Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Gut Microbes ; 15(1): 2185034, 2023.
Article En | MEDLINE | ID: mdl-36919522

Probiotics are used for both generally healthy consumers and in clinical settings. However, theoretical and proven adverse events from probiotic consumption exist. New probiotic strains and products, as well as expanding use of probiotics into vulnerable populations, warrants concise, and actionable recommendations on how to work toward their safe and effective use. The International Scientific Association for Probiotics and Prebiotics convened a meeting to discuss and produce evidence-based recommendations on potential acute and long-term risks, risks to vulnerable populations, the importance for probiotic product quality to match the needs of vulnerable populations, and the need for adverse event reporting related to probiotic use. The importance of whole genome sequencing, which enables determination of virulence, toxin, and antibiotic resistance genes, as well as clear assignment of species and strain identity, is emphasized. We present recommendations to guide the scientific and medical community on judging probiotic safety.


What is the context? Probiotics, available to healthy consumers as both dietary supplements and foods, are also used by some patient populations. The goal of this paper is to determine if any new factors have emerged that would impact current views about probiotic safety for both these populations.What is new? The authors conclude that established practices are sensibly addressing factors important to the safety of traditional probiotics used by the general population. They also make recommendations regarding emerging safety considerations. Probiotics targeted for patient populations should undergo stringent testing to meet quality standards appropriate for that population, preferably verified by an independent third party. The safety of probiotics derived from species without a history of safe use must be considered on a case-by-case basis. Research is needed to address some gaps, for example which best animal models to use for safety assessment of live microbes, the possibility of antibiotic resistance gene transfer via transformation, and potential impact of probiotic-induced changes in microbiomes, interactions with drugs, and probiotic colonization.What is the impact? Probiotics of sufficient quality for patient populations are being developed and should be used accordingly. Long-term safety assessments for probiotics should be consistent with, and not more stringent than, current regulatory requirements for biologic drugs, including fecal microbial transplants. Rigor in collecting and reporting data on adverse events is needed. The authors confirm the need for understanding the entire genetic makeup of a probiotic as a cornerstone for assessing its safety.


Gastrointestinal Microbiome , Probiotics , Anti-Bacterial Agents/adverse effects , Prebiotics , Probiotics/adverse effects
2.
Cell ; 185(18): 3307-3328.e19, 2022 09 01.
Article En | MEDLINE | ID: mdl-35987213

Non-nutritive sweeteners (NNS) are commonly integrated into human diet and presumed to be inert; however, animal studies suggest that they may impact the microbiome and downstream glycemic responses. We causally assessed NNS impacts in humans and their microbiomes in a randomized-controlled trial encompassing 120 healthy adults, administered saccharin, sucralose, aspartame, and stevia sachets for 2 weeks in doses lower than the acceptable daily intake, compared with controls receiving sachet-contained vehicle glucose or no supplement. As groups, each administered NNS distinctly altered stool and oral microbiome and plasma metabolome, whereas saccharin and sucralose significantly impaired glycemic responses. Importantly, gnotobiotic mice conventionalized with microbiomes from multiple top and bottom responders of each of the four NNS-supplemented groups featured glycemic responses largely reflecting those noted in respective human donors, which were preempted by distinct microbial signals, as exemplified by sucralose. Collectively, human NNS consumption may induce person-specific, microbiome-dependent glycemic alterations, necessitating future assessment of clinical implications.


