechanoelectric transducers of sound and head motion. In mammals, damage to sensory hair cells leads to hearing or balance deficits. Nonmammalian vertebrates such as birds can regenerate hair cells after injury. In a previous study, we characterized transcription factor gene expression during chicken hair cell regeneration. In those studies, a laser microbeam or ototoxic antibiotics were used to damage the sensory epithelia (SE). The current study focused on 27 genes that were upregulated in regenerating SEs compared to untreated SEs in the previous study. Those genes were knocked down by siRNA to determine their requirement for supporting cell proliferation and to measure resulting changes in the larger network of gene expression. We identified 11 genes necessary for proliferation and also identified novel interactive relationships between many of them. Defined components of the WNT, PAX, and AP1 pathways were shown to be required for supporting cell proliferation. These pathways intersect on WNT4, which is also necessary for proliferation. Among the required genes, the CCAAT enhancer binding protein, CEBPG, acts downstream of Jun Kinase and JUND in the AP1 pathway. The WNT coreceptor LRP5 acts downstream of CEBPG, as does the transcription factor BTAF1. Both of these genes are also necessary for supporting cell proliferation. This is the first large-scale screen of its type and suggests an important intersection between the AP1 pathway, the PAX pathway, and WNT signaling in the regulation of supporting cell proliferation during inner ear hair cell regeneration.


Subject(s)
Ear, Inner/physiology , Hair Cells, Auditory, Inner/physiology , Nerve Regeneration/physiology , RNA Interference/physiology , Transcription Factors/genetics , Animals , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/physiology , Cell Proliferation , Chickens , Epithelium/physiology , Gene Knockdown Techniques , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/physiology , Lasers , Microarray Analysis , Paxillin/genetics , Paxillin/physiology , Saccule and Utricle/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/physiology , Transcription Factor TFIID/genetics , Transcription Factor TFIID/physiology , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt Proteins/physiology , Wnt4 Protein
14.
PLoS One ; 16(7): e0248792, 2021.
Article in English | MEDLINE | ID: mdl-34288921

ABSTRACT

Whole genome sequencing of viral specimens following molecular diagnosis is a powerful analytical tool of molecular epidemiology that can critically assist in resolving chains of transmission, identifying of new variants or assessing pathogen evolution and allows a real-time view into the dynamics of a pandemic. In Cyprus, the first two cases of COVID-19 were identified on March 9, 2020 and since then 33,567 confirmed cases and 230 deaths were documented. In this study, viral whole genome sequencing was performed on 133 SARS-CoV-2 positive samples collected between March 2020 and January 2021. Phylogenetic analysis was conducted to evaluate the genomic diversity of circulating SARS-CoV-2 lineages in Cyprus. 15 different lineages were identified that clustered into three groups associated with the spring, summer and autumn/winter wave of SARS-CoV-2 incidence in Cyprus, respectively. The majority of the Cypriot samples belonged to the B.1.258 lineage first detected in September that spread rapidly and largely dominated the autumn/winter wave with a peak prevalence of 86% during the months of November and December. The B.1.1.7 UK variant (VOC-202012/01) was identified for the first time at the end of December and spread rapidly reaching 37% prevalence within one month. Overall, we describe the changing pattern of circulating SARS-CoV-2 lineages in Cyprus since the beginning of the pandemic until the end of January 2021. These findings highlight the role of importation of new variants through travel towards the emergence of successive waves of incidence in Cyprus and demonstrate the importance of genomic surveillance in determining viral genetic diversity and the timely identification of new variants for guiding public health intervention measures.


Subject(s)
COVID-19/epidemiology , SARS-CoV-2/genetics , Cyprus/epidemiology , Humans , Molecular Epidemiology , Phylogeny , SARS-CoV-2/physiology
15.
Microorganisms ; 10(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35056533

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has hit its second year and continues to damage lives and livelihoods across the globe. There continues to be a global effort to present serological data on SARS-CoV-2 antibodies in different individuals. As such, this study aimed to characterize the seroprevalence of SARS-CoV-2 antibodies in the Cypriot population for the first time since the pandemic started. Our results show that a majority of people infected with SARS-CoV-2 developed IgG antibodies against the virus, whether anti-NP, anti-S1RBD, or both, at least 20 days after their infection. Additionally, the percentage of people with at least one antibody against SARS-CoV-2 in the group of volunteers deemed SARS-CoV-2 negative via RT-PCR or who remain untested/undetermined (14.43%) is comparable to other reported percentages worldwide, ranging anywhere from 0.2% to 24%. We postulate that these percentages reflect the underreporting of true infections in the population, and also show the steady increase of herd immunity. Additionally, we showed a significantly marked decrease in anti-NP IgG antibodies in contrast to relatively stable levels of anti-S1RBD IgG antibodies in previously infected individuals across time.

16.
Water Sci Technol ; 58(3): 647-51, 2008.
Article in English | MEDLINE | ID: mdl-18725734

ABSTRACT

The last case of polio in Cyprus caused by a wild-type poliovirus occurred in 1995. Since then Cyprus belongs to the countries considered poliovirus-free by the WHO. The aim of this study was to confirm the absence of any circulating wild-type polioviruses and to monitor vaccine-derived polioviruses in Cyprus by analysis of sewages. During the course of this study no wild-type polioviruses were identified, although the identification of viable oral poliovirus vaccine isolates confirmed the presence and circulation of poliovirus vaccine strains in sewage in Cyprus.