Microbiota , Non-Nutritive Sweeteners , Adult , Animals , Aspartame/pharmacology , Blood Glucose , Humans , Mice , Non-Nutritive Sweeteners/analysis , Non-Nutritive Sweeteners/pharmacology , Saccharin/pharmacology
4.
BMC Med ; 20(1): 56, 2022 02 09.
Article En | MEDLINE | ID: mdl-35135549

BACKGROUND: Dietary modifications are crucial for managing newly diagnosed type 2 diabetes mellitus (T2DM) and preventing its health complications, but many patients fail to achieve clinical goals with diet alone. We sought to evaluate the clinical effects of a personalized postprandial-targeting (PPT) diet on glycemic control and metabolic health in individuals with newly diagnosed T2DM as compared to the commonly recommended Mediterranean-style (MED) diet. METHODS: We enrolled 23 adults with newly diagnosed T2DM (aged 53.5 ± 8.9 years, 48% males) for a randomized crossover trial of two 2-week-long dietary interventions. Participants were blinded to their assignment to one of the two sequence groups: either PPT-MED or MED-PPT diets. The PPT diet relies on a machine learning algorithm that integrates clinical and microbiome features to predict personal postprandial glucose responses (PPGR). We further evaluated the long-term effects of PPT diet on glycemic control and metabolic health by an additional 6-month PPT intervention (n = 16). Participants were connected to continuous glucose monitoring (CGM) throughout the study and self-recorded dietary intake using a smartphone application. RESULTS: In the crossover intervention, the PPT diet lead to significant lower levels of CGM-based measures as compared to the MED diet, including average PPGR (mean difference between diets, - 19.8 ± 16.3 mg/dl × h, p < 0.001), mean glucose (mean difference between diets, - 7.8 ± 5.5 mg/dl, p < 0.001), and daily time of glucose levels > 140 mg/dl (mean difference between diets, - 2.42 ± 1.7 h/day, p < 0.001). Blood fructosamine also decreased significantly more during PPT compared to MED intervention (mean change difference between diets, - 16.4 ± 37 µmol/dl, p < 0.0001). At the end of 6 months, the PPT intervention leads to significant improvements in multiple metabolic health parameters, among them HbA1c (mean ± SD, - 0.39 ± 0.48%, p < 0.001), fasting glucose (- 16.4 ± 24.2 mg/dl, p = 0.02) and triglycerides (- 49 ± 46 mg/dl, p < 0.001). Importantly, 61% of the participants exhibited diabetes remission, as measured by HbA1c < 6.5%. Finally, some clinical improvements were significantly associated with gut microbiome changes per person. CONCLUSION: In this crossover trial in subjects with newly diagnosed T2DM, a PPT diet improved CGM-based glycemic measures significantly more than a Mediterranean-style MED diet. Additional 6-month PPT intervention further improved glycemic control and metabolic health parameters, supporting the clinical efficacy of this approach. TRIAL REGISTRATION: ClinicalTrials.gov number, NCT01892956.


Diabetes Mellitus, Type 2 , Diet, Mediterranean , Adult , Blood Glucose/metabolism , Blood Glucose Self-Monitoring , Diabetes Mellitus, Type 2/diagnosis , Female , Glycemic Control , Humans , Male , Middle Aged , Pilot Projects
5.
Nature ; 600(7890): 713-719, 2021 12.
Article En | MEDLINE | ID: mdl-34880502

Cigarette smoking constitutes a leading global cause of morbidity and preventable death1, and most active smokers report a desire or recent attempt to quit2. Smoking-cessation-induced weight gain (SCWG; 4.5 kg reported to be gained on average per 6-12 months, >10 kg year-1 in 13% of those who stopped smoking3) constitutes a major obstacle to smoking abstinence4, even under stable5,6 or restricted7 caloric intake. Here we use a mouse model to demonstrate that smoking and cessation induce a dysbiotic state that is driven by an intestinal influx of cigarette-smoke-related metabolites. Microbiome depletion induced by treatment with antibiotics prevents SCWG. Conversely, fecal microbiome transplantation from mice previously exposed to cigarette smoke into germ-free mice naive to smoke exposure induces excessive weight gain across diets and mouse strains. Metabolically, microbiome-induced SCWG involves a concerted host and microbiome shunting of dietary choline to dimethylglycine driving increased gut energy harvest, coupled with the depletion of a cross-regulated weight-lowering metabolite, N-acetylglycine, and possibly by the effects of other differentially abundant cigarette-smoke-related metabolites. Dimethylglycine and N-acetylglycine may also modulate weight and associated adipose-tissue immunity under non-smoking conditions. Preliminary observations in a small cross-sectional human cohort support these findings, which calls for larger human trials to establish the relevance of this mechanism in active smokers. Collectively, we uncover a microbiome-dependent orchestration of SCWG that may be exploitable to improve smoking-cessation success and to correct metabolic perturbations even in non-smoking settings.


Gastrointestinal Microbiome , Smoking Cessation , Weight Gain , Animals , Cross-Sectional Studies , Dysbiosis/etiology , Dysbiosis/metabolism , Dysbiosis/pathology , Mice , Models, Animal , Smoking/metabolism , Smoking/pathology
6.
Nat Microbiol ; 6(8): 1043-1054, 2021 08.
Article En | MEDLINE | ID: mdl-34226711

Antimicrobial resistance poses a substantial threat to human health. The gut microbiome is considered a reservoir for potential spread of resistance genes from commensals to pathogens, termed the gut resistome. The impact of probiotics, commonly consumed by many in health or in conjunction with the administration of antibiotics, on the gut resistome is elusive. Reanalysis of gut metagenomes from healthy antibiotics-naïve humans supplemented with an 11-probiotic-strain preparation, allowing direct assessment of the gut resistome in situ along the gastrointestinal (GI) tract, demonstrated that probiotics reduce the number of antibiotic resistance genes exclusively in the gut of colonization-permissive individuals. In mice and in a separate cohort of humans, a course of antibiotics resulted in expansion of the lower GI tract resistome, which was mitigated by autologous faecal microbiome transplantation or during spontaneous recovery. In contrast, probiotics further exacerbated resistome expansion in the GI mucosa by supporting the bloom of strains carrying vancomycin resistance genes but not resistance genes encoded by the probiotic strains. Importantly, the aforementioned effects were not reflected in stool samples, highlighting the importance of direct sampling to analyse the effect of probiotics and antibiotics on the gut resistome. Analysing antibiotic resistance gene content in additional published clinical trials with probiotics further highlighted the importance of person-specific metagenomics-based profiling of the gut resistome using direct sampling. Collectively, these findings suggest opposing person-specific and antibiotic-dependent effects of probiotics on the resistome, whose contribution to the spread of antimicrobial resistance genes along the human GI tract merit further studies.


Anti-Bacterial Agents/administration & dosage , Bacteria/drug effects , Bacteria/genetics , Bacterial Proteins/genetics , Drug Resistance, Bacterial , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Adult , Bacteria/classification , Bacteria/isolation & purification , Bacterial Proteins/metabolism , Cohort Studies , Fecal Microbiota Transplantation , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Humans , Male , Metagenome/drug effects , Middle Aged , Young Adult
7.
Diabetes Care ; 44(9): 1980-1991, 2021 09.
Article En | MEDLINE | ID: mdl-34301736

OBJECTIVE: To compare the clinical effects of a personalized postprandial-targeting (PPT) diet versus a Mediterranean (MED) diet on glycemic control and metabolic health in prediabetes. RESEARCH DESIGN AND METHODS: We randomly assigned adults with prediabetes (n = 225) to follow a MED diet or a PPT diet for a 6-month dietary intervention and additional 6-month follow-up. The PPT diet relies on a machine learning algorithm that integrates clinical and microbiome features to predict personal postprandial glucose responses. During the intervention, all participants were connected to continuous glucose monitoring (CGM) and self-reported dietary intake using a smartphone application. RESULTS: Among 225 participants randomized (58.7% women, mean ± SD age 50 ± 7 years, BMI 31.3 ± 5.8 kg/m2, HbA1c, 5.9 ± 0.2% [41 ± 2.4 mmol/mol], fasting plasma glucose 114 ± 12 mg/dL [6.33 ± 0.67 mmol/L]), 200 (89%) completed the 6-month intervention. A total of 177 participants also contributed 12-month follow-up data. Both interventions reduced the daily time with glucose levels >140 mg/dL (7.8 mmol/L) and HbA1c levels, but reductions were significantly greater in PPT compared with MED. The mean 6-month change in "time above 140" was -0.3 ± 0.8 h/day and -1.3 ± 1.5 h/day for MED and PPT, respectively (95% CI between-group difference -1.29 to -0.66, P < 0.001). The mean 6-month change in HbA1c was -0.08 ± 0.19% (-0.9 ± 2.1 mmol/mol) and -0.16 ± 0.24% (-1.7 ± 2.6 mmol/mol) for MED and PPT, respectively (95% CI between-group difference -0.14 to -0.02, P = 0.007). The significant between-group differences were maintained at 12-month follow-up. No significant differences were noted between the groups in a CGM-measured oral glucose tolerance test. CONCLUSIONS: In this clinical trial in prediabetes, a PPT diet improved glycemic control significantly more than a MED diet as measured by daily time of glucose levels >140 mg/dL (7.8 mmol/L) and HbA1c. These findings may have implications for dietary advice in clinical practice.


Diabetes Mellitus, Type 2 , Diet, Mediterranean , Prediabetic State/diet therapy , Adult , Blood Glucose , Blood Glucose Self-Monitoring , Female , Glucose , Glycated Hemoglobin/analysis , Glycemic Control , Humans , Male , Middle Aged
8.
Nutrients ; 13(2)2021 Jan 28.
Article En | MEDLINE | ID: mdl-33525593

The soar in COVID-19 cases around the globe has forced many to adapt to social distancing and self-isolation. In order to reduce contact with healthcare facilities and other patients, the CDC has advocated the use of telemedicine, i.e., electronic information and telecommunication technology. While these changes may disrupt normal behaviors and routines and induce anxiety, resulting in decreased vigilance to healthy diet and physical activity and reluctance to seek medical attention, they may just as well be circumvented using modern technology. Indeed, as the beginning of the pandemic a plethora of alternatives to conventional physical interactions were introduced. In this Perspective, we portray the role of SmartPhone applications (apps) in monitoring healthy nutrition, from their basic functionality as food diaries required for simple decision-making and nutritional interventions, through more advanced purposes, such as multi-dimensional data-mining and development of machine learning algorithms. Finally, we will delineate the emerging field of personalized nutrition and introduce pioneering technologies and concepts yet to be incorporated in SmartPhone-based dietary surveillance.


COVID-19/epidemiology , Mobile Applications , Pandemics , SARS-CoV-2 , Smartphone , Telemedicine , COVID-19/therapy , Exercise , Humans , Nutritional Status
9.
Nutr Rev ; 78(12 Suppl 2): 75-78, 2020 12 01.
Article En | MEDLINE | ID: mdl-33259609

Precision medicine has become the mainstay of modern therapeutics, especially for neoplastic disease, but this paradigm does not commonly prevail in dietary planning. Compelling evidence suggests that individual features, including the structure and function of the gut microbiota, contribute to harvesting and metabolizing energy from food, and thereby modulate the host metabolic phenotype and glucose homeostasis. Here, the concept of precision to dietary planning is highlighted by demonstrating the role of the microbiota in glucose intolerance in response to noncaloric artificial sweeteners, and by linking the microbiota and other host features to postprandial increases in blood glucose. These findings highlight the heterogeneity that exists among humans, which translates into divergent metabolic responses to similar food and warrants the adoption of next-generation sequencing technologies and advanced bioinformatics to revolutionize nutrition studies, laying the groundwork for an individually focused tailor-made practice.


Gastrointestinal Microbiome , Metabolic Diseases/prevention & control , Precision Medicine , Glucose/metabolism , Homeostasis , Humans , Postprandial Period , Sweetening Agents
10.
Nat Med ; 26(12): 1899-1911, 2020 12.
Article En | MEDLINE | ID: mdl-33106666

Acute liver failure (ALF) is a fulminant complication of multiple etiologies, characterized by rapid hepatic destruction, multi-organ failure and mortality. ALF treatment is mainly limited to supportive care and liver transplantation. Here we utilize the acetaminophen (APAP) and thioacetamide (TAA) ALF models in characterizing 56,527 single-cell transcriptomes to define the mouse ALF cellular atlas. We demonstrate that unique, previously uncharacterized stellate cell, endothelial cell, Kupffer cell, monocyte and neutrophil subsets, and their intricate intercellular crosstalk, drive ALF. We unravel a common MYC-dependent transcriptional program orchestrating stellate, endothelial and Kupffer cell activation during ALF, which is regulated by the gut microbiome through Toll-like receptor (TLR) signaling. Pharmacological inhibition of MYC, upstream TLR signaling checkpoints or microbiome depletion suppress this cell-specific, MYC-dependent program, thereby attenuating ALF. In humans, we demonstrate upregulated hepatic MYC expression in ALF transplant recipients compared to healthy donors. Collectively we demonstrate that detailed cellular/genetic decoding may enable pathway-specific ALF therapeutic intervention.


Liver Failure, Acute/genetics , Microbiota/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcriptome/drug effects , Acetaminophen/toxicity , Animals , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/pathology , Liver Transplantation/adverse effects , Mice , Microbiota/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Single-Cell Analysis , Thioacetamide/toxicity , Toll-Like Receptors/genetics
11.
Annu Rev Nutr ; 40: 189-219, 2020 09 23.
Article En | MEDLINE | ID: mdl-32520640

Nutrient content and nutrient timing are considered key regulators of human health and a variety of diseases and involve complex interactions with the mucosal immune system. In particular, the innate immune system is emerging as an important signaling hub that modulates the response to nutritional signals, in part via signaling through the gut microbiota. In this review we elucidate emerging evidence that interactions between innate immunity and diet affect human metabolic health and disease, including cardiometabolic disorders, allergic diseases, autoimmune disorders, infections, and cancers. Furthermore, we discuss the potential modulatory effects of the gut microbiota on interactions between the immune system and nutrition in health and disease, namely how it relays nutritional signals to the innate immune system under specific physiological contexts. Finally, we identify key open questions and challenges to comprehensively understanding the intersection between nutrition and innate immunity and how potential nutritional, immune, and microbial therapeutics may be developed into promising future avenues of precision treatment.


Gastrointestinal Microbiome , Immunity, Innate/physiology , Metabolic Diseases/prevention & control , Nutritional Status , Diet , Humans
12.
Nat Rev Immunol ; 20(1): 40-54, 2020 01.
Article En | MEDLINE | ID: mdl-31388093

Low-grade inflammation is the hallmark of metabolic disorders such as obesity, type 2 diabetes and nonalcoholic fatty liver disease. Emerging evidence indicates that these disorders are characterized by alterations in the intestinal microbiota composition and its metabolites, which translocate from the gut across a disrupted intestinal barrier to affect various metabolic organs, such as the liver and adipose tissue, thereby contributing to metabolic inflammation. Here, we discuss some of the recently identified mechanisms that showcase the role of the intestinal microbiota and barrier dysfunction in metabolic inflammation. We propose a concept by which the gut microbiota fuels metabolic inflammation and dysregulation.


Gastrointestinal Microbiome , Inflammation/microbiology , Animals , Chronic Disease , Dysbiosis/metabolism , Humans , Intestinal Mucosa/metabolism , Metabolic Diseases/microbiology
13.
Gut Microbes ; 11(1): 77-93, 2020.
Article En | MEDLINE | ID: mdl-30951391

Technological developments, including massively parallel DNA sequencing, gnotobiotics, metabolomics, RNA sequencing and culturomics, have markedly propelled the field of microbiome research in recent years. These methodologies can be harnessed to improve our in-depth mechanistic understanding of basic concepts related to consumption of probiotics, including their rules of engagement with the indigenous microbiome and impacts on the human host. We have recently demonstrated that even during probiotic supplementation, resident gut bacteria in a subset of individuals resist the mucosal presence of probiotic strains, limiting their modulatory effect on the microbiome and on the host gut transcriptional landscape. Resistance is partly alleviated by antibiotics treatment, which enables probiotics to interact with the host at the gut mucosal interface, although rather than promoting reconstitution of the indigenous microbiome and of the host transcriptional profile, they inhibit these components from returning to their naïve pre-antibiotic configurations. In this commentary, we discuss our findings in the context of previous and recent works, and suggest that incorporating the state-of-the-art methods currently utilized in microbiome research into the field of probiotics may lead to improved understanding of their mechanisms of activity, as well as their efficacy and long-term safety.


Anti-Bacterial Agents/adverse effects , Gastrointestinal Microbiome , Probiotics/therapeutic use , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Humans , Metagenomics
14.
Cell ; 178(3): 686-698.e14, 2019 07 25.
Article En | MEDLINE | ID: mdl-31257031

Immune cells residing in white adipose tissue have been highlighted as important factors contributing to the pathogenesis of metabolic diseases, but the molecular regulators that drive adipose tissue immune cell remodeling during obesity remain largely unknown. Using index and transcriptional single-cell sorting, we comprehensively map all adipose tissue immune populations in both mice and humans during obesity. We describe a novel and conserved Trem2+ lipid-associated macrophage (LAM) subset and identify markers, spatial localization, origin, and functional pathways associated with these cells. Genetic ablation of Trem2 in mice globally inhibits the downstream molecular LAM program, leading to adipocyte hypertrophy as well as systemic hypercholesterolemia, body fat accumulation, and glucose intolerance. These findings identify Trem2 signaling as a major pathway by which macrophages respond to loss of tissue-level lipid homeostasis, highlighting Trem2 as a key sensor of metabolic pathologies across multiple tissues and a potential therapeutic target in metabolic diseases.


Macrophages/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diet, High-Fat , Glucose Intolerance , Humans , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lipid Metabolism/genetics , Lipids/analysis , Macrophages/cytology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Obesity/metabolism , Obesity/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Single-Cell Analysis
15.
Nature ; 572(7770): 474-480, 2019 08.
Article En | MEDLINE | ID: mdl-31330533

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.


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
16.
Nat Med ; 25(5): 716-729, 2019 05.
Article En | MEDLINE | ID: mdl-31061539

Consumption of over-the-counter probiotics for promotion of health and well-being has increased worldwide in recent years. However, although probiotic use has been greatly popularized among the general public, there are conflicting clinical results for many probiotic strains and formulations. Emerging insights from microbiome research enable an assessment of gut colonization by probiotics, strain-level activity, interactions with the indigenous microbiome, safety and impacts on the host, and allow the association of probiotics with physiological effects and potentially useful medical indications. In this Perspective, we highlight key advances, challenges and limitations in striving toward an unbiased interpretation of the large amount of data regarding over-the-counter probiotics, and propose avenues to improve the quality of evidence, transparency, public awareness and regulation of their use.


Probiotics/adverse effects , Probiotics/therapeutic use , Animals , Clinical Trials as Topic , Clostridium Infections/therapy , Gastroenteritis/therapy , Gastrointestinal Microbiome , Host Microbial Interactions , Humans , Immunomodulation , Infant, Newborn , Irritable Bowel Syndrome/therapy , Neonatal Sepsis/therapy , Probiotics/standards , Respiratory Tract Infections/therapy , Safety , Treatment Outcome
17.
Sci Transl Med ; 11(477)2019 01 30.
Article En | MEDLINE | ID: mdl-30700573

Advances in microbiome research are spurring the development of new therapeutics for a variety of diseases, but translational challenges remain.


Microbiota , Precision Medicine , Animals , Diet , Fecal Microbiota Transplantation , Gastrointestinal Tract/drug effects , Humans , Microbiota/drug effects , Probiotics/pharmacology
18.
Nat Rev Gastroenterol Hepatol ; 16(1): 35-56, 2019 01.
Article En | MEDLINE | ID: mdl-30262901

Since the renaissance of microbiome research in the past decade, much insight has accumulated in comprehending forces shaping the architecture and functionality of resident microorganisms in the human gut. Of the multiple host-endogenous and host-exogenous factors involved, diet emerges as a pivotal determinant of gut microbiota community structure and function. By introducing dietary signals into the nexus between the host and its microbiota, nutrition sustains homeostasis or contributes to disease susceptibility. Herein, we summarize major concepts related to the effect of dietary constituents on the gut microbiota, highlighting chief principles in the diet-microbiota crosstalk. We then discuss the health benefits and detrimental consequences that the interactions between dietary and microbial factors elicit in the host. Finally, we present the promises and challenges that arise when seeking to incorporate microbiome data in dietary planning and portray the anticipated revolution that the field of nutrition is facing upon adopting these novel concepts.


Diet , Gastrointestinal Microbiome/physiology , Animals , Homeostasis , Humans
19.
PLoS One ; 13(9): e0204479, 2018.
Article En | MEDLINE | ID: mdl-30261024

INTRODUCTION: Typhoid fever (TF) continues to cause considerable morbidity and mortality in Nepal, but only limited epidemiologic data is available about TF outside Kathmandu. METHODS: As part of an interventional trial, we performed a prospective cohort study of bacteremic TF patients in Dhulikhel Hospital between October 2012 and October 2014. Demographic, epidemiological, clinical, and microbiologic data were recorded. RESULTS: 116 bacteremic typhoid patients were included in the study. Most were young, healthy, adults (mean age 27.9±12 years), 41.4% of whom were female. More than 70% of patients were employed in non-manual services or were university students. Salmonella Typhi accounted for 64/115 (55.7%) of all isolates, while Salmonella Paratyphi accounted for 51/115 (44.3%), of which 42 were Paratyphi A and 9 Paratyphi B. A significant proportion of TF cases occurred also during the dry season (48/116, 41.6%). The clinical presentation of Salmonella Typhi and Paratyphi infections was similar, except for a greater proportion of arthralgia in patients with Salmonella Typhi. Most Salmonella Typhi and Paratyphi isolates were resistant to nalidixic acid and susceptible to older antibiotics. One Salmonella Paratyphi isolate was resistant to ceftriaxone. CONCLUSIONS: TF remains common in the Dhulikhel area, even among those with a high level of education. Public health measures aimed at reducing the incidence of TF in the Dhulikhel area are warranted. The relative burden of TF caused by Salmonella Paratyphi is rising; a vaccine with activity against Salmonella Paratyphi is needed. Since Salmonella Paratyphi B was more prevalent in this cohort than in large cohorts of patients from Kathmandu, it is likely that there are significant regional variations in the epidemiology of TF outside Kathmandu.


Typhoid Fever/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Bacteremia/microbiology , Child , Child, Preschool , Cohort Studies , Female , Humans , Male , Middle Aged , Nepal/epidemiology , Paratyphoid Fever/epidemiology , Paratyphoid Fever/microbiology , Prevalence , Prospective Studies , Salmonella paratyphi A/classification , Salmonella paratyphi A/isolation & purification , Salmonella typhi/isolation & purification , Typhoid Fever/microbiology , Young Adult
20.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Article En | MEDLINE | ID: mdl-30193112

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.


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
...