Subject(s)
Poliomyelitis/prevention & control , Poliovirus/isolation & purification , Sewage/virology , Cyprus , Environmental Monitoring , Humans , Poliomyelitis/virology , Poliovirus/genetics , Poliovirus/immunology , Poliovirus Vaccine, Oral/immunology , Reverse Transcriptase Polymerase Chain Reaction
17.
Curr Opin Biotechnol ; 51: 57-63, 2018 06.
Article in English | MEDLINE | ID: mdl-29223004

ABSTRACT

Generalized dietary and lifestyle guidelines have been formulated and published for decades now from a variety of relevant agencies in an attempt to guide people towards healthy choices. As the pandemic rise in metabolic diseases continues to increase, it has become clear that the one-fit-for-all diet approach does not work and that there is a significant variation in inter-individual responses to diet and lifestyle interventions. Recent technological advances have given an unprecedented insight into the sources of this variation, pointing towards our genome and microbiome as potentially and previously under-explored culprits contributing to individually unique dietary responses. Variations in our genome influence the bioavailability and metabolism of nutrients between individuals, while inter-individual compositional variation of commensal gut microbiota leads to different microbe functional potential, metabolite production and metabolism modulation. Quantifying and incorporating these factors into a comprehensive personalized nutrition approach may enable practitioners to rationally incorporate individual nutritional recommendations in combating the metabolic syndrome pandemic.


Subject(s)
Gastrointestinal Microbiome/genetics , Genome, Human , Metabolic Syndrome/prevention & control , Microbiota , Nutrition Therapy , Precision Medicine , Diet , Humans , Life Style , Nutrigenomics
18.
Eur J Med Genet ; 50(1): 73-8, 2007.
Article in English | MEDLINE | ID: mdl-17194633

ABSTRACT

We report on a 13-year-old girl with normal karyotype and a de novo cryptic terminal deletion of chromosome 2q, detected by subtelomeric FISH analysis. Further investigation with array-CGH analysis using the 1Mb resolution Spectral Chip 2600 (Spectral Genomics) confirmed the deletion and also showed a deletion of four additional clones. No other abnormalities were detected by array-CGH. FISH studies using 8 BAC-probes were performed for fine mapping of the deletion and confirmed the array results. FISH analysis showed that the deletion breakpoint lies between clones RP11-84G18 and RP11-83N2 (physical distance between clones 0.36Mb) and extends to the telomere. The size of the deletion was estimated to be about 6.4-6.7Mb. Clinical findings include: developmental delay, severe behavioural disturbances, growth-pubertal retardation, congenital conductive mild hearing loss, growth hormone deficiency, compensate hypothyroidism, dysmorphic facial features, excessive joint hypermobility, brachymetaphalangy, abnormal dermatoglyphics and a history of neonatal laryngomalacia, hypotonia and umbilical hernia. The phenotype of our patient is in keeping with those of the literature, with the exception of cardiovascular, urogenital, neurological anomalies and eczema, which were not observed. The report of the clinical and molecular presentation of similar cases will allow accurate phenotype-genotype correlation and proper genetic counseling of the family.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 2/genetics , In Situ Hybridization, Fluorescence , Oligonucleotide Array Sequence Analysis , Telomere/genetics , Adolescent , Child , Female , Humans , Intellectual Disability/genetics , Joint Instability/genetics , Muscle Hypotonia/genetics
19.
Cell Metab ; 25(3): 506-521, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28273474

ABSTRACT

In addition to the immune system's traditional roles of conferring anti-infectious and anti-neoplastic protection, it has been recently implicated in the regulation of systemic metabolic homeostasis. This cross-talk between the immune and the metabolic systems is pivotal in promoting "metabolic health" throughout the life of an organism and plays fundamental roles in its adaptation to ever-changing environmental makeups and nutritional availability. Perturbations in this intricate immune-metabolic cross-talk contribute to the tendency to develop altered metabolic states that may culminate in metabolic disorders such as malnutrition, obesity, type 2 diabetes mellitus (T2DM), and other features of the metabolic syndrome. Regulators of immune-metabolic interactions include host genetics, nutritional status, and the intestinal microbiome. In this Perspective, we highlight current understanding of immune-metabolism interactions, illustrate differences among individuals and between populations in this respect, and point toward future avenues of research possibly enabling immune harnessing as means of personalized treatment for common metabolic disorders.


Subject(s)
Health , Metabolic Diseases/immunology , Aging/physiology , Animals , Humans , Metabolic Diseases/genetics , Microbiota , Models, Biological , Translational Research, Biomedical
20.
Cell Metab ; 23(3): 393-4, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26959178

ABSTRACT

Malnutrition is a global health burden affecting the development of millions of children worldwide, but the effects of current treatment strategies are modest. Charbonneau et al. (2016) identify sialylated oligosaccharides in breast milk as microbiota-dependent growth-promoting metabolites, paving the way for a new rational treatment of severe infant stunting.


Subject(s)
Child Development , Malnutrition/diet therapy , Milk, Human/chemistry , Milk/chemistry , Oligosaccharides/metabolism , Animals , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL